Microbial Metabolomics
Microbial Metabolomics
J. Beale
Konstantinos A. Kouremenos
Enzo A. Palombo Editors
Microbial
Metabolomics
Applications in Clinical, Environmental,
and Industrial Microbiology
Microbial Metabolomics
David J. Beale Konstantinos A. Kouremenos
•
Enzo A. Palombo
Editors
Microbial Metabolomics
Applications in Clinical, Environmental,
and Industrial Microbiology
123
Editors
David J. Beale Enzo A. Palombo
Land and Water Swinburne University of Technology
CSIRO Hawthorn, VIC
Dutton Park, QLD Australia
Australia
Konstantinos A. Kouremenos
Metabolomics Australia, Bio21 Molecular
Science & Biotechnology Institute
The University of Melbourne
Parkville, VIC
Australia
Systems biology has greatly increased our understanding of many cellular functions
and cell–cell interactions. Our awareness of the enormous range of activities per-
formed by microorganisms has been greatly enhanced by major developments in
the fields of genomics, proteomics and transcriptomics. However, understanding
these functions does not fully describe the global metabolic features of single cells
or cell populations (mono-specific or multi-species). Characterization of the
metabolome brings us closer to the phenotype of the cell and more closely reflects
the activities of microbes in response to environmental stimuli.
Microbial metabolomics is an emerging field that has developed rapidly in recent
years. This development has been paralleled and supported by important advances
in analytical instrumentation and technologies, in particular chromatographic and
mass spectrometric methods, coupled with new and more powerful computational
tools.
This book brings together contributions from global experts from diverse areas
that have facilitated the exciting advances in microbial metabolomics, with special
attention given to the development of relevant hardware and software platforms.
Thus, the principles of these technologies will be a major focus of the book.
The main application of metabolomics is likely to be in the field of clinical and
veterinary microbiology with a focus on disease-causing microorganisms.
However, there is a great potential to apply metabolomics to help better understand
complex biological systems that are dominated by multi-species microbial popu-
lations exposed to changing growth and nutritional conditions. In particular,
environmental (e.g. water and soil), food (e.g. microbial spoilage and food patho-
gens), agriculture and industrial applications are seen as developing. As such, this
book looks at the application metabolomics from clinical, environmental and
industrial perspectives.
v
Contents
vii
Chapter 1
Introduction to Microbial Metabolomics
Silas G. Villas-Boas
1 Introduction
The ultimate goal of metabolomics is the unbiased and nontargeted analysis of cell
metabolites combined with the detection, identification, and sometimes also the
absolute quantification of a multitude of compounds in biological samples. Despite
this being a very ambitious goal due to all the analytical challenges associated with
the analysis of metabolomes (and our inability to design completely unbiased
analytical methods), there has been an enormous development in the field of
metabolomics in the past 16 years that has placed metabolomics in the spotlight of
cutting-edge technologies for life sciences in the twenty-first century (Reeves and
Rabinowtiz 2011; Nguyen et al. 2012; Russell and Duncan 2013; Zhang et al.
2014).
Microbiology greatly benefited from developments in metabolomics since its
conception (Allen et al. 2003; Castrillo et al. 2003; Villas-Bôas et al. 2005a, b,
2006; Wang et al. 2006; van der Werf et al. 2007). Microbial systems are usually
simpler and highly controllable compared to higher (multicellular) organisms,
which make them the ideal platform for developing, applying, and validating
metabolomics tools. Moreover, the development of genome-scale metabolic models
for a wide range of microbial species—based on genome sequence data—has
offered the best scaffold for interpretation of metabolomics data and, more
importantly, for integration of metabolomics data with other omics (Lee et al.
2005).
However, microbial metabolomics is not free of its challenges. The microbial
metabolome is sparsely distributed into three very distinct matrices: (i) inside the
cell (intracellular); (ii) outside the cell, in the medium (extracellular); and (iii) in the
Fiehn 2001, 2002; Roessner et al. 2000, 2001) and paved the way for modern
metabolomics. However, the microbial metabolomics field was also particularly
influenced by proposed approaches and methodologies developed by the group of
Professor Stephen Oliver at that time at the University of Manchester, UK (Allen
et al. 2003; Castrillo et al. 2003). Oliver’s group was not only the first group to
propose the concept of metabolic fingerprinting using direct infusion mass spec-
trometry and S. cerevisiae as the model organism (Castrillo et al. 2003); they also
demonstrated the importance of extracellular metabolites found in the culture media
for phenotyping purposes, referring to it as the metabolic footprint (Allen et al.
2003). Other important contributions to early developments in microbial metabo-
lomics include the global metabolite profiling of intra- and extracellular metabolites
of microbial cells and cultures by methyl chloroformate (MCF) derivatization and
gas chromatography–mass spectrometry (GC-MS) analysis (Villas-Bôas et al. 2003,
2005a, b); assessment of sampling and sample preparation for microbial metabo-
lomics (Maharjan and Ferenci 2003; Wittmann et al. 2004; Villas-Bôas et al. 2005a,
b); quantitative microbial metabolomics using capillary electrophoresis–mass
spectrometry (CE-MS) (Soga and Ross 1999; Soga and Heiger 2000; Soga and
Imaizumi 2001; Soga et al. 2002a, b); and the application of stable isotope dilution
theory in metabolome characterization of S. cerevisiae cultures, where each
metabolite concentration is quantified relative to the concentration of its
U-13C-labeled equivalent (Mashego et al. 2003).
In the past 14 years, metabolomics has been successfully applied to a wide range
of microbiological applications, from microbial functional genomics (Allen et al.
2003; Castrillo et al. 2003; Moxley et al. 2009), phenotyping (Bundy et al. 2005;
Villas-Bôas et al. 2008), to the detection of microbial contaminations in foods
(Jahangir et al. 2008; Cevallos-Cevallos et al. 2011), fermentation broth (Sue et al.
2011); and to characterize microbial symbiotic associations with different hosts
(Martin et al. 2007; Russell and Duncan 2013). In addition, the microbial meta-
bolomics field includes two important microbial metabolome databases developed
at the University of Alberta (Canada): The Yeast Metabolome Database (YMDB)
(http://www.ymdb.ca/) and the E. coli Metabolome Database (ECMDB) (http://
ecmdb.ca/) (Jewison et al. 2012 and Guo et al. 2013). These are databases of
metabolites found in cultures or cells of S. cerevisiae and E. coli, respectively.
The YMDB contains 2027 small molecules with 980 associated enzymes and 138
associated transporters and the ECMDB contains 3755 small molecules with 1402
associated enzymes and 387 associated transporters. For each metabolite there is a
description of the compound, its chemical characteristics, synonyms and links to
spectral and chemical databases. Therefore, these two databases represent a land-
mark for microbial metabolomics, offering powerful Web-based tools to aid in
microbial metabolomics data analysis and interpretation.
1 Introduction to Microbial Metabolomics 5
3 Challenges
and other substrates to the cells, but to also remove metabolic waste products in
order to maintain intracellular homeostasis. Therefore, the secretion of metabolites
is also an essential biochemical function to all living cells, but it is very little studied
if compared to the process of nutrient uptake.
The secretion of macromolecules such as proteins (enzymes) and nucleic acids
have been widely studied in microorganisms (Alvarez-Martinez and Christie 2009;
Chapon-Hervé et al. 1997; Genin and Boucher 1994). The best known mechanisms
of macromolecule secretion are the xcp secretion pathway (Chapon-Hervé et al.
1997), the transmembrane protein family PulD (Genin and Boucher 1994), and the
type IV secretion systems (T4SS) (Alvarez-Martinez and Christie 2009). On the
other hand, recent studies have shown that ABC transmembrane proteins
(ATP-binding cassette) and MFS (Major Facilitator Superfamily) play a very
important role in the secretion of secondary/toxic metabolites in microbial systems
such as endogenous and exogenous antibiotics (Stergiopoulos et al. 2002; Martín
et al. 2005). It is believed that small metabolites, mainly those end products of
microbial fermentation (e.g., acetate, lactate, butyrate, ethanol, butanol, acetone,
etc.) are excreted passively through the plasma membrane, or secreted by spe-
cialized mechanisms such as response reaction to hypo-osmotic stress (Krämer
1994) and uniport, synport, and antiport transport systems (Krämer 1994).
Although there is little experimental data to validate these mechanisms as being
responsible for the metabolic efflux, all of them are based on the concept of
metabolic overflow, which says that under specific metabolic conditions, a massive
excretion of some metabolic intermediates is observed due to intracellular accu-
mulation of one or more intermediates from one or more metabolic pathways
(Krämer 1994). Although this concept seems to be appropriate to explain the
secretion of some metabolic intermediates, it does not apply to many cases studied
during continuous culture (chemostats) (Krämer 1994). Microbial metabolomics
data obtained during continuous culture in our laboratory challenge the concept of
metabolic overflow (Granucci et al. 2015). For instance, in several studies in which
microbial samples were collected before, during and after an environmental per-
turbation (during continuous culture), we observed that some intracellular
metabolites are actively secreted to the extracellular medium in response to an
environmental stimulus, demonstrating that the secretion of a given metabolite to
the extracellular medium does not take place exclusively as a result of its accu-
mulation in the intracellular medium. Our results suggest that microbial cells very
often remove some intracellular metabolites, even though these metabolites could
be key intermediates of central carbon metabolism such as pyruvate, phospho-
enolpyruvate, and several amino acids. Therefore, a better understanding about the
relationship between intra- and extracellular metabolites is essential if we want to
make better use of extracellular metabolomics data.
1 Introduction to Microbial Metabolomics 7
4 Cutting-Edge Applications
It is evident today that the metabolite profile of a pure microbial culture is very
distinct from that of a mixed culture and the metabolite profile of a mixed culture is
not a simple sum up of the metabolite profiles of the different species composing the
community (Sue et al. 2011; Kimes et al. 2013; Lv 2013; Ponnusamy et al. 2013).
Microbial cells sense the presence of a different microbial species in their envi-
ronment and immediately respond to the foreign presence by changing their
metabolic state. This change in metabolic state can be easily detected through
metabolomics (e.g., metabolic footprint analysis) (Sue et al. 2011). Therefore,
metabolomics has become an important tool for assessing the metabolic state of
microbial communities both in vitro (Sue et al. 2011; Ponnusamy et al. 2013) as
well as in nature (Badri et al. 2013; Kimes et al. 2013). Nevertheless, it is essential
to any metabolomic study involving microbial communities to keep track of the
microbial community dynamics in terms of its composition both qualitative and
quantitatively.
The identification of the cause of microbial infection is not always simple and
usually requires days of culturing and sometimes evasive sampling procedures. The
discovery of metabolic biomarkers specific to different infective agents through
8 S.G. Villas-Boas
metabolomics can pave the way for development of fast, sensitive, and accurate
methods of diagnosis through metabolite profiles. Metabolomics has been suc-
cessfully applied to distinguish different causative agents of bacterial pneumonia in
animal models (Slupsky 2010; Hoerr et al. 2012) as well as for early diagnosis of
pediatric septic shock (Mickiewicz et al. 2013). In addition, the opportunity to use
noninvasive sampling procedures such as the use of exhaled breath for diagnostic of
infectious diseases through metabolite profiling of volatile metabolites promises to
be the future of clinical diagnosis (Boots et al. 2012).
5 Future Perspective
References
Han J, Antunes LCM, Finlay BB, Borcherst CH (2010) Metabolomics: towards understanding
host-microbe interactions. Future Microbiol 5:153–161
Hoerr V, Zbytnuik L, Leger C, Tam PPC, Kubes P, Vogel HJ (2012) Gram-negative and
Gram-positive bacterial infections give rise to a different metabolic response in a mouse model.
J Proteome Res 11:3231–3245
Hofman-Bang J (1999) Nitrogen catabolite repression in Saccharomyces cerevisiae. Mol
Biotechnol 12:35–73
Jahangir M, Kim HK, Choi YH, Verpoorte R (2008) Metabolomic response of Brassica rapa
submitted to pre-harvest bacterial contamination. Food Chem 107:362–368
Jewison T, Knox C, Neveu V, Djoumbou Y, Guo AC, Lee J, Liu P, Mandal R, Krishnamurthy R,
Sinelnikov I, Wilson M, Wishart DS (2012) YMDB: the yeast metabolome database. Nucleic
Acid Res 40:D815–D820
Jojima T, Omumasaba CA, Inui M, Yukawa H (2010) Sugar transporters in efficient utilization of
mixed sugar substrates: current knowledge and outlook. Appl Microbiol Biotechnol 85:471–
480
Kimes NE, Callaghan AV, Aktas DF, Smith WL, Sunner J, Golding BT, Drozdowska M,
Hazen TC, Suflita JM, Morris PJ (2013) Metagenomic analysis and metabolite profiling of
deep-sea sediments from the Gulf of Mexico following the Deepwater Horizon oil spill. Front
Microbiol 4:50
Krämer R (1994) Secretion of amino acids by bacteria: physiology and mechanism. FEMS
Microbiol Rev 13:75–94
Lee SY, Lee D, Kim TY (2005) Systems biotechnology for strain improvement. Trends
Biotechnol 23:349–358
Lv HT (2013) Mass spectrometry-based metabolomics towards understanding of gene functions
with a diversity of biological contexts. Mass Spectrom Rev 32:118–128
Maharjan RP, Ferenci T (2003) Global metabolite analysis: the influence of extraction
methodology on metabolome profiles of Escherichia coli. Anal Biochem 313:145–154
Marcobal A, Kashyap PC, Nelson TA, Aronov PA, Donia MS, Spormann A, Fischbach MA,
Sonnenburg JL (2013) A metabolomic view of how the human gut microbiota impacts the host
metabolome using humanized and gnotobiotic mice. ISME J 7:1933–1943
Martín JF, Casqueiro J, Liras P (2005) Secretion systems for secondary metabolites: how producer
cells send out messages of intracellular communication. Curr Opin Microbiol 8:282–293
Martin FPJ, Dumas ME, Wang YL, Legido-Quigley C, Yap IKS, Tang HR, Zirah S, Murphy GM,
Cloarec O, Lindon JC, Sprenger N, Fay LB, Kochhar S, van Bladeren P, Holmes E,
Nicholson JK (2007) A top-down systems biology view of microbiome-mammalian metabolic
interactions in a mouse model. Mol Syst Biol 112:112
Mashego MR, van Gulik WM, Vinke JL, Heijnen JJ (2003) Critical evaluation of sampling
techniques for residual glucose determination in carbon limited chemostat culture of
Saccharomyces cerevisiae. Biotech Bioeng 83:395–399
Mickiewicz B, Vogel HJ, Wong HR, Winston BW (2013) Metabolomics as a novel approach for
early diagnosis of pediatric septic shock and its mortality. Am J Respir Crit Care Med
187:967–976
Ming X, Stein TTP, Barnes V, Rhodes N, Guo LN (2012) Metabolic perturbance in autism
spectrum disorders: a metabolomics study. J Proteomic Res 11:5856–5862
Montel E, Bridge PD, Sutton BC (1991) An integrated approach to Phoma systematics.
Mychopathologia 115:89–103
Moxley JF et al (2009) Linking high-resolution metabolic flux phenotypes and transcriptional
regulation in yeast modulated by the global regulator Gcn4p. Proc Natl Acad Sci USA
106:6477–6482
Nguyen QT, Merlo ME, Medema MH, Jankevics A, Breitling R, Takano E (2012) Metabolomics
methods for the synthetic biology of secondary metabolism. FEBS Lett 586:2177–2183
Oliver SG (1997) Yeast as a navigational aid in genome analysis. Microbiology 7:405–409
Oliver SG, Winson MK, Kell DB, Baganz F (1998) Systematic functional analysis of the yeast
genome. Trends Biotechnol 16:373–378
1 Introduction to Microbial Metabolomics 11
Ponnusamy K, Lee S, Lee CH (2013) Time-dependent correlation of the microbial community and
the metabolomics of traditional Barley Nuruk starter fermentation. Biosci Biotechnol Biochem
77:683–690
Reeves ML, Rabinowtiz JD (2011) Metabolomics in systems microbiology. Curr Opin Biotechnol
22:17–25
Roessner U, Wagner C, Kopka J, Trethewey RN, Willmitzer L (2000) Simultaneous analysis of
metabolites in potato tuber by gas chromatography-mass spectrometry. Plant J 23:131–142
Roessner U, Luedemann A, Brust D, Fiehn O, Linke T, Willmitzer L, Fernie AR (2001) Metabolic
profiling allows comprehensive phenotyping of genetically or environmentally modified plant
systems. Plant Cell 13:11–29
Russell WR, Duncan SH (2013) Advanced analytical methodologies to study the microbial
metabolome of the human gut. Trac-Trends Anal Chem 52:54–60
Slupsky CM (2010) NMR-based analysis of metabolites in urine provides rapid diagnosis and
etiology of pneumonia. Biomark Med 4:195–197
Smedsgaard J, Frisvad JC (1996) Using direct electrospray mass spectrometry in taxonomy and
secondary metabolite profiling of crude fungal extracts. J Microbiol Meth 25:5–17
Soga T, Ross GA (1999) Simultaneous determination of inorganic anions, organic acids, amino
acids and carbohydrates by capillary electrophoresis. J Chromatogr A 837:231–239
Soga T, Heiger DN (2000) Amino acid analysis by capillary electrophoresis electrospray
ionization mass spectrometry. Anal Chem 72:1236–1241
Soga T, Imaizumi M (2001) Capillary electrophoresis method for the analysis of inorganic anions,
organic acids, amino acids, nucleotides, carbohydrates and other anionic compounds.
Electrophoresis 22:3418–3425
Soga T, Ueno Y, Naraoka H, Ohashi Y, Tomita M, Nishioka T (2002a) Simultaneous
determination of anionic intermediates for Bacillus subtilis metabolic pathways by capillary
electrophoresis electrospray ionization mass spectrometry. Anal Chem 74:2233–2239
Soga T, Ueno Y, Naraoka H, Matsuda K, Tomita M, Nishioka T (2002b) Pressure-assisted
capillary electrophoresis electrospray ionization mass spectrometry for analysis of multivalent
anions. Anal Chem 74:6224–6229
Stergiopoulos I, Zwiers L-H, Waard, MAD (2002) Secretion of natural and synthetic toxic
compounds from filamentous fungi by membrane transporters of the ATP-binding cassette and
major facilitator superfamily. Eur J Plant Pathol 108:719–734
Storr M, Vogel HJ, Schicho R (2013) Metabolomics: is it useful for inflammatory bowel diseases?
Curr Opin Gastroenterol 29:378–383
Sue T, Obolonkin V, Griffiths H, Villas-Bôas SG (2011) An exometabolomics approach to
monitoring microbial contamination in microalgal fermentation processes by using metabolic
footprint analysis. Appl Environ Microbiol 77:7605–7610
Tredwell GD et al (2011) The development of metabolomic sampling procedures for Pichia
pastoris and baseline metabolome data. Plos One 6(1):e16286
Tweeddale H, Notley-McRobb L, Ferenci T (1998) Effect of slow growth on metabolism of
Escherichia coli, as revealed by global metabolite pool (Metabolome) analysis. J Bacteriol
180:5109–5116
Tweeddale H, Notley-McRobb L, Ferenci T (1999) Assessing the effect of reactive oxygen species
on Escherichia coli using a metabolome approach. Redox Rep 4:237–241
van der Werf MJ, Overkamp KM, Muilwijk B, Coulier L, Hankemeier T (2007) Microbial
metabolomics: toward a platform with full metabolome coverage. Anal Biochem 370:17–25
Villas-Bôas SG, Delicado DG, Åkesson M, Nielsen J (2003) Simultaneous analysis of amino and
nonamino organic acids as methyl chloroformate derivatives using gas chromatography-mass
spectrometry. Anal Biochem 322:134–138
Villas-Bôas SG, Moxley JF, Åkesson M, Stephanopoulos G, Nielsen J (2005a) High-throughput
metabolic state analysis: the missing link in integrated functional genomics of yeasts.
Biochem J 388:669–677
Villas-Bôas SG, Højer-Pedersen J, Åkesson M, Smedsgaard J, Nielsen J (2005b) Global
metabolite analysis of yeasts: evaluation of sample preparation methods. Yeast 22:1155–1169
12 S.G. Villas-Boas
Villas-Bôas SG, Noel S, Lane GA, Attwood G, Cookson A (2006) Extracellular metabolomics: a
metabolic footprinting approach to assess fiber degradation in complex media. Anal Biochem
349:297–305
Villas-Bôas SG, Bruheim P (2007) Cold glycerol-saline: the promising quenching solution for
accurate intracellular metabolite analysis of microbial cells. Anal Biochem 370:87–97
Villas-Bôas SG et al (2008) Phenotypic characterization of transposon-inserted mutants of
Clostridium proteoclasticum B316T using extracellular metabolomics. J Biotech 134:55–63
Wang Q, Wu C, Chen T, Chen X, Zhao X (2006) Integrating metabolomics into systems biology
framework to exploit metabolic complexity: strategies and applications in microorganisms.
Appl Microbiol Biotech 70:151–161
Wittmann C, Krömer JO, Kiefer P, Binz T, Heinzle E (2004) Impact of the cold shock
phenomenon on quantification of intracellular metabolites in bacteria. Anal Biochem 327:135–
139
Xie BG, Waters MJ, Schirra HJ (2012) Investigating potential mechanisms of obesity by
metabolomics. J Biomed Biotech #805683.
Zenobi R (2013) Single-cell metabolomics: analytical and biological perspectives. Science
342:1243259
Zhang AH, Sun H, Yan GL, Wang P, Han Y, Wang XJ (2014) Metabolomics in diagnosis and
biomarker discovery of colorectal cancer. Cancer Lett 345:17–20
Chapter 2
Microbes, Metabolites and Health
1 Introduction
diverse microbial populations could deliver very similar functional outcomes for
both the microbial community and the host (Huttenhower et al. 2012).
There is growing evidence that the gut microbiota impacts significantly on the
health of the host. Animal studies have shown that the gut microbiota plays a key role
in the post-natal maturation of both the gut itself and the gut immune system (Smith
et al. 2007; Chung et al. 2012). In humans, changes in gut microbiota structure have
been associated with gut-associated disorders including: inflammatory bowel dis-
eases (Gevers et al. 2014; Frank et al. 2007; De Cruz et al. 2015) and colorectal
cancer (Garrett 2015); metabolism-related disorders such as obesity (Ley et al. 2006),
metabolic syndrome (Le Chatelier et al. 2013; Haro et al. 2016) and diabetes (Qin
et al. 2012) but also with systemic disorders including cardiovascular disease, neu-
rodegenerative diseases including Alzheimer’s (Alam et al. 2014; Naseer et al. 2014)
and Parkinson’s (Goldman et al. 2014; Scheperjans et al. 2015) diseases and
neuro-developmental and psychiatric conditions including autism spectrum disorder
(Louis 2012; Mulle et al. 2013) and schizophrenia (Nemani et al. 2015).
Of these disorders, the role of the microbiota has probably been studied most
extensively in obesity which is also a well-recognized risk factor for many of the
other chronic diseases mentioned above. Obese humans, when compared to normal
weight individuals, displayed a reduced ratio of microbes from the Bacteroidetes
phylum relative to those of the Firmicutes phylum in their faeces and this ratio
increased in obese individuals who lost weight (Ley et al. 2006). Important insights
into the functional significance of these associations have come from the use germ
free, gnotobiotic and gene mutant animal models. This same relationship of
Bacteroidetes to Firmicutes was also observed in obese mice carrying two copies of
the ob mutation in the leptin gene (ob/ob mice) relative to their ob/+ and +/+ litter
mates. In this animal model, biochemical analyses revealed that these different
microbiota varied in the efficiency of energy harvest from the diet and that both the
obese-prone and lean phenotypes was transferrable to germ free mice by colo-
nization with the microbiota from obese and lean donors respectively (Turnbaugh
et al. 2006). This same result was observed when human faecal microbiota from
female twins discordant for obesity were used to colonize germ free mice (Ridaura
et al. 2013). Mice practise coprophagy. Co-housing of mice, conventionalised with
the obese twin’s microbiota, with other mice conventionalised with the lean twin’s
microbiota prevented acquisition of the obese phenotype. Microbial community
analysis revealed that this prevention of obesity was associated with an invasion of
Bacteroidetes from the lean microbiota into the microbiota of the
obese-conventionalised animals (Ridaura et al. 2013). Interestingly this effect was
diet-dependent and was most marked when the animals were fed a diet high in fruit
and vegetables. When the animals were fed a diet high in saturated fat and low in
fruit and vegetables, levels of Bacteroidetes invasion from the lean convention-
alised animals was very low (Ridaura et al. 2013) highlighting the importance of
diet as a selective force in gut microbiota structure/function determination. While
microbial population changes associated with diseases have been extensively
studied, the mechanisms by which these changes elicit their physiological effects on
the host are just beginning to be unraveled.
2 Microbes, Metabolites and Health 15
Metabolites and other products made by the gut microbiota play a key role in the
dialogue between the gut microbiota and the host. Contributions of the gut micro-
biota to the nutritional wellbeing of the host are well known and include the provision
of vitamin K, B group vitamins, B12, biotin, riboflavin, thiamine and folate (Koenig
et al. 2011; Said 2011) and amino acids (Yatsunenko et al. 2012). However, gut
microbes also make a vast array of proteins, peptides and low molecular weight
metabolites and chemically transform others of host or bacterial origin, all of which
impact on the structure and functionality of the microbiota and the health of the host
(Nicholson et al. 2012). Some bacteria help shape their local community by pro-
ducing bacteriophage or bacteriocins to kill off specific competitors. The production
of bacteriocins by commensal Escherichia coli can, for example, inhibit the invasion
of their niche by pathogenic E. coli O157:H7 (Schamberger and Diez-Gonzalez
2002). Others form feeding chains where one species performs an initial break down
of a starting nutrient source to release intermediate breakdown products that can
themselves provide substrates for other types of bacteria as exemplified using in vitro
fermentation studies to examine the degradation of dietary glycans (Leitch et al.
2007a, b) and host-derived mucins (Png et al. 2010). Others can induce the host to
create local environments that provide a colonisation and proliferation advantage for
themselves and potentially other bacteria (Collier et al. 2008), while others can have
direct effects on host functions e.g. the production of gamma amino butyric acid
(GABA) (Barrett et al. 2012), the major inhibitory neuro transmitter in the mam-
malian central nervous system (CNS) with a major role in the regulation of muscle
tone in humans. Although GABA produced peripherally does not cross the blood
brain barrier and hence is unlikely to impact on CNS functions, it may modify gut
motility and blood pressure (Inoue et al. 2003).
Likewise the host helps shape the gut microbiota through the secretion of
bioactive molecules into the lumen of the gut. Defensins, (Gallo and Hooper 2012)
secreted from the Paneth cells and most enterocytes of the gut mucosa, and IgA
(Cerutti 2008), secreted from plasma cells in the lamina propria and through the gut
epithelium, provide protection against overgrowth of many pathogenic microbial
species. Further, using a germ free/gnotobiotic maternal separation mouse model of
stress it has been demonstrated recently that stress can alter the colonic environment
leading to an altered colonic microbiota. While this altered microbiota was insuf-
ficient to recapitulate the stress-associated changes in host physiology upon
microbial transplantation to non-stressed germ free animals, transplantation was
associated with a transfer of anxiety-like behavior and behavioral despair but the
molecular effectors are yet to be defined.
So what are the key influencers of gut microbiota structure and function and how
might they elicit their impacts on health? While the gut of the near term foetus may
have some microbes present, the numbers are very low (Jimenez et al. 2008). The
mode of delivery (vaginal vs. caesarean section) impacts significantly on the
structure of the gut microbiota of the neonate (Dominguez-Bello et al. 2010) with
vaginally delivered infants developing bacterial communities resembling those of
their mother’s vaginal microbiota, while the gut microbiota of caesarean-delivered
infants’ reflected more closely those of their mother’s skin. Prior to weaning the gut
16 T.J. Lockett et al.
The high community awareness of the health potential of dietary fibre is relatively
new. This interest was triggered by early work of a number of British physicians in
South and East Africa. They saw that native Africans ate a diet high in whole grain
foods and were essentially free of the non-infectious diseases which affected
Europeans living in the same geographical area who ate highly refined foods
(Burkitt 1973). Initially, they focused on large bowel disorders including consti-
pation, diverticular disease, appendicitis and related problems. They linked these
differences to their dietary fibre intakes and suggested that the disease profile of the
Europeans reflected a simple dietary fibre deficiency. These early studies relied on
observation (not measurement) and were quite limited in scope. They were limited
also by the early definition of dietary fibre as plant cell wall material resistant to
human small intestinal enzymes i.e. indigestible and largely insoluble plant mate-
rial. The total indigestibility of these non-starch polysaccharides (NSP) explained
their faecal bulking and laxating effects and protection against large bowel diseases
very neatly (Cummings et al. 1992). However, fibre (as NSP) has proved very
disappointing for protecting against other diseases of affluence, especially colorectal
cancer (CRC), where it was expected to lower risk substantially. While some
studies (e.g. the European Prospective Investigation into Cancer and Nutrition,
EPIC) have shown a dose-dependent reduction in CRC risk (Murphy et al. 2012),
others have not (Park et al. 2005; Lanza et al. 2007). The Australian paradox also
fits with these inconsistencies. Australia seems to be one of the few countries where
2 Microbes, Metabolites and Health 17
fibre consumption has increased substantially over time with a reported population
average of 28 g/person/day (Baghurst et al. 1996) but CRC morbidity and mortality
rates remain high (AIHW 2012). This paradox is leading to a revision of the relative
importance of the different components which contribute to fibre intake in low risk
populations and also the way in which dietary fibre actually works in the large
bowel.
Much of the credit for the popularization of the dietary fibre hypothesis is due to
Dennis Burkitt (Trowell and Burkitt 1987) but it seems that the focus on fibre as an
indigestible bulking agent may be misplaced. It is now impossible to quantify actual
food consumption rates in the original African population. Changes due to
urbanization and higher incomes have altered the traditional rural lifestyle patterns
of native South Africans so that their activity levels and total dietary fibre
(TDF) consumption have fallen. TDF intakes have declined from an estimated 25–
35 g/person/day (2 generations ago) to 15–20 g/person/day (now) (Segal et al.
2000). One would have expected this to lead to an increase in CRC rates but this
does not seem to have happened. Comparison of the diet of indigenous Africans in
South Africa and African Americans in the United States shows that they are quite
similar in their low TDF content (O’Keefe et al. 2007) but the latter have one of the
highest global rates of CRC of any ethnic group. One suggestion is that the high
content of animal products in the African American diet is responsible. It is true that
CRC risk has been linked to greater consumption of red and processed meat (Norat
et al. 2005) but the work of Walker and others suggest very strongly that the type of
fibre consumed by Africans may be equally important (Walker et al. 1986). The
original concept underpinning the differential in disease risks was the high whole
grain consumption of the Africans. Put simply, the notion of “fibre” came later,
essentially as roughage (Topping and Illman 1986). However, it has emerged that
the culinary habits of the Africans are an important factor. Many native South
Africans consume reheated or cold maize porridge on a daily basis, particularly in
rural areas. Cooking gelatinises the starch, increasing its small intestinal
digestibility but cooling leads to self-association of the chains. This process (ret-
rogradation) leads to resistance to amylolysis i.e., the formation of resistant starch
(RS). Resistance to amylase is a key feature of TDF meaning that RS is also a fibre
component. It has been shown that hot traditionally cooked maize meal contains
18 g of RS/100 g while cooked and cooled maize contains a higher RS content than
that of a hot maize meal (Heneker et al. 1998). Cooking is not the only factor. The
amylose content of South African maize is 37–40 % of total starch which is rela-
tively high (37.1–39.9 %) (van der Merwe et al. 2001). Amylose is a relatively
compact starch polymer which is slower to gelatinize than amylopectin and also
quicker to retrograde meaning that it is digested more slowly than low amylose
starches and also yields more RS in food products.
One of the most important observations driving the reappraisal of the relative
importance of fibre (i.e., NSP and RS) was the lack of relationship between TDF
and bowel habit in native South African children. This has led to the current focus
on the role of the microbiota in human health, specifically through the production of
short chain fatty acids (SCFA). Knowledge of the existence of RS is not new nor is
18 T.J. Lockett et al.
the presence of SCFA in the large bowel of omnivores (Elsden et al. 1946). It was
known that TDF polysaccharides are lost during whole of gut transit (Schneeman
1986). For NSP, the extent of loss varies by source and ranges from *0 (for
cellulose) to >90 % for pectin. In contrast very little starch appears in normal
human faeces which, coupled with its susceptibility to amylolysis, gave the obvious
conclusion that small intestinal starch digestion was complete. The existence of
potentially significant levels of indigestible starch (i.e., amylase resistant starch—
RS) in foods was shown by Englyst and colleagues (Englyst et al. 1982). There was
earlier (indirect) evidence from Levitt’s laboratory (Anderson et al. 1981) in intact
humans showing that, following consumption of a low-fibre convenience cereal
product, breath H2 evolution rose significantly. Breath H2 is a marker for large
bowel fermentation and their observations can now be interpreted as showing the
presence of RS and also that fermentation was occurring. Both are now established
as significant factors in human gut physiology (Topping and Clifton 2001). RS is
defined in terms of starch which escapes into the large bowel and the products of its
fermentation. RS is today recognized as a component of fibre and not a separate
entity.
There are striking similarities between large bowel fermentation by the micro-
biota of humans and other omnivores and that of obligate herbivores (Topping and
Clifton 2001). The end products are similar i.e., SCFA (principally acetate, pro-
pionate and butyrate), gases (CH4, H2 and CO2) and the energy to fuel bacterial
growth. In humans and relevant model species (most notably pigs), SCFA levels are
highest in the proximal large bowel and decline with passage of the digesta stream.
This is consistent with the high production rates in the caecum and proximal colon
i.e., where substrate availability is greatest. The fall in SCFA levels along the colon
reflects substrate depletion but also their uptake by the large bowel so that <10 % of
total production is excreted in faeces. This distributional profile has very important
implications for gut health and is believed to relate to the higher prevalence of
non-infectious disease in the distal colon and rectum (Topping and Clifton 2001;
Cats et al. 1996).
SCFA have a number of general effects which contribute to large bowel health
(Topping and Clifton 2001; Brouns et al. 2002; Wong et al. 2006). Their production
acidifies the digesta which inhibits the overgrowth of potential pathogens and limits
the absorption of potentially toxic compounds e.g. NH3. The latter cannot enter
colonocytes as NH4+ so lower pH values limit their exposure to this and other
cytotoxic and genotoxic agents (Topping 2007). A study in normal humans con-
suming their usual diets with supplements of NSP or NSP + RS showed an increase
in faecal NH3 excretion which correlated closely with greater butyrate excretion
(McOrist et al. 2011). This is consistent with diminished absorption by (and less
exposure to) colonocytes. SCFA enhance visceral blood flow through dilation of
resistance vessels in the colon which facilitates O2 supply (Topping and Clifton
2001). The history of research in dietary fibre is littered with misconceptions,
revisions and reappraisals. The current focus on the microbiome is another case in
point. The emergence of modern technologies has given us an opportunity to
identify the species which are present and how they are altered by diet. Previously,
2 Microbes, Metabolites and Health 19
Dietary fibre is not a single entity but an umbrella term for a large, diverse group of
substances, predominately but not exclusively carbohydrate polymers of plant
origin, that pass through the small intestine and undergo partial or complete fer-
mentation in the colon.
The global consensus definition of dietary fibre (Codex 2010), has broadened
from a botanical and structural focus (plant cell wall polymers) and now captures
other salient properties of fibre, notably compositional diversity and physiological
functionality.
The major classes of dietary fibre are NSP, RS and non-digestible oligosac-
charides although inclusion of the latter can vary with jurisdiction (Codex 2010).
Fibre also includes analogous compounds, non-digestible carbohydrate polymers,
of animal origin, and plant secondary metabolites that are intimately associated with
plant cell wall polysaccharides, notably lignin.
Fibres, both natural and synthetic, differ markedly in their physiological prop-
erties and capacity to promote human health (Dahl and Stewart 2015; Bird 2007;
Cummings and Stephen 2007; Buttriss and Stokes 2008; Brownlee 2014; Edwards
et al. 2015). While physiological functionality is included in the definition of fibre,
it is applied only to foods containing carbohydrate polymers sourced from either
raw food materials or chemically synthesized. A demonstrable beneficial physio-
logical effect in humans is a requirement for foods to be advertised as containing
added/functional fibre.
A prerequisite property and defining feature of fibre is that it is potentially
fermentable by colonic microbiota. In recent times there has been growing scientific
and commercial interest in highly fermentable fibres, notably non-digestible
oligosaccharides and resistant starches, owing to their favourable effects on gut
microbiota composition and activity, and host health (Bird 2007; Conlon and Bird
2015; Bird and Topping 2001; Broekaert et al. 2011, de Menezes et al. 2013).
NSP, mainly cellulose and hemicelluloses, and lignin, are the fundamental
structural elements of plant cell walls and have been integral to all descriptions of
dietary fibre (Miller Jones 2014). Cellulose and hemicelluloses are widely dis-
tributed in the plant kingdom and are ubiquitous in the plant foods that humans eat.
NSP typically comprise long chains of monosaccharides, mostly glucose moieties,
joined by b-1,4 linkages. Cellulose is a high molecular weight homopolymer
consisting of linear b-1,4 linked-chain of glucose units. The chains are closely
packed and the hydrogen bonds that form between adjacent chains ensure that this
20 T.J. Lockett et al.
between 2 and 60 fructose residues whereas FOS comprises smaller molecules with
a DP between 2 and 8. ‘Oligofructose’ is a partial enzymatic hydrolysate of inulin.
Fibre also encompasses a range of other plant substances, including lignin, cutin,
waxes, suberin, tannins, saponins and phytosterols, that are naturally associated
with botanical cellular structures (Bird 2007; Englyst et al. 2007; Miller Jones
2014). Lignin is a heterogenous polyphenolic ether, not a carbohydrate, that is
covalently bonded to cellulose (Tungland and Meyer 2002). Most of these sub-
stances are quantitatively minor constituents of human diets and are not metabo-
lized extensively by the gut microbiota (Topping and Clifton 2001). However, they
may impair fermentation of plant structural and storage polysaccharides (Stephen
1994; Edwards et al. 2012).
In certain disease settings, dietary carbohydrates that are normally digestible in
the upper gut (‘available carbohydrate’) can also make their way to the colon and
essentially function as dietary fibre. For instance, lactose can be an important
substrate for the microbiota in those individuals with lactase deficiency (Deng et al.
2015; Lukito et al. 2015; Wahlqvist 2015; Windey et al. 2015). The contribution of
this carbohydrate is dependent on the degree of lactase deficiency. Other bowel
diseases and conditions that compromise the functional capacity of the upper gut,
such as celiac and Crohn’s diseases, will also increase the amount of malabsorbed
carbohydrate, and other dietary constituents, reaching the colonic microbiota
(Conlon and Bird 2015).
Physicochemical and functional properties of fibre, including solubility in water,
viscosity and gel formation, and water holding capacity, have formed the basis of
various classification schemes. Soluble/insoluble fibre classification has proven
popular, however it is inconsistent with the accepted definitions for fibre and is an
unreliable predictor of physiological function. ‘Solubility’ relates to an in vitro
environment, not the gut lumen. The proportion of fibre that is solubilised is often a
function of the assay conditions used and in many instances the results are method
specific (Cummings and Stephen 2007; Englyst et al. 2007). The proportion of
soluble fibre can also change during food processing and passage of the fibre
through the upper gut (Arrigoni 2001; Comino et al. 2016).
Dietary fibres can be classified on their fermentability (Dahl and Stewart 2015;
Slavin 2001). But, gauging the rate and extent of fibre fermentation in vivo is
challenging, which is why knowledge of fermentation of fibre-rich substrates is
based largely on studies deploying in vitro techniques. These commonly use human
faecal inocula in static and flow-through incubation systems to simulate conditions
and events in the complex, anaerobic and highly dynamic micro-ecosystem of the
large bowel. As such, they provide limited indicative data on fibre degradation by
the colonic microbiota.
Generally, about 70 % of the fibre that is consumed by people on mixed diets is
considered to be fermented to completion in the colon (Slavin 2001; Topping and
Clifton 2001). However, individual fibres vary markedly in their fermentability
although most are partially fermentable. Water soluble fibres, such as those from
vegetables & fruits, are usually more easily fermented compared to insoluble fibres,
such as cellulose, which is quite resistant to fermentation. For instance, only about
22 T.J. Lockett et al.
fermentable substrates but the amount and type is very much diet dependent
(Grembecka 2015; Halmos et al. 2015; Muir et al. 2009; Tennant 2014).
Interactions between different fibre components in a food as well as its physical
form (matrix), in particular the structural integrity of plant cell walls, will influence
the rate and extent of fermentation of the component fibres. Milling, processing,
preparation and storage conditions of foods can alter their fibre content and func-
tionality and hence their impact on the gut microbiota (Tuohy et al. 2012). For
instance, soluble fibre may be lost through leaching during the cooking process.
Heat treatment may solubilise certain fibres, thereby increasing the ratio of soluble
to insoluble fibre. Short chain fructans can be degraded by yeast enzymes and also
exposure to acidic conditions. A sufficiently large particle size is needed to ensure
an intact cell wall structure resists extensive fermentation. Coarse bran loses both its
water holding capacity and some of its laxative effects when it is finely ground
simply because it is more extensively fermented in the large bowel (Topping and
Bird 1999).
Another important consideration is functional capacity of the upper gut. Small
intestinal digesta transit rate and nutrient assimilation can differ markedly among
people. For instance, some individuals are especially efficient in digesting and
fermenting starch (Thornton et al. 1987). Furthermore, some fibres may undergo
degradation in the upper digestive tract owing to the action of bacterial glycanases
in that region of the gut (Bach Knudsen 2015). However, the loss of fermentable
substrate is probably small.
Poorly fermented carbohydrates may alter the metabolic capacity of the gut
microbiota through various mechanisms. For instance, by acting as a physical
scaffold for colonic microbes, resident and transitory, it may facilitate the fer-
mentation process. Fibres in general also influence fermentation events by aug-
menting intestinal motility and flow of the faecal stream (Topping and Bird 1999;
James et al. 2015).
The dietary fibres described in the previous section have distinct effects on the gut
microbiome. This can be because of their solubility, structure or where in the gut
the fibre is fermented. This differential effect will lead to selection of certain groups
of bacteria, specific fermentation outputs, like SCFA and gas production, and likely
inhibition of pathogenic bacteria.
Some of the most widely studied prebiotic fibres are inulin, fructooligosaccharies
(FOS) and galactooligosaccharides (GOS) with the latter two encompassing
numerous different types of fibres. All of these fibres are soluble and highly fer-
mentable which means they are fermented in the terminal ileum or the proximal
colon (Eswaran et al. 2013). FOS and GOS were both found to have a bifidogenic
24 T.J. Lockett et al.
effect and increased levels of Lactobacillus in both animals and humans (Davis
et al. 2011; Djouzi and Andiueux 1997; Kleessen et al. 2007a). These effects were
accompanied with higher levels of SCFA and a lower pH in the large bowel,
however FOS were found to favour an increase in butyrate whereas GOS advocated
for higher propionate levels (Djouzi and Andiueux 1997). FOS and GOS also had
differential effects on gas production in rats, with FOS feeding increasing both
hydrogen and methane excretion, but GOS only induced higher methane excretions
(Djouzi and Andiueux 1997). Inulin seems to have differential effects in animals
and humans. This difference was found albeit using a human microbiota-associated
rat model. In the rat model, no difference in Bifidobacterium spp. was observed after
inulin intake, but higher levels of fibre degrading bacteria Roseburia spp. and
Eubacterium rectale were observed and a reduction in potentially pathogenic
bacteria (Clostridium spp.) (Kleessen et al. 2007a; Van den Abbeele et al. 2011).
SCFA concentrations increased and pH levels reduced with inulin intake in ani-
mals. Human intervention trials have not been able to replicate these changes. In
these studies, higher levels of Bifidobacterium, Faecalibacterium prasnitzii,
reduced numbers of Bacteroides-Prevotella and potentially pathogenic bacteria
were observed (Kleessen et al. 2007b; Ramirez-Farias et al. 2009; Costabile et al.
2010). In a later study, Ramirez-Farias et al. (2009) showed that it was certain
species of Bifidobacterium; B. adolescentis, B. longum and B. bifidum that changed
with inulin intake. Another significant difference between the animals and human
studies was the absences of changes observed in faecal SCFA levels in humans
given inulin (Kleessen et al. 2007b; Costabile et al. 2010). Another way to evaluate
the effect of certain fibres is to remove them from the diet. This was done by
Halmos et al. (2015) in a randomised control crossover trial of a low Fermentable
Oligo- Di- and Monosaccharides And Polyols (FODMAP) diet (no inulin) and a
diet containing a typical level of FODMAP’s found in an Australian diet. This study
showed that by avoiding FODMAP the overall bacterial abundance, and the relative
abundance of Clostridium cluster XIVa (butyrate-producing bacteria) and the
mucus-associated bacterium Akkermansia muciniphila was reduced. However,
these changes did not result in differences in faecal SCFA levels, although pH was
higher on the low FODMAP diet.
A possible reason for not noticing any differences in faecal SCFA levels may
well be because >95 % of the SCFAs produced in the human colon are taken up by
the gut epithelia (Kleessen et al. 2007b; Costabile et al. 2010). This does not
exclude that there might be a difference in SCFA levels in the proximal colon
however, when SCFAs are measured in faeces, levels are diminished due to uptake
along the colon.
RS, can also change the gut microbiome and the metabolic components pro-
duced, however the RS type plays an important role. All of the RS types are
generally digested in the proximal and the transverse colon, but some are digested
throughout the colon like RS type 1 and 4 (Clarke et al. 2011b; Eswaran et al.
2013).
Animal studies have revealed great benefit and potential for RS as a prebiotic
fibre. A limited number of studies have been conducted using RS type 1; in animals
2 Microbes, Metabolites and Health 25
(rats), this type of RS did promote higher abundances of Bifidobacterium spp. and
increased SCFA levels at month five compared to control (Kleessen et al. 1997).
The RS type 2 fibre is often used in animal studies and seems to have a broader effect
on the gut microbiota. It was shown in rat and pig studies that RS type 2 can increase
the abundances of Bifidobacterium spp., Lactobacillus spp., Enterobacteriaceae and
Ruminococcus bromii compared to control animals (Conlon et al. 2012; Abell et al.
2011; Kleessen et al. 1997; Le Blay et al. 1999; Martin et al. 1998). These microbial
changes were accompanied by higher SCFA levels due to RS type 2 feeding with
certain studies observing specific increases in propionate (Conlon et al. 2012; Abell
et al. 2011) and butyrate (Conlon et al. 2012; Abell et al. 2011; Le Blay et al. 1999;
Martin et al. 1998) excretion. RS type 3 have also been shown in rat and pig studies
to select for Bifidobacterium spp., Lactobacillus spp. and Faecalibacterium
prausnitzii, but lower the abundances of gamma-Proteobacteria (Conlon et al. 2012;
Haenen et al. 2013). Again, these microbial changes were in conjuction with
increased SCFA levels, especially butyrate and propionate. The gut microbiota can
also be modified with RS type 4 with a rat study conducted with chemically modified
RS showing increases in Lactobacillus spp. and Parabacteroides distasonis com-
pared to control (Abell et al. 2011). This study also recorded increases in SCFA
levels.
Human studies have confirmed many of the changes observed in the animal gut,
however with a high inter-individual variation for both microbial and biochemical
composition. Studies in humans are generally conducted using RS type 2, 3 and 4.
RS type 2 and 3 have very similar effects in the human gut promoting higher levels
of R. bromii, F. prausnitzii and E. rectale compared to a Non-Starch Polysaccharide
(NSP) control (Abell et al. 2008; Martinez et al. 2010; Walker et al. 2011). For two
of these studies, SCFA were also measured and the RS consumption was associated
with higher levels of faecal SCFA levels, especially butyrate (Abell et al. 2008;
Walker et al. 2011; McOrist et al. 2011). This increase in faecal butyrate levels
successfully correlated with the increasing faecal abundance of F. prausnitzii and E.
rectale (Abell et al. 2008; Walker et al. 2011). Human research studies have been
conducted with RS type 4 showing beneficial effects on the gut microbiota with
higher abundances of Actinobacteria, Bacteroidetes and a reduction in Firmicutes
(Martinez et al. 2010), more specifically Clostridium cluster XIVa and IV,
Lactobacillus, B. adolescentis, P. distasonis and R. bromii (Clarke et al. 2011b,
Martinez et al. 2010; West et al. 2013; Le Leu et al. 2015). Reductions were also
observed in bacteria, Ruminococcus gnavis, R. torques and E. coli, previously
associated with gut diseases (Le Leu et al. 2015). RS type 4 consumption also
resulted in higher SCFA level in the faeces of volunteers consuming these fibres.
These substantial shifts in bacterial composition imply that the different RS types
have the ability to select for certain groups of bacteria, despite a relative high
inter-individual variation. Combined, these studies strongly suggest a key role for
R. bromii in the degradation of RS and these bacteria have also been found to be the
predominant ones to colonise RS (Leitch et al. 2007a; Ze et al. 2012). It is also
worthwhile keeping in mind that these prebiotic effects are not universal, as some
volunteers are non-responders. The response in any volunteer will always depend
26 T.J. Lockett et al.
on the initial composition of the gut microbiota which helps to explain the often
large inter-individual variation seen in human intervention studies examining the
gut microbiome.
Fibres, like NSP, with a very limited prebiotic effect because up to 75 % of the
ingested fibre is not fermented in the gut (Backman 2009; Roberts et al. 2010) and
no selective promotion of specific bacterial populations (Abell et al. 2008; Walker
et al. 2011) can still affect the gut microbiota. It is suggested that NSP can interact
with the intestinal bacteria via contrabiotic effects, whereby they prevent potentially
harmful interactions between bacteria and the gut epithelium that occur upon
dysbiosis (Backman 2009; Parsons et al. 2014; Roberts et al. 2010, 2013). The
contrabiotic effect has been tested for a range of soluble NSP’s and they are not all
equally effective. NSP from plantain bananas and broccoli seems to have greater
effect than NSP from apple and leek (Roberts et al. 2010). These tests have mainly
been conducted in vitro testing the ability of the fibres to block the attachment of
adherent, invasive E. coli (AIEC). Soluble plantain NSP’s have not only been able
to block AIEC but also adherent enteric pathogens, like Shigella spp., Clostridium
difficile, enterotoxigenic E. coli and Salmonella spp. (Parsons et al. 2014; Roberts
et al. 2013). Parsons et al. (2014) also demonstrated that NSP from plantain bananas
can block Salmonellosis in chicken when added to their feed and translocation of
Salmonella Typhimurium across isolated follicle-associated epithelium from human
ileum. It is believed that microfold (M)-cells overlying Peyer’s patches in the
human ileum and lymphoid follicles in the colon are potential points of entry for
AIEC (Roberts et al. 2010). It is suggested that the contrabiotic effect is mediated
via an interaction between the soluble NSP and the epithelial cell causing an
electrogenic chloride secretion preventing the adhesion of the pathogens (Parsons
et al. 2014). Another and more simple explanation is the direct interaction between
plantain NSP and E. coli, C. difficile and Salmonellae as they can all utilise plantain
NSP as an energy source (Roberts et al. 2010, 2013).
Collectively, the microbes present within the GI tract can produce a vast array of
bioactive compounds, many of which can influence the functions of host tissues.
A comprehensive review of the many known gut microbe-derived metabolites and
their functions can be found elsewhere (Nicholson et al. 2012). We will focus our
discussion on the impacts of some of the primary fermentation end-products, par-
ticularly the SCFA, which are considered beneficial and largely a result of the
breakdown of dietary polysaccharides, and some toxic products such as ammonia,
phenols and hydrogen sulphide which are largely derived from fermentation of
dietary protein.
2 Microbes, Metabolites and Health 27
butyrate has been shown to improve diversion colitis symptoms (Harig et al.
1989), helping to prevent atrophy of colonic tissues (Luceri et al. 2016).
Enteropathogenic microbes such as certain strains of E. coli and Enterococcus
may contribute significantly to IBD and other gut conditions. In addition to the
evidence of raised numbers in affected individuals there is experimental evidence to
support such activities. To add weight to an important role for SCFA production,
and the microbes which mediate this, in helping to protect against effects of these
bacteria, the toxic effects of enterohaemorrhagic E. coli O157:H7 can be inhibited
in vivo through increased production of acetate (Fukuda et al. 2011b). The acetate
derived from bacteria of the genus Bifidobacterium was able to mediate this pro-
tection, suggesting potential for the use of appropriate strains as probiotics to treat
conditions where E. coli or other similar pathogens contribute to disease. It is
important to note that some strains of E. coli may benefit health e.g. by maintaining
insulin-like growth factor signalling to muscle and preventing wasting associated
with intestinal infections by certain pathogenic microbes (Schieber et al. 2015).
Nevertheless, increasing numbers of gram negative bacteria such as E. coli can
increase the risk of inflammation as components of their cell well, lipopolysac-
charide, promote inflammatory responses in tissues.
Despite numerous microbial species having been linked to IBD, it has not been
possible to consistently pinpoint any particular microbes as a cause. It is quite likely
that many gut microbes have the potential to trigger IBD under the right conditions,
especially those where host defences, particular the mucus gut barrier, are weakened
(Swidsinski et al. 2009). A large proportion of host immune defences reside within the
gut to deal with the threat of luminal microbes invading the tissues. One of the first lines
of defence is the mucus barrier which lines the gastrointestinal tract. Microbes are
present within the outer layer of this mucus, and many use it as a nutritional substrate,
but they are not normally present within the innermost layer. A breakdown of mucus
barrier integrity is implicated in IBD, in part because the numbers of bacteria that
contribute to large bowel mucus turnover have been shown to be altered in those with
the disease, i.e. A. muciniphila, Ruminococcus torques and Ruminococcus gnavus (Png
et al. 2010). Interestingly, some of these bacteria have also been found to be altered in
the stool of children with autism spectrum disorder, who also often report a significant
level of GI discomfort (Wang et al. 2011).
The maintenance of tight junctions between cells of the GI tract is also a critical
line of defence by helping to prevent passage of unwanted microbes or molecules
into host tissues. SCFA, but particularly butyrate, appear to play an important role
in the maintenance of tight junctions (Fukuda et al. 2011b; Suzuki et al. 2008).
A breakdown of this defence, a so-called ‘leaky gut’, may be a significant con-
tributor to inflammation in IBD and is also linked to a growing number of other
conditions (Michielan and D’Inca 2015). The recognition that a ‘dysbiosis’ of gut
microbe populations is a likely contributor to IBD has recently led to the testing of a
seemingly radical new treatment, faecal microbial transplantation (FMT), in which
an individual receives stool donated by healthy individuals. It is hypothesised that
the healthy complement of microbes will become established in the large bowel,
promote re-establishment of tight junctions, a protective mucus lining, repopulate
2 Microbes, Metabolites and Health 29
the mucus layer with commensal bacteria and thereby curtail microbe-mediated
inflammation. Although relatively unproven for treatment of IBD and other con-
ditions where microbes contribute to the etiology, FMT is now firmly established as
a successful means of treating C. difficile infections (which often resist all other
known treatments) through rectal administration and through ingestion of capsules
containing stool (Borody and Khoruts 2012; Mattila et al. 2012).
Gut microbes and their products also appear to play a role in the aetiology of
colorectal cancer (CRC). Individuals with IBD have an elevated risk of CRC,
suggesting that microbe-associated inflammation can contribute to oncogenesis. In
otherwise healthy individuals there is a significantly greater risk of CRC when diets
high in red or processed meat and low in fibre, which are typical of western style
diets, are consumed (Norat et al. 2005; Murphy et al. 2012). A range of mechanisms
has been proposed to explain this. Dietary protein can escape digestion and undergo
fermentation in the large bowel, generating toxic by-products which can include
ammonia, phenols, cresols, amines, and hydrogen sulphide (Windey et al. 2012).
N-nitroso compounds (NOC) are alkylating agents which can also be produced
from meat proteins by the action of gut microbes and have also been suggested as
contributing to CRC through their ability to form DNA adducts in human colonic
cells (Bingham et al. 1996). There is growing experimental evidence in animals and
humans that consuming high levels of dietary protein, particularly as red meat,
increases the level of DNA damage in the colon (which is regarded as a required
step in oncogenesis) (Toden et al. 2007; Winter et al. 2011; Conlon et al. 2012;
O’Callaghan et al. 2012; Le Leu et al. 2015). These studies also show that addition
of fermentable dietary fibre to the diet in the form of resistant starch protects against
this damage through mechanisms that appear to be at least partly mediated by the
actions of SCFA produced by the microbiota. Gut microbes, including members of
Lactobacillus, Bifidobacterium and Bacteroides genera, mediate secondary bile
acid formation and turnover (Nicholson et al. 2012). These acids, particularly
deoxycholic acid, are potentially carcinogenic and there is evidence that their levels
are higher when SCFA levels are lower (Ou et al. 2012). Numerous species of gut
microbes have been implicated in CRC, and these may act through a variety of
mechanisms, such as the production of compounds like toxins or forms of reactive
oxygen which damage tissues, or through exclusion of beneficial bacteria (Yu and
Fang 2015). Some of the bacteria implicated include Fusobacterium nucleatum,
invasive bacteria found to be strongly associated with the neoplastic tissues of CRC
patients (Castellarin et al. 2012), Bacteroides fragilis, Enterococcus faecalis,
Helicobacter pylori and Strepococcus bovis (Yu and Fang 2015).
The SCFA products of dietary fibre fermentation by the gut microbiota are pleio-
tropic metabolites that elicit their biological effects via both intra- and extra-cellular
mechanisms. While un-ionized SCFA can cross cellular membranes by simple
30 T.J. Lockett et al.
diffusion, with pKa values of around 4.8, the majority of these acids exist in the
ionised state at normal colonic pH. Therefore, most uptake of SCFA by the tissues
of the colonic mucosa is facilitated by membrane bound transporter proteins. These
include the low affinity/ high capacity transporters SLC16A1 (proton-coupled
monocarboxylate transporter-1, MCT-1), SLC16A3 (MCT-3) and SLC16A7
(MCT-16) which are important when SCFA concentrations are relatively high and
high affinity/ low capacity sodium-coupled monocarboxylate transporter-1
(SMCT-1; or SLC5A8) which are important when digesta and tissue SCFA con-
centrations are low. Low affinity receptors such as MCT-1 are distributed
throughout the gut with expression highest in the large intestine of most mammals
and reside on the apical (luminal) surface of the cell. In contrast, the high affinity
transporter SMCT-1, which also promotes water absorption, is expressed on the
apical epithelial membrane of the distal ileum and large intestine of mice, with
expression highest in the colo-rectum (Iwanaga and Kishimoto 2015). SMCT-1 is
also found on cells within the lamina propria (the tissue immediately below the
mucosal layers) where SCFA, particularly butyrate, influences immune cells and
enteric neurons (Vadivel et al. 2014). The relative affinity of SMCT-1 for its dif-
ferent SCFA substrates is: butyrate > propionate > lactate > acetate, which may
contribute to its proposed tumour suppressor role (Ganapathy et al. 2005).
Past research has focused on the ability of SCFA to modulate biological
responses by directly inhibiting histone deacetylases (HDACs) which results in
increased histone acetylation and altered gene expression. The potent HDAC
inhibitory effects of butyrate on the expression of genes that regulate proteins
involved in cellular apoptosis, cell cycle regulation and DNA repair that may
protect from colorectal oncogenesis (Fung et al. 2012) has received considerable
attention, although HDAC inhibition may be SCFA and tissue specific. Acetate has
recently been found to increase brain histone acetylation-state in rodents (Soliman
and Rosenberger 2011). The effects on the immune response of SCFA on HDAC
inhibition are largely anti-inflammatory (Tan et al. 2014) affecting not only cells of
the innate immune system (Lin et al. 2015) but also increasing Foxp3 gene
expression and the numbers and immunosuppressive function of T regulatory cells
in mice (Tao et al. 2007).
SCFA also modulate biological responses by activating G-protein-coupled
receptors (GPCRs), mainly GPR41 (FFAR3), GPR43 (FFAR2) and GPR109A,
which are expressed on the apical membrane of colonic epithelial and enteroen-
docrine cells (see reviews by Vadivel et al. 2014; Tan et al. 2014). GPR41 and
GPR43 are activated by all three SCFA with propionate the most potent agonist for
both GPR41 and GPR43, and with acetate more selective for GPR43 (Le Poul et al.
2003a). GPR109A is only activated by butyrate (Thangaraju et al. 2009) and is
expressed in adipocytes, intestinal epithelial cells and some immune cells (see
review by Singh et al. 2014). GPR43 was first described in neutrophils (Le Poul
et al. 2003a) and has since been described in other immune cells, the gastroin-
testinal tract including endocrine L-cells of the ileum and colon producing intestinal
peptide YY (PYY) and glucagon-like peptide 1 (GLP-1) (Tolhurst et al. 2012).
GPR41 is more broadly expressed in a number of tissues (Le Poul et al. 2003b).
2 Microbes, Metabolites and Health 31
The gut intestinal barrier consists of a layer of epithelial cells and products
including mucus, immunoglobulins and other antimicrobial agents and microbes
that interact to provide a selective barrier to protect the host. The barrier enables the
uptake of water, electrolytes, nutrients and other molecules while preventing the
entry of antigens and microorganisms (Camilleri et al. 2012). The epithelium forms
the main physical barrier between the gut lumen and the mucosal tissues with
absorption occurring either transcellularly by active transport or passive diffusion,
or through the paracellular spaces between the cells by passive movement. The
selective paracellular barrier is maintained by three adhesive protein complexes:
desmosomes, adherens junctions, and tight junctions (Groschwitz and Hogan
2009). The tight junctions seal the intercellular space and involve claudins and
occludins that form the paracellular pore, and scaffold and other cytoplasmic pro-
teins including zonula occludens (ZO-1, 2 and 3).
Intestinal barrier dysfunction enabling gut translocation of bacteria causing
inflammation and autoimmune disease has also been suggested as a causative factor
in a range of diseases including irritable bowel syndrome (Piche 2014;
Vivinus-Nebot et al. 2014), inflammatory bowel disease (Michielan and D’Inca
2015; Ploger et al. 2012) (IBD), food allergies (Perrier and Corthesy 2011), celiac
disease, asthma (Hijazi et al. 2004) and types 1 (T1D) and 2 (T2D) diabetes (de
Kort et al. 2011). Butyrate, but not acetate, decreased bacterial translocation in cell
models (Lewis et al. 2010) and enhanced intestinal barrier function in vitro by
modifying the expression of the tight junction protein Claudin-1 (Wang et al. 2012).
Elsewhere, acetate has been shown to enhance gut barrier function and protect mice
treated with E. coli O157:H7 from translocation of Shiga toxin from the gut lumen
(Fukuda et al. 2011a).
Impaired tight junction function has been found in patients with Crohn’s disease
(Zeissig et al. 2007) (CD) and ulcerative colitis (Heller et al. 2005) (UC) and first
degree relatives of patients with Crohn’s disease have increased intestinal perme-
ability (Peeters et al. 1997). Paracellular permeability is increased in patients with
quiescent IBD and IBS-like symptoms associated with persistent subclinical
intestinal inflammation (Vivinus-Nebot et al. 2014). Butyrate-producing bacteria
are reduced in the faeces of CD (Takahashi et al. 2016) and UC (Machiels et al.
2013) patients compared to healthy controls. The physical and chemical protective
mucus layer is thinner and more variable in thickness in UC disease patients (Pullan
et al. 1994); in rodents, diets containing resistant starch delivering high concen-
trations of butyrate to the large bowel increased mucus thickness (Toden et al.
2014). However, the use of butyrate enemas to control or maintain relapse in IBD
patients has had mixed results (Breuer et al. 1997; Scheppach 1996; Steinhart et al.
1996; Hamer et al. 2010) which may be due to difficulty maintaining prolonged
mucosal contact. Butyrate metabolism is impaired in the inflamed mucosa of IBD
patients, which may be due to a reduction in butyrate uptake due to a downregu-
lation of the MCT-1 butyrate transporter in IBD (Thibault et al. 2007), leading to
the suggestion that butyrate deficiency is a causative factor for the inflammation
(Thibault et al. 2010). Butyrate delivered to the colon by butyrylated starch
32 T.J. Lockett et al.
7 Future Prospects
there remains much to be done. Expanding our knowledge of the gut microbiome
and its metabolic potential will be important. Only some 10 % of the currently
sequenced open reading frames in the microbiome are recognized as encoding
proteins with an assignable or predicted structural or enzymatic function. Further
our understanding of the metabolome is in its infancy while attribution of the
physiological impacts of metabolites continues to be assessed on a candidate by
candidate basis (Martin et al. 2007). While there is much important work to be done
in the expansion of these individual catalogues (genes, their function; metabolites
and their impact on physiology) there is now a real need to bring these areas closer
together. With the increasing use of high resolution spectroscopy methods to
analyse metabolite profiles of microbial origin, opportunities now exist to statisti-
cally integrate large and complex metabolic and metagenomic data sets, using
informatics to reveal pathways and putative functional relationships previously
inaccessible. However, validation of these proposed relationships will be crucial. In
this regard, the development of improved in vitro fermentation systems that more
accurately represent the processes occurring in the gut digesta and the availability of
gut organoid culture systems that accurately recapitulate biological responses of the
gut mucosa to bioactive metabolites are providing an exciting prospect of a medium
throughput screening of these in silico predictions en route to more classical animal
and human substantiation of the strongest candidates. While we are only in the early
stages of this new, data-driven revolution, if it lives up to its promise then the path
to a more detailed understanding of the gut, its metabolites and their impact on
human health becomes clearer and the promise of translating this knowledge to help
inform the personalization of health care, a tangible and exciting prospect.
References
Abell GC, Cooke CM, Bennett CN, Conlon MA, Mcorist AL (2008) Phylotypes related to
Ruminococcus bromii are abundant in the large bowel of humans and increase in response to a
diet high in resistant starch. FEMS Microbiol Ecol 66:505–515
Abell GCJ, Christophersen CT, McOrist AL, Clarke JM (2011) Dietary resistant and butyrylated
starches have different effects on the faecal bacterial flora of azoxymethane-treated rats. Br J
Nutr 105:1480–1485
Agbenorhevi JK, Kontogiorgos V, Kirby AR, Morris VJ, Tosh SM (2011) Rheological and
microstructural investigation of oat beta-glucan isolates varying in molecular weight. Int J Biol
Macromol 49:369–377
Australian Institute of Health and Welfare & Australasian Association of Cancer Registries
(AIHW) (2012) Cancer in Australia: an overview, 2012 Cancer Series No. 74. Cat. No. CAN
70
Alam MZ, Alam Q, Kamal MA, Abuzenadah AM, Haque A (2014) A possible link of gut
microbiota alteration in type 2 diabetes and alzheimer’s disease pathogenicity: an update. CNS
Neurol Disord-Drug Targets 13:383–390
Anderson IH, Levine AS, Levitt MD (1981) Incomplete absorption of the carbohydrate in
all-purpose wheat-flour. N Engl J Med 304:891–892
36 T.J. Lockett et al.
Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, Deroos P, Liu H, Cross JR, Pfeffer K,
Coffer PJ, Rudensky AY (2013) Metabolites produced by commensal bacteria promote
peripheral regulatory T-cell generation. Nature 504:451–455
Arrigoni E (2001) Enzymatic modification of dietary fiber sources. Marcel Dekker Inc, New York
Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, Fukuda S, Saito T,
Narushima S, Hase K, Kim S, Fritz JV, Wilmes P, Ueha S, Matsushima K, Ohno H, Olle B,
Sakaguchi S, Taniguchi T, Morita H, Hattori M, Honda K (2013) Treg induction by a rationally
selected mixture of Clostridia strains from the human microbiota. Nature 500:232–236
Bach Knudsen KE (2015) Microbial degradation of whole-grain complex carbohydrates and
impact on short-chain fatty acids and health. Adv Nutr (Bethesda, Md.) 6:206–213
Backhed F, Roswall J, Peng YQ, Feng Q, Jia HJ, Kovatcheva-Datchary P, Li Y, Xia Y, Xie HL,
Zhong HZ, Khan MT, Zhang JF, Li JH, Xiao L, Al-Aama J, Zhang DY, Lee YS, Kotowska D,
Colding C, Tremaroli V, Yin Y, Bergman S, Xu X, Madsen L, Kristiansen K, Dahlgren J,
Jun W (2015) Dynamics and stabilization of the human gut microbiome during the first year of
life. Cell Host Microbe 17:690–703
Backman RV (2009) The effects of plantain non-starch polysaccharide upon the gut bacteria. Ph.
D., University of Aberdeen, UK
Baghurst PA, Baghurst KI, Record SJ (1996) Dietary fibre, non-starch polysaccharides and
resistant starch—a review. Food Aust 48:S3–S35
Barrett E, Ross RP, O’Toole PW, Fitzgerald GF, Stanton C (2012) c-Aminobutyric acid
production by culturable bacteria from the human intestine. J Appl Microbiol 113:411–417
Binder HJ (2009) Mechanisms of diarrhea in inflammatory bowel diseases. Ann NY Acad Sci
1165:285–293
Binder HJ (2010) Role of colonic short-chain fatty acid transport in diarrhea. Annu Rev Physiol
72:297–313
Bingham SA, Pignatelli B, Pollock JRA, Ellul A, Malaveille C, Gross G, Runswick S,
Cummings JH, O’Neill IK (1996) Does increased endogenous formation of N-nitroso
compounds in the human colon explain the association between red meat and colon cancer?
Carcinogenesis 17:515–523
Bird AR (2007) Dietary fibre, health and chronic disease. Diab Manage J 12
Bird AR, Brown IL, Topping DL (2000) Starches, resistant starches, the gut microflora and human
health. Curr Issues Intest Microbiol 1:25–37
Bird AR, Conlon MA, Christophersen CT, Topping DL (2010) Resistant starch, large bowel
fermentation and a broader perspective of prebiotics and probiotics. Benef Microbes 1:423–431
Bird AR, Topping DL (2001) Resistant starches, fermentation, and large bowel health. Marcel
Dekker Inc, New York
Bird AR, Usher S, May B, Topping DL, Morell MK (2012) Resistant starch measurement, intakes,
and dietary targets. CRC Press, Taylor & Francis Group, Boca Raton
Borody TJ, Khoruts A (2012) Fecal microbiota transplantation and emerging applications. Nat Rev
Gastroenterol Hepatol 9:88–96
Breuer RI, Soergel KH, Lashner BA, Christ ML, Hanauer SB, Vanagunas A, Harig JM,
Keshavarzian A, Robinson M, Sellin JH, Weinberg D, Vidican DE, Flemal KL,
Rademaker AW (1997) Short chain fatty acid rectal irrigation for left-sided ulcerative colitis:
a randomised, placebo controlled trial. Gut 40:485–491
Broekaert WF, Courtin CM, Verbeke K, van de Wiele T, Verstraete W, Delcour JA (2011)
Prebiotic and other health-related effects of cereal-derived arabinoxylans,
arabinoxylan-oligosaccharides, and xylooligosaccharides. Crit Rev Food Sci Nutr 51:178–194
Brouns F, Kettlitz B, Arrigoni E (2002) Resistant starch and “the butyrate revolution”. Trends
Food Sci Technol 13:251–261
Brownlee I (2014) The impact of dietary fibre intake on the physiology and health of the stomach
and upper gastrointestinal tract. Bioact Carbohydr Diet Fibre 4:155–169
Burkitt DP (1973) Some diseases characteristic of modern western civilization. Br Med J 1:274–
278
Buttriss JL, Stokes CS (2008) Dietary fibre and health: an overview. Nutr Bull 33:186–200
2 Microbes, Metabolites and Health 37
Cummings JH, Bingham SA, Heaton KW, Eastwood MA (1992) Fecal weight, colon cancer risk,
and dietary-intake of nonstarch polysaccharides (dietary fiber). Gastroenterology 103:1783–
1789
Cummings JH, Stephen AM (2007) Carbohydrate terminology and classification. Eur J Clin Nutr
61:S5–S18
Dahl WJ, Stewart ML (2015) Position of the academy of nutrition and dietetics: health
implications of dietary fiber. J Acad Nutr Diet 115:1861–1870
Davis LM, Martinez I, Walter J, Goin C, Hutkins RW (2011) Barcoded pyrosequencing reveals
that consumption of galactooligosaccharides results in a highly specific bifidogenic response in
humans. PLoS One 6:e25200
De Cruz P, Kang S, Wagner J, Buckley M, Sim WH, Prideaux L, Lockett T, McSweeney C,
Morrison M, Kirkwood CD, Kamm MA (2015) Association between specific
mucosa-associated microbiota in Crohn’s disease at the time of resection and subsequent
disease recurrence: a pilot study. J Gastroenterol Hepatol 30:268–278
de Cruz P, Prideaux L, Wagner J, Ng SC, McSweeney C, Kirkwood C, Morrison M, Kamm MA
(2012) Characterization of the gastrointestinal microbiota in health and inflammatory bowel
disease. Inflamm Bowel Dis 18:372–390
de Kort S, Keszthelyi D, Masclee AA (2011) Leaky gut and diabetes mellitus: what is the link?
Obes Rev 12:449–458
de Menezes EW, Giuntini EB, Dan MCT, Sarda FAH, Lajolo FM (2013) Codex dietary fibre
definition—justification for inclusion of carbohydrates from 3 to 9 degrees of polymerisation.
Food Chem 140:581–585
Deng YY, Misselwitz B, Dai N, Fox M (2015) Lactose intolerance in adults: biological
mechanism and dietary management. Nutrients 7:8020–8035
Djouzi A, Andiueux C (1997) Compared effects of three oligosaccharides on metabolism of
intestinal microflora in rats inoculated with a human faecal flora. Br J Nutr 78:313–324
Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, Fierer N, Knight R
(2010) Delivery mode shapes the acquisition and structure of the initial microbiota across
multiple body habitats in newborns. Proc Natl Acad Sci USA 107:11971–11975
Eaton SB (2006) The ancestral human diet: what was it and should it be a paradigm for
contemporary nutrition? Proc Nutr Soc 65:1–6
Edwards CA, Garcia AL, Combet E (2012) Colonic metabolism of bioactive molecules potential
impact of dietary fiber. CRC Press, Taylor & Francis Group, Boca Raton
Edwards CA, Xie CR, Garcia AL (2015) Dietary fibre and health in children and adolescents. Proc
Nutr Soc 74:292–302
Elsden SR, Hitchcock MWS, Marshall RA, Phillipson AT (1946) Volatile acid in the digesta of
ruminants and other animals. J Exp Biol 22:191–202
Englyst H, Wiggins HS, Cummings JH (1982) Determination of the non-starch polysaccharides in
plant foods by gas-liquid-chromatography of constituent sugars as alditol acetates. Analyst
107:307–318
Englyst K, Quigley M, Englyst H, Parmar B, Damant A, Elahi S, Lawrance P (2013) Evaluation of
methods of analysis for dietary fibre using real foods and model foods. Food Chem 140:568–
573
Englyst KN, Liu S, Englyst HN (2007) Nutritional characterization and measurement of dietary
carbohydrates. Eur J Clin Nutr 61:S19–S39
Eswaran S, Muir J, Chey WD (2013) Fiber and functional gastrointestinal disorders. Am J
Gastroenterol 108:718–727
Favier CF, Vaughan EE, de Vos WM, Akkermans ADL (2002) Molecular monitoring of
succession of bacterial communities in human neonates. Appl Environ Microbiol 68:219–226
Fleming SE, Choi SY, Fitch MD (1991) Absorption of short-chain fatty acids from the rat cecum
in vivo. J Nutr 121:1787–1797
Flint HJ, Scott KP, Louis P, Duncan SH (2012) The role of the gut microbiota in nutrition and
health. Nat Rev Gastroenterol Hepatol 9:577–589
2 Microbes, Metabolites and Health 39
Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR (2007)
Molecular-phylogenetic characterization of microbial community imbalances in human
inflammatory bowel diseases. Proc Natl Acad Sci USA 104:13780–13785
Fukuda S, Toh H, Hase K, Oshima K, Nakanishi Y, Yoshimura K, Tobe T, Clarke JM,
Topping DL, Suzuki T, Taylor TD, Itoh K, Kikuchi J, Morita H, Hattori M, Ohno H (2011a)
Bifidobacteria can protect from enteropathogenic infection through production of acetate.
Nature 469:543–547
Fukuda S, Toh H, Hase K, Oshima K, Nakanishi Y, Yoshimura K, Tobe T, Clarke JM,
Topping DL, Suzuki T, Taylor TD, Itoh K, Kikuchi J, Morita H, Hattori M, Ohno H (2011b)
Bifidobacteria can protect from enteropathogenic infection through production of acetate.
Nature 469:543–547
Fung KY, Cosgrove L, Clarke J, Lockett T, Morita T, Hase K, Topping DL (2014) Large bowel
short chain fatty acid production, immune status and immune cell function: implications for
chronic inflammatory disease. Nova Science Publishers, New York
Fung KY, Ooi CC, Lewanowitsch T, Tan S, Tan HT, Lim TK, Lin Q, Williams DB, Lockett TJ,
Cosgrove LJ, Chung MC, Head RJ (2012) Identification of potential pathways involved in
induction of apoptosis by butyrate and 4-benzoylbutyrate in HT29 colorectal cancer cells.
J Proteome Res 11:6019–6029
Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, Nakanishi Y, Uetake C,
Kato K, Kato T, Takahashi M, Fukuda NN, Murakami S, Miyauchi E, Hino S, Atarashi K,
Onawa S, Fujimura Y, Lockett T, Clarke JM, Topping DL, Tomita M, Hori S, Ohara O,
Morita T, Koseki H, Kikuchi J, Honda K, Hase K, Ohno H (2013) Commensal
microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature
504:446–450
Galecka M, Szachta P, Bartnicka A, Lykowska-Szuber L, Eder P, Schwiertz A (2013)
Faecalibacterium prausnitzii and Crohn’s disease—is there any connection? Pol J Microbiol
62:91–95
Gallo RL, Hooper LV (2012) Epithelial antimicrobial defence of the skin and intestine. Nat Rev
Immunol 12:503–516
Ganapathy V, Gopal E, Miyauchi S, Prasad PD (2005) Biological functions of SLC5A8, a
candidate tumour suppressor. Biochem Soc Trans 33:237–240
Garrett WS (2015) Cancer and the microbiota. Science 348:80–86
Gevers D, Kugathasan S, Denson LA, Vazquez-Baeza Y, van Treuren W, Ren B, Schwager E,
Knights D, Song SJ, Yassour M, Morgan XC, Kostic AD, Luo C, Gonzalez A, McDonald D,
Haberman Y, Walters T, Baker S, Rosh J, Stephens M, Heyman M, Markowitz J,
Baldassano R, Griffiths A, Sylvester F, Mack D, Kim S, Crandall W, Hyams J,
Huttenhower C, Knight R, Xavier RJ (2014) The treatment-naive microbiome in new-onset
Crohn’s disease. Cell Host Microbe 15:382–392
Gibson GR, Probert HM, Van Loo J, Rastall RA, Roberfroid MB (2004) Dietary modulation of the
human colonic microbiota: updating the concept of prebiotics. Nutr Res Rev 17:259–275
Gibson GR, Roberfroid MB (1995) Dietary modulation of the human colonic microbiota—
introducing the concept of prebiotics. J Nutr 125:1401–1412
Goldman SM, Kamel F, Ross GW, Jewell SA, Marras C, Hoppin JA, Umbach DM,
Bhudhikanok GS, Meng C, Korell M, Comyns K, Hauser RA, Jankovic J, Factor SA,
Bressman S, Lyons KE, Sandler DP, Langston JW, Tanner CM (2014) Peptidoglycan
recognition protein genes and risk of Parkinson’s disease. Mov Disord 29:1171–1180
Grembecka M (2015) Sugar alcohols-their role in the modern world of sweeteners: a review. Eur
Food Res Technol 241:1–14
Groschwitz KR, Hogan SP (2009) Intestinal barrier function: molecular regulation and disease
pathogenesis. J Allergy Clin Immunol 124:3–20 (quiz 21-2)
Group NHW, Peterson J, Garges S, Giovanni M, McInnes P, Wang L, Schloss JA, Bonazzi V,
McEwen JE, Wetterstrand KA, Deal C, Baker CC, di Francesco V, Howcroft TK, KARP RW,
Lunsford RD, Wellington CR, Belachew T, Wright M, Giblin C, David H, Mills M,
Salomon R, Mullins C, Akolkar B, Begg L, Davis C, Grandison L, Humble M, Khalsa J,
40 T.J. Lockett et al.
Little AR, Peavy H, Pontzer C, Portnoy M, Sayre MH, Starke-Reed P, Zakhari S, Read J,
Watson B, Guyer M (2009) The NIH human microbiome project. Genome Res 19:2317–2323
Haenen D, Zhang J, Souza da Silva C, Bosch G, van der Meer IM, van Arkel J, van den Borne JJ,
Perez Gutierrez O, Smidt H, Kemp B, Muller M, Hooiveld GJ (2013) A diet high in resistant
starch modulates microbiota composition, SCFA concentrations, and gene expression in pig
intestine. J Nutr 143:274–283
Halmos EP, Christophersen CT, Bird AR, Shepherd SJ, Gibson PR, Muir JG (2015) Diets that
differ in their FODMAP content alter the colonic luminal microenvironment. Gut 64:93–100
Hamer HM, Jonkers DMAE, Vanhoutvin SALW, Troost FJ, Rijkers G, de Bruine A, Bast A,
Venema K, Brummer RJM (2010) Effect of butyrate enemas on inflammation and antioxidant
status in the colonic mucosa of patients with ulcerative colitis in remission. Clin Nutr 29:
738–744
Harig JM, Soergel KH, Komorowski RA, Wood CM (1989) Treatment of diversion colitis with
short-chain fatty-acid irrigation. N Engl J Med 320:23–28
Haro C, Garcia-Carpintero S, Alcala-Diaz JF, Gomez-Delgado F, Delgado-Lista J, Perez-Martinez
P, Zuniga OAR, Quintana-Navarro GM, Landa BB, Clemente JC, Lopez-Miranda J,
Camargo A, Perez-Jimeanez F (2016) The gut microbial community in metabolic syndrome
patients is modified by diet. J Nutr Biochem 27:27–31
Heller F, Florian P, Bojarski C, Richter J, Christ M, Hillenbrand B, Mankertz J, Gitter AH,
Burgel N, Fromm M, Zeitz M, Fuss I, Strober W, Schulzke JD (2005) Interleukin-13 is the key
effector Th2 cytokine in ulcerative colitis that affects epithelial tight junctions, apoptosis, and
cell restitution. Gastroenterology 129:550–564
Heneker T, Edwards CA, Preston T (1998) Digestion and colonic fermentation of maize porridge
using a 13CO2 breath test. South African Congress of Gastroenterology
Hijazi Z, Molla AM, Al-Habashi H, Muawad WM, Molla AM, Sharma PN (2004) Intestinal
permeability is increased in bronchial asthma. Arch Dis Child 89:227–229
Hoogerwerf WA, Tsao SC, Devuyst O, Levine SA, Yun CH, Yip JW, Cohen ME, Wilson PD,
Lazenby AJ, Tse CM, Donowitz M (1996) NHE2 and NHE3 are human and rabbit intestinal
brush-border proteins. Am J Physiol 270:G29–G41
Hou JK, Abraham B, El-Serag H (2011) Dietary intake and risk of developing inflammatory bowel
disease: a systematic review of the literature. Am J Gastroenterol 106:563–573
Huttenhower C, Gevers D, Knight R, Abubucker S, Badger JH, Chinwalla AT, Creasy HH,
Earl AM, Fitzgerald MG, Fulton RS, Giglio MG, Hallsworth-Pepin K, Lobos EA, Madupu R,
Magrini V, Martin JC, Mitreva M, Muzny DM, Sodergren EJ, Versalovic J, Wollam AM,
Worley KC, Wortman JR, Young SK, Zeng QD, Aagaard KM, Abolude OO, Allen-Vercoe E,
Alm EJ, Alvarado L, Andersen GL, Anderson S, Appelbaum E, Arachchi HM, Armitage G,
Arze CA, Ayvaz T, Baker CC, Begg L, Belachew T, Bhonagiri V, Bihan M, Blaser MJ,
Bloom T, Bonazzi V, Brooks JP, Buck GA, Buhay CJ, Busam DA, Campbell JL, Canon SR,
Cantarel BL, Chain PSG, Chen IMA, Chen L, Chhibba S, Chu K, Ciulla DM, Clemente JC,
Clifton SW, Conlan S, Crabtree J, Cutting MA, Davidovics NJ, Davis CC, Desantis TZ,
Deal C, Delehaunty KD, Dewhirst FE, Deych E, Ding Y, Dooling DJ, Dugan SP, Dunne WM,
Durkin AS, Edgar RC, Erlich RL, Farmer CN, Farrell RM, Faust K, Feldgarden M, Felix VM,
Fisher S, Fodor AA, Forney LJ, Foster L, di Francesco V, Friedman J, Friedrich DC,
Fronick CC, Fulton LL, Gao HY, Garcia N, Giannoukos G, Giblin C, Giovanni MY,
Goldberg JM, Goll J, Gonzalez A, Griggs A et al (2012) Structure, function and diversity of the
healthy human microbiome. Nature 486:207–214
Inan MS, Rasoulpour RJ, Yin L, Hubbard AK, Rosenberg DW, Giardina C (2000) The luminal
short-chain fatty acid butyrate modulates NF-kappaB activity in a human colonic epithelial cell
line. Gastroenterology 118:724–734
Inoue K, Shirai T, Ochiai H, Kasao M, Hayakawa K, Kimura M, Sansawa H (2003)
Blood-pressure-lowering effect of a novel fermented milk containing gamma-aminobutyric
acid (GABA) in mild hypertensives. Eur J Clin Nutr 57:490–495
Iwanaga T, Kishimoto A (2015) Cellular distributions of monocarboxylate transporters: a review.
Biomed Res 36:279–301
2 Microbes, Metabolites and Health 41
James SL, Christophersen CT, Bird AR, Conlon MA, Rosella O, Gibson PR, Muir JG (2015)
Abnormal fibre usage in UC in remission. Gut 64:562–570
Jenkins CLD, Lewis D, Bushell R, Belobrajdic DP, Bird AR (2011) Chain length of cereal
fructans isolated from wheat stem and barley grain modulates in vitro fermentation. J Cereal
Sci 53:188–191
Jimenez E, Marin ML, Martin R, Odriozola JM, Olivares M, Xaus J, Fernandez L, Rodriguez JM
(2008) Is meconium from healthy newborns actually sterile? Res Microbiol 159:187–193
Keenan MJ, Zhou J, Hegsted M, Pelkman C, Durham HA, Coulon DB, Martin RJ (2015) Role of
resistant starch in improving gut health, adiposity, and insulin resistance. Adv Nutr 6:198–205
Khan KJ, Ullman TA, Ford AC, Abreu MT, Abadir A, Marshall JK, Talley NJ, Moayyedi P
(2011) Antibiotic therapy in inflammatory bowel disease: a systematic review and
meta-analysis. Am J Gastroenterol 106:661–673
Kimberg DV, Field M, Johnson J, Henderson A, Gershon E (1971) Stimulation of intestinal
mucosal adenyl cyclase by cholera enterotoxin and prostaglandins. J Clin Invest 50:1218–1230
Kleessen B, Hartmann L, Blaut M (2007a) Oligofructose and long-chain inulin: influence on the
gut microbial ecology of rats associated with a human faecal flora. Br J Nutr 86:291
Kleessen B, Schwarz S, Boehm A, Fuhrmann H, Richter A, Henle T, Krueger M (2007b)
Jerusalem artichoke and chicory inulin in bakery products affect faecal microbiota of healthy
volunteers. Br J Nutr 98:540–549
Kleessen B, Stoof G, Proll J, Schmiedl D, Noack J, Blaut M (1997) Feeding resistant starch affects
fecal and cecal microflora and short-chain fatty acids in rats. J Anim Sci 75:2453–2462
Koenig JE, Spor A, Scalfone N, Fricker AD, Stombaugh J, Knight R, Angenent LT, Ley RE
(2011) Succession of microbial consortia in the developing infant gut microbiome. Proc Natl
Acad Sci USA 108:4578–4585
Komiyama Y, Andoh A, Fujiwara D, Ohmae H, Araki Y, Fujiyama Y, Mitsuyama K, Kanauchi O
(2011) New prebiotics from rice bran ameliorate inflammation in murine colitis models through
the modulation of intestinal homeostasis and the mucosal immune system. Scand J
Gastroenterol 46:40–52
Lanza E, Yu BB, Murphy G, Albert PS, Caan B, Marshall JR, Lance P, Paskett ED, Weissfeld J,
Slattery M, Burt R, Iber F, Shike M, Kikendall JW, Brewer BK, Schatzkin A, Polyp Prevention
Trial Study Group (2007) The polyp prevention trial-continued follow-up study: no effect of a
low-fat, high-fiber, high-fruit, and—vegetable diet on adenoma recurrence eight years after
randomization. Cancer Epidemiol Biomark Prev 16:1745–1752
le Blay G, Michel C, Blottiere HM, Cherbut C (1999) Enhancement of butyrate production in the
rat caecocolonic tract by long-term ingestion of resistant potato starch. Br J Nutr 82:419–426
le Chatelier E, Nielsen T, Qin JJ, Prifti E, Hildebrand F, Falony G, Almeida M, Arumugam M,
Batto JM, Kennedy S, Leonard P, Li JH, Burgdorf K, Grarup N, Jorgensen T, Brandslund I,
Nielsen HB, Juncker AS, Bertalan M, Levenez F, Pons N, Rasmussen S, Sunagawa S, Tap J,
Tims S, Zoetendal EG, Brunak S, Clement K, Dore J, Kleerebezem M, Kristiansen K,
Renault P, Sicheritz-Ponten T, de Vos WM, Zucker JD, Raes J, Hansen T, Bork P, Wang J,
Ehrlich SD, Pedersen O, Meta HITC (2013) Richness of human gut microbiome correlates with
metabolic markers. Nature 500:541
le Leu R, Young G, Hu Y, Winter J, Conlon M (2013) Dietary red meat aggravates dextran sulfate
sodium-induced colitis in mice whereas resistant starch attenuates inflammation. Dig Dis Sci
58:3475–3482
le Leu RK, Winter JM, Christophersen CT, Young GP, Humphreys KJ, Hu Y, Gratz SW,
Miller RB, Topping DL, Bird AR, Conlon MA (2015) Butyrylated starch intake can prevent
red meat-induced O6-methyl-2-deoxyguanosine adducts in human rectal tissue: a randomised
clinical trial. Br J Nutr 114:220–230
le Poul E, Loison C, Struyf S, Springael JY, Lannoy V, Decobecq ME, Brezillon S, Dupriez V,
Vassart G, van Damme J, Parmentier M, Detheux M (2003a) Functional characterization of
human receptors for short chain fatty acids and their role in polymorphonuclear cell activation.
J Biol Chem 278:25481–25489
42 T.J. Lockett et al.
le Poul E, Loison C, Struyf S, Springael JY, Lannoy V, Decobecq ME, Brezillon S, Dupriez V,
Vassart G, van Damme J, Parmentier M, Detheux M (2003b) Functional characterization of
human receptors for short chain fatty acids and their role in polymorphonuclear cell activation.
J Biol Chem 278:25481–25489
Leach JD (2007) Evolutionary perspective on dietary intake of fibre and colorectal cancer. Eur J
Clin Nutr 61:140–142
Lecona E, Olmo N, Turnay J, Santiago-Gomez A, de Silanes IL, Gorospe M, Lizarbe MA (2008)
Kinetic analysis of butyrate transport in human colon adenocarcinoma cells reveals two
different carrier-mediated mechanisms. Biochem J 409:311–320
Lee WJ, Hase K (2014) Gut microbiota-generated metabolites in animal health and disease. Nat
Chem Biol 10:416–424
Leitch EC, Walker AW, Duncan SH, Holtrop G, Flint HJ (2007a) Selective colonization of
insoluble substrates by human faecal bacteria. Environ Microbiol 9:667–679
Leitch ECM, Walker AW, Duncan SH, Holtrop G, Flint HJ (2007b) Selective colonization of
insoluble substrates by human faecal bacteria. Environ Microbiol 9:667–679
Lewis K, Lutgendorff F, Phan V, Soderholm JD, Sherman PM, McKay DM (2010) Enhanced
translocation of bacteria across metabolically stressed epithelia is reduced by butyrate. Inflamm
Bowel Dis 16:1138–1148
Ley RE, Turnbaugh PJ, Klein S, Gordon JI (2006) Microbial ecology—human gut microbes
associated with obesity. Nature 444:1022–1023
Lin MY, de Zoete MR, Van Putten JP, Strijbis K (2015) Redirection of epithelial immune
responses by short-chain fatty acids through inhibition of histone deacetylases. Front Immunol
6:554
Louis P (2012) Does the human gut microbiota contribute to the etiology of autism spectrum
disorders? Dig Dis Sci 57:1987–1989
Luceri C, Femia AP, Fazi M, di Martino C, Zolfanelli F, Dolara P, Tonelli F (2016) Effect of
butyrate enemas on gene expression profiles and endoscopic/histopathological scores of
diverted colorectal mucosa: a randomized trial. Dig Liver Dis 48:27–33
Lukito W, Malik SG, Surono IS, Wahlqvist ML (2015) From ‘lactose intolerance’ to ‘lactose
nutrition’. Asia Pac J Clin Nutr 24:S1–S8
Machiels K, Joossens M, Sabino J, De PV, Arijs I, Eeckhaut V, Ballet V, Claes K, Van IF,
Verbeke K, Ferrante M, Verhaegen J, Rutgeerts P, Vermeire S (2013) A decrease of the
butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines
dysbiosis in patients with ulcerative colitis. Gut 63:1275–1283
Machiels K, Joossens M, Sabino J, de Preter V, Arijs I, Eeckhaut V, Ballet V, Claes K, Van
Immerseel F, Verbeke K, Ferrante M, Verhaegen J, Rutgeerts P, Vermeire S (2014) A decrease
of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines
dysbiosis in patients with ulcerative colitis. Gut 63:1275–1283
Manichanh C, Rigottier-Gois L, Bonnaud E, Gloux K, Pelletier E, Frangeul L, Nalin R, Jarrin C,
Chardon P, Marteau P, Roca J, Dore J (2006) Reduced diversity of faecal microbiota in
Crohn’s disease revealed by a metagenomic approach. Gut 55:205–211
Martin FPJ, Dumas ME, Wang YL, Legido-Quigley C, Yap IKS, Tang HR, Zirah S, Murphy GM,
Cloarec O, Lindon JC, Sprenger N, Fay LB, Kochhar S, van Bladeren P, Holmes E,
Nicholson JK (2007) A top-down systems biology view of microbiome-mammalian metabolic
interactions in a mouse model. Mol Syst Biol 3:112
Martin LJM, Dumon HJW, Champ MMJ (1998) Production of short-chain fatty acids from
resistant starch in a pig model. J Sci Food Agric 77:71–80
Martinez I, Kim J, Duffy PR, Schlegel VL, Walter J (2010) Resistant starches types 2 and 4 have
differential effects on the composition of the fecal microbiota in human subjects. PLoS One 5:
e15046
Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, Schilter HC, Rolph MS, Mackay F,
Artis D, Xavier RJ, Teixeira MM, Mackay CR (2009) Regulation of inflammatory responses
by gut microbiota and chemoattractant receptor GPR43. Nature 461:1282–1286
2 Microbes, Metabolites and Health 43
Ou JH, Delany JP, Zhang M, Sharma S, O’Keefe SJD (2012) Association between low colonic
short-chain fatty acids and high bile acids in high colon cancer risk populations. Nutr Cancer
Int J 64:34–40
Pal S, Murugan C, Clarke JM, Young GP, Ramakrishna BS, Lockett T (2013) Esterified starch as a
treatment for acute gastroenteritis—a double blind randomized controlled trial.
Gastroenterology 144:S904
Park Y, Hunter DJ, Spiegelman D, Bergkvist L, Berrino F, Van den Brandt PA, Buring JE,
Colditz GA, Freudenheim JL, Fuchs CS, Giovannucci E, Goldbohm RA, Graham S,
Harnack L, Hartman AM, Jacobs DR, Kato I, Krogh V, Leitzmann MF, Mccullough ML,
Miller AB, Pietinen P, Rohan TE, Schatzkin A, Willett WC, Wolk A, Zeleniuch-Jacquotte A,
Zhang SMM, Smith-Warner SA (2005) Dietary fiber intake and risk of colorectal cancer—a
pooled analysis of prospective cohort studies. Jama-J Am Med Assoc 294:2849–2857
Parsons BN, Wigley P, Simpson HL, Williams JM, Humphrey S, Salisbury AM, Watson AJ,
Fry SC, O’Brien D, Roberts CL, O’Kennedy N, Keita AV, Soderholm JD, Rhodes JM,
Campbell BJ (2014) Dietary supplementation with soluble plantain non-starch polysaccharides
inhibits intestinal invasion of Salmonella Typhimurium in the chicken. PLoS One 9:e87658
Peeters M, Geypens B, Claus D, Nevens H, Ghoos Y, Verbeke G, Baert F, Vermeire S,
Vlietinck R, Rutgeerts P (1997) Clustering of increased small intestinal permeability in families
with Crohn’s disease. Gastroenterology 113:802–807
Perrier C, Corthesy B (2011) Gut permeability and food allergies. Clin Exp Allergy 41:20–28
Piche T (2014) Tight junctions and IBS–the link between epithelial permeability, low-grade
inflammation, and symptom generation? Neurogastroenterol Motil 26:296–302
Ploger S, Stumpff F, Penner GB, Schulzke JD, Gabel G, Martens H, Shen Z, Gunzel D,
Aschenbach JR (2012) Microbial butyrate and its role for barrier function in the gastrointestinal
tract. Ann NY Acad Sci 1258:52–59
Png CW, Linden SK, Gilshenan KS, Zoetendal EG, McSweeney CS, Sly LI, McGuckin MA,
Florin THJ (2010) Mucolytic bacteria with increased prevalence in IBD mucosa augment
in vitro utilization of mucin by other bacteria. Am J Gastroenterol 105:2420–2428
Puddu A, Sanguineti R, Montecucco F, Viviani GL (2014) Evidence for the gut microbiota
short-chain fatty acids as key pathophysiological molecules improving diabetes. Mediators
Inflamm 2014:162021
Pullan RD, Thomas GA, Rhodes M, Newcombe RG, Williams GT, Allen A, Rhodes J (1994)
Thickness of adherent mucus gel on colonic mucosa in humans and its relevance to colitis. Gut
35:353–359
Qin JJ, Li RQ, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F,
Yamada T, Mende DR, Li JH, Xu JM, Li SC, Li DF, Cao JJ, Wang B, Liang HQ, Zheng HS,
Xie YL, Tap J, Lepage P, Bertalan M, Batto JM, Hansen T, le Paslier D, Linneberg A,
Nielsen HB, Pelletier E, Renault P, Sicheritz-Ponten T, Turner K, Zhu HM, Yu C, Li ST,
Jian M, Zhou Y, Li YR, Zhang XQ, Li SG, Qin N, Yang HM, Wang J, Brunak S, Dore J,
Guarner F, Kristiansen K, Pedersen O, Parkhill J, Weissenbach J, Bork P, Ehrlich SD, Wang J,
Meta HITC (2010) A human gut microbial gene catalogue established by metagenomic
sequencing. Nature 464:59–65
Qin JJ, Li YR, Cai ZM, Li SH, Zhu JF, Zhang F, Liang SS, Zhang WW, Guan YL, Shen DQ,
Peng YQ, Zhang DY, Jie ZY, Wu WX, Qin YW, Xue WB, Li JH, Han LC, Lu DH, Wu PX,
Dai YL, Sun XJ, Li ZS, Tang AF, Zhong SL, Li XP, Chen WN, Xu R, Wang MB, Feng Q,
Gong MH, Yu J, Zhang YY, Zhang M, Hansen T, Sanchez G, Raes J, Falony G, Okuda S,
Almeida M, Lechatelier E, Renault P, Pons N, Batto JM, Zhang ZX, Chen H, Yang RF,
Zheng WM, Li SG, Yang HM, Wang J, Ehrlich SD, Nielsen R, Pedersen O, Kristiansen K,
Wang J (2012) A metagenome-wide association study of gut microbiota in type 2 diabetes.
Nature 490:55–60
Raghupathy P, Ramakrishna BS, Oommen SP, Ahmed TS, Priyaa T, Dziura J, Young GP,
Binder HJ (2006) Amylase-resistant starch as adjunct to oral rehydration therapy in children
with diarrhea. J Pediatr Gastroenterol Nutr 42:362–368
2 Microbes, Metabolites and Health 45
Rajendran VM, Binder HJ (1994) Apical membrane Cl-butyrate exchange: mechanism of short
chain fatty acid stimulation of active chloride absorption in rat distal colon. J Membr Biol
141:51–58
Rajilic-Stojanovic M, Shanahan F, Guarner F, de Vos WM (2013) Phylogenetic analysis of
dysbiosis in ulcerative colitis during remission. Inflamm Bowel Dis 19:481–488
Ramakrishna BS, Nance SH, Roberts-Thomson IC, Roediger WE (1990) The effects of
enterotoxins and short-chain fatty acids on water and electrolyte fluxes in ileal and colonic
loops in vivo in the rat. Digestion 45:93–101
Ramakrishna BS, Venkataraman S, Srinivasan P, Dash P, Young GP, Binder HJ (2000)
Amylase-resistant starch plus oral rehydration solution for cholera. N Engl J Med 342:308–313
Ramirez-Farias C, Slezak K, Fuller Z, Duncan A, Holtrop G, Louis P (2009) Effect of inulin on the
human gut microbiota: stimulation of Bifidobacterium adolescentis and Faecalibacterium
prausnitzii. Br J Nutr 101:541–550
Ridaura VK, Faith JJ, Rey FE, Cheng JY, Duncan AE, Kau AL, Griffin NW, Lombard V,
Henrissat B, Bain JR, Muehlbauer MJ, Ilkayeva O, Semenkovich CF, Funai K, Hayashi DK,
Lyle BJ, Martini MC, Ursell LK, Clemente JC, Van Treuren W, Walters WA, Knight R,
Newgard CB, Heath AC, Gordon JI (2013) Gut microbiota from twins discordant for obesity
modulate metabolism in mice. Science 341:1079
Roberfroid MB (2005) Introducing inulin-type fructans. Br J Nutr 93:S13–S25
Roberfroid MB (2007) Inulin-type fructans: functional food ingredients. J Nutr 137:2493S–2502S
Roberts CL, Keita AV, Duncan SH, O’Kennedy N, Soderholm JD, Rhodes JM, Campbell BJ
(2010) Translocation of Crohn’s disease Escherichia coli across M-cells: contrasting effects of
soluble plant fibres and emulsifiers. Gut 59:1331–1339
Roberts CL, Keita AV, Parsons BN, Prorok-Hamon M, Knight P, Winstanley C, O’Kennedy N,
Soderholm JD, Rhodes JM, Campbell BJ (2013) Soluble plantain fibre blocks adhesion and
M-cell translocation of intestinal pathogens. J Nutr Biochem 24:97–103
Said HM (2011) Intestinal absorption of water-soluble vitamins in health and disease. Biochem J
437:357–372
Sakaguchi S, Yamaguchi T, Nomura T, Ono M (2008) Regulatory T cells and immune tolerance.
Cell 133:775–787
Savage DC (1977) Microbial ecology of gastrointestinal-tract. Annu Rev Microbiol 31:107–133
Schamberger GP, Diez-Gonzalez F (2002) Selection of recently isolated colicinogenic Escherichia
coli strains inhibitory to Escherichia coli O157: H7. J Food Prot 65:1381–1387
Scheperjans F, Aho V, Pereira PAB, Koskinen K, Paulin L, Pekkonen E, Haapaniemi E,
Kaakkola S, Eerola-Rautio J, Pohja M, Kinnunen E, Murros K, Auvinen P (2015) Gut
microbiota are related to Parkinson’s disease and clinical phenotype. Mov Disord 30:350–358
Scheppach W (1996) Treatment of distal ulcerative colitis with short-chain fatty acid enemas. a
placebo-controlled trial. German-Austrian SCFA Study Group. Dig Dis Sci 41:2254–2259
Schieber AM, Lee YM, Chang MW, Leblanc M, Collins B, Downes M, Evans RM, Ayres JS
(2015) Disease tolerance mediated by microbiome E. coli involves inflammasome and IGF-1
signaling. Science 350:558–563
Schneeman BO (1986) Dietary fiber—physical and chemical-properties, methods of analysis, and
physiological-effects. Food Technol 40:104–110
Segal I, Edwards CA, Walker ARP (2000) Continuing low colon cancer incidence in African
populations. Am J Gastroenterol 95:859–860
Sellon RK, Tonkonogy S, Schultz M, Dieleman LA, Grenther W, Balish E, Rennick DM, Sartor RB
(1998) Resident enteric bacteria are necessary for development of spontaneous colitis and
immune system activation in interleukin-10-deficient mice. Infect Immun 66:5224–5231
Singh N, Gurav A, Sivaprakasam S, Brady E, Padia R, Shi H, Thangaraju M, Prasad PD,
Manicassamy S, Munn DH, Lee JR, Offermanns S, Ganapathy V (2014) Activation of
Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic
inflammation and carcinogenesis. Immunity 40:128–139
Slavin JL (2001) Dietary fiber and colon cancer. Marcel Dekker Inc, New York
46 T.J. Lockett et al.
Smith K, McCoy KD, Macpherson AJ (2007) Use of axenic animals in studying the adaptation of
mammals to their commensal intestinal microbiota. Semin Immunol 19:59–69
Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly Y, Glickman JN,
Garrett WS (2013) The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell
homeostasis. Science 341:569–573
Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermudez-Humaran LG, Gratadoux JJ, Blugeon S,
Bridonneau C, Furet JP, Corthier G, Grangette C, Vasquez N, Pochart P, Trugnan G,
Thomas G, Blottiere HM, Dore J, Marteau P, Seksik P, Langella P (2008) Faecalibacterium
prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis
of Crohn disease patients. Proc Natl Acad Sci USA 105:16731–16736
Soliman ML, Rosenberger TA (2011) Acetate supplementation increases brain histone acetylation
and inhibits histone deacetylase activity and expression. Mol Cell Biochem 352:173–180
Steinhart AH, Hiruki T, Brzezinski A, Baker JP (1996) Treatment of left-sided ulcerative colitis
with butyrate enemas: a controlled trial. Aliment Pharmacol Ther 10:729–736
Stephen AM (1994) Whole grains—impact of consuming whole grains on physiological-effects of
dietary fiber and starch. Crit Rev Food Sci Nutr 34:499–511
Subramanya S, Ramakrishna BS, Binder HJ, Farthing MJ, Young GP (2006) Evaluation of oral
rehydration solution by whole-gut perfusion in rats: effect of osmolarity, sodium concentration
and resistant starch. J Pediatr Gastroenterol Nutr 43:568–575
Subramanya SB, Rajendran VM, Srinivasan P, Kumar NSN, Ramakrishna BS, Binder HJ (2007)
Differential regulation of cholera toxin-inhibited Na-H exchange isoforms by butyrate in rat
ileum. Am J Physiol Gastrointest. Liver Physiol 293:G857–G863
Suzuki T, Yoshida S, Hara H (2008) Physiological concentrations of short-chain fatty acids
immediately suppress colonic epithelial permeability. Br J Nutr 100:297–305
Swidsinski A, Loening-Boecke V, Herber A (2009) Mucosal flora in Crohn’s disease and
ulcerative colitis—an overview. J Physiol Pharmacol 60(suppl 6):61–71
Takahashi K, Nishida A, Fujimoto T, Fujii M, Shioya M, Imaeda H, Inatomi O, Bamba S,
Andoh A, Sugimoto M (2016) Reduced abundance of butyrate-producing bacteria species in
the fecal microbial community in Crohn’s disease. Digestion 93:59–65
Tan J, Mckenzie C, Potamitis M, Thorburn AN, Mackay CR, Macia L (2014) The role of
short-chain fatty acids in health and disease. Adv Immunol 121:91–119
Tao R, de Zoeten EF, Ozkaynak E, Chen C, Wang L, Porrett PM, Li B, Turka LA, Olson EN,
Greene MI, Wells AD, Hancock WW (2007) Deacetylase inhibition promotes the generation
and function of regulatory T cells. Nat Med 13:1299–1307
Tennant DR (2014) Potential intakes of total polyols based on UK usage survey data. Food Addit
Contam Part A-Chem Anal Control Eposure Risk Assess 31:574–586
Thangaraju M, Cresci GA, Liu K, Ananth S, Gnanaprakasam JP, Browning DD, Mellinger JD,
Smith SB, Digby GJ, Lambert NA, Prasad PD, Ganapathy V (2009) GPR109A is a
G-protein-coupled receptor for the bacterial fermentation product butyrate and functions as a
tumor suppressor in colon. Cancer Res 69:2826–2832
Thibault R, Blachier F, Darcy-Vrillon B, De CP, Bourreille A, Segain JP (2010) Butyrate
utilization by the colonic mucosa in inflammatory bowel diseases: a transport deficiency.
Inflamm Bowel Dis 16:684–695
Thibault R, de Coppet P, Daly K, Bourreille A, Cuff M, Bonnet C, Mosnier JF, Galmiche JP,
Shirazi-Beechey S, Segain JP (2007) Down-regulation of the monocarboxylate transporter 1 is
involved in butyrate deficiency during intestinal inflammation. Gastroenterology 133:1916–1927
Thorburn AN, Mckenzie CI, Shen S, Stanley D, Macia L, Mason LJ, Roberts LK, Wong CH,
Shim R, Robert R, Chevalier N, Tan JK, Marino E, Moore RJ, Wong L, McConville MJ,
Tull DL, Wood LG, Murphy VE, Mattes J, Gibson PG, Mackay CR (2015) Evidence that
asthma is a developmental origin disease influenced by maternal diet and bacterial metabolites.
Nat Commun 6:7320
Thornton JR, Dryden A, Kelleher J, Losowsky MS (1987) Super-efficient starch absorption—a
risk factor for colonic neoplasia. Dig Dis Sci 32:1088–1091
2 Microbes, Metabolites and Health 47
Toden S, Bird AR, Topping DL, Conlon MA (2007) High red meat diets induce greater numbers
of colonic DNA double-strand breaks than white meat in rats: attenuation by high-amylose
maize starch. Carcinogenesis 28:2355–2362
Toden S, Lockett TJ, Topping DL, Scherer BL, Watson EJ, Southwood JG, Clarke JM (2014)
Butyrylated starch affects colorectal cancer markers beneficially and dose-dependently in
genotoxin-treated rats. Cancer Biol Ther 15:1515–1523
Tolhurst G, Heffron H, Lam YS, Parker HE, Habib AM, Diakogiannaki E, Cameron J, Grosse J,
Reimann F, Gribble FM (2012) Short-chain fatty acids stimulate glucagon-like peptide-1
secretion via the G-protein-coupled receptor FFAR2. Diabetes 61:364–371
Topping D (2007) Cereal complex carbohydrates and their contribution to human health. J Cereal
Sci 46:220–229
Topping DL, Bird AR (1999) Foods, nutrients and digestive health. Aust J Nutr Diet 56:S22–S34
Topping DL, Clifton PM (2001) Short-chain fatty acids and human colonic function: roles of
resistant starch and nonstarch polysaccharides. Physiol Rev 81:1031–1064
Topping DL, Illman RJ (1986) Bacterial fermentation in the human large-bowel—time to change
from the roughage model of dietary fiber. Med J Austral 144:307–309
Tosh SM, Brummer Y, Wolever TMS, Wood PJ (2008) Glycemic response to oat bran muffins
treated to vary molecular weight of beta-glucan. Cereal Chem 85:211–217
Trowell HC, Burkitt DP (1987) The development of the concept of dietary fiber. Mol Aspects Med
9:7–15
Tungland BC, Meyer D (2002) Nondigestible oligo—and polysaccharides (Dietary fiber): their
physiology and role in human health and food. Compr Rev Food Sci Food Saf 1:73–92
Tuohy KM, Conterno L, Gasperotti M, Viola R (2012) Up-regulating the human intestinal
microbiome using whole plant foods, polyphenols, and/or fiber. J Agric Food Chem 60:8776–
8782
Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI (2006) An
obesity-associated gut microbiome with increased capacity for energy harvest. Nature
444:1027–1031
Vadivel G, Prasad PD, Thangaraju M, Martin PM, Singh N (2014) Butyrate-mediated protection
against colonic inflammation and colon carcinogenesis: role of butyrate transporters and
butyrate receptors. Food sources, functions and health benefits, Butyrate
Van den Abbeele P, Gerard P, Rabot S, Bruneau A, El Aidy S, Derrien M, Kleerebezem M,
Zoetendal EG, Smidt H, Verstraete W, Van de Wiele T, Possemiers S (2011) Arabinoxylans
and inulin differentially modulate the mucosal and luminal gut microbiota and
mucin-degradation in humanized rats. Environ Microbiol 13:2667–2680
Van der Merwe B, Erasmus C, Taylor JRN (2001) African maize porridge: a food with slow
in vitro starch digestibility. Food Chem 72:347–353
Vieira ELM, Leonel AJ, Sad AP, Beltrao NRM, Costa TF, Ferreira TMR, Gomes-Santos AC,
Faria AMC, Peluzio MCG, Cara DC, Alvarez-Leite JI (2012) Oral administration of sodium
butyrate attenuates inflammation and mucosal lesion in experimental acute ulcerative colitis.
J Nutr Biochem 23:430–436
Vinolo MA, Rodrigues HG, Hatanaka E, Hebeda CB, Farsky SH, Curi R (2009) Short-chain fatty
acids stimulate the migration of neutrophils to inflammatory sites. Clin Sci (Lond) 117:331–
338
Vinolo MA, Rodrigues HG, Hatanaka E, Sato FT, Sampaio SC, Curi R (2011a) Suppressive effect
of short-chain fatty acids on production of proinflammatory mediators by neutrophils. J Nutr
Biochem 22:849–855
Vinolo MA, Rodrigues HG, Nachbar RT, Curi R (2011b) Regulation of inflammation by short
chain fatty acids. Nutrients 3:858–876
Vivinus-Nebot M, Frin-Mathy G, Bzioueche H, Dainese R, Bernard G, Anty R, Filippi J,
Saint-Paul MC, Tulic MK, Verhasselt V, Hebuterne X, Piche T (2014) Functional bowel
symptoms in quiescent inflammatory bowel diseases: role of epithelial barrier disruption and
low-grade inflammation. Gut 63:744–752
Wahlqvist ML (2015) Lactose nutrition in lactase nonpersisters. Asia Pac J Clin Nutr 24:S21–S25
48 T.J. Lockett et al.
Walker ARP, Walker BF, Walker AJ (1986) Fecal pH, dietary fiber intake, and proneness to colon
cancer in 4 South-African populations. Br J Cancer 53:489–495
Walker AW, Ince J, Duncan SH, Webster LM, Holtrop G, Ze X, Brown D, Stares MD, Scott P,
Bergerat A, Louis P, Mcintosh F, Johnstone AM, Lobley GE, Parkhill J, Flint HJ (2011)
Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J
5:220–230
Wang HB, Wang PY, Wang X, Wan YL, Liu YC (2012) Butyrate enhances intestinal epithelial
barrier function via up-regulation of tight junction protein Claudin-1 transcription. Dig Dis Sci
57:3126–3135
Wang L, Christophersen CT, Sorich MJ, Gerber JP, Angley MT, Conlon MA (2011) Low relative
abundances of the mucolytic bacterium Akkermansia muciniphila and Bifidobacterium spp. in
feces of children with autism. Appl Environ Microbiol 77:6718–6721
West NP, Christophersen CT, Pyne DB, Cripps AW, Conlon MA, Topping DL, Kang S,
McSweeney CS, Fricker PA, Aguirre D, Clarke JM (2013) Butyrylated starch increases colonic
butyrate concentration but has limited effects on immunity in healthy physically active
individuals. Exerc Immunol Rev 19:102–119
Windey K, de Preter V, Verbeke K (2012) Relevance of protein fermentation to gut health. Mol
Nutr Food Res 56:184–196
Windey K, Houben E, Deroover L, Verbeke K (2015) Contribution of colonic fermentation and
fecal water toxicity to the pathophysiology of lactose-intolerance. Nutrients 7:7505–7522
Winter J, Nyskohus L, Young GP, Hu Y, Conlon MA, Bird AR, Topping DL, Le Leu RK (2011)
Inhibition by resistant starch of red meat-induced promutagenic adducts in mouse colon.
Cancer Prev Res 4:1920–1928
Wong JMW, de Souza R, Kendall CWC, Emam A, Jenkins DJA (2006) Colonic health:
fermentation and short chain fatty acids. J Clin Gastroenterol 40:235–243
Xu H, McCoy A, Li J, Zhao Y, Ghishan FK (2015) Sodium butyrate stimulates NHE8 expression
via its role on activating NHE8 basal promoter activity. Am Physiol-Gastrointest Liver Physiol
309:G500–G505
Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M,
Hidalgo G, Baldassano RN, Anokhin AP, Heath AC, Warner B, Reeder J, Kuczynski J,
Caporaso JG, Lozupone CA, Lauber C, Clemente JC, Knights D, Knight R, Gordon JI (2012)
Human gut microbiome viewed across age and geography. Nature 486:222
Yetukuri L, Ekroos K, Vidal-Puig A, Oresic M (2008) Informatics and computational strategies for
the study of lipids. Mol Biosyst 4:121–127
Yu YN, Fang JY (2015) Gut microbiota and colorectal cancer. Gastrointest Tumors 2:26–32
Ze X, Duncan SH, Louis P, Flint HJ (2012) Ruminococcus bromii is a keystone species for the
degradation of resistant starch in the human colon. ISME J 6:1535–1543
Zeissig S, Burgel N, Gunzel D, Richter J, Mankertz J, Wahnschaffe U, Kroesen AJ, Zeitz M,
Fromm M, Schulzke JD (2007) Changes in expression and distribution of claudin 2, 5 and 8
lead to discontinuous tight junctions and barrier dysfunction in active Crohn’s disease. Gut
56:61–72
Chapter 3
Exploring the Bioactive Landscape
of the Gut Microbiota to Identify
Metabolites Underpinning Human Health
1 Introduction
North America, Europe and Australasia have amongst the highest incidences of
chronic gastrointestinal and metabolic diseases including inflammatory bowel dis-
eases (IBD), colorectal cancer (CRC) and obesity (Molodecky et al. 2012; Stevens
et al. 2012; Bray et al. 2013). Although once considered rare in large parts of the
world the incidences of IBD and obesity in particular have also been steadily
increasing in Asia, South America and the Middle East (Ng et al. 2014; Kaplan
2015). These diseases are associated with considerable socioeconomic costs; for
example, the estimated costs to the global economy from obesity approaches US$2
trillion per annum, which equates to 2.8 % of global gross domestic product (Dobbs
et al. 2014). Thus, there is an urgent need to develop more effective preventative
and therapeutic strategies to ameliorate the impacts of these diseases.
Genomic studies have revealed that IBD, CRC and obesity are underpinned by
specific host genetic susceptibilities that are considered to be necessary but often
not sufficient for disease to develop (Jostins et al. 2012; Peters et al. 2015; Locke
et al. 2015), and it is now recognised that environmental factors and lifestyle
choices also affect disease risk. Epidemiological studies also suggest that host
genetic, environmental factors and lifestyle choices either alone or in combination
does not fully explain disease risk implying that other risk factors remain to be
identified. With that context, the human gastrointestinal tract harbours a diverse
microbial community (gut microbiota) that provides a range of ecological and
metabolic functions relevant to host health and well-being (reviewed by Backhed
et al. 2005). Human- and animal-based studies have now also identified the gut
microbiota as an important risk factor in the aetiology of chronic gut diseases. First,
human studies have revealed that the microbiota varies between healthy and dis-
eased individuals (e.g. Turnbaugh et al. 2009; Qin et al. 2010; Nakatsu et al. 2015)
and these variations are associated with changes in the disease state (Cotillard et al.
2013; Nakatsu et al. 2015; De Cruz et al. 2015). Second, germ-free animals are
protected from disease but become susceptible following microbiota transfer
(Turnbaugh et al. 2006; Zackular et al. 2013; Schaubeck et al. 2016). Third, both
human- and animal-based studies have revealed these diseases are responsive to
interventions that modulate the activity of the gut microbiota including antibiotics
(Zackular et al. 2013; Murphy et al. 2013; Schaubeck et al. 2016), diet (Donohoe
et al. 2014; Quince et al. 2015), probiotics (Kadooka et al. 2010; Bassaganya-Riera
et al. 2012) and faecal microbiota transfers (Suskind et al. 2015).
Even though IBD, CRC and obesity are a heterogeneous group of diseases, they
are all characterised by an activated inflammatory response. Nuclear factor-kappa
B (NF-κB) is a master regulator of gut epithelial integrity and inflammation, and
activation of the NF-κB signalling pathway plays a key role in driving the
inflammatory response during the onset and progression of these diseases.
Consistent with this, the NF-κB signalling pathway is a validated therapeutic target
for the treatment of IBD (Atreya et al. 2008) (Fig. 1), and it is also a recognised
therapeutic target for CRC (Sakamoto and Maeda 2010), and for obesity and its
co-morbidities (Donath 2014; Esser et al. 2015). The NF-κB pathway is particularly
well recognised as a therapeutic target for IBD, however, many of the current
therapeutics are only partially effective and/or have significant side effects. For
instance, glucocorticosteroids can affect linear growth and bone health in paediatric
subjects; methotrexate can cause hepatotoxicity, and as a teratogen, the treatment of
female subjects is complicated; salicylates are associated with an increased risk of
bleeding. Similarly, the newer biologics (e.g. anti-TNFα factors) are expensive,
increase the risk of infection and suffer from a loss of response. Interestingly, the
gut microbiota plays a central role in modulating the host immune response and
specific gut microbes have been shown to possess potent NF-κB suppressive
capabilities that can ameliorate the inflammatory response (Ménard et al. 2004;
Sokol et al. 2008; Heuvelin et al. 2009; Petrof et al. 2009; Eeckhaut et al. 2012;
Khokhlova et al. 2012; Kaci et al. 2013). This suggests that exploiting gut
microbe-derived NF-κB suppressive bioactives may provide new opportunities to
maintain host health. In this Chapter, we examine our current understanding of the
host-microbiota interaction and outline strategies to identify and characterise the
NF-κB suppressive capabilities of the gut microbiota. In particular, we propose that
an integrated approach combining culture-dependent and independent approaches
with a more mechanistic dissection of the microbiota provided by improved cul-
tivation techniques, high-throughput functional screens and metabolomic and
genetic dissections is necessary to transform our understanding of gut health and
support the development of new preventative and therapeutic strategies.
3 Exploring the Bioactive Landscape of the Gut Microbiota … 51
Fig. 1 The NF-κB pathway as a validated drug target for the treatment of chronic gut diseases.
The NF-κB pathway can be activated by several mechanisms including microbe-associated
molecular patterns (MAMP; e.g. via lipopolysaccharide, flagellin from the gut microbiota),
damage associated molecular patterns (DAMP; e.g. via extracellular detection of normally
intracellular proteins) or cytokines. Targeting of the NF-κB pathway for the treatment of chronic
gut diseases is best recognised for IBD with glucocorticosteroids (corticosteroids), methotrexate,
salicylates (e.g. mesalazine, sulfasalazine) and anti-TNFα biologics interfering with pathway
signalling. However, this pathway is also increasingly targeted for CRC, and obesity and its
co-morbidities
The publication of the human genome sequence was a seminal milestone in our
history. Published with much excitement in 2001, it promised new insights and
understanding of what it means to be human (Venter et al. 2001; Lander et al.
2001). Initial estimates of the number of protein-coding genes deemed necessary to
explain the biological and phenotypic complexity characteristic of humans varied
widely, however, there was considerable surprise when it was revealed that the
human genome is comprised of as few as 25,000 genes (International Human
Genome Sequencing Consortium 2004). Humans are not autonomous and fol-
lowing a period of introspection it was increasingly recognised that our associated
microbiota provides a range of functions relevant to health and disease. Thus, in its
aftermath, there was an increasing call to sequence our second genome—that of the
human microbiota (Davies 2001; Relman and Falkow 2001). This international
effort to sequence the human microbiome has principally been led by the Human
Microbiome Project (HMP) funded by the US National Institutes of Health
(Peterson et al. 2009) and the MetaHIT Project (Ehrlich 2010) funded by the
European Union, with additional coordination of other global efforts mediated
through the International Human Microbiome Consortium.
Humans and their associated microbiota co-exist as a symbiotic multispecies
assemblage termed a “holobiont” that is defined as a physical association between a
52 P. Ó Cuív et al.
host and its associated microbiota for signifıcant portions of their life history
(Bordenstein and Theis 2015). The emergence of the holobiont concept has dra-
matically altered our perception of human health—where the role of microbes was
traditionally viewed from the perspective of infectious diseases—to one where the
microbiota is viewed as an integral component that contributes essential function-
alities relevant to the fitness of the holobiont. The assembly of a holobiont is a
dynamic process that impacts both the host and microbiota (Gilbert 2014). For
instance, the human gut provides a wide variety of ecological niches that are
characterised by a constant temperature, oxygen tension, humidity and nutrient
supply. This supports colonisation by a numerically abundant and diverse micro-
biota that in return helps prevent colonisation by potential pathogens, detoxifies
harmful compounds, produces essential nutrients and catalyses the biotransforma-
tion of dietary substrate so they can be utilised by the host (Fig. 2). The “holo-
genome” then is comprised of the genetic potential encoded by the host’s genome
and their associated microbiota (microbiome) and can thus be considered as an
extension of the host genotype itself. Notably, the hologenome is dynamic in terms
of its composition with the potential to change more rapidly than the host genome
alone via gene acquisition or loss which also confers a greater adaptive potential to
the holobiont (Quercia et al. 2014).
The holobiont concept provides a new paradigm for a more holistic under-
standing of the aetiology of chronic gut diseases. For instance, some of the genetic
Fig. 2 The emergent holobiont model of human health. The holobiont assembly impacts the
fitness of the host and its associated microbiota and is characterised by specific host–microbiota
interdependencies. The hologenome is comprised of the genetic capacity of both the human
genome and microbiome. The holobiont provides a new model to examine the impact of
environmental factors and lifestyle choices on host health and disease risk
3 Exploring the Bioactive Landscape of the Gut Microbiota … 53
susceptibility loci for these diseases also affect the ability of specific microbes to
colonise the gut (reviewed by Spor et al. 2011) suggesting that the contribution of
genetic susceptibility and microbiota composition to disease risk may be intrinsi-
cally linked. In addition, the holobiont has provided a framework on which the
impact of environmental factors and lifestyle choices on health and disease risk can
be dissected and this has informed the development of new strategies to rationally
modulate the holobiont phenotype to improve host health (Zeevi et al. 2015). The
holobiont may also represent an optimum biological system to bioprospect for
novel NF-κB suppressive bioactives as the gut microbiota has co-evolved with the
development of the host mucosal immune system. In particular, we hypothesise that
select microbes produce bioactives that actively suppress the NF-κB-mediated
immune response perhaps as an essential capability to allow for successful
colonisation and persistence. These NF-κB suppressive bioactives may have
specific attributes that are relevant to the development of new therapeutics including
high bioactivity, bioavailability and target site specificity, as well as stability in the
gut environment. Thus, these bioactives could potentially be used directly or serve
as lead molecules for the development of novel NF-κB suppressive therapeutics.
Alternatively, determining the mechanism by which they exert their suppressive
effects could help to identify new cellular targets that could be drugged by existing
or new therapeutics. Taken together the identification and characterisation of these
bioactives may help realise new opportunities to prevent or treat chronic gut
diseases.
Our understanding and appreciation of the diversity and functional capacity of the
gut microbiota are largely based on distinct yet complementary culture-dependent
and culture-independent analyses of the gut environment (Fig. 3). Many of the
current reference strains used in gut microbiota research were first isolated in the
mid-twentieth century following the advent of techniques in anaerobic microbiol-
ogy (for a historical perspective see Rajilić-Stojanović and de Vos 2014). However,
it has long been recognised that the vast majority of gut microbes are resistant to
cultivation as revealed by the discordance between microscopic counts of microbial
cells and those recovered using traditional laboratory based cultivation. This phe-
nomenon was first described in aquatic environments and termed “the great plate
count anomaly” (reviewed by Staley and Konopka 1985). Instead, advances in
molecular biology and DNA sequencing technology culminated in the establish-
ment of culture-independent approaches to study the microbiota, based largely on
16S rRNA gene community profiling and metagenomics. A crucial discovery was
that the microbial 16S rRNA gene could be used as a molecular clock to infer
phylogeny and provide an estimate of microbial diversity (Woese and Fox 1977).
54 P. Ó Cuív et al.
The 16S rRNA gene is approximately 1550 bp in length and has a divergence rate
of 1–2 % per 50 million years (Ochman et al. 1999). The gene is comprised of
conserved and (hyper)variable regions and this architecture has been exploited in
culture-independent studies to assess microbial diversity. Here, near full-length or
subsections of the 16S rRNA gene are amplified by polymerase chain reaction
(PCR) using primers targeting the conserved regions and the intervening variable
regions are used to infer phylogeny (Klindworth et al. 2013). The length of the 16S
rRNA gene sequence can affect phylogenetic assignment (Kim et al. 2011; Franzén
et al. 2015) and it does not provide any information on the functional potential of
the taxa identified. However, the development of metagenomic approaches, facil-
itating the sequencing of bulk DNA recovered from microbial communities has
now provided new opportunities to both assess microbial diversity through
sequencing of defined phylogenetic marker genes (Sunagawa et al. 2013) and the
functional capacity of the microbiome (Qin et al. 2010; Li et al. 2014a). In a
landmark study Qin et al. (2010) examined the microbiome of 124 subjects and
determined that it is comprised of a genetic pool of up to 3.3 million non-redundant
genes that is as much as 150× that of the human genome. In practical terms the
3 Exploring the Bioactive Landscape of the Gut Microbiota … 55
ease of propagation and its amenability to genetic dissection. In contrast, the vast
majority of gut microbes are fastidious anaerobes that are not known to be
amenable to genetic dissection and hence their genetic potential remains cryptic.
This has led to suggestions that an increased effort must be expended to functionally
characterise existing gene sets as this will provide new insights into the microbial
factors supporting gut health or driving disease (Roberts 2004; Galperin and
Koonin 2010; Anton et al. 2014; Joice et al. 2014).
Based on these collective observations, we contend that new advances in
microbial isolation coupled with parallel developments in functional characterisa-
tion and dissection approaches will provide the best opportunities to develop
streamlined strategies to identify NF-κB suppressive and other types of bioactives
produced by gut microbes. In particular, considering the complexity of the gut
microbiota these strategies must be cost-effective, scalable and amenable to
automation, and the following sections provide an overview of each of these
aspects.
which further reduce the risk of inadvertent oxygen contamination and allow many
standard techniques (e.g. spread plates, streak plates) to be used to isolate target
microbes.
The distinct ecological niches present along the human gut can be challenging to
replicate in a laboratory environment particularly as the nutritional requirements of
many target microbes are unknown. The use of “habitat simulating” media has been
widely used to circumvent this challenge and typically includes sterile aqueous
extracts of faecal or rumen digesta, in addition to sources of amino acids, carbo-
hydrates and other nutrients (Eller et al. 1971; Barcenilla et al. 2000; McSweeney
et al. 2005; Lagier et al. 2012), leading to the isolation of phylogenetically diverse
gut microbes including bacteria that have specific host dependencies such as
Akkermansia muciniphila (Derrien et al. 2004) and the obligate symbiont seg-
mented filamentous bacterium (Schnupf et al. 2015). Although habitat simulating
media often support the growth of subdominant populations, their enrichment and
isolation is often complicated because of rapid overgrowth by fast growing,
numerically abundant microbes. More selective media have been developed for the
isolation of specific gut taxa including Bacteroides spp. (Livingston et al. 1978),
Bifidobacterium spp. (Ferraris et al. 2010) and Enterococcus spp. (Isenberg et al.
1970) by identifying specific nutritional dependencies, and promoters/inhibitors of
growth (e.g. antibiotics, bile salts, sodium azide). Alternatively, subdominant
populations can be enriched by selecting for a specific phenotype (e.g. spore for-
mation) and this has enabled taxonomically novel microbes to be directly recovered
on nutrient-rich habitat simulating media (Atarashi et al. 2013; Browne et al. 2016).
3 Exploring the Bioactive Landscape of the Gut Microbiota … 59
Accordingly, based on these, our own (Ó Cuív et al. 2011b, 2015) and other
(Rettedal et al. 2014; Ma et al. 2014) observations, many “uncultured” microbes
grow reproducibly well in vitro when isolated as axenic cultures. Thus, many more
novel gut microbes could be recovered if the practical considerations involved with
screening large numbers of microbial isolates under strict anaerobic conditions
could be overcome.
To improve the throughput of microbial isolation, Stevenson et al. (2004)
developed an approach called “Plate wash PCR” to recover axenic isolates of
previously uncultured bacteria from agricultural soil and the guts of wood-feeding
termites. Briefly, an inoculum is plated in duplicate on solid medium and following
growth the colonies are re-suspended en masse from one of the replicate plates and
the sample extracted DNA is screened using specific PCR primers. By this
approach, a broad range of growth parameters can be rapidly screened to determine
conditions supporting the growth of target taxa. Once identified, colonies from the
matching replica plate are grown in multiwell plates and screened with specific
primers to identify the target isolate. Plate wash PCR was successfully used to
isolate a Lachnospiraceae affiliated bacterium that inhibits colonisation of the
murine gut by Clostridium difficile VPI 10463 (Reeves et al. 2012), and it has been
adapted to support the isolation of human gut bacteria affiliated with the HMP’s
most-wanted taxa using a microfluidic platform (Ma et al. 2014). Goodman et al.
(2011) described a similar approach but determined the diversity of microbial
isolates recovered on the replica culture plate by 16S rRNA-based microbial pro-
filing. In addition, to further improve the throughput of the isolation process, a most
probable number (MPN) approach was used to create, in 384 well plates, person-
alised archived culture collections of axenic isolates directly from faecal samples
without picking individual colonies. The MPN approach is based on extinction
culturing, whereby diluting microbial cells so that ≤1 culturable cell is used as an
inoculum supports the production of axenic cultures (Button et al. 1993). This
favours the isolation of the most abundant rather than the fastest growing or most
culturable microbes and the MPN method has also been used to produce axenic
cultures of previously uncultured rumen bacteria (Kenters et al. 2011). Rettedal
et al. (2014) also used 16S rRNA profiling to profile gut bacteria recovered on a
broad range of solid growth media. Then, by a process termed cultivation-based
multiplex phenotyping, they combined growth on solid medium with antibiotic
selection and 16S rRNA profiling to selectively target and recover target bacteria
including members of the HMP’s most-wanted taxa (Fodor et al. 2012). Recently,
Browne et al. (2016) applied a similar approach to isolate spore forming bacteria
from the human gut.
Separately, Raoult and colleagues (Lagier et al. 2012) coined the term “cultur-
omics” and demonstrated that increasing the throughput of microbial isolation
greatly extended the number of cultured isolates from the human gut. By this
approach, 32,500 colonies representing 340 bacterial species and including 31
previously unidentified species were obtained using 212 culture conditions and
60 P. Ó Cuív et al.
In 2014 it was reported that over 1000 cultured gut microbial species had been
described in the scientific literature and this number continues to increase rapidly
due to the new advances in microbial cultivation techniques (Rajilić-Stojanović and
de Vos 2014). The NF-κB suppressive activities of the vast majority of existing
isolates have not been assessed but taken together with the increasing rate of
microbial isolation there is a need for improved functional screening strategies to
effectively identify these strains. Strategies to identify immunomodulatory microbes
should address three key criteria. First, the assays should be biologically relevant,
sensitive and specific, facilitating the identification of virulent or cytotoxic microbes
at an early point in the screening process. Second, the assays should allow the
extent of immunomodulatory activity to be quantified and the host pathways
affected to be readily identified and dissected to determine the target of the
bioactive. Third, the assays should be cost-effective and robust, easy to perform and
amenable to scaling to an automated high-throughput format. Historically, the use
of well-established cell lines to identify NF-κB suppressive microbial isolates
broadly fulfils these criteria.
The NF-κB pathway has been extensively characterised and transcription can be
activated via two alternate pathways, called the canonical and non-canonical path-
ways. These pathways can be activated either independently (e.g. TNFα/IL-1β
activates the canonical pathway, B-cell activating factor activates the non-canonical
pathway) or in tandem (e.g. CD40L/Lipopolysaccharide activate both pathways).
Many gut bacteria are considered to be pathobionts—symbionts that are capable of
acting as pathogens under certain environmental conditions—and are capable of
stimulating an immune response. Consequently, the ability of gut bacteria to supress
NF-κB activation is often initially assessed using peripheral blood mononuclear cells
(PBMC) as several studies have reported that peripheral blood cells predict the
in vivo immunomodulatory potential of different bacteria (Foligne et al. 2007; Sokol
et al. 2008). Alternatively, NF-κB suppressive capability can be assessed using
peripheral blood derived cell lines (e.g. human monocyte-like THP-1 cell line,
murine RAW macrophage cell line) stimulated with a specific NF-κB pathway
agonist. These cell lines have rapid and reproducible growth characteristics and they
express a broad range of Toll-like receptors (TLR) [e.g. THP-1 cells expresses all
TLRs including the surface TLRs (i.e. TLR1/2, TLR2, TLR4, TLR5 and TLR6/2)].
These characteristics can be used to identify microbes that modulate NF-κB activity,
or that express virulence or cytotoxic factors, in a high-throughput manner.
Despite the usefulness of immune cell lines the ability of gut microbes to sup-
press NF-κB activity is typically assessed using intestinal epithelial cell culture
lines. Numerous epithelial cell lines are widely used by researchers, however, the
HT-29, Caco-2 and T84 cell lines and their derivatives are amongst the most widely
used to assess immunomodulatory activity. Gut epithelial cells are constantly
3 Exploring the Bioactive Landscape of the Gut Microbiota … 63
facing a single internal lumen compartment or, alternatively, they can be grown as
monolayers in a transwell system. These cell cultures can also be stably maintained
through repeated rounds of propagation and freezing thus recapitulating the main
elements of cancer cell culture lines and providing a superior in vitro model to
assess NF-κB suppressive capabilities. For instance, the impact of NF-κB sup-
pressive bioactives on individual epithelial cell subtypes could be assessed by
fluorescence-activated cell sorting using antibodies targeting the NF-κB complex
and lineage specific markers. Enteroids/colonoids can be generated from animals
carrying reporter genes or, alternatively, Schwank et al. (2013) reported that the
CRISPR/Cas9 system could be used to edit organoid genome sequences. Along
with new developments in CRISPR/Cas9-based large fragment deletions and
insertions (Wang et al. 2015; Zhang et al. 2015a), this may provide new oppor-
tunities to produce patient-specific reporter cell lines. Together, these developments
offer new opportunities to identify and dissect disease-specific pathways as well as
assess their responsiveness to different therapeutics.
The healthy gut microbiota produces a diverse array of factors including proteins
(Rieu et al. 2014), peptides (Kaci et al. 2011; Quevrain et al. 2016),
polysaccharide-peptidoglycans (Matsumoto et al. 2009) and secondary metabolites
(Bansal et al. 2010; Gonzalez-Sarrias et al. 2010; Lim et al. 2015; Lee et al. 2015)
that are capable of suppressing NF-κB, revealing this capability is characterised by
a high degree of functional redundancy. Metabolomic approaches have played a
central role in the identification of these factors although they have been challenged
by the sheer diversity of metabolites produced by gut microbes. In addition, many
of these metabolites are produced at low concentrations and include novel
metabolites that are not represented in existing databases, further hindering iden-
tification efforts. Nonetheless, effective bioassay guided fractionation strategies that
typically involve successive fractionation coupled with functional assays to track
the fraction(s) retaining suppressive activity can be devised to identify NF-κB
suppressive bioactive factors (Fig. 4).
Microbes in the healthy gut environment are physically separated from epithelial
cells by a mucus layer and bioactive factors must be capable of traversing this
barrier to reach their cellular target. Many NF-κB bioactives are secreted into the
extracellular milieu and the first stage of the screening process involves the
preparation of a cell-free supernatant fraction of spent medium that can be assessed
for suppressive activity. The supernatant fraction of most fastidious gut microbes is
likely to contain SCFA which are produced by anaerobes as an end product of
fermentation, and are amongst the most abundant metabolites produced. Acetate,
propionate and butyrate are produced at the highest concentrations with other SCFA
3 Exploring the Bioactive Landscape of the Gut Microbiota … 65
Fig. 4 Bioassay guided fractionation strategy to enrich and purify NF-κB suppressive bioactives.
The bioactive fractions are successively fractionated and the NF-κB suppressive activity in the
fractions is assessed after each treatment. Fractions enriched in primary and secondary metabolites
(1° and 2° respectively), and peptides are typically produced by size fractionation.
Fractions >3 kDa are typically not considered to be suitable for drug development but may help
to identify new drugable targets. Metabolites and peptide bioactives can be further fractionated
using biochemical treatments (e.g. protease, denaturant, thermal treatment). Peptide bioactives can
be further fractionated and used to identify new drugable targets or as lead molecules for drug
development. The 1° and 2° metabolites can be further fractionated (e.g. solid phase extractions,
HPLC based fractionation) to identify the bioactive factor. Secondary metabolites can also be used
to identify new drugable targets or as lead molecules for drug development
basis of size and a simple 3 kDa molecular weight cut-off filter allows peptides up
to 27 amino acids long (assuming an average amino acid size of 110 Da) to be
easily separated from larger molecules. Bioactive secondary metabolites and pep-
tides can subsequently be distinguished using routine (e.g. protease, denaturing,
thermal) treatments and the sample can subsequently be further fractionated (e.g.
solid phase extractions, HPLC based fractionation) to further reduce the complexity
of the samples. This approach has been used effectively to identify
NF-κB suppressive peptides produced by Faecalibacterium prausnitzii (Quevrain
et al. 2016). However, the identification of secondary metabolites can be more
challenging and it can be necessary to fractionate large sample volumes to identify
the metabolites of interest, although this process can be expedited if isogenic mutant
or non-suppressive strains can be processed in parallel (Donia et al. 2014). Once
sufficiently enriched and concentrated the bioactives can be identified using spe-
cialist metabolomic methodologies and equipment.
Much of our understanding of the functional capacity of the microbial world has
been provided by the genetic dissection of clinically and agriculturally relevant
bacteria that are only distantly related to the microbes that typically inhabit the
human gut. The ability to conclusively link genes and function is a central challenge
in elucidating the functional potential of the microbiota. However, with limited
exceptions (Rey et al. 2013; Ichimura et al. 2013), few molecular tools have been
described for the characterisation of gut microbes. The vast majority of the currently
available microbial isolates are not known to be amenable to genetic transformation
although (meta)genomics has revealed evidence of extensive lateral gene transfer
within the gut microbiome. To address this challenge, we recently developed an
innovative approach termed metaparental mating that expedites the directed isola-
tion of genetically tractable gut bacteria from mixed microbial communities (Ó
Cuív et al. 2015). The metaparental mating approach is based on the
well-established biparental mating approach (Simon et al. 1983; Simon et al. 1986)
and uses RP4 (RK2)-mediated bacterial conjugation and a broad host range
mobilisable shuttle vector. Metaparental mating has several advantages over
alternative natural (i.e. transduction, transformation) or contrived (e.g. electropo-
ration, sonoporation) genetic transformation approaches. First, RP4-based conju-
gation is very promiscuous and has been shown to mediate the transfer of DNA to a
diverse range of bacteria (Whitehead and Hespell 1990; Picardeau 2008; Tolonen
et al. 2009; Dominguez and O’Sullivan 2013) and also to archaea (Dodsworth et al.
2010), fungi (Nishikawa et al. 1990) and animal cells (Waters 2001). Second, the
metaparental mating can be performed under anaerobic conditions and stably
transformed recipients can be recovered by selection of a vector encoded marker. In
3 Exploring the Bioactive Landscape of the Gut Microbiota … 67
addition, as the antibiotic resistance phenotype of the recipients may not be known,
the laboratory E. coli ST18 donor strain can be efficiently counter selected without
antibiotics by nutritional auxotrophy (i.e. the omission of δ-aminolevulinic acid
from the selection medium). Third, the RP4-based conjugation can be readily
scaled and automated (Clarke et al. 2005) to increase the throughput of the meta-
parental mating mediated isolation process.
We used the metaparental mating approach to specifically target bacteria affili-
ated with the Firmicutes as these comprise the majority of the human gut microbial
core although they are underrepresented in microbial culture collections.
Furthermore, few of these bacteria, and in particular those affiliated with the
Clostridia, have been genetically characterised although many strains are capable of
modulating the host immune response (Sokol et al. 2008; Ivanov et al. 2009;
Atarashi et al. 2011, 2013; Li et al. 2014b; Quevrain et al. 2016). In support of this
effort we developed a series of modular vectors termed pEHR5 that can be con-
jugated from an E. coli host to a pool of potential recipients. As the efficiency of
conjugation can be affected by the size of the vectors, their modular architecture
helps minimise their overall size. In addition, it allows individual modules to be
easily exchanged ensuring that the base vectors are flexible and can be readily
re-purposed. Similar modular vectors have been used in a broad range of non-
E. coli hosts to support protein expression and the construction of fluorescently
labelled bacterial strains (Herrero et al. 1990; Charpentier et al. 2004; Fodor et al.
2004; Heap et al. 2009; Dammeyer et al. 2013; Wright et al. 2015). By this
approach, we recovered a broad suite of axenic fastidious gut bacteria affiliated with
the Firmicutes that were stably transformed with pEHR5-based vectors. In addition,
we demonstrated that the metaparental mating approach and the pEHR vectors can
be used for heterologous protein expression by constructing fluorescently labelled
gut bacteria (Ó Cuív et al. 2015).
The pEHR5 vector system is freely available to the research community without
the need for a restrictive material transfer agreement and it offers a basis for the
development of a uniform and streamlined set of molecular tools for the isolation
and functional genetic characterisation of fastidious microbes. Nonetheless, the
metaparental mating approach can plausibly be applied with any RP4 mobilisable
vector bearing an appropriate resistance marker(s) and origin(s) of replication, thus
allowing genetically tractable bacteria to be recovered from complex microbial
communities. We confirmed this hypothesis by using the narrow host range vector
pJQ200sk(+) (Quandt and Hynes 1993) to demonstrate that E. coli transconjugants
bearing pJQ200sk(+) could be selectively recovered from an anaerobic enrichment
from human faeces by metaparental mating (Fig. 5). In addition, we used the vector
pGusAmob [(Girbal et al. 2003), pGusA modified to carry an oriT] to target the
recovery of Firmicutes affiliated bacteria and isolated transconjugants affiliated with
Blautia hathewayi, Streptococcus pleomorphus and Anaerococcus vaginalis on
M10-based medium. We have now also demonstrated that the pEHR vectors can be
cured using standard molecular techniques to yield naïve strains (Pottenger and Ó
Cuív, Unpublished data).
68 P. Ó Cuív et al.
The metaparental mating approach and the pEHR vector series are significant
developments for the genetic dissection of the gut microbiota by forward and
reverse genetic approaches. For instance, it is now known that many gut bacteria
carry putative biosynthetic gene clusters for secondary metabolites (Letzel et al.
2013; Donia et al. 2014; Cimermancic et al. 2014; Donia and Fischbach 2015;
Hadjithomas et al. 2015), some of which may encode for NF-κB suppressive
bioactives. The specific gene clusters underpinning the production of NF-κB sup-
pressive bioactives could potentially be identified by comparative genomics of
suppressive and non-suppressive strains, however, reverse genetic approaches can
now also plausibly be applied to specifically disrupt target genes and conclusively
confirm their role in the production of specific bioactives (Donia et al. 2014).
Consistent with this, mutagenesis strategies based on homologous recombination
(Al-Hinai et al. 2012; Heap et al. 2012; Faulds-Pain and Wren 2013) and the Ll.
ltrB group II intron (Chen et al. 2005; Heap et al. 2007; Tolonen et al. 2009) have
been described for a diverse range bacteria affiliated with the Firmicutes. While the
specific factors underpinning NF-κB suppressive activity have been identified in
some cases (e.g. Rieu et al. 2014; Quevrain et al. 2016) in most instances, they
3 Exploring the Bioactive Landscape of the Gut Microbiota … 69
The butyrate producing gut bacterium, F. prausnitzii, comprises part of the core
microbiota in healthy adult humans and is ubiquitously found in the gut of mam-
mals and insects (Foglesong et al. 1984; Bjerrum et al. 2006; Castillo et al. 2007;
Qin et al. 2010; Nava and Stappenbeck 2011; Miquel et al. 2013; Oikonomou et al.
2013). This suggests that F. prausnitzii plays a critical role in host metabolism and
physiology and consequently it is widely considered to be a model gut bacterium
with relevance to health and disease. In that context, much progress has been made
in identifying the true metabolic potential of F. prasunitzii and its contribution to
health and well being (Sokol et al. 2008; Quevrain et al. 2016). Swidsinski et al.
(2008) first reported a reduced population of F. prausnitzii in CD subjects and
Sokol et al. (2008) subsequently demonstrated a low abundance of F. prausnitzii in
ileal biopsies from CD subjects at the time of surgery was associated with recur-
rence six months postoperatively, and that the abundance at six months was
70 P. Ó Cuív et al.
metabolites. Using a gnotobiotic mouse model, Miquel et al. (2015) revealed that
the protective effect of F. prausnitzii following colonisation was associated with the
presence of salicylic acid and shikimic acid in gut and serum metabolomic profiles.
Salicylic acid is a precursor of 5-aminosalycilic acid and is capable of suppressing
IL-8 secretion from TNFα stimulated HT-29 cells. In contrast, shikimic acid is not
capable of suppressing IL-8 secretion from TNFα stimulated HT-29 cells, however,
this molecule is a precursor of anti-inflammatory aromatic compounds including
salicylic acid and 3,4-oxo-eisopropylideneshikimic acid (Xing et al. 2013).
These observations underline the role played by F. prausnitzii in the mainte-
nance of gut homeostasis and reveal the evolution of a variety of strategies to affect
specific aspects of gut function and the immune response. Consistent with this, F.
prausnitzii has been suggested as a candidate next generation probiotic for the
treatment of gut inflammatory diseases (Sokol et al. 2008; Neef and Sanz 2013).
Critically, the characterisation of F. prausnitzii has provided a template by which
the contribution of other microbes to gut health can be examined. It should be noted
that although F. prausnitzii A2-165 has been known to suppress NF-κB since 2008
and its genome was sequenced in 2009, the specific bioactives supporting this
activity remained unidentified until 2015, highlighting the limited capacity of -omic
approaches to identify novel functional capabilities. We have now used meta-
parental mating to isolate genetically tractable strains of F. prausnitzii (Ó Cuív et al.
2015), and we anticipate that these will further expedite the functional dissection of
this important gut bacterium.
The healthy gut microbiota plays a vital role in helping to maintain gut homeostasis
and preventing the onset of chronic gut disease. Surprisingly, little is known about
the essential functionalities that underlie this capability and how they might be
exploited to develop more effective therapeutic interventions. The rapid advances in
DNA sequencing technologies continue to provide an unprecedented insight into
structure-functional activity of the gut microbiome. In contrast, the development of
complementary approaches including microbial culturing, functional assays,
metabolomics and genetic technologies have not kept pace with these develop-
ments. This has hindered efforts to realise the functional potential of the microbiota,
however, the successful metabolomic dissection of F. prausnitzii will encourage
and inform the development of improved methodologies for other gut bacteria.
Similarly, new advances in microbial culturing and genetic techniques will provide
new opportunities to support a more mechanistic dissection of these functionalities.
We anticipate that the effective integration of these disparate yet complementary
approaches will afford the best opportunity to effectively bioprospect the gut
microbiota and support the discovery of novel bioactives, and the development of
new therapeutics.
72 P. Ó Cuív et al.
References
Bryant MP (1972) Commentary on the Hungate technique for culture of anaerobic bacteria. Am J
Clin Nutr 25:1324–1328
Button DK, Schut F, Quang P et al (1993) Viability and isolation of marine bacteria by dilution
culture: theory, procedures, and initial results. Appl Env Microbiol 59:881–891
Canani RB, Costanzo MD, Leone L et al (2011) Potential beneficial effects of butyrate in intestinal
and extraintestinal diseases. World J Gastroenterol 17:1519–1528
Cario E, Rosenberg IM, Brandwein SL et al (2000) Lipopolysaccharide activates distinct signaling
pathways in intestinal epithelial cell lines expressing toll-like receptors. J Immunol 164:966–
972
Carlsson AH, Yakymenko O, Olivier I et al (2013) Faecalibacterium prausnitzii supernatant
improves intestinal barrier function in mice DSS colitis. Scand J Gastroenterol 48:1136–1144
Castillo M, Skene G, Roca M et al (2007) Application of 16S rRNA gene-targetted fluorescence
in situ hybridization and restriction fragment length polymorphism to study porcine microbiota
along the gastrointestinal tract in response to different sources of dietary fibre. FEMS Microbiol
Ecol 59:138–146
Charpentier E, Anton AI, Barry P et al (2004) Novel cassette-based shuttle vector system for
gram-positive bacteria. Appl Environ Microbiol 70:6076–6085
Chen Y, Mcclane BA, Fisher DJ et al (2005) Construction of an alpha toxin gene knockout mutant
of Clostridium perfringens type A by use of a mobile group II intron. Appl Environ Microbiol
71:7542–7547
Cimermancic P, Medema MH, Claesen J et al (2014) Insights into secondary metabolism from a
global analysis of prokaryotic biosynthetic gene clusters. Cell 158:412–421
Clarke L, Carbon J (1976) A colony bank containing synthetic Col El hybrid plasmids
representative of the entire E. coli genome. Cell 9:91–99
Clarke P, Ó Cuív P, O’Connell M (2005) Novel mobilizable prokaryotic two-hybrid system
vectors for high-throughput protein interaction mapping in Escherichia coli by bacterial
conjugation. Nucleic Acids Res 33:e18
Cohen LJ, Kang HS, Chu J et al (2015) Functional metagenomic discovery of bacterial effectors in
the human microbiome and isolation of commendamide, a GPCR G2A/132 agonist. Proc Natl
Acad Sci USA 112:E4825–E4834
Cotillard A, Kennedy SP, Kong LC et al (2013) Dietary intervention impact on gut microbial gene
richness. Nature 500:585–588
Dammeyer T, Timmis KN, Tinnefeld P (2013) Broad host range vectors for expression of proteins
with (Twin-) Strep-tag, His-tag and engineered, export optimized yellow fluorescent protein.
Microb Cell Fact 12:49
Davies J (2001) In a map for human life, count the microbes, too. Science 291:2316
De Cruz P, Kang S, Wagner J et al (2015) Association between specific mucosa-associated
microbiota in Crohn’s disease at the time of resection and subsequent disease recurrence: a
pilot study. J Gastroenterol Hepatol 30:268–278
De Palma G, Nadal I, Medina M et al (2010) Intestinal dysbiosis and reduced
immunoglobulin-coated bacteria associated with coeliac disease in children. BMC Microbiol
10:63
Derrien M, Vaughan EE, Plugge CM et al (2004) Akkermansia muciniphila gen. nov., sp. nov., a
human intestinal mucin-degrading bacterium. Int J Syst Evol Microbiol 54:1469–1476
Dey N, Soergel DA, Repo S et al (2013) Association of gut microbiota with post-operative clinical
course in Crohn’s disease. BMC Gastroenterol 13:131
Dione N, Khelaifia S, La Scola B et al (2015) A quasi-universal medium to break the
aerobic/anaerobic bacterial culture dichotomy in clinical microbiology. Clin Microbiol Infect
Dobbs R, Sawers C, Thompson F et al (2014) Overcoming obesity: an initial economic analysis.
McKinsey Global Institute, Europe
Dodsworth JA, Li L, Wei S et al (2010) Interdomain conjugal transfer of DNA from bacteria to
archaea. Appl Environ Microbiol 76:5644–5647
Dominguez W, O’sullivan DJ (2013) Developing an efficient and reproducible conjugation-based
gene transfer system for bifidobacteria. Microbiology 159:328–338
74 P. Ó Cuív et al.
Donaldson GP, Lee SM, Mazmanian SK (2015) Gut biogeography of the bacterial microbiota. Nat
Rev Micro 14:20–32 (advance online publication)
Donath MY (2014) Targeting inflammation in the treatment of type 2 diabetes: time to start. Nat
Rev Drug Discov 13:465–476
Donia MS, Cimermancic P, Schulze CJ et al (2014) A systematic analysis of biosynthetic gene
clusters in the human microbiome reveals a common family of antibiotics. Cell 158:1402–1414
Donia MS, Fischbach MA (2015) Small molecules from the human microbiota. Science
349:1254766
Donohoe DR, Holley D, Collins LB et al (2014) A gnotobiotic mouse model demonstrates that
dietary fiber protects against colorectal tumorigenesis in a microbiota-and butyrate-dependent
manner. Cancer Discov 4:1387–1397
Dowell VR, Hawkins TM, Control CFD (1981) Laboratory methods in anaerobic bacteriology. U.
S. Department of Health and Human Services, Public Health Service, Centers for Disease
Control, USA
Dubourg G, Lagier JC, Armougom F et al (2013) The gut microbiota of a patient with resistant
tuberculosis is more comprehensively studied by culturomics than by metagenomics. Eur J
Clin Microbiol Infect Dis 32:637–645
Eckburg PB, Bik EM, Bernstein CN et al (2005) Diversity of the human intestinal microbial flora.
Science 308:1635–1638
Eeckhaut V, Machiels K, Perrier C et al (2012) Butyricicoccus pullicaecorum in inflammatory
bowel disease. Gut 62:1745–1752
Ehrlich SD (2010) Metagenomics of the intestinal microbiota: potential applications. Gastroenterol
Clin Biol 34(Supplement 1):S23–S28
Eller C, Crabill MR, Bryant MP (1971) Anaerobic roll tube media for nonselective enumeration
and isolation of bacteria in human feces. Appl Microbiol 22:522–529
Ellrott K, Jaroszewski L, Li W et al (2010) Expansion of the protein repertoire in newly explored
environments: human gut microbiome specific protein amilies. PLoS Comput Biol 6:e1000798
Esser N, Paquot N, Scheen AJ (2015) Anti-inflammatory agents to treat or prevent type 2 diabetes,
metabolic syndrome and cardiovascular disease. Expert Opin Investig Drugs 24:283–307
Faulds-Pain A, Wren BW (2013) Improved bacterial mutagenesis by high-frequency allele
exchange, demonstrated in Clostridium difficile and Streptococcus suis. Appl Environ
Microbiol 79:4768–4771
Ferraris L, Aires J, Waligora-Dupriet AJ et al (2010) New selective medium for selection of
bifidobacteria from human feces. Anaerobe 16:469–471
Fish JA, Chai B, Wang Q et al (2013) FunGene: the functional gene pipeline and repository. Front
Microbiol 4:291
Fodor AA, Desantis TZ, Wylie KM et al (2012) The “most wanted” taxa from the human
microbiome for whole genome sequencing. PLoS ONE 7:e41294
Fodor BD, Kovacs AT, Csaki R et al (2004) Modular broad-host-range expression vectors for
single-protein and protein complex purification. Appl Environ Microbiol 70:712–721
Foglesong MA, Cruden DL, Markovetz AJ (1984) Pleomorphism of fusobacteria isolated from the
cockroach hindgut. J Bacteriol 158:474–480
Foligne B, Nutten S, Grangette C et al (2007) Correlation between in vitro and in vivo
immunomodulatory properties of lactic acid bacteria. World J Gastroenterol 13:236–243
Fosgerau K, Hoffmann T (2015) Peptide therapeutics: current status and future directions. Drug
Discov Today 20:122–128
Franzén O, Hu J, Bao X et al (2015) Improved OTU-picking using long-read 16S rRNA gene
amplicon sequencing and generic hierarchical clustering. Microbiome 3:1–14
Galperin MY, Koonin EV (2010) From complete genome sequence to “complete” understanding?
Trends Biotechnol 28:398–406
Gilbert SF (2014) A holobiont birth narrative: the epigenetic transmission of the human
microbiome. Front Genet 5:282
Gill SR, Pop M, Deboy RT et al (2006) Metagenomic analysis of the human distal gut
microbiome. Science 312:1355–1359
3 Exploring the Bioactive Landscape of the Gut Microbiota … 75
Li K, Bihan M, Methe BA (2013) Analyses of the stability and core taxonomic memberships of the
human microbiome. PLoS ONE 8:e63139
Li J, Jia H, Cai X et al (2014a) An integrated catalog of reference genes in the human gut
microbiome. Nat Biotechnol 32:834–841
Li YN, Huang F, Cheng HJ et al (2014b) Intestine-derived Clostridium leptum induces murine
tolerogenic dendritic cells and regulatory T cells in vitro. Hum Immunol 75:1232–1238
Lim SM, Jeong JJ, Kang GD et al (2015) Timosaponin AIII and its metabolite sarsasapogenin
ameliorate colitis in mice by inhibiting NF-kappaB and MAPK activation and restoring
Th17/Treg cell balance. Int Immunopharmacol 25:493–503
Liu H, Bouillaut L, Sonenshein AL et al (2013) Use of a mariner-based transposon mutagenesis
system to isolate Clostridium perfringens mutants deficient in gliding motility. J Bacteriol
195:629–636
Liu X, Peng D, Luo Y et al (2009) Construction of an Escherichia coli to Bacillus thuringiensis
shuttle vector for large DNA fragments. Appl Microbiol Biotechnol 82:765–772
Livingston SJ, Kominos SD, Yee RB (1978) New medium for selection and presumptive
identification of the Bacteroides fragilis group. J Clin Microbiol 7:448–453
Locke AE, Kahali B, Berndt SI et al (2015) Genetic studies of body mass index yield new insights
for obesity biology. Nature 518:197–206
Loman NJ, Constantinidou C, Chan JZM et al (2012) High-throughput bacterial genome
sequencing: an embarrassment of choice, a world of opportunity. Nat Rev Micro 10:599–606
Lozupone CA, Stombaugh JI, Gordon JI et al (2012) Diversity, stability and resilience of the
human gut microbiota. Nature 489:220–230
Ma L, Kim J, Hatzenpichler R et al (2014) Gene-targeted microfluidic cultivation validated by
isolation of a gut bacterium listed in human microbiome project’s most wanted taxa. Proc Natl
Acad Sci USA 111:9768–9773
Macy JM, Snellen JE, Hungate RE (1972) Use of syringe methods for anaerobiosis. Am J Clin
Nutrition 25:1318–1323
Mahe MM, Sundaram N, Watson CL et al (2015) Establishment of human epithelial enteroids and
colonoids from whole tissue and biopsy. J Vis Exp. doi:10.3791/52483
Martin R, Chain F, Miquel S et al (2014) The commensal bacterium Faecalibacterium prausnitzii
is protective in DNBS-induced chronic moderate and severe colitis models. Inflamm Bowel Dis
20:417–430
Martin R, Miquel S, Chain F et al (2015) Faecalibacterium prausnitzii prevents physiological
damages in a chronic low-grade inflammation murine model. BMC Microbiol 1:67
Martínez I, Muller CE, Walter J (2013) Long-term temporal analysis of the human fecal
microbiota revealed a stable core of dominant bacterial species. PLoS ONE 8:e69621
Mastropietro G, Tiscornia I, Perelmuter K et al (2015) HT-29 and Caco-2 reporter cell lines for
functional studies of nuclear factor kappa b activation. Mediators Inflamm 2015:13
Matsumoto S, Hara T, Nagaoka M et al (2009) A component of polysaccharide peptidoglycan
complex on Lactobacillus induced an improvement of murine model of inflammatory bowel
disease and colitis-associated cancer. Immunology 128:e170–e180
McGovern DPB, Gardet A, Torkvist L et al (2010) Genome-wide association identifies multiple
ulcerative colitis susceptibility loci. Nat Genet 42:332–337
McSweeney CS, Denman SE, Mackie RI (2005) Rumen bacteria. In: Makkar HPS,
McSweeney CS (eds) Methods in gut microbial ecology for ruminants. Springer, Dordrecht
Melmed G, Thomas LS, Lee N et al (2003) Human intestinal epithelial cells are broadly
unresponsive to Toll-like receptor 2-dependent bacterial ligands: implications for
host-microbial interactions in the gut. J Immunol 170:1406–1415
Ménard S, Candalh C, Bambou JC et al (2004) Lactic acid bacteria secrete metabolites retaining
anti-inflammatory properties after intestinal transport. Gut 53:821–828
Meng J, Kanzaki G, Meas D et al (2012) A genome-wide inducible phenotypic screen identifies
antisense RNA constructs silencing Escherichia coli essential genes. FEMS Microbiol Lett
329:45–53
78 P. Ó Cuív et al.
Relman DA, Falkow S (2001) The meaning and impact of the human genome sequence for
microbiology. Trends Microbiol 9:206–208
Remely M, Aumueller E, Jahn D et al (2014) Microbiota and epigenetic regulation of
inflammatory mediators in type 2 diabetes and obesity. Benef Microbes 5:33–43
Rettedal EA, Gumpert H, Sommer MOA (2014) Cultivation-based multiplex phenotyping of
human gut microbiota allows targeted recovery of previously uncultured bacteria. Nat
Commun 5
Rey FE, Gonzalez MD, Cheng J et al (2013) Metabolic niche of a prominent sulfate-reducing
human gut bacterium. Proc Natl Acad Sci USA. 110:13582–13587
Rieu-Lesme F, Delbes C, Sollelis L (2005) Recovery of partial 16S rDNA sequences suggests the
presence of Crenarchaeota in the human digestive ecosystem. Curr Microbiol 51:317–321
Rieu A, Aoudia N, Jego G et al (2014) The biofilm mode of life boosts the anti-inflammatory
properties of Lactobacillus. Cell Microbiol 16:1836–1853
Roberts RJ (2004) Identifying protein function—a call for community action. PLoS Biol 2:E42
Rose C, Parker A, Jefferson B et al (2015) The characterization of feces and urine: a review of the
literature to inform advanced treatment technology. Crit Rev Environ Sci Technol 45:1827–1879
Rossi O, Van Berkel LA, Chain F et al (2016) Faecalibacterium prausnitzii A2-165 has a high
capacity to induce IL-10 in human and murine dendritic cells and modulates T cell responses.
Sci Rep 6:18507
Sabat G, Rose P, Hickey WJ et al (2000) Selective and sensitive method for PCR amplification of
Escherichia coli 16S rRNA genes in soil. Appl Environ Microbiol 66:844–849
Sakamoto K, Maeda S (2010) Targeting NF-kappaB for colorectal cancer. Expert Opin Ther
Targets 14:593–601
Sandrin TR, Goldstein JE, Schumaker S (2013) MALDI TOF MS profiling of bacteria at the strain
level: a review. Mass Spectrom Rev 32:188–217
Santos Rocha C, Lakhdari O, Blottiere HM et al (2012) Anti-inflammatory properties of dairy
lactobacilli. Inflamm Bowel Dis 18:657–666
Sato T, Clevers H (2013) Growing self-organizing mini-guts from a single intestinal stem cell:
mechanism and applications. Science 340:1190–1194
Savage DC (1977) Microbial ecology of the gastrointestinal tract. Annu Rev Microbiol 31:107–
133
Scanlan PD, Shanahan F, Marchesi JR (2008) Human methanogen diversity and incidence in
healthy and diseased colonic groups using mcrA gene analysis. BMC Microbiol 8:79
Schaubeck M, Clavel T, Calasan J et al (2016) Dysbiotic gut microbiota causes transmissible
Crohn’s disease-like ileitis independent of failure in antimicrobial defence. Gut 65:225–237
Schnoes AM, Brown SD, Dodevski I et al (2009) Annotation error in public databases:
misannotation of molecular function in enzyme superfamilies. PLoS Comput Biol 5:e1000605
Schnupf P, Gaboriau-Routhiau V, Gros M et al (2015) Growth and host interaction of mouse
segmented filamentous bacteria in vitro. Nature 520:99–103
Schwank G, Koo BK, Sasselli V et al (2013) Functional repair of CFTR by CRISPR/Cas9 in
intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell 13:653–658
Sekelja M, Berget I, Naes T et al (2011) Unveiling an abundant core microbiota in the human adult
colon by a phylogroup-independent searching approach. ISME J 5:519–531
Simon R, O’connell M, Labes M et al (1986) Plasmid vectors for the genetic analysis and
manipulation of rhizobia and other gram-negative bacteria. Methods Enzymol 118:640–659
Simon R, Priefer U, Puhler A (1983) A broad host range mobilization system for in vivo genetic
engineering: transposon mutagenesis in gram negative bacteria. Nat Biotech 1:784
Sokol H, Pigneur B, Watterlot L et al (2008) Faecalibacterium prausnitzii is an anti-inflammatory
commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl
Acad Sci USA 105:16731–16736
Sokol H, Seksik P, Furet JP et al (2009) Low counts of Faecalibacterium prausnitzii in colitis
microbiota. Inflamm Bowel Dis 15:1183–1189
Sonnenberg GF, Monticelli LA, Alenghat T et al (2012) Innate lymphoid cells promote anatomical
containment of lymphoid-resident commensal bacteria. Science 336:1321–1325
3 Exploring the Bioactive Landscape of the Gut Microbiota … 81
Spor A, Koren O, Ley R (2011) Unravelling the effects of the environment and host genotype on
the gut microbiome. Nat Rev Micro 9:279–290
Staley JT, Konopka A (1985) Measurement of in situ activities of nonphotosynthetic
microorganisms in aquatic and terrestrial habitats. Annu Rev Microbiol 39:321–346
Stelzner M, Helmrath M, Dunn JCY et al (2012) A nomenclature for intestinal in vitro cultures.
Am Physiol—Gastrointest Liver Physiol 302:G1359–G1363
Stephen AM, Cummings JH (1980) The microbial contribution to human faecal mass. J Med
Microbiol 13:45–56
Stevens GA, Singh GM, Lu Y et al (2012) National, regional, and global trends in adult
overweight and obesity prevalences. Popul Health Metr 10:22
Stevenson BS, Eichorst SA, Wertz JT et al (2004) New strategies for cultivation and detection of
previously uncultured microbes. Appl Environ Microbiol 70:4748–4755
Sunagawa S, Mende DR, Zeller G et al (2013) Metagenomic species profiling using universal
phylogenetic marker genes. Nat Meth 10:1196–1199
Suskind DL, Brittnacher MJ, Wahbeh G et al (2015) Fecal microbial transplant effect on clinical
outcomes and fecal microbiome in active Crohn’s disease. Inflamm Bowel Dis 21:556–663
Swidsinski A, Loening-Baucke V, Vaneechoutte M et al (2008) Active Crohn’s disease and
ulcerative colitis can be specifically diagnosed and monitored based on the biostructure of the
fecal flora. Inflamm Bowel Dis 14:147–161
Tap J, Mondot S, Levenez F et al (2009) Towards the human intestinal microbiota phylogenetic
core. Environ Microbiol 11:2574–2584
Tazoe H, Otomo Y, Karaki S et al (2009) Expression of short-chain fatty acid receptor GPR41 in
the human colon. Biomed Res 30:149–156
Tedelind S, Westberg F, Kjerrulf M et al (2007) Anti-inflammatory properties of the short-chain
fatty acids acetate and propionate: a study with relevance to inflammatory bowel disease.
World J Gastroenterol 13:2826–2832
The human microbiome project consortium (2012) Structure, function and diversity of the healthy
human microbiome. Nature 486:207–214
Thangaraju M, Cresci GA, Liu K et al (2009) GPR109A is a G-protein-coupled receptor for the
bacterial fermentation product butyrate and functions as a tumor suppressor in colon. Cancer
Res 69:2826–2832
Tolonen AC, Chilaka AC, Church GM (2009) Targeted gene inactivation in Clostridium
phytofermentans shows that cellulose degradation requires the family 9 hydrolase Cphy3367.
Mol Microbiol 74:1300–1313
Turnbaugh PJ, Hamady M, Yatsunenko T et al (2009) A core gut microbiome in obese and lean
twins. Nature 457:480–484
Turnbaugh PJ, Ley RE, Mahowald MA et al (2006) An obesity-associated gut microbiome with
increased capacity for energy harvest. Nature 444:1027–1031
Uhlig T, Kyprianou T, Martinelli FG et al (2014) The emergence of peptides in the pharmaceutical
business: from exploration to exploitation. EuPA Open Proteomics 4:58–69
Vandussen KL, Marinshaw JM, Shaikh N et al (2015) Development of an enhanced human
gastrointestinal epithelial culture system to facilitate patient-based assays. Gut 64:911–920
Veeranagouda Y, Husain F, Wexler HM (2012) Transposon mutagenesis of the anaerobic
commensal, Bacteroides fragilis, using the EZ:TN5 transposome. FEMS Microbiol Lett
333:94–100
Venter JC, Adams MD, Myers EW et al (2001) The sequence of the human genome. Science
291:1304–1351
Vidal JE, Chen J, Li J et al (2009) Use of an EZ-Tn5-Based Random Mutagenesis System to
Identify a Novel Toxin Regulatory Locus in Clostridium perfringens Strain 13. PLoS ONE 4:
e6232
Vinolo MA, Ferguson GJ, Kulkarni S et al (2011) SCFAs induce mouse neutrophil chemotaxis
through the GPR43 receptor. PLoS ONE 6:e21205
82 P. Ó Cuív et al.
Virginia Polytechnic Institute and State University Anaerobe Laboratory (1975) Anaerobe
laboratory manual. Virginia Polytechnic Institute and State University, Anaerobe Laboratory,
Virginia
Wang B, Li K, Wang A et al (2015) Highly efficient CRISPR/HDR-mediated knock-in for mouse
embryonic stem cells and zygotes. Biotechniques 59:201–208
Waters VL (2001) Conjugation between bacterial and mammalian cells. Nat Genet 29:375–376
Whitehead TR, Hespell RB (1990) Heterologous expression of the Bacteroides ruminicola
xylanase gene in Bacteroides fragilis and Bacteroides uniformis. FEMS Microbiol Lett 54:
61–65
Whitfield-Cargile CM, Cohen ND, Chapkin RS et al (2016) The microbiota-derived metabolite
indole decreases mucosal inflammation and injury in a murine model of NSAID enteropathy.
Gut Microbes 7(3):246–261
Woese CR, Fox GE (1977) Phylogenetic structure of the prokaryotic domain: the primary
kingdoms. Proc Natl Acad Sci USA 74:5088–5090
Wright O, Delmans M, Stan GB et al (2015) GeneGuard: a modular plasmid system designed for
biosafety. ACS Synth Biol 4:307–316
Xing J, You C, Dong K et al (2013) Ameliorative effects of 3,4-oxo-isopropylidene-shikimic acid
on experimental colitis and their mechanisms in rats. Int Immunopharmacol 15:524–531
Yang X, Xie L, Li Y et al (2009) More than 9,000,000 unique genes in human gut bacterial
community: estimating gene numbers inside a human body. PLoS ONE 4:e6074
Yassour M, Lim MY, Yun HS et al (2016) Sub-clinical detection of gut microbial biomarkers of
obesity and type 2 diabetes. Genome Med 8:17
Zackular JP, Baxter NT, Iverson KD et al (2013) The gut microbiome modulates colon
tumorigenesis. MBio 4
Zeevi D, Korem T, Zmora N et al (2015) Personalized nutrition by prediction of glycemic
responses. Cell 163:1079–1094
Zhang L, Jia R, Palange NJ et al (2015a) Large Genomic Fragment Deletions and Insertions in
Mouse Using CRISPR/Cas9. PLoS ONE 10:e0120396
Zhang Y, Grosse-Honebrink A, Minton NP (2015b) A universal mariner transposon system for
forward genetic studies in the genus Clostridium. PLoS ONE 10:e0122411
Zhang Z, Geng J, Tang X et al (2014) Spatial heterogeneity and co-occurrence patterns of human
mucosal-associated intestinal microbiota. ISME J 8:881–893
Zoetendal EG, von Wright A, Vilpponen-Salmela T et al (2002) Mucosa-associated bacteria in the
human gastrointestinal tract are uniformly distributed along the colon and differ from the
community recovered from feces. Appl Environ Microbiol 68:3401–3407
Chapter 4
Using Metabolomic Approaches
to Characterize the Human Pathogen
Leishmania in Macrophages
Leishmania spp., are protozoan parasites that belong to the order Kinetoplastida
(Phylum Euglenozoa). The Kinetoplastida encompass a large group of flagellated
protists including the uniflagellated Trypanosomatidae, which all have parasitic life
styles, and the biflagellated Bodonidae which are typically free living. The family
Trypanosomatidae comprises the medically important genera Trypanosoma, which
includes Trypanosoma brucei and Trypanosoma cruzi, the causative agent of
human African trypanosomiasis and Chagas disease, respectively, as well as
Leishmania spp., which are the cause of a spectrum of diseases collectively termed
the leishmaniases (Stuart et al. 2008). T. brucei, T. cruzi, and Leishmania spp., all
rely on an insect vector as well as a mammalian host to complete their complex
lifecycle. While T. brucei and T. cruzi are transmitted by the tsetse fly and tri-
atomine bugs, respectively, Leishmania spp., are transmitted by phlebotomine
sandflies.
The pathology of leishmanial diseases range from localized self-healing cutaneous
or diffuse cutaneous infections (CL and DCL, respectively) to mucocutaneous
leishmaniasis (MCL) and visceral leishmaniasis (VL, also called kala-azar) (Pearson
and Sousa 1996). The pathology can be linked to different species—e.g., Leishmania
donovani, Leishmania infantum, and Leishmania chagasi cause VL, while
Leishmania major, Leishmania mexicana, Leishmania amazonensis, and Leishmania
aethiopica cause CL or DCL, and Leishmania braziliensis, Leishmania peruviana,
and Leishmania guyanensis cause MCL. CL is characterized by open sores around the
site of the sandfly bite, which can take years to heal and leave disfiguring scars.
Furthermore, CL can recur years after patients have seemingly healed from the initial
infection (Marovich et al. 2001; Gangneux et al. 2007). DCL is a more severe form of
CL that results in the formation of several hundred nodules or ulcers. The marring
lesions and disfiguring scars caused by CL and DCL can result in stigmatization and
lead to social exclusion and economic disadvantage (Kassi et al. 2008). MCL results
from the dissemination of parasites to mucosal membranes around the mouth and nose
and, in some cases, to the genital or optical mucosa (Huna-Baron et al. 2000). This
severe form of leishmaniasis can result in devastating destruction and deformation of
the face with high risk of secondary infections. VL, or kala-azar, occurs when para-
sites disseminate to the bone marrow, liver, and spleen, resulting in anemia, fever,
weightloss, and enlargement of liver and spleen. If left untreated, VL leads to death in
nearly 100 % of cases within a two-year period (WHO.int).
Leishmaniasis is endemic in 88 countries throughout the tropics, subtropics, and
the Mediterranean Basin with over 350 million people at risk of infection (WHO.int).
Disease prevalence is estimated at 12 million people with more than 2 million new
infections occurring annually (WHO.int). Mortalities from VL are increasing
worldwide and currently stand at >20,000 deaths annually, making it the second
deadliest parasitic disease after malaria (WHO.int; Desjeux 2004; Reithinger 2008;
Alvar et al. 2012). The current war in Syria and instability in the Middle East have led
to increased incidence and spread of leishmaniasis in the area due to a number of
factors including greater refugee migration and an inability to access staff and
facilities for diagnosis and treatment (Alawieh et al. 2014).
2 Treatment of Leishmaniasis
Fig. 1 Leishmania undergo a complex digenetic lifecycle developing within the sandfly vector
and the mammalian host. Transmission occurs when an infected sandfly takes a blood meal.
Amastigotes/infected macrophages are taken up by the sandfly during a blood meal (1) (Anjili
et al. 2006) and are initially enclosed within a digestive peritrophic sac in the sandfly gut (red)
(Pimenta et al. 1997). Amastigotes differentiate to flagellated procyclic promastigotes (2) which
undergo multiple rounds of division before slowing their replication and differentiating into highly
motile, but nondividing, nectomonads which escape the peritrophic sac (Gossage et al. 2003;
Dostalova and Volf 2012). Nectomonads adhere to the mid-gut wall (3) and develop into
leptomonads in the thoracic mid-gut (4). Leptomonads undergo further rounds of rapid replication
and are immobilized in a gel-like matrix which is secreted by this life-cycle stage. Leptomonads
differentiate into either nondividing haptomonads, which form a plug by attaching to each other
and the stomodeal valve (5), or nondividing metacyclics, which are highly infectious and are
transmitted during a blood meal (6) (Sacks and Perkins 1984, 1985; da Silva and Sacks 1987;
Sacks and da Silva 1987; Sacks 1989). In the mammalian host, metacyclic promastigotes are
phagocytosed by polymorphic neutrophils (PMN), which are first to reach the site of inflammation
following the sandfly bite (7, 8) (Laskay et al. 2003; van Zandbergen et al. 2004). PMNs have a
short life span and macrophages phagocytose released parasites and/or infected apoptotic PMNs
(8, 9) (Laskay et al. 2003; van Zandbergen et al. 2004; Ritter et al. 2009). Within the
phagolysosome of macrophages, Leishmania differentiate into the amastigote stage, adapt to their
new milieu (10) and start replicating (11) (Kaye and Scott 2011). Amastigotes are released when
heavily infected macrophages rupture (12) allowing infection of other cells (Noronha et al. 2000).
The lifecycle begins again when sandflies ingest free amastigotes or infected cells during a blood
meal (1)
parasites do not replicate) represents a ‘Trojan Horse’ strategy for targeting mac-
rophages, the preferred host cell for these pathogens (Laskay et al. 2003; van
Zandbergen et al. 2004, 2007). Alternatively, extracellular Leishmania may be taken
up incidentally during the phagocytosis of necrotic PMNs (the ‘Trojan rabbit’
model) (Ritter et al. 2009). Following uptake by macrophages and the maturation of
the vacuole into a phagolysosomal compartment, promastigotes differentiate to
4 Using Metabolomic Approaches to Characterize the Human Pathogen … 87
The paucity of good treatment options and no vaccine means that there is an urgent
need to identify new therapeutic targets in Leishmania. Many of these targets will
ideally be specific to parasite metabolism, targeting pathways critical for the sur-
vival and proliferation of the intracellular amastigote stage. Understanding the
interplay of host and parasite metabolism is critical in identifying targets that will
not affect the host metabolism. Finally, by mapping parasite metabolism it is
anticipated that the mode of action (MOA) of current chemotherapies will be
revealed and new insights into drug resistance mechanisms gained. The metabolism
of Leishmania has been studied for several decades using a range of approaches and
techniques. Here we discuss these techniques, the insights they have provided into
Leishmania metabolism as well as their limitations.
Vanegas et al. 2007; Collingridge et al. 2010; Tonkin et al. 2015), complicating the
interpretation of proteomics analyses.
These studies suggest that Leishmania may be more dependent on posttransla-
tional regulatory mechanisms during differentiation or adaptation to changing
environmental circumstance than other eukaryotes. Indeed gene families for pro-
teins involved in protein posttranslational modifications (kinases, phosphatases,
etc.) are commonly amplified in these parasites. In particular, phosphoproteomic
studies have revealed major changes in protein phosphorylation during pro-
mastigote to amastigote differentiation (Rosenzweig et al. 2008b; Morales et al.
2010a; Tsigankov et al. 2013, 2014). Interestingly, some of the most abundant
proteins to be phosphorylated during differentiation were heat-shock proteins which
are otherwise constitutively expressed (Morales et al. 2010a). Rosenzweig et al.
(2008b) utilized isobaric tags for relative and absolute quantification/liquid
chromatography-mass spectrometry/mass spectrometry (iTRAQ/LC-MS/MS) to
characterize a number of posttranslational modifications in Leishmania pro-
mastigotes and Amaaxenic. The Leishmania genomes also encode large numbers of
protein kinases and phosphatases, several of which have been shown to be essential
for differentiation and/or amastigote survival (Ivens et al. 2005; Morales et al.
2010b; Cayla et al. 2014). To date, however, no complete signaling pathways in
Leishmania (from surface receptor/sensor to downstream effector(s)) have been
delineated and the function of most of these posttranslational modifications remains
poorly defined. Nonetheless, studies on proteins involved in regulating mitochon-
drial proteins through the covalent attachment of ubiquitin-like proteins
(Gannavaram et al. 2011, 2012) suggest that this will be a rewarding endeavor. In a
recent study, Goldman-Pinkovich et al. (2016) described a coordinated arginine
deprivation response (ADR) that is activated in response to low arginine levels
which are expected to occur in the macrophage phagolysosome. L. donovani
responded to low external arginine levels by rapidly upregulating the arginine
transporter, LdAAP3. This response was dependent on expression of the mitogen
activated protein kinase 2 (MAPK2)-dependent signaling pathway
(Goldman-Pinkovich et al. 2016). Similarly, stage-specific regulation of expression
of the major plasma membrane glucose transporters in L. mexicana was found to be
regulated by ubiquitination of the cytoplasmic tails of the transporter proteins and
their internalization and degradation in the lysosome (Vince et al. 2011).
revealed that changes in the metabolome were highly varied. For some combina-
tions, the changes were not additive (amphotericin-B/paromomycin, amphotericin/
miltefosine and amphotericin-B/antimonial), while in other cases changes to the
metabolome were greater than what would be predicted from singly resistant lines
(miltefosine/paromomycin and antimonial/paromomycin). Quantitative analysis of
amphotericin B/antimonial (AS) and antimonial/paromomycin (SP) resistant lines
identified several shared changes in metabolites of proline biosynthesis and the
transsulfuration pathway which the researchers suggest is indicative of increased
protection against oxidative stress in these CTR lines (proline as a general
stress-response metabolite and free radical scavenger, products of transsulfuration
pathway feeding into trypanothione biosynthesis). Changes in the abundance of
lipid and sterol pathway intermediates were also observed in the tested CTR lines
suggesting alterations in membrane composition. Importantly, the researchers
sought to validate these conclusions using functional assays (susceptibility to
drug-induced and extracellular reactive oxygen stress, genomic DNA damage and
membrane fluidity).
As with many other metabolomics studies in Leishmania, the bulk of
MOA/resistance experiments have been undertaken only in the promastigote stage
(Vincent and Barrett 2015). Ultimately, however, any conclusions made about a
given drug’s MOA or proposed mechanism of resistance need to be verified in the
disease causing amastigote stage, such as those generated in vitro (Amaaxenic) or
isolated from in vitro infected macrophages (AmaMΦ) or from murine lesions
(Amalesion) (Vincent and Barrett 2015). As detailed in the following sections, each
Leishmania developmental stage is characterized by unique metabolic features and
growth rate which may result in significantly different drug efficacy, MOA, and
mechanisms of resistance. Furthermore, the host environment may additionally alter
a drug’s effectiveness by altering the parasite’s environment (e.g. nutrient restriction
and stress induced by host microbicidal responses), modulate a drug’s availability
(e.g. membrane permeability and prodrug catabolism), or indirectly kill the
amastigote by inhibiting targets in the macrophage host (e.g. immunomodulatoy
effects). Indeed, compound screening experiments have revealed significant dif-
ferences in the sensitivity of promastigote and amastigote stages to many drugs (De
Muylder et al. 2011; De Rycker et al. 2013). These findings indicate that under-
standing the nutritional environment and metabolism of the host cell is also criti-
cally important when delineating a drug’s MOA and the parasite’s development of
resistance. For example, it has been suggested that the accumulation of amino acids
in drug-resistant lines preserves the parasite’s fitness for the invasion and prolif-
eration in the nutritionally limited environment of the host macrophage by pro-
viding an alternative carbon source (Vermeersch et al. 2009; Berg et al. 2013;
Canuto et al. 2014). Finally, care needs to be taken when studying field isolates so
that detected genetic differences can be aligned with drug susceptibility/resistance
measures, virulence, and metabolomics data.
4 Using Metabolomic Approaches to Characterize the Human Pathogen … 97
Fig. 2 Central carbon metabolism in Leishmania. The schematic shows the acquisition of carbon
sources and key metabolic pathways. Glycolysis and gluconeogenesis as well as the GSF pathway occur
in specialized peroxisomes termed glycosomes, while the TCA cycle and β-oxidation occur in
mitochondria (enzymes for β-oxidation also localize to the glycosomes but mitochondria are likely the
main site of β-oxidation). The major carbohydrate reserve material of Leishmania is comprised of short
oligosaccharides (4–40 mannose residues long), termed mannogen which accumulates in the cytosol and
may play a major role in regulating substrate availability for gluconeogenesis and glycolysis. The role of
several enzymes/pathways in Leishmania metabolism was assessed by generating gene knock-outs or
using metabolic inhibitors. Essential pathways are indicated by red arrows. In many cases, the effects of
gene depletions or metabolic inhibitors on Leishmania viability differ between the promastigote and
amastigote stage (see text), highlighting the distinct metabolism of the two life-cycle stages. For example
parasites lacking the gluconeogenic enzyme FBPase grow normally as promastigotes in rich media but
display attenuated virulence in macrophages and mice. Other pathways appear to be essential in both
life-cycle stages—e.g., Luque-Ortega et al. (2008) showed that inhibition of the ATP synthase in
Leishmania through histatin 5 is lethal for promastigotes and amastigotes. The salvage of some essential
metabolites such as purine and vitamins are discussed elsewhere (e.g. McConville and Naderer 2011).
AcCoA Acetylcoenzyme A; C Cytochrome C; ETC Electron transport chain; Fru6P
Fructose-6-phosphate; Fru1,6P2, Fructose-1,6-bisphosphate; GDP-Man Guanosine diphosphate man-
nose; Glc6P Glucose-6-phosphate; GlcN6P Glucosamine-6-phosphate; Hst5 Histatin 5; Man6P
Mannose-6-phosphate; Man1P Mannose-1-phosphate; NaFAc Sodium fluoroacetate; KG Ketoglutarate;
Mann Mannogen oligosaccharides; NAD Nicotinamide dinucleotide; PM Plasma membrane; PPP
Pentose phosphate pathway; Rib5P Ribose-5-phosphate; TCA Tricarboxylic acid; Triose-P Triose
phosphate; UQ Ubiquinone; I Complex I (NADH dehydrogenase); II Complex II (fumarate reductase);
III Complex III (cytochrome bc1 complex); IV Complex IV (cytochrome c oxidase); V Complex V (ATP
synthase)
While metabolic footprinting has provided important insights into the distinct
metabolism of the Leishmania life-cycle stages, the information gained from ana-
lyzing extracellular metabolites is limited. An alternative approach is to measure
4 Using Metabolomic Approaches to Characterize the Human Pathogen … 99
stringent response was not just a consequence of reduced replication but rather a
hard-wired response that is induced during amastigote differentiation. Amastigotes
also exhibited increased fatty acid β-oxidation and increased reliance on the TCA
cycle metabolism (Saunders et al. 2014), consistent with results from earlier pro-
teomic analyses and metabolomics analysis (see above).
These labeling studies were used to predict pathways essential for amastigote
survival in vivo. In particular, the finding that amastigotes stages exhibited negli-
gible rates of glutamate uptake (compared to promastigotes) while the carbon
backbones derived from 13C-glucose and 13C-fatty acids were incorporated into
glutamate/glutamine, suggested that the TCA cycle may have an important role in
generating precursors, such as α-ketoglutarate for glutamate/glutamine synthesis.
These amino acids serve as essential amino donors for a number of other essential
pathways including aminosugar synthesis, pyrimidine synthesis, and
glutathione/trypanothione synthesis. Consistent with the proposal, treatment of
infected macrophages with either sodium fluoroacetate (NaFAc, an inhibitor of
TCA cycle aconitase enzyme) or methionine sulfoxime (MSO, an inhibitors of
glutamine synthetase) resulted in death of intracellular L. mexicana amastigotes
(Saunders et al. 2014). While all developmental stages exhibited some growth
sensitivity to these inhibitors in axenic cultures, particularly when exogenous
glutamate or glutamine was absent, Amaaxenic were much more sensitive to these
inhibitors than Prolog. While Prolog ceased growth during treatment with 5 mM
NaFAc, Amaaxenic lost viability at 50–100-times lower concentrations. The effect of
NaFAc on Amaaxenic was only partially rescued by addition of very high concen-
trations of glutamate (5 mM). Similarly, MSO treatment led to a growth arrest in
Prolog which was rescued by supplementation with glutamine. Interestingly, growth
102 J. Kloehn et al.
All of the studies described above rely on the analysis of axenically cultured
amastigotes or isolated Amalesion that have been incubated in rich media which may
not reflect the nutrient levels in vivo (Saunders et al. 2014). Indeed, the nutrients
4 Using Metabolomic Approaches to Characterize the Human Pathogen … 103
Fig. 4 The physiological state of Leishmania life-cycle stages. Differences in the replication rate
as well as protein, RNA, and lipid turnover were measured using 2H2O-labeling in vitro and
in vivo (Kloehn et al. 2015). Leishmania Prolog show the fastest turnover rate for all
macromolecules indicating rapid replication, protein and RNA, and lipid synthesis. Strikingly,
the protein, RNA, and lipid turnover rates of Amalesion are reduced about 15–30-fold compared to
Prolog and are markedly reduced even compared to nondividing Prostat. These results indicate that
Leishmania enter a semiquiescent state in the lesion environment which is characterized by slow
growth and low activity of other energy intensive cellular processes such as protein synthesis
providing further support for the notion that amastigotes enter into a distinct state of
slow growth and metabolic quiescence.
Several studies on Amaaxenic have suggested that global rates of protein synthesis
are downregulated, based on measurements of rates of 35S-methionine incorpora-
tion and polysome profile analysis (Lahav et al. 2011; Cloutier et al. 2012). Another
interesting approach for estimating protein turnover/metabolic activity in Amalesion
in vivo has recently been developed using L. major that express a photoconvertible
fluorescent protein (Muller et al. 2013). The authors suggest that protein synthesis
in situ is reduced due to the suppression of metabolism and cell division by sub-
lethal levels of NO. However, a broader application of this approach is limited
given that the fluorescence readout is difficult to calibrate to overall protein turnover
rates and also requires the generation of transgenic parasites lines with possible
associated virulence reduction (da Silva and Sacks 1987; Moreira et al. 2012; Ali
et al. 2013).
Additionally, 2H2O-labeling was shown to delineate specific metabolic pathways
in culture and in vivo. Analysis of 2H-incorporation into total cellular fatty acid
pools in Amalesion showed that this stage largely relies on the scavenging of fatty
acids, in contrast to the situation in promastigotes. However, Amalesion were still
dependent on the synthesis of linoleic acid (C18:2), as shown by the labeling of this
fatty acid in parasite extracts but not host cell serum and tissue (Kloehn et al. 2015).
Linoleic acid is the major polyunsaturated fatty acid of these stages and is syn-
thesized by desaturation of oleic acid. The enzyme oleate desaturase is absent in
mammals, and may therefore be a promising drug target.
Metabolomic approaches have provided important new insights into the biology of
Leishmania and will continue to be an essential tool for investigating the meta-
bolism of these parasites given their dependence on posttranslational regulatory
processes. In particular, metabolomics has been used to map metabolic networks in
different developmental stages and parasite mutant lines, as well as to define the
mode of action of drugs and understand resistance mechanisms. Importantly, var-
ious 13C/2H labeling strategies have now been used to map parasite metabolism and
physiology in vitro and in infected tissues. We suggest that metabolomic mea-
surements using noninvasive stable isotope tracers will be increasingly important to
understand Leishmania metabolism in the mammalian host. Combining metabo-
lomic approaches with other ‘-omics’ approaches as well as molecular biology (e.g.
gene depletion), biochemistry (e.g. metabolic inhibitors), and biological data (e.g.
virulence) are expected to advance our understanding of Leishmania metabolism.
Last, phenotypically heterogeneous cells and the mechanism underlying cell-to-cell
variability have been identified and studied in a number of bacterial infections
(Lewis 2010; Helaine and Holden 2013; Helaine et al. 2014; Bumann 2015; Kopf
et al. 2016) but are poorly understood in protozoan infections, despite their crucial
4 Using Metabolomic Approaches to Characterize the Human Pathogen … 107
References
Alawieh A et al (2014) Revisiting leishmaniasis in the time of war: the Syrian conflict and the
Lebanese outbreak. Int J Infect Dis 29:115–119
Ali KS, Rees RC, Terrell-Nield C, Ali SA (2013) Virulence loss and amastigote transformation
failure determine host cell responses to Leishmania mexicana. Parasite Immunol 35(12):441–
456
Alvar J et al (2012) Leishmaniasis worldwide and global estimates of its incidence. PLoS One 7
(5):e35671
Amaral V, Pirmez C, Goncalves A, Ferreira V, Grimaldi G Jr (2000) Cell populations in lesions of
cutaneous leishmaniasis of Leishmania (L.) amazonensis—infected rhesus macaques, Macaca
mulatta. Mem Inst Oswaldo Cruz 95(2):209–216
Andrews KT, Fisher G, Skinner-Adams TS (2014) Drug repurposing and human parasitic
protozoan diseases. Int J Parasitol Drugs Drug Resist 4(2):95–111
Anjili C et al (2006) Estimation of the minimum number of Leishmania major amastigotes
required for infecting Phlebotomus duboscqi (Diptera: Psychodidae). East Afr Med J 83(2):68–
71
Antoniewicz MR, Kelleher JK, Stephanopoulos G (2011) Measuring deuterium enrichment of
glucose hydrogen atoms by gas chromatography/mass spectrometry. Anal Chem 83(8):3211–
3216
Arjmand M et al (2016) Metabolomics-based study of logarithmic and stationary phases of
promastigotes in Leishmania major by 1H NMR spectroscopy. Iran Biomed J 20(2):77–83
Ashutosh Sundar S, Goyal N (2007) Molecular mechanisms of antimony resistance in Leishmania.
J Med Microbiol 56(Pt 2):143–153
Bakker BM et al (2000) Compartmentation protects Trypanosomes from the dangerous design of
glycolysis. Proc Natl Acad Sci USA 97(5):2087–2092
Barhoumi M, Tanner NK, Banroques J, Linder P, Guizani I (2006) Leishmania infantum LeIF
protein is an ATP-dependent RNA helicase and an eIF4A-like factor that inhibits translation in
yeast. FEBS J 273(22):5086–5100
Bates PA (2007) Transmission of Leishmania metacyclic promastigotes by phlebotomine sand
flies. Int J Parasitol 37(10):1097–1106
Beach R, Kiilu G, Hendricks L, Oster C, Leeuwenburg J (1984) Cutaneous leishmaniasis in
Kenya: transmission of Leishmania major to man by the bite of a naturally infected
Phlebotomus duboscqi. Trans R Soc Trop Med Hyg 78(6):747–751
Beach R, Kiilu G, Leeuwenburg J (1985) Modification of sand fly biting behavior by Leishmania
leads to increased parasite transmission. Am J Trop Med Hyg 34(2):278–282
Bente M et al (2003) Developmentally induced changes of the proteome in the protozoan parasite
Leishmania donovani. Proteomics 3(9):1811–1829
Berg M et al (2015) Experimental resistance to drug combinations in Leishmania donovani:
metabolic and phenotypic adaptations. Antimicrob Agents Chemother 59(4):2242–2255
108 J. Kloehn et al.
Croft SL, Sundar S, Fairlamb AH (2006) Drug resistance in leishmaniasis. Clin Microbiol Rev 19
(1):111–126
Cull B et al (2014) Glycosome turnover in Leishmania major is mediated by autophagy.
Autophagy 10(12):2143–2157
da Silva R, Sacks DL (1987) Metacyclogenesis is a major determinant of Leishmania promastigote
virulence and attenuation. Infect Immun 55(11):2802–2806
Davidson RN, den Boer M, Ritmeijer K (2009) Paromomycin. Trans R Soc Trop Med Hyg 103
(7):653–660
De Muylder G et al (2011) A screen against Leishmania intracellular amastigotes: comparison to a
promastigote screen and identification of a host cell-specific hit. PLoS Negl Trop Dis 5(7):
e1253
de Oliveira CI, Nascimento IP, Barral A, Soto M, Barral-Netto M (2009) Challenges and
perspectives in vaccination against leishmaniasis. Parasitol Int 58(4):319–324
De Rycker M et al (2013) Comparison of a high-throughput high-content intracellular Leishmania
donovani assay with an axenic amastigote assay. Antimicrob Agents Chemother 57(7):2913–
2922
De Souza DP, Saunders EC, McConville MJ, Likic VA (2006) Progressive peak clustering in
GC-MS Metabolomic experiments applied to Leishmania parasites. Bioinformatics 22
(11):1391–1396
Dereure J et al (2003) Visceral leishmaniasis. Persistence of parasites in lymph nodes after clinical
cure. J Infect 47(1):77–81
Dermine JF, Desjardins M (1999) Survival of intracellular pathogens within macrophages.
Protoplasma 210(1–2):11–24
Dermine JF, Goyette G, Houde M, Turco SJ, Desjardins M (2005) Leishmania donovani
lipophosphoglycan disrupts phagosome microdomains in J774 macrophages. Cell Microbiol 7
(9):1263–1270
Dermine JF, Scianimanico S, Prive C, Descoteaux A, Desjardins M (2000) Leishmania
promastigotes require lipophosphoglycan to actively modulate the fusion properties of
phagosomes at an early step of phagocytosis. Cell Microbiol 2(2):115–126
Desjardins M, Descoteaux A (1997) Inhibition of phagolysosomal biogenesis by the Leishmania
lipophosphoglycan. J Exp Med 185(12):2061–2068
Desjeux P (2004) Leishmaniasis: current situation and new perspectives. Comp Immunol
Microbiol Infect Dis 27(5):305–318
Dostalova A, Volf P (2012) Leishmania development in sand flies: parasite-vector interactions
overview. Parasit Vectors 5:276
Downing T et al (2011) Whole genome sequencing of multiple Leishmania donovani clinical
isolates provides insights into population structure and mechanisms of drug resistance. Genome
Res 21(12):2143–2156
Doyle MA et al (2009) LeishCyc: a biochemical pathways database for Leishmania major. BMC
Syst Biol 3:57
Dufner D, Previs SF (2003) Measuring in vivo metabolism using heavy water. Curr Opin Clin
Nutr Metab Care 6(5):511–517
Eibl H, Unger C (1990) Hexadecylphosphocholine: a new and selective antitumor drug. Cancer
Treat Rev 17(2–3):233–242
El Fakhry Y, Ouellette M, Papadopoulou B (2002) A proteomic approach to identify
developmentally regulated proteins in Leishmania infantum. Proteomics 2(8):1007–1017
El-Sayed NM et al (2005) The genome sequence of Trypanosoma cruzi, etiologic agent of Chagas
disease. Science 309(5733):409–415
Englander SW, Mayne L, Bai Y, Sosnick TR (1997) Hydrogen exchange: the modern legacy of
Linderstrom-Lang. Protein Sci 6(5):1101–1109
Evans KJ, Kedzierski L (2012) Development of Vaccines against Visceral Leishmaniasis. J Trop
Med 2012:892817
110 J. Kloehn et al.
Lahav T et al (2011) Multiple levels of gene regulation mediate differentiation of the intracellular
pathogen Leishmania. FASEB J 25(2):515–525
Lamour SD, Choi BS, Keun HC, Muller I, Saric J (2012) Metabolic characterization of
Leishmania major infection in activated and nonactivated macrophages. J Proteome Res 11
(8):4211–4222
Landau BR et al (1995) Use of 2H2O for estimating rates of gluconeogenesis. Application to the
fasted state. J Clin Invest 95(1):172–178
Lang T, Goyard S, Lebastard M, Milon G (2005) Bioluminescent Leishmania expressing
luciferase for rapid and high throughput screening of drugs acting on amastigote-harbouring
macrophages and for quantitative real-time monitoring of parasitism features in living mice.
Cell Microbiol 7(3):383–392
Laskay T, van Zandbergen G, Solbach W (2003) Neutrophil granulocytes–Trojan horses for
Leishmania major and other intracellular microbes? Trends Microbiol 11(5):210–214
Lau SK et al (2015) Metabolomic profiling of Burkholderia pseudomallei using
UHPLC-ESI-Q-TOF-MS reveals specific biomarkers including 4-methyl-5-thiazoleethanol
and unique thiamine degradation pathway. Cell Biosci 5:26
Leifso K, Cohen-Freue G, Dogra N, Murray A, McMaster WR (2007) Genomic and proteomic
expression analysis of Leishmania promastigote and amastigote life stages: the Leishmania
genome is constitutively expressed. Mol Biochem Parasitol 152(1):35–46
Lester W Jr, Sun SH, Seber A (1960) Observations on the influence of deuterium on bacterial
growth. Ann NY Acad Sci 84:667–677
Lewis K (2010) Persister cells. Annu Rev Microbiol 64:357–372
Lorenz MC, Fink GR (2002) Life and death in a macrophage: role of the glyoxylate cycle in
virulence. Eukaryot Cell 1(5):657–662
Luque-Ortega JR, van’t Hof W, Veerman EC, Saugar JM, Rivas L (2008) Human antimicrobial
peptide histatin 5 is a cell-penetrating peptide targeting mitochondrial ATP synthesis in
Leishmania. FASEB J 22(6):1817–1828
Magkos F, Mittendorfer B (2009) Stable isotope-labeled tracers for the investigation of fatty acid
and triglyceride metabolism in humans in vivo. Clin Lipidol 4(2):215–230
Maher EA et al (2012) Metabolism of [U-13 C]glucose in human brain tumors in vivo. NMR
Biomed 25(11):1234–1244
Maier T, Guell M, Serrano L (2009) Correlation of mRNA and protein in complex biological
samples. FEBS Lett 583(24):3966–3973
Marovich MA et al (2001) Leishmaniasis recidivans recurrence after 43 years: a clinical and
immunologic report after successful treatment. Clin Infect Dis 33(7):1076–1079
McCall LI, Zhang WW, Ranasinghe S, Matlashewski G (2013) Leishmanization revisited:
immunization with a naturally attenuated cutaneous Leishmania donovani isolate from Sri
Lanka protects against visceral leishmaniasis. Vaccine 31(10):1420–1425
McConville M (2014) Open questions: microbes, metabolism and host-pathogen interactions.
BMC Biol 12:18
McConville MJ, de Souza D, Saunders E, Likic VA, Naderer T (2007) Living in a phagolysosome;
metabolism of Leishmania amastigotes. Trends Parasitol 23(8):368–375
McConville MJ, Naderer T (2011) Metabolic pathways required for the intracellular survival of
Leishmania. Annu Rev Microbiol 65:543–561
McConville MJ, Saunders EC, Kloehn J, Dagley MJ (1988) Leishmania carbon metabolism in the
macrophage phagolysosome—feast or famine? F1000Research 4(F1000 Faculty Rev):938
McElrath MJ, Murray HW, Cohn ZA (1988) The dynamics of granuloma formation in
experimental visceral leishmaniasis. J Exp Med 167(6):1927–1937
Michel G et al (2011) Luciferase-expressing Leishmania infantum allows the monitoring of
amastigote population size, in vivo, ex vivo and in vitro. PLoS Negl Trop Dis 5(9):e1323
Michels PA, Bringaud F, Herman M, Hannaert V (2006) Metabolic functions of glycosomes in
Trypanosomatids. Biochim Biophys Acta 1763(12):1463–1477
Millington OR, Myburgh E, Mottram JC, Alexander J (2010) Imaging of the host/parasite
interplay in cutaneous leishmaniasis. Exp Parasitol 126(3):310–317
4 Using Metabolomic Approaches to Characterize the Human Pathogen … 113
Mittra B et al (2013) Iron uptake controls the generation of Leishmania infective forms through
regulation of ROS levels. J Exp Med 210(2):401–416
Miyamoto S et al (2001) Discrepancies between the gene expression, protein expression, and
enzymatic activity of thymidylate synthase and dihydropyrimidine dehydrogenase in human
gastrointestinal cancers and adjacent normal mucosa. Int J Oncol 18(4):705–713
Moore EM, Lockwood DN (2010) Treatment of visceral leishmaniasis. J Glob Infect Dis 2
(2):151–158
Moradin N, Descoteaux A (2012) Leishmania promastigotes: building a safe niche within
macrophages. Front Cell Infect Microbiol 2:121
Morales MA et al (2010a) Phosphoproteome dynamics reveal heat-shock protein complexes
specific to the Leishmania donovani infectious stage. Proc Natl Acad Sci USA 107(18):8381–
8386
Morales MA, Pescher P, Spath GF (2010b) Leishmania major MPK7 protein kinase activity
inhibits intracellular growth of the pathogenic amastigote stage. Eukaryot Cell 9(1):22–30
Moreira D et al (2012) Impact of continuous axenic cultivation in Leishmania infantum virulence.
PLoS Negl Trop Dis 6(1):e1469
Moreira D et al (2015) Leishmania infantum modulates host macrophage mitochondrial
metabolism by hijacking the SIRT1-AMPK axis. PLoS Pathog 11(3):e1004684
Muller AJ et al (2013) Photoconvertible pathogen labeling reveals nitric oxide control of
Leishmania major infection in vivo via dampening of parasite metabolism. Cell Host Microbe
14(4):460–467
Munoz-Elias EJ et al (2005) Replication dynamics of Mycobacterium tuberculosis in chronically
infected mice. Infect Immun 73(1):546–551
Murphy EJ (2006) Stable isotope methods for the in vivo measurement of lipogenesis and
triglyceride metabolism. J Anim Sci 84(Suppl):E94–E104
Myler PJ et al (2000) Genomic organization and gene function in Leishmania. Biochem Soc Trans
28(5):527–531
Naderer T et al (2006) Virulence of Leishmania major in macrophages and mice requires the
gluconeogenic enzyme fructose-1,6-bisphosphatase. Proc Natl Acad Sci USA 103(14):5502–
5507
Naderer T, Heng J, McConville MJ (2010) Evidence that intracellular stages of Leishmania major
utilize amino sugars as a major carbon source. PLoS Pathog 6(12):e1001245
Naderer T et al (2015) Intracellular survival of Leishmania major depends on uptake and
degradation of extracellular matrix glycosaminoglycans by macrophages. PLoS Pathog 11(9):
e1005136
Nasereddin A, Schweynoch C, Schonian G, Jaffe CL (2010) Characterization of Leishmania
(Leishmania) tropica axenic amastigotes. Acta Trop 113(1):72–79
Neese RA et al (2002) Measurement in vivo of proliferation rates of slow turnover cells by 2H2O
labeling of the deoxyribose moiety of DNA. Proc Natl Acad Sci USA 99(24):15345–15350
Neurohr KJ, Barrett EJ, Shulman RG (1983) In vivo carbon-13 nuclear magnetic resonance studies
of heart metabolism. Proc Natl Acad Sci USA 80(6):1603–1607
Noronha FS, Cruz JS, Beirao PS, Horta MF (2000) Macrophage damage by Leishmania
amazonensis cytolysin: evidence of pore formation on cell membrane. Infect Immun 68
(8):4578–4584
Nugent PG, Karsani SA, Wait R, Tempero J, Smith DF (2004) Proteomic analysis of Leishmania
mexicana differentiation. Mol Biochem Parasitol 136(1):51–62
Olliaro PL et al (2005) Treatment options for visceral leishmaniasis: a systematic review of clinical
studies done in India, 1980–2004. Lancet Infect Dis 5(12):763–774
Oppenheim RD et al (2014) BCKDH: the missing link in apicomplexan mitochondrial metabolism
is required for full virulence of Toxoplasma gondii and Plasmodium berghei. PLoS Pathog 10
(7):e1004263
Ouellette M, Drummelsmith J, Papadopoulou B (2004) Leishmaniasis: drugs in the clinic,
resistance and new developments. Drug Resist Updat 7(4–5):257–266
114 J. Kloehn et al.
Rojo D et al (2015) A multiplatform metabolomic approach to the basis of antimonial action and
resistance in Leishmania infantum. PLoS One 10(7):e0130675
Rosenzweig D et al (2008a) Retooling Leishmania metabolism: from sand fly gut to human
macrophage. FASEB J 22(2):590–602
Rosenzweig D, Smith D, Myler PJ, Olafson RW, Zilberstein D (2008b) Post-translational
modification of cellular proteins during Leishmania donovani differentiation. Proteomics 8
(9):1843–1850
Russell DG, Vanderven BC, Glennie S, Mwandumba H, Heyderman RS (2009) The macrophage
marches on its phagosome: dynamic assays of phagosome function. Nat Rev Immunol 9
(8):594–600
Saar Y et al (1998) Characterization of developmentally-regulated activities in axenic amastigotes
of Leishmania donovani. Mol Biochem Parasitol 95(1):9–20
Sacks DL (1989) Metacyclogenesis in Leishmania promastigotes. Exp Parasitol 69(1):100–103
Sacks DL, da Silva RP (1987) The generation of infective stage Leishmania major promastigotes
is associated with the cell-surface expression and release of a developmentally regulated
glycolipid. J Immunol 139(9):3099–3106
Sacks DL, Perkins PV (1984) Identification of an infective stage of Leishmania promastigotes.
Science 223(4643):1417–1419
Sacks DL, Perkins PV (1985) Development of infective stage Leishmania promastigotes within
phlebotomine sand flies. Am J Trop Med Hyg 34(3):456–459
Saunders EC et al (2015) Use of (13)C stable isotope labelling for pathway and metabolic flux
analysis in Leishmania parasites. Methods Mol Biol 1201:281–296
Saunders EC et al (2012) LeishCyc: a guide to building a metabolic pathway database and
visualization of metabolomic data. Methods Mol Biol 881:505–529
Saunders EC et al (2014) Induction of a stringent metabolic response in intracellular stages of
Leishmania mexicana leads to increased dependence on mitochondrial metabolism. PLoS
Pathog 10(1):e1003888
Saxena A et al (2007) Analysis of the Leishmania donovani transcriptome reveals an ordered
progression of transient and permanent changes in gene expression during differentiation. Mol
Biochem Parasitol 152(1):53–65
Schlein Y, Jacobson RL, Messer G (1992) Leishmania infections damage the feeding mechanism
of the sandfly vector and implement parasite transmission by bite. Proc Natl Acad Sci USA 89
(20):9944–9948
Schriefer A, Wilson ME, Carvalho EM (2008) Recent developments leading toward a paradigm
switch in the diagnostic and therapeutic approach to human leishmaniasis. Curr Opin Infect Dis
21(5):483–488
Seco-Hidalgo V, Osuna A, Pablos LM (2015) To bet or not to bet: deciphering cell to cell variation
in protozoan infections. Trends Parasitol 31(8):350–356
Seifert K (2011) Structures, targets and recent approaches in anti-leishmanial drug discovery and
development. Open Med Chem J 5:31–39
Seifert K et al (2003) Characterisation of Leishmania donovani promastigotes resistant to
hexadecylphosphocholine (miltefosine). Int J Antimicrob Agents 22(4):380–387
Seifert K et al (2007) Inactivation of the miltefosine transporter, LdMT, causes miltefosine
resistance that is conferred to the amastigote stage of Leishmania donovani and persists in vivo.
Int J Antimicrob Agents 30(3):229–235
Shalwitz RA et al (1989) Hepatic glycogen synthesis from duodenal glucose and alanine. An
in situ 13C NMR study. J Biol Chem 264(7):3930–3934
Silva LA, Vinaud MC, Castro AM, Cravo PV, Bezerra JC (2015) In silico search of energy
metabolism inhibitors for alternative leishmaniasis treatments. Biomed Res Int 2015:965725
Sindermann H, Engel J (2006) Development of miltefosine as an oral treatment for leishmaniasis.
Trans R Soc Trop Med Hyg 100(Suppl 1):S17–20
Sollelis L et al (2015) First efficient CRISPR-Cas9-mediated genome editing in Leishmania
parasites. Cell Microbiol 17(10):1405–1412
116 J. Kloehn et al.
Online Reference
1 Introduction
Microbial metabolism has helped shape the world into what it is today, and con-
tinues to drive biogeochemical cycles (Falkowski et al. 2008) including the carbon
cycle. Soil microorganisms play a central role in the global carbon cycle, with an
estimated soil carbon pool of 2500 Gt, over three times the size of the atmospheric
carbon pool (Lal 2004). Inputs of organic carbon into soil is largely plant and
microbial biomass-derived, and carbon is released from soil into the atmosphere
mainly as CO2, the product of plant root, and microbial respiration (Johnston et al.
2004). While we are able to measure emergent properties such as the total release of
CO2 from soil and total organic carbon in soil at a particular time, the underlying
processes that occur between input and release are not well defined. This limits our
ability to understand how human activities are altering the balance of the global
carbon cycle, and how this will affect soil carbon dynamics (Lal 2004) that are
mediated by soil microbial community metabolism.
The bulk of microbial community studies have been based on metagenomics
approaches, where total genomic DNA from soil is sequenced (Delmont et al. 2011;
Fierer et al. 2012; Roesch et al. 2007). This culture-independent approach yields
insights into the microbial community structure (phylogenetic makeup), and has
become a very active field of research due to advances in sequencing technologies
(Franzosa et al. 2015). Besides community structure, metagenomes also reveal the
complement of genes present in soil microbial communities, reflecting their
potential metabolic functions which are inferences based on often poorly annotated
Fig. 1 Schematic representation of three competing models for the fate of organic inputs to soil
(top), and the recently proposed soil continuum model (below). Selective preservation assumes that
some organic materials are preferentially mineralized, leaving intrinsically ‘stable’ decomposition
products behind. Progressive decomposition reflects the concept of microbial processing of large
plant biopolymers to smaller molecules. In the proposed SCM, a continuum of organic fragments
is continuously processed by the decomposer community from large plant and animal residues
toward smaller molecular size. At the same time, greater oxidation of the organic materials
increases solubility in water as well as the opportunity for protection against further decomposition
through greater reactivity toward mineral surfaces and incorporation into aggregates. Dashed
arrow lines denote mainly abiotic transfer; solid lines denote mainly biotic transfer; thicker lines
indicate more rapid rates; larger boxes and ends of wedges illustrate greater pool sizes; all
differences are illustrative. All arrows represent processes that are a function of temperature,
moisture, and the biota present. Reprinted from Lehmann and Kleber (2015), with permission
122 A. Lubbe and T. Northen
but performed aqueous extraction of soils for one h at 4 °C to slow any metabolic
activity present. Another concern with aqueous extraction is enrichment of the
metabolite profile by intracellular metabolites. Osmotic shock can potentially lyze
microbial cells and cause leakage of metabolites (Gregorich et al. 2000). Swenson
et al. (2015b) compared aqueous soil extracts to samples extracted in 10 mM
K2SO4 and 10 mM NH4HCO3, and found no significant qualitative differences in
metabolites detected. It was concluded that water is a suitable extractant for soil
exometabolomics of DOM and that these extracts would be most representative of
the types of resources available for soil microbes.
In some cases, an experiment may require analysis of soil intracellular and
extracellular metabolites. In this case, cell lysis is an important and desirable step in
sample preparation. To access intracellular metabolites, a traditional approach used
in soil science involves chloroform fumigation of soil to lyze microbial cells,
followed by extraction (Brookes et al. 1985; Vance et al. 1987). Swenson et al.
(2015b) compared metabolite profiles of water extracts of fumigated and unfumi-
gated soil samples (Fig. 2). A significant increase in the number and abundance of
metabolites was observed, however, the fumigation technique requires long times
of exposure to chloroform vapors, which raise concerns about continued metabolic
activity or increased enzymatic degradation of metabolites (Warren 2013a, b). The
use of organic solvents which are able to lyze microbial cells is another way to
obtain soil extracts containing intracellular and extracellular metabolites. This was
demonstrated by Swenson et al. (2015b) who used hierarchical cluster analysis to
show similarity in metabolite patterns between fumigated soil extracts and organic
solvent extracts of unfumigated soil. Soil was also directly extracted with chloro-
form and K2SO4 solution (1:4, v/v) to obtain extracts containing intracellular
metabolites (Kakumanu et al. 2013). Rochfort et al. (2015) extracted freeze-dried,
finely ground soil with an 8:2 methanol-water solution by sonication for 10 min.
Since this method breaks up soil aggregates in addition to using organic solvents, it
is not surprising that it provided extracts with a wide coverage of metabolite classes,
derived from the intracellular and extracellular soil metabolite pools.
The major analytical methods used in the field of metabolomics are based on
Mass Spectrometry (MS) and Nuclear Magnetic Resonance (NMR) (Dettmer et al.
2007; Dunn et al. 2005; Forseth and Schroeder 2011). Each method has advantages
and disadvantages related to sensitivity, structural information, ease of quantitation,
breadth of metabolite coverage, and availability of structural databases for identi-
fication (Lenz and Wilson 2007). Although studies on the soil microbial exome-
tabolome are limited, a few recent examples demonstrate the utility of these
methods to this field.
Gas Chromatography coupled with Mass Spectrometry (GC-MS) has previously
been applied to targeted analyses of particular chemical classes of small soil
metabolites such as sugars or amino acids (Kakumanu et al. 2013). GC-MS is also
well suited for measuring a broad range of small metabolite classes, and has been
widely used in untargeted metabolomics studies in plants (Jenkins et al. 2004),
human biofluids (Garcia and Barbas 2011), and microbial biomass (Koek et al.
2006). Analysis of hydrophilic/polar metabolites by GC-MS requires derivatization
124 A. Lubbe and T. Northen
Fig. 2 a Workflow for soil DOM extraction: A Soil is sieved through 2 mm and fumigated with
chloroform for 24 h to access intracellular metabolites or left unfumigated for extracellular
metabolites. B Soil is extracted with the appropriate solution in a 1:4 ratio (2 g soil: 8 mL
extractant) on an orbital shaker for 1 h at 4 °C. C Extract is centrifuged to pellet soil and the
supernatant filtered through 0.45 lm filter discs. D Dried down and derivatized for GC-MS.
E Data are analyzed for metabolite identification. b Relative intensity of metabolites in extracts of
unfumigated and fumigated soil prepared with different extractants, and analyzed by GC-MS.
Reprinted from (Swenson et al. 2015b), with permission
5 Exometabolomics for Linking Soil Carbon Dynamics … 125
Fig. 3 Sorption of small metabolites from a soil bacterial lysate on an iron oxide mineral,
ferrihydrite. For each metabolite, the percent sorption (relative to the non-mineral control) is
displayed as mineral concentration increased from 0.5–32 mg. Metabolites were analyzed by
LC-MS. Putatively identified metabolites are indicated by parentheses. Reprinted from Swenson
et al. (2015a), with permission
in the acid mine drainage exometabolome were found to be secreted by the cells in
laboratory cultures. This suggested an important role in organic matter production by
E. mutabilis for consumption by other microbial strains in this ecosystem.
Baran et al. (2015) extended this approach by characterizing multiple isolates
from a biological soil crust (biocrust) community. The primary producer in this
community, the filamentous cyanobacterium Microcoleus vaginatus, was cultured
in the laboratory. Exo- and endo-metabolite profiling revealed that many metabo-
lites were released into the culture medium by this strain. Seven bacterial isolates,
representing diverse phyla from the biocrust environment, were cultured individ-
ually in different-rich media to characterize their substrate preferences. Only a small
proportion of metabolites detected in the media were taken up by any given strain,
and there was little overlap between the strains’ preferred substrates. Metabolite
130 A. Lubbe and T. Northen
Fig. 4 Comparison of levels of selected metabolites in the growth media following growth of
Synechococcus (full bars) against their levels in control media (open bars, n = 4), as determined
by LC-MS. The peak areas axis was scaled with a square root to improve the visualization of
smaller peaks. Statistically significant differences are indicated as “*” (p < 0.05), “**” (p < 0.01),
or “***” (p < 0.001). An arrow is shown next to the name of a metabolite if it was found to be
significantly consumed (←), released (!), or both consumed and released ($). Reprinted from
Baran et al. (2011), with permission
profiling of the biocrust soil water was also performed to link the observed patterns
from the isolates to the intact microbial community. Changes in metabolite profiles
at different times following wet up of desiccated biocrust showed patterns similar to
those observed in the individual isolate experiments. This study revealed the par-
ticular substrate preferences of sympatric isolates from a soil community, which
suggest that exometabolite niche partitioning may be an important driver in
5 Exometabolomics for Linking Soil Carbon Dynamics … 131
Stable isotope probing (SIP) techniques involves addition of a stable isotope enri-
ched substrate, and tracking its fate as it is transformed by the metabolism of
community members into labeled molecules/biomarkers (Dumont and Murrell
2005). Variations of SIP target different biomarkers that become labeled as a result
of growth on the labeled substrate. One approach targets microbial phospholipid
fatty acids (PLFAs). Since different microbial classes possess characteristic fatty
acids as part of their cell membranes, selective extraction and analysis of PLFA
patterns is an established approach for determining the composition of microbial
communities (Zelles 1999). In PLFA-SIP, tracking which is the characteristic
phospholipid fatty acids become labeled with the stable isotope yields information
about which groups of microbes were responsible for metabolizing the labeled
substrate. This approach has been used to identify groups of microorganisms per-
forming particular functions in soils based on labeling with substrates such as 13CH4
(Bull et al. 2000), 13C-acetate (Boschker et al. 1998), 13C- glucose, and -ribose
(Apostel et al. 2015). Uniformly labeled 13C-cellulose and 13C-lignin were substrates
in a PLFA-SIP study on the role of diverse microbial groups in plant polymer
degradation (Torres et al. 2014). Treonis et al. (2004) combined a 13CO2-labeling
experiment with PLFA analysis to identify microbes assimilating plant root
132 A. Lubbe and T. Northen
Labeling experiments with stable isotopes can also aid analysis of the highly
complex soil exometabolome. For example, in NMR-based studies, the low natural
abundance of the magnetic isotope of carbon (13C) results in low sensitivity of
detection in unlabeled systems. Signals are dramatically enhanced as metabolites
become labeled with 13C isotopes, thereby facilitating identification of metabolites
downstream of the labeled substrate (Schneider et al. 2003). In mass spectrometry
based methods coupled to chromatographic separations, labeled metabolites can be
detected at the same retention time as their unlabeled counterparts, with charac-
teristic shifts in mass spectral features corresponding to the number of incorporated
labeled isotopes (Rodgers et al. 2000). Computational methods have been devel-
oped for the quantitative detection of features derived from a particular labeled
compound, even when not all metabolites are identified (Hiller et al. 2010).
However, stable isotope labeling can greatly facilitate unambiguous assignment of
chemical formulas, and thereby identification of unknown features (Baran et al.
2010). Thus, SIP methods show great promise for reducing noise by highlighting
relevant metabolites and pathways, and identifying unknown metabolites in com-
plex datasets such as those generated in soil exometabolomics experiments.
Soil is a very heterogeneous matrix, in which biotic and abiotic factors combine to
create diverse microclimates. Studies on soil microbial communities are often
conducted on homogenized bulk soil samples, in which the spatial structure of soil
and soil microorganisms are disrupted (Becker et al. 2006). Yet, observations of
microbial communities at the micron scale have revealed defined spatial organi-
zation. For example, in dental plaque, a nine taxon microbial consortium was
observed to be radially arranged around cells of filamentous bacteria (Welch et al.
2016). Obligate aerobes were arranged around the periphery of the consortium,
anaerobes were found in the interior, and others were localized in ways suggestive
of their functional roles in the consortium. Such structured assemblages have been
observed in biofilms and consortia occurring in aquatic systems, on leaf and root
surfaces, and in pathogenic or commensal associations with humans (Almstrand
et al. 2013; Wessel et al. 2013). It is believed that the spatial arrangement of soil
microbial communities is a very important driver of microbial diversity in soil,
thereby also of microbial community functioning (Ettema and Wardle 2002).
Therefore, the next level of detail required to understand microbial soil commu-
nities is characterizing their functioning in space. To achieve this, experimental
methods are needed to observe specific community members and their activities in
relation to other community members in their native spatial arrangement.
134 A. Lubbe and T. Northen
(a)
P. stutzeri
RCH2
S. oneidensis
MR1
(b)
2.5 mm
m/z 575.5 576.5 523.5
Fig. 5 Mass spectrometry imaging of P. stutzeri RCH2 and S. oneidensis MR1 coculture.
a Optical image of coculture on solid medium. b Tricolor mass spectrometry image of coculture
with m/z corresponding to species-specific lipids (Katherine Louie, unpublished)
136 A. Lubbe and T. Northen
as a mixed biofilm of these strains. A range of peptides, lipids, and small molecules
were detected (Watrous et al. 2013). A REX-NIMS approach was used by Louie
et al. (2013) to identify ions localized to regions within and between bacterial
colonies cultured individually and in coculture on agar media. In a study on
methanotrophic microbial mats from sea shelf methane seeps, Time-of-Flight
Secondary Ion Mass Spectrometry (TOF-SIMS) was used to characterize microbial
lipid biomarkers (Thiel et al. 2007). Characteristic lipid classes were detected in
distinguished areas, which matched the presence of different microbial colonies as
determined by conventional microscopic techniques.
Mass Spectrometry Imaging techniques show great promise for characterization
of microbial communities in space, however, technical challenges limit their
application to complex microbial communities in soil environments. Benefits and
challenges of different imaging mass spectrometry techniques were comprehen-
sively reviewed by Watrous and Dorrestein (2011). While able to detect metabolites
with a wide mass range, the resolution of methods such as MALDI-IMS and
nanoDESI currently does not allow imaging down to the single-cell level.
Dynamic SIMS offers the best spatial resolution (sub-µm scale) but does not pro-
vide molecular information beyond elements and small atomic clusters. Another
challenge of imaging experiments targeting small metabolites lies in the sample
preparation. Methods for preparing thin soil sections as used in soil sciences typ-
ically involve fixing in formaldehyde, washing and impregnation in resin (Nunan
et al. 2001), which would not be suitable for IMS. Tissue samples (e.g. mammalian,
plant) are usually embedded in a substance such as OTC polymer, gelatin, or
agarose gel for stabilization during cryosectioning (Cornett et al. 2007; Lee et al.
2012). Even if such treatment would not cause delocalization of metabolites in soil
samples, the heterogeneous physical structure of soil may hamper cutting thin
sections for imaging. Most IMS experiments to date involve laboratory cultured
agar samples which can be analyzed fresh or mounted and dehydrated in prepa-
ration for imaging experiments (Traxler et al. 2013; Yang et al. 2012). Studies
where microbial consortia from a natural environment were used either involved
smears of soil on the sample target (Orphan et al. 2001), or cryosectioning of
well-structured communities such as microbial mats (Thiel et al. 2007) or sym-
bionts associated with other organisms (Lechene et al. 2007; Schoenian et al. 2011).
Given the great potential of these approaches, there is an urgent need for improved
sample preparation methods that will enable small metabolite imaging of soil
microbial communities in their natural spatial arrangement.
molecular methods and SIP approaches have improved understanding of the par-
ticular members of the soil community’s metabolic capabilities. What members of a
particular soil microbial community actually do, will depend on the substrates that
are available in their environment. Therefore, exometabolomics is a very promising
approach in that it provides direct evidence of the soil metabolites available to soil
microorganisms and how the available substrates are transformed by microbial
community metabolism.
This chapter reviewed the handful of reports where an exometabolomics
approach was applied to the study of intact soil microbial communities, or to
laboratory experiments focusing on a particular aspect of such complex systems.
Soil microbial communities are very complex, and soils are extremely heteroge-
neous matrices, so it is not surprising that there are many technical challenges that
remain to be resolved in this field. Care should be taken to use sample preparation
methods appropriate for the specific question being asked and analytical method
being used. There is no single analytical method that can detect the massive
diversity of metabolites across large dynamic ranges in an unbiased way. The
choice of analytical method will depend on the focus and needs of the study, and
combinations of complementary techniques may offer a more comprehensive
coverage of diverse chemical classes (Simpson et al. 2004; Werf et al. 2007). It is
important to keep in mind that the mineral composition and other factors may
confound analysis by preferentially sorbing certain metabolites (e.g. ferrihydrite
sorbing phosphate containing metabolites) making it challenging to compare soil
types.
As with any metabolomics workflow, soil community exometabolomics
experiments generate large datasets. Untargeted metabolomics results usually
include many detected features that remain unidentified. There are several
well-established mass spectrometry and NMR databases that can aid in identifica-
tion of such unknowns (Kind et al. 2009; Smith et al. 2005; Wishart 2007). Many of
these target intracellular metabolism of organisms such as yeast or plants (Bais et al.
2010; Hummel et al. 2007; Jewison et al. 2011). Since much soil organic matter is
derived from plant and microbial biomass, these are useful to soil organic matter
characterizations. There is currently great interest in secondary metabolites from
soil microorganisms, and increasing the number of entries in databases of such
compounds will also be very helpful in the context of soil exometabolomics
(Hadjithomas et al. 2015). Many workflows have been developed for the analysis of
large metabolomics datasets, which are also applicable to exometabolomics data
analysis (Bowen and Northen 2010; Rübel et al. 2013; Tautenhahn et al. 2012; Xia
et al. 2012). Data analysis tools for interpreting data in SIP experiments will be of
particular value (Hiller et al. 2010; Huang et al. 2014). Any experimental setup and
data reported should meet the quality and reporting standards as set by the larger
metabolomics community (Goodacre et al. 2007).
The full potential of soil microbial community exometabolomics will be realized
when it can be integrated with other approaches such as metagenomics, meta-
transcriptomics, and metaproteomics. A recent review describes examples where
such multiomics approaches were applied to understanding microbial communities
138 A. Lubbe and T. Northen
(Franzosa et al. 2015). Careful planning of experimental design and data integration
strategies are needed to derive the most value out of such combined approaches
(Muller et al. 2013). Such data integration should result in improved mechanistic
models of the structure and functioning of soil microbial communities that can be
tested in combinations of laboratory and field experiments (Franzosa et al. 2015).
This will enable better predictions of the effects of environmental perturbations on
soil carbon cycling by soil microorganisms.
References
Baran R, Bowen BP, Northen TR (2011) Untargeted metabolic footprinting reveals a surprising
breadth of metabolite uptake and release by Synechococcus sp. PCC 7002. Mol BioSyst
7:3200. doi:10.1039/c1mb05196b
Baran R, Bowen BP, Bouskill NJ, Brodie EL, Yannone SM, Northen TR (2010) Metabolite
identification in synechococcus sp. PCC 7002 using untargeted stable isotope assisted
metabolite profiling. Anal Chem 82:9034–9042. doi:10.1021/ac1020112
Bardgett RD, Freeman C, Ostle NJ (2008) Microbial contributions to climate change through
carbon cycle feedbacks. ISME J 2:805–814. doi:10.1038/ismej.2008.58
Beale DJ, Barratt R, Marlow DR, Dunn MS, Palombo EA, Mor-rison PD, Key C (2013)
Application of metabolomics to understanding biofilms in water distribution systems: a pilot
study. Biofouling 29:283–294. doi:10.1080/08927014.2013.772140
Beale DJ, Dunn MS, Marney D (2010) Application of GC–MS metabolic profiling to “blue-green
water” from microbial influenced corrosion in copper pipes. Corros Sci 52:3140–3145. doi:10.
1016/j.corsci.2010.04.039
Beale DJ, Dunn MS, Morrison PD, Porter NA, Marlow DR (2012) Characterisation of bulk water
samples from copper pipes undergoing microbially influenced corrosion by diagnostic
metabolomic pro-filing. Corros Sci 55:272–279. doi:10.1016/j.corsci.2011.10.026
Becker JM, Parkin T, Nakatsu CH, Wilbur JD, Konopka A (2006) Bacterial activity, community
structure, and centimeter-scale spatial heterogeneity in contaminated soil. Microb Ecol 51:220–
231. doi:10.1007/s00248-005-0002-9
Behrends V, Williams H, Bundy J (2014) Metabolic footprinting: extracellular metabolomic
analysis. In: Filloux A, Ramos J-L (eds) Pseudomonas methods and protocols. Methods in
molecular biology. Springer, New York, pp 281–292
Behrens S, Lösekann T, Pett-Ridge J, Weber PK, Ng WO, Stevenson BS, Hutcheon ID,
Relman DA, Spormann AM (2008) Linking microbial phylogeny to metabolic activity at the
single-cell level by using enhanced element labeling-catalyzed reporter deposition fluorescence
in situ hybridization (El-Fish) and nanosims. Appl Environ Microbiol 74:3143–3150. doi:10.
1128/AEM.00191-08
Bellon-Maurel V, McBratney A (2011) Near-infrared (NIR) and mid-infrared (MIR) spectroscopic
techniques for assessing the amount of carbon stock in soils–critical review and research
perspectives. Soil Biol Biochem 43:1398–1410. doi:10.1016/j.soilbio.2011.02.019
Boschker HTS, Nold SC, Wellsbury P, Bos D, de Graaf W, Pel R, Parkes RJ, Cappenberg TE
(1998) Direct linking of microbial populations to specific biogeochemical processes by
13C-labelling of biomarkers. Nature 392:801–805. doi:10.1038/33900
Bowen BP, Northen TR (2010) Dealing with the unknown: metabolomics and metabolite atlases.
J Am Soc Mass Spectrom 21:1471–1476. doi:10.1016/j.jasms.2010.04.003
Brookes PC, Landman A, Pruden G, Jenkinson DS (1985) Chloroform fumigation and the release
of soil nitrogen: a rapid direct extraction method to measure microbial biomass nitrogen in soil.
Soil Biol Biochem 17:837–842. doi:10.1016/0038-0717(85)90144-0
Bull ID, Parekh NR, Hall GH, Ineson P, Evershed RP (2000) Detection and classification of
atmospheric methane oxidizing bacteria in soil. Nature 405:175–178. doi:10.1038/35012061
Castrillo JI, Zeef LA, Hoyle DC, Zhang N, Hayes A, Gardner DC, Cornell MJ, Petty J, Hakes L,
Wardleworth L, Rash B, Brown M, Dunn WB, Broadhurst D, O’Donoghue K, Hester SS,
Dunkley TP, Hart SR, Swainston N, Li P, Gaskell SJ, Pa-ton NW, Lilley KS, Kell DB,
Oliver SG (2007) Growth control of the eukaryote cell: a systems biology study in yeast. J Biol
6:4. doi:10.1186/jbiol54
Chandra S, Pumphrey G, Abraham JM, Madsen EL (2008) Dynamic SIMS ion microscopy
imaging of individual bacterial cells for studies of isotopically labeled molecules. Appl Surf Sci
255:847–851. doi:10.1016/j.apsusc.2008.05.129 (Proceedings of the Sixteenth International
Conference on Secondary Ion Mass Spectrometry, SIMS XVI)
Cliff JB, Gaspar DJ, Bottomley PJ, Myrold DD (2002) Exploration of inorganic C and N
assimilation by soil microbes with time-of-flight secondary ion mass spectrometry. Appl
Environ Microbiol 68:4067–4073. doi:10.1128/AEM.68.8.4067-4073.2002
140 A. Lubbe and T. Northen
Cornett DS, Reyzer ML, Chaurand P, Caprioli RM (2007) MALDI imaging mass spectrometry:
molecular snapshots of biochemical systems. Nat Methods 4:828–833. doi:10.1038/nmeth1094
Corvasce M, Zsolnay A, D’Orazio V, Lopez R, Miano TM (2006) Characterization of water
extractable organic matter in a deep soil pro-file. Chemosphere 62:1583–1590. doi:10.1016/j.
chemosphere.2005.07.065
Date Y, Nakanishi Y, Fukuda S, Kato T, Tsuneda S, Ohno H, Kikuchi J (2010) New monitoring
approach for metabolic dynamics in microbial ecosystems using stable-isotope-labeling
technologies. J Biosci Bioeng 110:87–93. doi:10.1016/j.jbiosc.2010.01.004
Delmont TO, Robe P, Cecillon S, Clark IM, Constancias F, Simonet P, Hirsch PR, Vogel TM
(2011) Accessing the soil metagenome for studies of microbial diversity. Appl Environ
Microbiol 77:1315–1324. doi:10.1128/AEM.01526-10
DeRito CM, Pumphrey GM, Madsen EL (2005) Use of field-based stable isotope probing to
identify adapted populations and track carbon flow through a phenol-degrading soil microbial
community. Appl Environ Microbiol 71:7858–7865. doi:10.1128/AEM.71.12.7858-7865.2005
Dettmer K, Aronov PA, Hammock BD (2007) Mass spectrometry-based metabolomics. Mass
Spectrom Rev 26:51–78. doi:10.1002/mas.20108
Devantier R, Scheithauer B, Villas-Bôas SG, Pedersen S, Olsson L (2005) Metabolite profiling for
analysis of yeast stress response during very high gravity ethanol fermentations. Biotechnol
Bioeng 90:703–714. doi:10.1002/bit.20457
Dumont MG, Murrell JC (2005) Stable isotope probing—linking microbial identity to function.
Nat Rev Microbiol 3:499–504. doi:10.1038/nrmicro1162
Dunn WB, Bailey NJC, Johnson HE (2005) Measuring the metabolome: current analytical
technologies. Analyst 130:606–625. doi:10.1039/B418288J
El Zahar Haichar F, Achouak W, Christen R, Heulin T, Marol C, Marais MF, Mougel C,
Ranjard L, Balesdent J, Berge O (2007) Identification of cellulolytic bacteria in soil by stable
isotope probing. Environ Microbiol 9:625–634. doi:10.1111/j.1462-2920.2006.01182.x
Ettema CH, Wardle DA (2002) Spatial soil ecology. Trends Ecol Evol 17:177–183. doi:10.1016/
S0169-5347(02)02496-5
Falkowski PG, Fenchel T, Delong EF (2008) The microbial engines that drive earth’s
biogeochemical cycles. Science 320:1034–1039. doi:10.1126/science.1153213
Fierer N, Leff JW, Adams BJ, Nielsen UN, Bates ST, Lauber CL, Owens S, Gilbert JA, Wall DH,
Caporaso JG (2012) Cross-biome metagenomic analyses of soil microbial communities and
their functional attributes. Proc Natl Acad Sci USA 109:21390–21395. doi:10.1073/pnas.
1215210110
Forseth RR, Schroeder FC (2011) NMR-spectroscopic analysis of mixtures: from structure to
function. Curr Opin Chem Biol 15:38–47. doi:10.1016/j.cbpa.2010.10.010
Franzosa EA, Hsu T, Sirota-Madi A, Shafquat A, Abu-Ali G, Morgan XC, Huttenhower C (2015)
Sequencing and beyond: integrating molecular “omics” for microbial community profiling. Nat
Rev Microbiol 13:360–372. doi:10.1038/nrmicro3451
Fu Z, Verderame TD, Leighton JM, Sampey BP, Appelbaum ER, Patel PS, Aon JC (2014)
Exometabolome analysis reveals hypoxia at the up-scaling of a Saccharomyces cerevisiae
high-cell density fed-batch biopharmaceutical process. Microb Cell Factories 13:32. doi:10.
1186/1475-2859-13-32
Garcia A, Barbas C (2011) Gas chromatography-mass spectrometry (GC-MS)-based metabo-
lomics. In: Metz TO (ed) Metabolic profiling, methods in molecular biology. Humana Press,
New York, pp 191–204
Goodacre R, Broadhurst D, Smilde AK, Kristal BS, Baker JD, Beger R, Bessant C, Connor S,
Capuani G, Craig A, Ebbels T, Kell DB, Manetti C, Newton J, Paternostro G, Somorjai R,
Sjöström M, Trygg J, Wulfert F (2007) Proposed minimum reporting standards for data
analysis in metabolomics. Metabolomics 3:231–241. doi:10.1007/s11306-007-0081-3
Gregorich EG, Liang BC, Drury CF, Mackenzie AF, McGill WB (2000) Elucidation of the source
and turnover of water soluble and microbial biomass carbon in agricultural soils. Soil Biol
Biochem 32:581–587. doi:10.1016/S0038-0717(99)00146-7
5 Exometabolomics for Linking Soil Carbon Dynamics … 141
Grivet JP, Delort AM, Portais JC (2003) NMR and microbiology: from physiology to
metabolomics. Biochimie Metabolic NMR 85:823–840. doi:10.1016/j.biochi.2003.08.004
Hadjithomas M, Chen IMA, Chu K, Ratner A, Palaniappan K, Szeto E, Huang J, Reddy TBK,
Cimermančič P, Fischbach MA, Ivanova NN, Markowitz VM, Kyrpides NC, Pati A (2015)
IMG-ABC: a knowledge base to fuel discovery of biosynthetic gene clusters and novel
secondary metabolites. mBio 6:e00932-15. doi:10.1128/mBio.00932-15
Halter D, Goulhen-Chollet F, Gallien S, Casiot C, Hamelin J, Gi-lard F, Heintz D, Schaeffer C,
Carapito C, Van Dorsselaer A, Tcherkez G, Arsène-Ploetze F, Bertin PN (2012) In situ
proteo-metabolomics reveals metabolite secretion by the acid mine drainage bio-indicator.
Euglena mutabilis. ISME J. 6:1391–1402. doi:10.1038/ismej.2011.198
Harrigan GG, Goodacre R (eds) (2003) Metabolic profiling: its role in biomarker discovery and
gene function analysis. Springer, Boston
Henriques IDS, Aga DS, Mendes P, O’Connor SK, Love NG (2007) Metabolic footprinting: a new
approach to identify physiological changes in complex microbial communities upon exposure
to toxic chemicals. Environ Sci Technol 41:3945–3951. doi:10.1021/es062796t
Hiller K, Metallo CM, Kelleher JK, Stephanopoulos G (2010) Non-targeted elucidation of
metabolic pathways using stable-isotope tracers and mass spectrometry. Anal Chem 82:6621–
6628. doi:10.1021/ac1011574
Hockaday WC, Grannas AM, Kim S, Hatcher PG (2006) Direct molecular evidence for the
degradation and mobility of black carbon in soils from ultrahigh-resolution mass spectral
analysis of dissolved organic matter from a fire-impacted forest soil. Org Geochem 37:501–
510. doi:10.1016/j.orggeochem.2005.11.003
Huang X, Chen Y-J, Cho K, Nikolskiy I, Crawford PA, Patti GJ (2014) X13CMS: Global tracking
of isotopic labels in untargeted metabolomics. Anal Chem 86:1632–1639. doi:10.1021/
ac403384n
Hummel J, Selbig J, Walther D, Kopka J (2007) The golm metabolome database: a database for
GC-MS based metabolite profiling. In: Nielsen J, Jewett MC (eds) Metabolomics, topics in
current genetics. Springer, Berlin, pp 75–95
Jenkins H, Hardy N, Beckmann M, Draper J, Smith AR, Taylor J, Fiehn O, Goodacre R, Bino RJ,
Hall R, Kopka J, Lane GA, Lange BM, Liu JR, Mendes P, Nikolau BJ, Oliver SG, Paton NW,
Rhee S, Roessner-Tunali U, Saito K, Smedsgaard J, Sumner LW, Wang T, Walsh S,
Wurtele ES, Kell DB (2004) A proposed framework for the description of plant metabolomics
experiments and their results. Nat Biotechnol 22:1601–1606. doi:10.1038/nbt1041
Jenkinson DS, Rayner JH (1977) The turnover of soil organic matter in some of the Rothamsted
classical experiments. Soil Sci 123:298–305
Jewison T, Knox C, Neveu V, Djoumbou Y, Guo AC, Lee J, Liu P, Mandal R, Krishnamurthy R,
Sinelnikov I, Wilson M, Wishart DS (2011) YMDB: the Yeast Metabolome Database. Nucleic
Acids Res. doi:10.1093/nar/gkr916
Johnston CA, Groffman P, Breshears DD, Cardon ZG, Currie W, Emanuel W, Gaudinski J,
Jackson RB, Lajtha K, Nadelhoffer K, Nelson D, Post WM, Retallack G, Wielopolski L (2004)
Carbon cycling in soil. Front Ecol Environ 2:522–528. doi:10.2307/3868382
Jones OAH, Sdepanian S, Lofts S, Svendsen C, Spurgeon DJ, Maguire ML, Griffin JL (2014)
Metabolomic analysis of soil communities can be used for pollution assessment. Environ
Toxicol Chem 33:61–64. doi:10.1002/etc.2418
Jones DL, Willett VB (2006) Experimental evaluation of methods to quantify dissolved organic
nitrogen (DON) and dissolved organic carbon (DOC) in soil. Soil Biol Biochem 38:991–999.
doi:10.1016/j.soilbio.2005.08.012
Kakumanu ML, Cantrell CL, Williams MA (2013) Microbial community response to varying
magnitudes of desiccation in soil: a test of the osmolyte accumulation hypothesis. Soil Biol
Biochem 57:644–653. doi:10.1016/j.soilbio.2012.08.014
Kalbitz K, Solinger S, Park JH, Michalzik B, Matzner E (2000) Controls on the dynamics of
dissolved organic matter in soils: a review. Soil Sci 165:277–304
142 A. Lubbe and T. Northen
Kell DB, Brown M, Davey HM, Dunn WB, Spasic I, Oliver SG (2005) Metabolic footprinting and
systems biology: the medium is the message. Nat Rev Microbiol 3:557–565. doi:10.1038/
nrmicro1177
Kelly AE, Ou HD, Withers R, Dötsch V (2002) Low-conductivity buffers for high-sensitivity
NMR measurements. J Am Chem Soc 124:12013–12019. doi:10.1021/ja026121b
Kido Soule MC, Longnecker K, Johnson WM, Kujawinski EB (2015) Environmental
metabolomics: analytical strategies. Mar Chem 177:374–387. doi:10.1016/j.marchem.2015.
06.029
Kim HK, Choi YH, Verpoorte R (2010) NMR-based metabolomic analysis of plants. Nat Protoc
5:536–549. doi:10.1038/nprot.2009.237
Kind T, Wohlgemuth G, Lee DY, Lu Y, Palazoglu M, Shahbaz S, Fiehn O (2009) FiehnLib: mass
spectral and retention index li-braries for metabolomics based on quadrupole and time-of-flight
gas chromatography/mass spectrometry. Anal Chem 81:10038–10048. doi:10.1021/ac9019522
Koek MM, Muilwijk B, van der Werf MJ, Hankemeier T (2006) Microbial metabolomics with gas
chromatography/mass spectrometry. Anal Chem 78:1272–1281. doi:10.1021/ac051683
Kögel-Knabner I (1997) 13C and 15N NMR spectroscopy as a tool in soil organic matter studies.
Geoderma 80:243–270. doi:10.1016/S0016-7061(97)00055-4
Kujawinski EB (2002) Electrospray ionization fourier transform ion cyclotron resonance mass
spectrometry (ESI FT-ICR MS): characterization of complex environmental mixtures. Environ
Forensics 3:207–216. doi:10.1080/713848382
Kujawinski EB, Del Vecchio R, Blough NV, Klein GC, Marshall AG (2004) Probing
molecular-level transformations of dissolved organic matter: insights on photochemical
degradation and protozoan modification of DOM from electrospray ionization Fourier
transform ion cyclotron resonance mass spectrometry. Mar Chem 92:23–37. doi:10.1016/j.
marchem.2004.06.038 (New Approaches in Marine Organic Biogeochemistry: A Tribute to the
Life and Science of John I Hedges)
Lal R (2004) Soil carbon sequestration impacts on global climate change and food security.
Science 304:1623–1627. doi:10.1126/science.1097396
Lechene CP, Luyten Y, McMahon G, Distel DL (2007) Quantitative imaging of nitrogen fixation
by individual bacteria within animal cells. Science 317:1563–1566. doi:10.1126/science.
1145557
Leenheer JA, Croué JP (2003) Characterizing aquatic dissolved organic matter. Environ Sci
Technol 37:18A–26A
Lee N, Nielsen PH, Andreasen KH, Juretschko S, Nielsen JL, Schleifer K-H, Wagner M (1999)
Combination of fluorescent in situ hybridization and microautoradiography—a new tool for
structure-function analyses in microbial ecology. Appl Environ Micro-biol 65:1289–1297
Lee YJ, Perdian DC, Song Z, Yeung ES, Nikolau BJ (2012) Use of mass spectrometry for imaging
metabolites in plants. Plant J 70:81–95. doi:10.1111/j.1365-313X.2012.04899.x
Lehmann J, Kleber M (2015) The contentious nature of soil organic matter. Nature 528:60–68.
doi:10.1038/nature16069
Lenz EM, Wilson ID (2007) Analytical strategies in metabonomics. J Proteome Res 6:443–458.
doi:10.1021/pr0605217
Louie KB, Bowen BP, Cheng X, Berleman JE, Chakraborty R, Deutschbauer A, Arkin A,
Northen TR (2013) “Replica-extraction-transfer” nanostructure-initiator mass spectrometry
imaging of acoustically printed bacteria. Anal Chem 85:10856–10862. doi:10.1021/ac402240q
Manefield M, Whiteley AS, Griffiths RI, Bailey MJ (2002) RNA stable isotope probing, a novel
means of linking microbial community function to phylogeny. Appl Environ Microbiol
68:5367–5373. doi:10.1128/AEM.68.11.5367-5373.2002
Mas S, Villas-Bôas SG, Edberg Hansen M, Åkesson M, Nielsen J (2007) A comparison of direct
infusion MS and GC-MS for metabolic footprinting of yeast mutants. Biotechnol Bioeng
96:1014–1022. doi:10.1002/bit.21194
Morales-Cid G, Gebefugi I, Kanawati B, Harir M, Hertkorn N, Rosselló-Mora R, Schmitt-Kopplin
P (2009) Automated microextraction sample preparation coupled on-line to FT-ICR-MS:
5 Exometabolomics for Linking Soil Carbon Dynamics … 143
application to desalting and concentration of river and marine dissolved organic matter. Anal
Bioanal Chem 395:797–807. doi:10.1007/s00216-009-3025-0
Morales SE, Holben WE (2011) Linking bacterial identities and eco-system processes: can “omic”
analyses be more than the sum of their parts? FEMS Microbiol Ecol 75:2–16. doi:10.1111/j.
1574-6941.2010.00938.x
Muller EEL, Glaab E, May P, Vlassis N, Wilmes P (2013) Con-densing the omics fog of microbial
communities. Trends Microbiol 21:325–333. doi:10.1016/j.tim.2013.04.009
Nicholson JK, Lindon JC (2008) Systems biology: metabonomics. Nature 455:1054–1056. doi:10.
1038/4551054a
Nunan N, Ritz K, Crabb D, Harris K, Wu K, Crawford JW, Young IM (2001) Quantification of the
in situ distribution of soil bacteria by large-scale imaging of thin sections of undisturbed soil.
FEMS Microbiol Ecol 37:67–77. doi:10.1111/j.1574-6941.2001.tb00854.x
Ohno T, Parr TB, Gruselle MI, Fernandez IJ, Sleighter RL, Hatcher PG (2014) Molecular
composition and biodegradability of soil organic matter: a case study comparing two new
england forest types. Environ Sci Technol 48:7229–7236. doi:10.1021/es405570c
Oliver SG, Winson MK, Kell DB, Baganz F (1998) Systematic functional analysis of the yeast
genome. Trends Biotechnol 16:373–378. doi:10.1016/S0167-7799(98)01214-1
Orphan VJ, House CH, Hinrichs KU, McKeegan KD, DeLong EF (2001) Methane-consuming
archaea revealed by directly coupled isotopic and phylogenetic analysis. Science 293:484–487.
doi:10.1126/science.1061338
Paczia N, Nilgen A, Lehmann T, Gätgens J, Wiechert W, Noack S (2012) Extensive
exometabolome analysis reveals extended overflow metabolism in various microorganisms.
Microb Cell Factories 11:122. doi:10.1186/1475-2859-11-122
Padmanabhan P, Padmanabhan S, DeRito C, Gray A, Gannon D, Snape JR, Tsai CS, Park W,
Jeon C, Madsen EL (2003) Respiration of 13C-labeled substrates added to soil in the field and
subsequent 16S rRNA gene analysis of 13C-labeled soil DNA. Appl Environ Microbiol
69:1614–1622
Piccolo A (2002) The supramolecular structure of humic substances: a novel understanding of
humus chemistry and implications in soil science. Adv Agronomy 75:57–134
Radajewski S, Ineson P, Parekh NR, Murrell JC (2000) Stable-isotope probing as a tool in
microbial ecology. Nature 403:646–649. doi:10.1038/35001054
Ramautar R, Mayboroda OA, Somsen GW, de Jong GJ (2011) CE-MS for metabolomics:
developments and applications in the period 2008–2010. Electrophoresis 32:52–65. doi:10.
1002/elps.201000378
Ramautar R, Somsen GW, de Jong GJ (2013) CE-MS for metabolomics: developments and
applications in the period 2010-2012. Electrophoresis 34:86–98. doi:10.1002/elps.201200390
Ramautar R, Somsen GW, de Jong GJ (2009) CE-MS in metabolomics. Electrophoresis 30:276–
291. doi:10.1002/elps.200800512
Resmer KL, White RL (2011) Metabolic footprinting of the anaerobic bacterium Fusobacterium
varium using 1H NMR spectroscopy. Mol Bio Syst 7:2220. doi:10.1039/c1mb05105a
Rettedal EA, Brözel VS (2015) Characterizing the diversity of active bacteria in soil by
comprehensive stable isotope probing of DNA and RNA with H218O. Microbiology Open
4:208–219. doi:10.1002/mbo3.230
Rochfort S, Ezernieks V, Mele P, Kitching M (2015) NMR metabolomics for soil analysis provide
complementary, orthogonal data to MIR and traditional soil chemistry approaches—a land use
study. Magn Reson Chem 53:719–725. doi:10.1002/mrc.4187
Rodgers RP, Blumer EN, Hendrickson CL, Marshall AG (2000) Stable isotope incorporation
triples the upper mass limit for determination of elemental composition by accurate mass
measurement. J Am Soc Mass Spectrom 11:835–840. doi:10.1016/S1044-0305(00)00158-6
Roesch LF, Fulthorpe RR, Riva A, Casella G, Hadwin AK, Kent AD, Daroub SH, Camargo FA,
Farmerie WG, Triplett EW (2007) Pyrosequencing enumerates and contrasts soil microbial
diversity. ISME J 1:283–290. doi:10.1038/ismej.2007.53
144 A. Lubbe and T. Northen
Rousk J, Jones DL (2010) Loss of low molecular weight dissolved organic carbon (DOC) and
nitrogen (DON) in H2O and 0.5 M K2SO4 soil extracts. Soil Biol Biochem 42:2331–2335.
doi:10.1016/j.soilbio.2010.08.017
Rübel O, Greiner A, Cholia S, Louie K, Bethel EW, Northen TR, Bowen BP (2013) OpenMSI: a
high-performance web-based platform for mass spectrometry imaging. Anal Chem 85:10354–
10361. doi:10.1021/ac402540a
Schloss PD, Handelsman J (2003) Biotechnological prospects from metagenomics. Curr Opin
Biotechnol 14:303–310. doi:10.1016/S0958-1669(03)00067-3
Schmidt MWI, Torn MS, Abiven S, Dittmar T, Guggenberger G, Janssens IA, Kleber M,
Kögel-Knabner I, Lehmann J, Manning DAC, Nannipieri P, Rasse DP, Weiner S,
Trumbore SE (2011) Persistence of soil organic matter as an ecosystem property. Nature
478:49–56. doi:10.1038/nature10386
Schneider B, Gershenzon J, Graser G, Hölscher D, Schmitt B (2003) One-dimensional 13C NMR
and HPLC-1H NMR techniques for observing carbon-13 and deuterium labelling in
biosynthetic studies. Phytochem Rev 2:31–43. doi:10.1023/B:PHYT.0000004196.73829.4e
Schoenian I, Spiteller M, Ghaste M, Wirth R, Herz H, Spiteller D (2011) Chemical basis of the
synergism and antagonism in microbial communities in the nests of leaf-cutting ants. Proc Natl
Acad Sci USA 108:1955–1960. doi:10.1073/pnas.1008441108
Sévin DC, Kuehne A, Zamboni N, Sauer U (2015) Biological in-sights through nontargeted
metabolomics. Curr Opin Biotechnol 34:1–8. doi:10.1016/j.copbio.2014.10.001
Silva LP, Northen TR (2015) Exometabolomics and MSI: deconstructing how cells interact to
transform their small molecule environment. Curr Opin Biotechnol 34:209–216. doi:10.1016/j.
copbio.2015.03.015
Simpson AJ, Tseng L-H, Simpson MJ, Spraul M, Braumann U, Kingery WL, Kelleher BP,
Hayes MHB (2004) The application of LC-NMR and LC-SPE-NMR to compositional studies
of natural organic matter. Analyst 129:1216. doi:10.1039/b408064e
Smith CA, Maille GO, Want EJ, Qin C, Trauger SA, Brandon TR, Custodio DE, Abagyan R,
Siuzdak G (2005) METLIN: a metabolite mass spectral database. Ther Drug Monit 27
Stevenson FJ (1994) Humus chemistry: genesis, composition, reactions. Wiley, New York
Swenson TL, Bowen BP, Nico PS, Northen TR (2015a) Competitive sorption of microbial
metabolites on an iron oxide mineral. Soil Biol Biochem 90:34–41. doi:10.1016/j.soilbio.2015.
07.022
Swenson TL, Jenkins S, Bowen BP, Northen TR (2015b) Untargeted soil metabolomics methods
for analysis of extractable organic matter. Soil Biol Biochem 80:189–198. doi:10.1016/j.
soilbio.2014.10.007
Szeto SSW, Reinke SN, Sykes BD, Lemire BD (2010) Mutations in the saccharomyces cerevisiae
succinate dehydrogenase result in distinct metabolic phenotypes revealed through 1H
NMR-based metabolic footprinting. J Proteome Res 9:6729–6739. doi:10.1021/pr100880y
Tautenhahn R, Patti GJ, Rinehart D, Siuzdak G (2012) XCMS online: a web-based platform to
process untargeted metabolomic data. Anal Chem 84:5035–5039. doi:10.1021/ac300698c
Theodoridis G, Gika HG, Wilson ID (2008) LC-MS-based methodology for global metabolite
profiling in metabonomics/metabolomics. TrAC Trends Anal Chem 27:251–260. doi:10.1016/
j.trac.2008.01.008
Thiel V, Heim C, Arp G, Hahmann U, Sjövall P, Lausmaa J (2007) Biomarkers at the microscopic
range: ToF-SIMS molecular imaging of Archaea-derived lipids in a microbial mat. Geobiology
5:413–421. doi:10.1111/j.1472-4669.2007.00119.x
Torres IF, Bastida F, Hernández T, Bombach P, Richnow HH, García C (2014) The role of lignin
and cellulose in the carbon-cycling of degraded soils under semiarid climate and their relation
to microbial biomass. Soil Biol Biochem 75:152–160. doi:10.1016/j.soilbio.2014.04.007
Traxler MF, Watrous JD, Alexandrov T, Dorrestein PC, Kolter R (2013) Interspecies interactions
stimulate diversification of the streptomyces coelicolor secreted metabolome. mBio 4:
e00459-13. doi:10.1128/mBio.00459-13
5 Exometabolomics for Linking Soil Carbon Dynamics … 145
Treonis AM, Ostle NJ, Stott AW, Primrose R, Grayston SJ, Ineson P (2004) Identification of
groups of metabolically-active rhizosphere microorganisms by stable isotope probing of
PLFAs. Soil Biol Biochem 36:533–537. doi:10.1016/j.soilbio.2003.10.015
Vance ED, Brookes PC, Jenkinson DS (1987) An extraction method for measuring soil microbial
biomass C. Soil Biol Biochem 19:703–707. doi:10.1016/0038-0717(87)90052-6
Villas-Bôas SG, Noel S, Lane GA, Attwood G, Cookson A (2006) Extracellular metabolomics: a
metabolic footprinting approach to assess fiber degradation in complex media. Anal Biochem
349:297–305. doi:10.1016/j.ab.2005.11.019
Warren CR (2014) Response of osmolytes in soil to drying and re-wetting. Soil Biol Biochem
70:22–32. doi:10.1016/j.soilbio.2013.12.008
Warren CR (2013a) High diversity of small organic N observed in soil water. Soil Biol Biochem
57:444–450. doi:10.1016/j.soilbio.2012.09.025
Warren CR (2013b) Quaternary ammonium compounds can be abundant in some soils and are
taken up as intact molecules by plants. New Phytol 198:476–485. doi:10.1111/nph.12171
Watrous JD, Dorrestein PC (2011) Imaging mass spectrometry in microbiology. Nat Rev
Microbiol 9:683–694. doi:10.1038/nrmicro2634
Watrous J, Roach P, Heath B, Alexandrov T, Laskin J, Dorrestein PC (2013) Metabolic profiling
directly from the Petri dish using nanospray desorption electrospray ionization imaging mass
spectrometry. Anal Chem 85:10385–10391. doi:10.1021/ac4023154
Weiss MS, Abele U, Weckesser J, Welte W, Schiltz E, Schulz GE (1991) Molecular architecture
and electrostatic properties of a bacterial porin. Science 254:1627–1630. doi:10.1126/science.
1721242
Welch JLM, Rossetti BJ, Rieken CW, Dewhirst FE, Borisy GG (2016) Biogeography of a human
oral microbiome at the micron scale. Proc Natl Acad Sci USA 113:E791–E800. doi:10.1073/
pnas.1522149113
van der Werf MJ, Overkamp KM, Muilwijk B, Coulier L, Hankemeier T (2007) Microbial
metabolomics: toward a platform with full metabolome coverage. Anal Biochem 370:17–25.
doi:10.1016/j.ab.2007.07.022
Wessel AK, Hmelo L, Parsek MR, Whiteley M (2013) Going local: technologies for exploring
bacterial microenvironments. Nat Rev Microbiol 11:337–348. doi:10.1038/nrmicro3010
Wishart DS (2007) Current Progress in computational metabolomics. Brief Bioinform 8:279–293.
doi:10.1093/bib/bbm030
Woo H-K, Northen TR, Yanes O, Siuzdak G (2008) Nanostructure-initiator mass spectrometry: a
protocol for preparing and applying NIMS surfaces for high-sensitivity mass analysis. Nat
Protoc 3:1341–1349. doi:10.1038/nprot.2008.110
Xia J, Mandal R, Sinelnikov IV, Broadhurst D, Wishart DS (2012) MetaboAnalyst 2.0—a
comprehensive server for metabolomic data analysis. Nucleic Acids Res 40:W127–W133.
doi:10.1093/nar/gks374
Yang JY, Phelan VV, Simkovsky R, Watrous JD, Trial RM, Fleming TC, Wenter R, Moore BS,
Golden SS, Pogliano K, Dorrestein PC (2012) Primer on agar-based microbial imaging mass
spectrometry. J Bacteriol 194:6023–6028. doi:10.1128/JB.00823-12
Zelles L (1999) Fatty acid patterns of phospholipids and lipopolysaccharides in the character-
isation of microbial communities in soil: a review. Biol Fertil Soils 29:111–129. doi:10.1007/
s003740050533
Zhou B, Xiao JF, Tuli L, Ressom HW (2012) LC-MS-based metabolomics. Mol Bio Syst 8:470–
481. doi:10.1039/c1mb05350g
Zsolnay Á (2003) Dissolved organic matter: artefacts, definitions, and functions. Geoderma,
Ecological aspects of dissolved organic matter in soils 113:187–209. doi:10.1016/S0016-7061
(02)00361-0
Chapter 6
Soil Microbial Metabolomics
1 Soil Complexity
The world demand for food is currently expected to increase by two- to fivefold by
2030 (Ewert et al. 2005; Food and Agriculture Organization (FAO) 2002). This
projection requires food production to increase from 60 to 70 % (Clair and Lynch
2010), although there is some confusion due to the types of food stuffs the global
population will be consuming at that time (Alexandratos and Bruinsma 2013) and
the exclusion from earlier studies of important food types (e.g. fruit and vegetables).
Agricultural practices over the last century have succeeded in significantly
increasing crop yields. For instance, global cereal production doubled between
1960 and 2000 (Tilman et al. 2002). However, the yield increases were driven
largely by intensification in the use of non-renewable synthetic fertilisers (Lynch
2007). They were seminal in improving western lifestyles but provided limited
relief in many regions of the world such as Africa, Asia and South America.
Moreover, the technologies were double edged, with gains in agricultural produc-
tion coinciding with increased soil erosion (Matson et al. 1997), industrial agri-
cultural pollution (Horrigan et al. 2002), declines in water quality (Foley et al.
2005) and, possibly most importantly, loss of biodiversity (including genetic ero-
sion) (Aguilar et al. 2008; Balestrini et al. 2015). Arguably, a sustainable way
forward is ‘ecological intensification’. This paradigm expands agricultural inten-
sification that promotes the development of food systems with enhanced nutrient
uptake and water use efficiency (Cassman 1999), reductions in pest and disease
control (Bommarco et al. 2013) and a restoration in soil fertility (Balestrini et al.
2015; Matson et al. 1997; Tittonell 2014). Many of these goals are only achievable
today due to the advances in analytical technologies and scientific knowledge that
underpins methodologies like metabolomics. As useful as the broad brush of tra-
ditional physicochemical analyses has been for agricultural systems (Sinsabaugh
et al. 2016) (to understand how to manipulate plant growth), it would be advan-
tageous to determine biogeochemical processes that occur in soil (Rockström et al.
2009; Sardans et al. 2011).
The soil matrix is one of the most diverse ecological systems on the planet
(Torsvik and Øvreås 2002). Home to a plethora of organisms, from the largest
redwood to the smallest microbe, there are numerous chemical (McBride 1994),
physical (Marshall and Holmes 1980) and biological (Barea et al. 2005; Lorenz and
Wackernagel 1994) interactions occurring in soil. Until recently, quantifying these
biogeochemical processes as a metabolome of “soil” was not considered feasible,
with only narrow components of the soil matrix, such as plants and animals, being
studied (Maron et al. 2011; Mendes et al. 2013; Mosier et al. 2013). However, a
holistic approach to understanding the soil community is something of increasing
interest with examples in organic nutrient pools [(Warren 2013, 2014), Table 1],
pollution assessment (Jones et al. 2014; Tremaroli et al. 2009), effects of climate
change [(Pang et al. 2016), Table 2], plant [(Badri et al. 2013a; van Dam and
Bouwmeester 2016), Table 3] and/or microbial metabolism (Ponomarova and Patil
2015; Warren 2015). Recent progress in performing untargeted metabolomics to
identify soil organic matter (SOM) metabolites (e.g. lipids and organic acids),
which can be linked to nutrient uptake [(Swenson et al. 2015b), Table 3], has
shown that “soil metabolomics” is now beyond theory. Still, the ability to study soil
holistically is at an early stage of development. The potential for using metabo-
lomics to advance study into how the soil matrix operates will increase as the
technologies underpinning the methodology (i.e. gas and liquid chromatography–
mass spectrometry (GC–MS and LC–MS), and nuclear magnetic resonance
(NMR)) improve (Singh 2006). Likewise, metabolomics can provide a holistic
understanding of the impact of the increasing resource demands has on soil
(Rockström et al. 2009). Through understanding of the important metabolomic
pools and fluxes in soil, so as to understand and monitor soil health as it is managed
by farmers, foresters and the community, is likely to improve productivity and the
environment while reducing costs (Abhilash et al. 2012; Desai et al. 2010).
Metabolomics of the soil would identify the largest component of metabolites
coming from the most varied and numerous collection of microbes known (Barea
et al. 2005; Huang et al. 2014; Mendes et al. 2013), with estimates of between 1 and
40 million species per gram of soil (Burns et al. 2013; Desai et al. 2010; Řezanka
and Sigler 2009). Despite the importance of microbes to the agricultural and
environmental communities, knowledge of the composition of the microbial bio-
mass is limited. For instance, estimates of fungi range from 700 000 to 1.5 million
(Lumbsch and Leavitt 2011; Ponomarova and Patil 2015; Rastogi and Sani 2011).
However, only 100,000 have been detailed. A large part of this struggle to
understand the composition of the soil microbial community is that <1 % has been
6 Soil Microbial Metabolomics 149
able to be cultured (Kirk et al. 2004; van den Berg et al. 2015). Metabolomics,
being a microbe-independent technique, could shed light on this rich network via
metabolite fluxes and identifying previously unknown metabolites and biogeo-
chemical processes, with genomic studies identifying numerous other microbial
clades that have been identified only by their molecular sequences (Cesco et al.
2012; Cheynier et al. 2013; Swenson et al. 2015b). There is potential to extract
useful information using metabolomics (Alivisatos et al. 2015); although it is
acknowledged that to date, studies have been limited due to the complexity of the
soil matrix (Ponomarova and Patil 2015).
This chapter uses the lens of metabolomics to discuss connections between soil
microbes and the complex interactions they undergo with plants, animals and
human intervention. Although studies of soil microbes using metabolomic tech-
niques are scant, associated studies that can shed light on the complex biogeo-
chemical interactions have been undertaken and provide direction. Due to the drive
to feed the planet and increase agricultural productivity, the rhizosphere of plants
and how to manipulate plant–microbe interactions has probably been of the greatest
research interest in this area (Bertin et al. 2003; De-la-Pena and Loyola-Vergas
Table 2 Typical metabolites found in soil-related samples using 1H-NMR
150
Sample; treatments Metabolite class Techniques used Metabolites Spectroscopic Why? Publication
range (ppm)
(a) Grape skin; soil, Amino acids 1H-NMR (D2O, Isoleucine, leucine, 0.90–1.10 Biomarkers (a) Pereira et al.
weather, cultivar. TSP), 1H-NMR valine (2006). (b) Liebeke
(b) Soil; bacterial (D2O, TSP or et al. (2009).
strain, location. CDCl3) (c) Rochfort et al.
(c) Worms; soil type, (2009)
organic versus
chemical fertiliser
(a) Plant cuts; time Amino acids 1H-NMR TOCSY, Threonine 1.30, 1.45 Biomarkers, (a) Bertram et al.
treatments. NOESY, HSCQ; Salinity (2010). (b) Pang
(b) Seedlings and (D2O with TSP), et al. (2016).
plants; NaCl and 1H-NMR NOESY (c) Rochfort et al.
NaHCO3. (D2O, Na2HPO4, (2009)
(c) Worms; soil type, NaH2PO4, TSP),
organic versus 1H-NMR (D2O, TSP
chemical fertiliser or CDCl3)
Worms; soil type, a-Hydroxy acids 1H-NMR (D2O, TSP Lactate 1.30 Biomarkers Rochfort et al.
organic vs. chemical or CDCl3) (2009)
fertiliser
Wine; bacterial strain Alcohols 1H-NMR 2,3-Butanediol 1.37 Vintage Lee et al. (2009)
NOESYPREAST
(D2O, oxalate, DSS)
(a) Grape skin; Soil, Amino acids 1H-NMR (D2O, Alanine 1.46–1.70 Biomarkers, (a) Pereira et al.
weather, cultivar. TSP), 1H-NMR salinity, (2006). (b) Bertram
(b) Plant cuts; time TOCSY, NOESY, vintage et al. (2010).
treatments. (c) Soil; HSCQ; (D2O with (c) Liebeke et al.
bacterial strain, TSP), 1H-NMR (2009). (d) Pang
location. NOESY (D2O, et al. (2016).
(continued)
M.W. Heaven and D. Benheim
Table 2 (continued)
Sample; treatments Metabolite class Techniques used Metabolites Spectroscopic Why? Publication
range (ppm)
(d) Seedlings and Na2HPO4, (e) Rochfort et al.
plants; NaCl and NaH2PO4, TSP), (2009). (f) Lee et al.
NaHCO3. 1H-NMR (D2O, TSP (2009)
(e) Worms; soil type, or CDCl3), 1H-NMR
organic versus NOESYPREAST
chemical fertiliser. (D2O, oxalate, DSS)
(f) Wine; bacterial
6 Soil Microbial Metabolomics
strain
Grape skin; soil, Amino acids 1H-NMR (D2O, Arginine 1.70 Biomarkers Pereira et al. (2006)
weather, cultivar TSP)
(a) Soil; bacterial Organic acids 1H-NMR (D2O, Acetic acid 1.92 Biomarkers, (a) Liebeke et al.
strain, location, TMSP), 1H-NMR ecotoxicity (2009). (b) Simpson
(b) Earthworms; (Various) and McKelvie
various (2009)
Grape skin; soil, Amino acids 1H-NMR (D2O, GABA + proline 1.94 Biomarkers Pereira et al. (2006)
weather, cultivar TSP)
Grape skin; soil, Amino acids 1H-NMR (D2O, Proline 1.98 Biomarkers Pereira et al. (2006)
weather, cultivar TSP)
Soil; bacterial strain, Amino acids 1H-NMR (D2O, Glutamic acid 2.06 Biomarkers Liebeke et al.
location TMSP) (2009)
Seedlings and plants; Amino acids 1H-NMR NOESY Glutamate 2.10 Salinity Pang et al. (2016)
NaCl and NaHCO3 (D2O, Na2HPO4,
NaH2PO4, TSP)
Seedlings and plants; Amino acids 1H-NMR NOESY Glutamine 2.20 Salinity Pang et al. (2016)
NaCl and NaHCO3 (D2O, Na2HPO4,
NaH2PO4, TSP)
(continued)
151
Table 2 (continued)
152
Sample; treatments Metabolite class Techniques used Metabolites Spectroscopic Why? Publication
range (ppm)
Grape skin; soil, Organic acid 1H-NMR (D2O, Shikimic acid 2.22 Biomarkers Pereira et al. (2006)
weather, cultivar TSP)
Earthworms; various Organic acids 1H-NMR (Various) Acetoacetate 2.23 Ecotoxicity Simpson and
McKelvie (2009)
Soil; bacterial strain, Organic acids 1H-NMR (D2O, Pyruvic acid 2.27 Biomarkers Liebeke et al.
location TMSP) (2009)
Wine; bacterial strain Organic acids 1H-NMR Acetate 2.28 Vintage Lee et al. (2009)
NOESYPREAST
(D2O, oxalate, DSS)
(a) Seedlings and Organic acid 1H-NMR NOESY Succinate 2.40, 2.42, Salinity, (a) Pang et al.
plants; NaCl and (D2O, Na2HPO4, 2.82 biomarkers, (2016), (b) Liebeke
NaHCO3. (b) Soil; NaH2PO4, TSP), ecotoxicity, et al. (2009).
bacterial strain, 1H-NMR (D2O, vintage (c) Simpson and
location. TMSP), 1H-NMR McKelvie (2009).
(c) Earthworms; NOESYPREAST (d) Lee et al. (2009)
various. (d) Wine; (D2O, oxalate, DSS)
bacterial strain
Grape skin; soil, Amino acids 1H-NMR (D2O, GABA + glutamine 2.46 Biomarkers Pereira et al. (2006)
weather, cultivar TSP)
(a) Earthworms; Amine 1H-NMR (Various), Dimethylamine 2.75, 2.80 Ecotoxicity, (a) Simpson and
various. 1H-NMR NOESY salinity McKelvie (2009).
(b) Seedlings and (D2O, Na2HPO4, (b) Pang et al.
plants; NaCl and NaH2PO4, TSP) (2016)
NaHCO3
Soil; bacterial strain, Amino acids 1H-NMR (D2O, Aspartic acid 2.79 Biomarkers Liebeke et al.
location TMSP) (2009)
(continued)
M.W. Heaven and D. Benheim
Table 2 (continued)
Sample; treatments Metabolite class Techniques used Metabolites Spectroscopic Why? Publication
range (ppm)
Seedlings and plants; Organic acid 1H-NMR NOESY 2-Oxoglutarate 3.00 Salinity Pang et al. (2016)
NaCl and NaHCO3 (D2O, Na2HPO4,
NaH2PO4, TSP)
(a) Grape skin; soil, Neurotransmitter 1H-NMR (D2O, GABA 1.85, 2.17, Biomarkers, (a) Pereira et al.
weather, cultivar. (Amino acid) TSP) 2.25, 2.84, vintage (2006). (b) Bertram
(b) Plant cuts; time 3.02, 3.06, et al. (2010). (c) Lee
treatments. (c) Plant 3.10, 3.28 et al. (2009)
6 Soil Microbial Metabolomics
Sample; treatments Metabolite class Techniques used Metabolites Spectroscopic Why? Publication
range (ppm)
(a) Soil; bacterial Amino acids 1H-NMR (D2O, Glycine 3.56–3.60 Biomarkers, (a) Liebeke et al.
strain, location. TMSP), 1H-NMR ecotoxicity, (2009). (b) Simpson
(b) Earthworms; (Various), 1H-NMR salinity and McKelvie
various. NOESY (D2O, (2009). (c) Pang
(c) Seedlings and Na2HPO4, et al. (2016)
plants; NaCl and NaH2PO4, TSP)
NaHCO3
in situ; Cytochrome Xenobiotics 1H-NMR Glycolate 3.95 Degradation Delort and
P450 inhibitor; (D2O + TSP), of Combourieu (2001)
xenobiotic ISP-MS xenobiotics
Seedlings and plants; Amino acids 1H-NMR NOESY Malate 4.40 Salinity Pang et al. (2016)
NaCl and NaHCO3 (D2O, Na2HPO4,
NaH2PO4, TSP)
(a) Grape skin; soil, Sugars 1H-NMR (D2O, Glucose, maltose, 4.62–5.42 Biomarkers (a) Pereira et al.
weather, cultivar. TSP), 1H-NMR sucrose (2006). (b) Liebeke
(b) Soil; bacterial (D2O, TMSP), et al. (2009).
strain, location. 1H-NMR TOCSY, (c) Bertram et al.
(c) Plant cuts; time NOESY, HSCQ; (2010). (d) Rochfort
treatments. (D2O with TSP), et al. (2009)
(d) Worms; soil type, 1H-NMR (D2O, TSP
organic versus or CDCl3)
chemical fertiliser
Wine; bacterial strain Organic acids 1H-NMR Tartrate 4.63 Biomarkers Lee et al. (2009)
NOESYPREAST
(D2O, oxalate, DSS)
Soil; bacterial strain, Sugars 1H-NMR (D2O, Trehalose 5.19 Biomarkers Liebeke et al.
location TMSP) (2009)
(continued)
M.W. Heaven and D. Benheim
Table 2 (continued)
Sample; treatments Metabolite class Techniques used Metabolites Spectroscopic Why? Publication
range (ppm)
Soil; bacterial strain, Organic acids 1H-NMR (D2O, Malic acid 6.03 Biomarkers Liebeke et al.
location TMSP) (2009)
(a) Worms; soil type, Organic acids 1H-NMR (D2O, TSP Fumarate 6.20–6.52 Biomarkers, (a) Rochfort et al.
organic versus or CDCl3), 1H-NMR ecotoxicity (2009). (b) Pang
chemical fertiliser. NOESY (D2O, et al. (2016).
(b) Seedlings and Na2HPO4, (c) Liebeke et al.
plants; NaCl and NaH2PO4, TSP), (2009). (d) Simpson
6 Soil Microbial Metabolomics
(c) Earthworms;
various
156
2014; van Dam and Bouwmeester 2016; Zahir et al. 2004). Other areas of dis-
cussion beyond the plant–microbe interactions include specific foci on how soil
microbes are affected by pollution, diseases, pests and potential climate change.
A theme of the chapter will be how metabolomics may be used to improve soil
management (Rochfort et al. 2015; Zhang et al. 2015), both for increasing pro-
ductivity of the soil and mitigating environmental effects.
Soil is often a secondary topic for metabolomics analyses. The focus of the research
is often on how microbes interact with plants such as grasses (e.g. Trifolium: clover)
or legumes [(Bertram et al. 2010), Table 2], weeds such as Arabidopsis [(Gamir
et al. 2014), Table 4], trees such as Aspen (Wallenstein et al. 2010) or invertebrates
that inhabit the soil such as earthworms Aporrectodea caliginosa [(Rochfort et al.
2009), Table 2] or Eisenia fetida (Simpson and McKelvie 2009). Recent reviews
have discussed soil microbial metabolites within the concept of environmental
metabolomics. For instance, a review on environmental metabolomics suggests the
field is fragmented, as this new “holistic” methodology was mainly being used to
study single species of the researchers’ interest, including those involving soil
microbes (Bundy et al. 2009). The authors reiterated that this misses one of envi-
ronmental metabolomics assets in gathering an understanding of the interactions
between species and their environment. Another review focused on using meta-
bolomics to assess soil contamination (Hernandez-Soriano and Jimenez-Lopez
2014). Earthworms, usually of the genus Eisenia, were found to be typical subjects
for metabolomic analyses. Studies related to microbial metabolomics were small,
with the only reference to work on the response to tellurite by Pseudomonas
pseudoalcaligenes (Tremaroli et al. 2009). Earthworms were also considered the
main target for soil metabolomics of another review on environmental sciences and
metabolomics (Lin et al. 2006). This review detailed the various effects of con-
taminants on the metabolic profiles of a variety of earthworms, particularly halo-
genated compounds and metal-contaminated soils. Another review discussed
‘eco-metabolomics’, the use of metabolomics and how it relates to ecology, with
regard to interactions between organisms, including those in the soil (Sardans et al.
2011). While mainly related to how plants and worms react to changes in envi-
ronment, some discussion was made on how fungi Magnaporthe grisea and
Sclerontinia sclerotiorum release metabolites to suppress plant defences.
Various studies that have examined microbes and/or the metabolites associated
with them. However, only in 2015 has there been research that specifically men-
tions the use of “soil metabolomics” to identify biogeochemical processes occurring
in soils [(Swenson et al. 2015a; Swenson et al. 2015b), Table 4]. The research
sought to understand the fluxes of microbial metabolites in SOM that might occur
due to climate change. Comparing fumigated and unfumigated soil samples using
158
3 The Rhizosphere
The rhizosphere, comprising the endorhizosphere, the rhizoplane and the ectorhi-
zosphere (Fig. 1), defines the narrow region between roots and soil directly influ-
enced by both root exudates and exfoliates, and associated microorganisms (Jones
1998). At the heart of the rhizosphere is the root of a plant that is undergoing
symbiosis. The root surface (epidermis and outer cortex) and its adhering soil are
collectively termed the rhizoplane: the interface where both microbial population
and biochemical plant–microbe interactions are at their maximum. The root systems
of plants serve critical roles in the provision of anchorage, water and mineral
absorption and conduction, lateral movement, reproduction, metabolite synthesis
and food storage centre (Kenrick 2013; Kramer and Boyer 1995; Selosse and
Strullu-Derrien 2015). Roots are linear units composed of multiple regions along
the root growth axis. The units—the root cap, root tip, elongation zone, root-hair
zone and mature zone—are uniquely differentiable and perform distinct functions
(Minz et al. 2013). Each of these units uses different libraries of metabolites for
communication to other units while releasing a multitude of metabolites that
modulate plant–microbe interactions in the rhizosphere (Huang et al. 2014). Root
exudates, used as both substrates and signalling molecules by soil microbes,
comprise of both low (e.g. amino acids, organic acids, carbohydrates, phenolics and
160 M.W. Heaven and D. Benheim
other secondary metabolites) and high molecular weight compounds (e.g. mucilage
and proteins) (Bais et al. 2008; Walker et al. 2003; Ziegler et al. 2013). The nature
of root exudates is observed to vary significantly between plant species.
Consequently, the rhizosphere microbiome differs with both plant species and soil
type (Tate 2000; Wieland et al. 2001). The influence of the rhizosphere can even
extend beyond the immediate area of the plant, as discovered with metabolomic
analyses of leaf litter around different tree species (Wallenstein et al. 2013;
Wallenstein et al. 2010).
Apart from the root, the rhizosphere’s main constituents are eukaryotic and
prokaryotic microbial species, with a diversity of microbes that can range from
thousands (Berendsen et al. 2012) to millions (Nihorimbere et al. 2011). The
dynamic microbiome which surrounds the roots is of critical importance to
6 Soil Microbial Metabolomics 161
ecosystem function (Bertin et al. 2003; Sardans et al. 2011), below-ground carbon
(Mendes et al. 2011) and nutrient cycling (Swenson et al. 2015b; Van Der Heijden
et al. 2008), affecting overall plant fitness and soil quality (Barea et al. 2005; Minz
et al. 2013). Plants spend up to half of their energy producing exudates into the
rhizosphere (van Dam and Bouwmeester 2016), though it has been argued that the
source of metabolites (e.g. plant vs. microbe) in the rhizosphere is still to be
elucidated convincingly (Dennis et al. 2010). A major role of exudates from roots is
the communication to soil-borne microbes, the most dominant class of soil biota
(Van Der Heijden et al. 2008). These microbes are phylogenetically diverse (e.g.
bacteria, archaea and fungi), and comprise symbionts, pathogens and saprotrophs
(Balestrini et al. 2015; Reynolds et al. 2003).
Saprotrophic microbes are crucial to nutrient cycling in terrestrial ecosystems,
generating the majority of nutrients required by terrestrial vegetation (Crowther
et al. 2012; Schlesinger 1991). The soil microbial community is able to do this
primarily via photosynthetically fixed carbon introduced to the soil ecosystem in the
form of plant biomass and root exudates (Badri et al. 2013a; Dennis et al. 2010;
Tate 2000). While there is competition between plants and microbes for available
nutrients (Kaye and Hart 1997; Kuzyakov and Xu 2013), two key plant–microbe
symbiotic associations, arbuscular mycorrhizal fungi (AMF) (Van Der Heijden
et al. 1998) and root nodule symbiosis (RNS) (Li et al. 2013), impart significant
benefits to both microbes and plants. AMF improve the supply of water and
nutrients (such as phosphate and nitrogen) to the plant via extraradical hyphae
(Kawaguchi and Minamisawa 2010; Parniske 2008). For RNS, nitrogen-fixing
bacteria enable enzymatic conversion of atmospheric nitrogen into bioavailable
ammonia for plant growth (Brewin 2010). Ecological benefits of symbioses are also
contributed as is the case of AMF, which increases host resilience toward drought
(Baum et al. 2015), decreased susceptibility to diseases (Saia et al. 2015b),
improved heavy metal (Schützendübel and Polle 2002), excess salinity tolerance
(Luo et al. 2009) and other abiotic factors (Habib et al. 2013).
The rhizosphere has undergone scrutiny using various omic techniques including
genomics, metagenomics, proteomics, transcriptomics and more recently metabo-
lomics (van Dam and Bouwmeester 2016). The principle metabolomic tools are
NMR spectroscopy [Table 2, (Sardans et al. 2011)], LC–MS [Table 4, (Allwood and
Goodacre 2010)], GC–MS [Table 3, (Kusano et al. 2011)], although other tech-
niques such as capillary electrophoresis–time of flight–MS (CE–TOF–MS, Table 1)
(Abhilash et al. 2012) have been utilised. For the symbiotic process to occur between
microbes and plants, there must be a series of metabolites that are transferred
between the two moieties (Rasmussen et al. 2012). Major bioactive metabolites
found in the rhizosphere include flavonoids (Cesco et al. 2012; Cheynier et al. 2013;
Narasimhan et al. 2003), phenolic compounds (Badri et al. 2013a;
162 M.W. Heaven and D. Benheim
4 Exudates
4.1 Rhizoengineering
The interactions of beneficial rhizosphere soil microbes with root systems have
pivotal roles in the growth, development and ecological fitness of their plant hosts.
The prevalences of intensive farming practices that are high yield and/or quality
centric are traditionally predicated on extensive use of environmentally harmful and
costly chemical fertilisers (Riding et al. 2015; Wissuwa et al. 2008). Subsequently,
166 M.W. Heaven and D. Benheim
this has led to increased industry interest in the use of sustainable and environ-
mentally ethical farming practices (Nihorimbere et al. 2011; Zahir et al. 2004). The
use of soil inoculants or ‘biofertilizers’ comprising beneficial soil microbes has
been observed to strongly fulfil this niche through enhancement of plant growth,
biological control of plant pathogens, nutrient supply and promotion of soil pro-
ductivity [(Cappellari et al. 2013), Table 1]. Examples of soil inoculants are
mycorrhizal fungi, the filamentous fungi Trichoderma spp. and plant
growth-promoting rhizobacteria (PGPR) (including, but not limited to, genera
Acetobacter, Azospirillum, Bacillus, Burkholderia, Herbaspirillum, Paenibacillus,
Phyllobacterium and Pseudomonas) [(Chamam et al. 2013; Saharan and Nehra
2011), Table 1].
For example, the effect of soil inoculation with PGPRs in the commercially
valuable wild marigold (Tagetes minuta) has been assessed (Cappellari et al. 2013).
Wild marigold produces an essential oil (EO) known as “Tagetes oil” sought for the
preparation of high-grade perfumes. The effect of single and co-inoculation of
Tagetes minuta with Pseudomonas fluorescens and Azospirillum brasilense on
plant growth parameters and essential oil production was assessed under glasshouse
conditions. Azospirillum has been used for growth and yield promotion in cereal
plants as rice, maize and wheat (Chamam et al. 2013). Both single and
co-inoculations showed an increase in shoot fresh weight by approximately 50 %.
Total phenolic content of shoots was upregulated by up to twofold with total EO
yield increased by 70 % in single and co-inoculated plants. Major components of
the EO that significantly increased (p < 0.05) included nine types of terpenoids,
such as tagetone and ocimenone, identified by GC–MS. While individual inocu-
lation with either Azospirillum brasilense or Pseudomonas fluorescens increased
plant growth and EO production, significant enhancement to both metrics was
observed in co-inoculated plants suggesting synergy between the two bacterial
genera.
PGPRs demonstrate significant capacity for plant enhancement. In some cases,
the magnitude of enhancement may be both bacterial strain and plant
cultivar-dependant (Chamam et al. 2013). This study showed a strain-dependent
effect in the association between two types of nitrogen-fixing bacterium
Azospirillum and Asian rice (Oryza sativa). The bacteria Azospirillum lipoferum 4B
(rhizosphere-colonising strain isolated from Oryza sativa cultivar Cigalon) and
Azospirillum sp. B510 (an endophytic strain isolated from Oryza sativa cultivar
Nipponbare) were observed to significantly increase (p < 0.05) growth of the
cultivar by up to 1.5 mg/plant over 10 days if they were used to inoculate the rice
strain from which they were isolated. Metabolic profiling data from reverse-phase
LC–MS demonstrated significant modification in rice secondary metabolites in
PGPR-inoculated plants, with 17 flavonoids, 10 hydroxycinnamic acid derivatives
and four alkylresorcinols the most affected metabolite classes. Moreover, the
metabolites were unique to the cultivar with only a few compounds, such as
tryptophan, found common to all cultivars. This research stands as a strong example
of how metabolomics techniques can be used to directly assess the nature of host–
PGPR symbiotic interactions, here being able to distinguish the physiological
6 Soil Microbial Metabolomics 167
responses of both rice cultivars to specific Azospirillum strains with opposing root
colonisation strategies. In another case, similar changes in plant secondary meta-
bolism were observed in a study investigating maize-Azospirillum interactions
(Walker et al. 2011). Major secondary metabolic changes were exclusively
observed in the roots of the Cigalon-4B pairing, predominantly for benzoxazinones
and benzoxazolinones, while the endophytic B510 strain elicited a systemic
response inducing metabolic shifts in both shoots and roots of both rice cultivars
tested.
Compared to bacterial soil inoculants, fungal soil inoculants including
Trichoderma spp., soil fungi that are often associated with plant root ecosystems
(Vinale et al. 2008), and AMF can impart significant benefit to host plants
(Contreras-Cornejo et al. 2011). Considered beneficial to plants, they protect them
from disease by attacking phytopathogenic fungi. Metabolomic studies have
identified that Trichoderma detect fungi due to the exudation of metabolites from
cell wall degrading enzymes and sensing the sugars released by degradation. More
importantly, Trichoderma releases secondary metabolites that are antifungal,
including antibiotics, water soluble acids and peptaibols. The non-polar nature of
the antibiotics (e.g. 6-pentyl-a-pyrone) suggests that these are used as a long-range
defence, while more polar metabolites (i.e. peptaibols) are used to attack fungi at
‘close quarters’. The positive relationship between Trichoderma and plants, which
includes increasing crop productivity by up to 300 %, along with activation of the
plant’s defences (Woo et al. 2006), has led to the use of this fungi as a natural
biocontrol agent.
Other research that has identified microbially related metabolites involved in
systemic acquired resistance and pathogen protection include the following:
• salicylic (SA) and jasmonic acid (JA) (Segarra et al. 2007) in cucumbers;
• SA, JA and indole-3-carboxaldehyde (Contreras-Cornejo et al. 2011), sugars,
amino acids, ethanolamine, tagatose, oxoproline, GABA and urea (Chaparro
et al. 2013) in Arabidopsis thaliana;
• the increased production of phytoalexins (e.g. flavonoids, terpenoids, indoles)
after inoculation by Trichoderma that showed increased root development and
biomass of Arabidopsis thaliana over 8 days (Contreras-Cornejo et al. 2011);
• 2,4-diacetylphloroglucinol, from AMF (Wehner et al. 2010);
Another reason for inoculation has been to counteract stressors on plants such as
drought or salinity. Abiotic stress adaptation has been shown to involve a range of
metabolites including the following:
• rhizospheric fungi Trichoderma harzianum Rifai strain T-22 using lipid per-
oxides in tomatoes as stress biomarkers (Mastouri et al. 2010);
• drought protection of plant through the increased production of hormones such
as indole-3-acetic acid, JA, SA, ethylene, auxins, cytokinins and gibberellins
and amino acids like proline (Azcón et al. 2013);
• reduced concentrations of malondialdehyde and increased concentrations in
proline and phenol metabolites in wheat due to salinity (Rawat et al. 2011); and,
168 M.W. Heaven and D. Benheim
• in the case of Trichoderma spp., the regulation of root system architecture that
was made possible with reactive oxygen species that convert into hydrogen
peroxide (Contreras-Cornejo et al. 2013; Mastouri et al. 2010; Samolski et al.
2012).
Metabolomics can also be used as a tool to find soil microbial metabolites that
are a signal of diseases occurring within the soil (Bundy et al. 2009). For example,
the fungi Trichoderma spp. (Lorito et al. 2010; Vinale et al. 2008) have been found
to improve plant defences through secondary metabolite communication.
A pathogen attacking a plant results in Trichoderma expressing amino acids to
create small proteins called hydrophobins that act to coat the plant as a barrier.
Other metabolites used for defence included heptelidic (koningic) acid (Itoh et al.
1980), along with isocyanide derivatives, proteins with a-aminoisobutyric acid and
a range of FAs (e.g. C8, C10, C10:1).
7 Humus
8 Terroir
In the viticulture industry, the word ‘terroir’ describes regional influences associ-
ated with climate, soil factors and plant genotype which strongly affect varietal
flavours and aromas (Styger et al. 2011). Biochemically, the same term relates to
modifications to the metabolome of a plant that impart a set of discrete desired
qualities, such as increased plant growth or reduced feeding by larvae (e.g. cabbage
“worm” caterpillar Trichoplusia ni) on plants (Badri et al. 2013b). The ‘terroir
effect’ is shown to manifest itself in cultivated grape varieties that have been
resident in the same location for extended time periods (van Leeuwen and Seguin
2006), and this concept has begun to be used to describe ‘microbial terroir’ (del
Mónaco et al. 2016). It has been suggested that this effect could likely be attributed
to soil microbes or the overall soil microbiome actively adjusting metabolite fluxes
in response to signals from plants (Badri et al. 2013b). In one study, GC–MS was
used to assess the effect of diverse soil microbiomes applied to the roots of
Arabidopsis thaliana on the leaf metabolome and, by extension, whether the
changes in leaf metabolome influenced the feeding behaviour of Trichoplusia ni
larvae. The study demonstrated that variations in soil microbiome composition
(primarily comprised >60 % actinobacteria and proteobacteria) produced a differ-
ential response to canopy and root biomass accumulation in A. thaliana plants. In
comparison to control soil slurries (absence of soil microbiome), all A. thaliana
plants showed an upregulation of amino acids, phenolics, sugars and sugar alcohols
in leaf material. In addition, presented evidence suggested a strong capacity of soil
microbial communities to either modulate above-ground feeding behaviour of
Trichoplusia ni or to enhance the herbivory resistance of 4-week-old A. thaliana
plants. It is surprising that in examining what makes a quality grape for wines that
soil and the microbial life has until recently been largely ignored (Burns et al.
2015). The reason behind this is that soil microbial effects are subtle versus climate
[(Pereira et al. 2006), Table 2], rainfall [(Lee et al. 2009), Table 2] and soil texture
(Pereira et al. 2006) which have been found to be larger drivers of terroir.
170 M.W. Heaven and D. Benheim
10 Extracellular Enzymes
As the previous sections attest, there is no doubt that it can be difficult to identify
microbial metabolites from the myriad of chemical compounds in soil (Ponomarova
and Patil 2015). This becomes more problematic when sampling away from the
microbial powerhouse of the rhizosphere. One example is extracellular enzymes
(EE), enzymes released by microbes or plants into the soil to facilitate biochemical
processes (Wallenstein and Weintraub 2008). These processes include degrading
recalcitrant fractions of SOM for uptake by the microbes, secretion of metabolites to
“sense” what predators or prey are in the immediate environment, and release of
antibiotics to attack other microbes (Burns et al. 2013; González-Fernández et al.
2015; Wallenstein and Weintraub 2008). For example, EE from fungi produce
metabolites such as the lactone-based botcinolides and the terpene-based botrydial
compounds (González-Fernández et al. 2015). These metabolites are thought to be
excreted to attack plants through decomposition of plant cell walls followed by
nutrient acquisition from the plants. EEs from soil bacteria and fungi are known to
consume carbon-rich biomolecules such as chitin (Roberts and Selitrennikoff 1988),
lignin (Burns et al. 2013), tannins (Joanisse et al. 2007) and pectins
(González-Fernández et al. 2015; Tepper and Anderson 1990). Sugars and amino
acids from glycoproteins are also found on microbial adhesives (Wang et al. 2014)
from EE-producing microorganisms after consumption of the plant and microbial
debris. Plant litter is also a rich source of nutrients for EE-producing microorgan-
isms. Monitoring of lignin decomposition has revealed the production of metabo-
lites such as quinines and radical lipids that may potentially form humic compounds
in soils with access to phenols, peptides and carbohydrates (Schmidt et al. 2011).
Quantitatively and qualitatively, there is still ambiguity in the sources and
consumption of metabolites in soil. Whether the source is via a combination of EE
excretion, plant decomposition or experimental error, metabolomics techniques
may be amenable to characterising the macromolecules produced by EEs (e.g.
polysomes). A potential solution described in the literature involves attaching
coloured marker molecules to EEs in a dilute soil slurry, as the enzymes tend to stay
fixed to the soil and unavailable for analyses through typical extraction techniques
(Burns et al. 2013). Limitations of the current methodology include no knowledge
of enzyme turnover rates, limited number of fluorescent markers available to attach
to a limited number of functional groups and selection of only those enzymes
capable of being stabilised in the slurry, versus in situ soil samples.
Metabolomics has been suggested as a way of removing these limitations by
detecting the entire metabolome of a soil sample. Both pyrolysis-GC-IRMS and
LC–MS analyses have provided examples of how this might work. For instance, a
general survey of EE metabolites involved soil from the USA and Germany
(Liebeke et al. 2009). Analyses of soil involved both untargeted metabolomics of
GC–MS and 1H-NMR of SOM, while targeting metabolites of microbe Bacillus
licheniformis for comparison. A range of FAs, sugars, amines, amino acids and
organic acids were identified. Most metabolites were species dependent though
174 M.W. Heaven and D. Benheim
metabolites acetic, fumaric, aspartic and glutamic acid, glycine, proline, serine,
glucose and sucrose were found ubiquitously across the varied agricultural types.
It is argued that we are in the Anthropocene age (Bundy et al. 2009; Desai et al.
2010; Rockström et al. 2009), so it is not surprising that soil microbes have had to
adapt to human endeavours, some of which can be deleterious to the environment.
Estimates made in 1998 include up to 100 000 chemicals which were available for
purchase (Rockström et al. 2009), many of which will end up in the environment.
Since then, material synthesis and engineering has advanced greatly, and so it
should be no surprise that many of these compounds end up affecting the soil
microbial food chain [(Simpson and McKelvie 2009), Table 2]. Indictor animals
such as earthworms are often used to determine soil health (Rochfort et al. 2009;
Whitfield et al. 2013), but there is an increasing amount of research into the effects
of synthetic chemicals on soil microbes (Hernandez-Soriano and Jimenez-Lopez
2014). The use of microbes to monitor or remediate contaminated sites is one of
particular interest (Desai et al. 2010; Jones et al. 2014). A soil metabolome would
allow for the ability to look across a range of biogeochemical factors that may affect
soil health. The discussion here will focus solely on metabolomics research as it
relates to microbes.
Agricultural changes of soil from pristine forest to farm land has been a major
change to the environment that has been occurring for thousands of years (Foley
et al. 2005). Changes occurring in soil management have been described through
the lens of metabolomics (Singh 2006). For instance, land use provided an
opportunity to use NMR metabolomics combined with mid-infrared spectroscopy
(MIR) to identify the effect of land management across four different regions of
Victoria, Australia (Rochfort et al. 2015). Comparing soil samples from relatively
untouched native land and adjacent farm land of oats or wheat, 1H-NMR was able
to identify a series of signals from lipid, terpene and sugar. The study identified that
these metabolites could be differentiated by NMR due to different concentrations
depending on land use, whereas soil location was differentiated using MIR. This is
similar to a 1H-NMR study of various mine sites across England (Jones et al. 2014)
which identified a similar series of metabolites, with differences between com-
pounds thought to have occurred due to a different solvent extraction system
(methanol for this study versus deuterium oxide). In both cases, the assumed
microbial source of these metabolites was explored, using a target microbe, Bacillus
subtilis, for the Australian study and the identification that most soil metabolites
were microbially based for the English analysis. Labelling techniques may give
enhanced information, as was shown in the effects of microbes feeding on mine
waste. Metabolite confirmation of mine waste and its effects on microbes was
conducted using stable isotope labelling (Mosier et al. 2013). Using this method
with 15N labelling allowed for the identification of 80 metabolites from 3500
6 Soil Microbial Metabolomics 175
Novel compounds, often of the size or smaller than the microbes themselves (e.g.
nanometre), are increasingly being developed, used and disposed of, on soils. The
design and use of engineered nanomaterials (ENM) is in part because they have
unusual and useful properties (e.g. strength and conductivity) that are different from
the bulk compound found in nature (Dinesh et al. 2012). ENM, so called to dis-
tinguish them from natural soil nanoparticles (e.g. colloids), are receiving
increasing attention in agricultural and environmental literature. As production of
ENM increases to meet the demand of high-tech materials, these products are more
likely to end up in soil. Products with ENM include sunscreens, cleaning products
and therapeutic goods. Some ENM, such as zero-valent Fe, are widely used in parts
of the world for cleaning up toxic chemicals (Lee et al. 2008). Until recently, the
effect of ENM on the environment had not been systematically studied, in part due
to the cost of manufacture, the similarity in size and composition of natural colloids
(Klaine et al. 2008), as well as the physicochemical aspects of the soil
(MacCormack and Goss 2008). Various studies have shown that ENM are poten-
tially hazardous in a laboratory setting, but there are few field studies (Dinesh et al.
2012; Johansen et al. 2008). To date, ENM have been predicted to be in the
environment (i.e. surface waters) in concentrations of 0.8 ng/L for carbon-based
ENM up to 10 µg/L for Ag-, Ti- and Zn-based compounds (Maurer-Jones et al.
2013). No such study has been conducted for concentrations of ENM in soil.
Adding ENM to soil to see how they affect the biosphere has been the main
method of determining their effects [(Jin et al. 2014; Johansen et al. 2008; Shah and
Belozerova 2009), Table 1]. This has enabled studies of how the dosage of ENM
affects the soil environment. As ENM are in a solid matrix, it can be difficult to
ascertain the dose actually received by microbes, and so studies may overestimate
their soil concentration. Although it is well recognised that they have the potential
176 M.W. Heaven and D. Benheim
metal ENM with unusual isotopes could improve the analyses of these particles
(Klaine et al. 2008). Most research of metal ENM has shown that they act as
bactericides (Lee et al. 2008). As these compounds have been used for water
treatment and fabric formation, some of the pollution may end up in soil (Stefaniuk
et al. 2016). Measuring microbial responses to metal ENM have usually focussed
on their toxicity through the use of fatty acid methyl esters (FAMEs) analysis. For
instance, a study on microbial response to Pd, Cu, Si and Au ENM used FAMEs
typical to microbes to determine how microbes survived in their presence [(Sasser
2006; Shah and Belozerova 2009), Table 1]. Despite using two comparative con-
centrations of metal ENM in soil over 15 days (0.013 and 0.066 % w/w), no
significant change in these microbial biomarkers metabolites was seen. However, it
was speculated that ENM may exhibit an indirect effect through interaction of
compounds required by microbes. This was shown in a concurrent experiment
where the growth of lettuce seeds was reduced in the presence of Pd and Au ENMs.
One positive review discussed how microbes could be used to create metal ENM
for commercial use, based on microbes being able to create metal organic com-
plexes such as iron oxide magnets, metal phosphate medicines and transition metal
catalysts (Lloyd et al. 2008).
Microbes such as AMF can be affected by heavy metal contamination (Karimi et al.
2011), and it has been shown that the metals reduce microbial biomass. Metal
wastes reaching soils are another environmental issue (Maurer-Jones et al. 2013;
Simpson and McKelvie 2009). Similarity in atomic size to nutrients, some heavy
metals can access microbes through channels designed to diffuse and transport
cations (Singh et al. 2016). This similarity has been put to use with Cu used as a
fungicide that has been shown to improve crop yield (Dhawi et al. 2015). As metals
cannot be degraded, microbial action tends to involve immobilising the contami-
nation so that its toxic effects are mitigated (Azcón et al. 2013). The success of
microbes and plants in resisting radiation effects (Stone 2009) and reducing con-
centrations of toxic metals has been identified in places of recent disasters such as
the Chernobyl and Fukishima nuclear accidents (Aung et al. 2015; Geras’kin et al.
2008). It has been noted that continuous exposure to heavy metals can result in
tolerance forming in microbes as they employ metabolic strategies to reduce metal
toxicity though reduction of the metal’s oxidation state or coordination of
metabolites to soluble metals ions (Jones 1998; Karimi et al. 2011). For instance,
the microbe Klebsiella mobilis CIAM 880 was able to release metabolites that bind
with Cd to promote plant growth in soil with high cadmium concentrations, pre-
cipitating the metal (Nies and Silver 1995; Pishchik et al. 2002). This was seen in
the plant having a larger root system and increased exudate concentrations released
into the soil.
178 M.W. Heaven and D. Benheim
The compound that probably typifies pollution since the industrial era is lead
(Nriagu and Pacyna 1988). One metabolomics study identified metabolites that
changed in concentration due to mycorrhizal microbe influence attenuating the
plants response to Pb contamination (Souza et al. 2014). The analyses identified a
series of amino acids that may complex heavy metal and remove the metal’s ability
to affect the plants. The amino acids include asparagine, histidine, proline and
glutamine. Using the AMF Glomus etunicatum, it was possible to identify potential
microbial interactions with plants (e.g. N and carbohydrate metabolism) that offer
protection from Pb. Pb was also identified as a factor in metabolomics analyses of a
series of mines in the UK [see the “Community Metabolomics” chapter by Jones
et al. (2014) this book]. Using 1H-NMR, metabolomics was used across 11 sites
that mined Pb and Zn (Jones et al. 2014). A simple methodology with minimal
extraction meant that, along with the microbial community, soil and invertebrates
were also sampled. The authors examined specific groups of metabolites (nu-
cleotides, sugars, lactate and amino acids) using multivariate statistics. They were
able to show that there were sites that had similar metabolic profiles and lower Fe
concentrations even though the mines were otherwise different in geochemical
makeup. The authors proposed that monitoring metabolomics could act as an early
warning to hazardous pollutant levels before any visible effects were seen. PLFA
was used in the Czech Republic to determine how mining waste materials fly ash
and mine digestate that contained high Pb, Si and Zn concentrations affected soil
microbes (Garcia-Sánchez et al. 2015). Organic compounds’ concentrations,
including phenolics, were identified as increasing under both contaminants, with the
researchers identifying this via increased microbial activity, particularly fungi,
under the digestate treatment (9.3 ± 1.4 µg/kg after 60 days, p 0.05). Other
compounds found due to contamination included carbohydrates, carboxylic and
amino acids, amines and polymers, although the researchers cautioned that the
metabolomics and genomics analyses could not differentiate between active and
passive microbes in the soil. A timed study showed that after two months, amine
compounds had become preferentially consumed. Digestate contaminated soil was
shown to result in a preference for microbes to consume carboxylic acids taken
from the SOM. The PLFA analyses found that metabolites associated with fungi
were most attenuated by the digestate and fly ash. Metabolites associated with
Gram-negative bacteria were also affected. The digestate increased the concentra-
tion of Gram-positive bacteria after two months. Fly ash was found to be beneficial
to soil community structure, with the metabolites associated with most types of
microbes increasing. Biofilms in mines have also proven to be a rich source of
metabolomic data on how microbes are affected by various metal wastes that
included sulphate, iron, zinc, copper and arsenic (Mosier et al. 2013).
Cadmium contamination is also a concern in soil as it affects the life cycles of
most species [(Sarry et al. 2006), Table 2]. Cd has been found to affect people
through kidney damage after consumption of contaminated food, with the suspicion
that the metal’s affinity to thiols is deleterious to organisms. This includes yeasts
and fungi that are found in a variety of forms in agriculture (Sláviková and
Vadkertiová 2003). Yeast under the effects of cadmium was studied using
6 Soil Microbial Metabolomics 179
metabolomic and proteomic techniques (Lafaye et al. 2005). The research identified
that the typical sulphur pathway in yeast for producing glutathione increased in rate
and the sulphur amino acid concentration reduced by 30 % as the microbe
attempted to remove cadmium. Proteome and metabolomic results were correlated
when Cd was used, but other treatments (e.g. sulphur starvation) that also result in
increased glutathione production were not, indicating an independent pathway is
initiated when Cd contamination is present.
A controlled study of increasing Fe concentrations was used to determine if the
microbe Pseudomonas stutzeri RCH2 was affected by metal-poor or metal-rich soils
(Swenson et al. 2015a). The idea was that Fe will affect the microbe’s metabolite
output as the competition for sites on either the metal or carbon of SOM in soil is
changed. As expected, increasing the Fe concentration increased the sorption of all
metabolites including those with phosphate, N-containing and carboxylate func-
tional groups. The research identified that concentration change in metabolites was
correlated to the charge of the anion (e.g. Phosphate− − Fe+) when it came to
sorption to Fe. The authors felt that it was important to conduct metabolomic
analyses to ascertain the rates of sorption due to a typical mix of microbial
metabolites, which may differ due to competing interactions compared to when
separate metabolites being tested. While the analyses did not bring many surprising
results (i.e. phosphates and dicarboxylates adsorb strongest to Fe), this paper is a
rare example of the application of metabolomics to understand the holistic system
of soil–microbe interactions.
Research on organic compound contamination has been presented that offers not
just problems but solutions using metabolomics of soil microbes. For instance,
metabolomics was used to determine how microbes might be able to recover pet-
roleum from reservoirs that are not cost effective by traditional extraction tech-
niques (Arora et al. 2014). This study used microbial enhanced oil recovery of an
oil well on soil samples from India to determine how efficient the microbes were
and what decomposition products they produced during the extraction. Using
indigenous hyperthermophilic Clostridium sp., they tested how these microbes may
extract oil at high temperature sites (>91 °C). The use of water from the oil well site
ensured no contamination of microbes from elsewhere in the soil. Metabolites
collected included biosurfactants, organic acids, solvents, exopolysaccharides and
volatile FAs. Following a targeted analysis of the metabolite mix from different
groups of bacteria, the researchers were able to optimise conditions to increase the
concentration of metabolites that would be suitable for extraction, including sugars
(particularly sucrose), nitrates and ammonium metabolites (particularly urea).
Metabolomics has also been used to better understand the bioremediation of soils or
soil models (Singh 2006). NMR studies have generally used soil samples rather
than solutions, although there are metabolomic studies using liquid-state NMR to
180 M.W. Heaven and D. Benheim
Potentially, the greatest anthropogenic effect of the current century is how the
planet responds to an increasingly variable climatic pattern of weather (Edenhofer
et al. 2014). Metabolomics has shown promise as one way to quickly quantitate
metabolites that microbes exude in response to climate stressors (Simpson et al.
2012). Expected changes in weather and climate have led to the call and use of
metabolomics techniques to understand how the denizens of the land will adapt
(Ahuja et al. 2010). Climatic effects on soil microbes are expected as changes in
182 M.W. Heaven and D. Benheim
temperature along with water and nutrient concentrations become more variable
(Rennenberg et al. 2009).
Salinity is one climate change issue that is already common. Salinity is
increasing and the replenishment of water tables is difficult. Exacerbating this
problem is research showing that replenishment of water tables is more difficult as
temperature rises (Lozupone and Knight 2007). Metabolomics approaches have
predominantly examined plants, but microbial interactions with plants due to
salinity have also been examined. Studies of a variety of plants (i.e. Arabidopsis
thaliana, Lotus japonicus and Oryza sativa) seem to suggest metabolites used to
communicate between microbes and plants in the rhizosphere will be significantly
affected (Sanchez et al. 2008). These metabolites include organic and amino acids,
along with sugars. This may be mitigated depending on the plant species, as was
found in the case of the highly salt-tolerant Brassicaceae, Thellungiella salsuginea,
where there appeared to be little change regardless of salinity levels [(Lugan et al.
2010), Table 3].
One issue with salinity is that the high salt concentration in these soils can
interfere with the sample, leading to loss of data through ion suppression (Oikawa
et al. 2011). To address this issue, the authors used capillary electrophoresis–mass
spectrometry (CE–MS) in combination with solid-phase extraction (SPE) to
selectively remove up to 17 cations from soil solution. They were also able to
differentiate between plant and microbial cations. Optimising the method with a
model soil solution of 78 organic and 12 inorganic compounds, the method was
then used on soil solutions from rice farms. Significant differences between soil
with and without rice were obtained, with microbial metabolites such as histamine
and tyrosine present only in the absence of plants, and leucine, isoleucine,
phenylalanine and serine significantly more concentrated without plants.
Climate change will probably result in increased periods of drought, and irri-
gation is one method to ensure crop survivability. This results in a dry–wet event
that can lead to various stresses to both plant and microbial life (Kakumanu et al.
2013). In particular, a change in osmotic potential between the soil and intracellular
contents of microorganism can result in reduced life expectancy of the microbial
biomass. Besides K+, which acts as a regulator of ionic strength in cells, amino
acids, carbohydrates, quaternary amines and tetrahydropyrimidine are regulated and
maintained within microbial cells when the amount of water available is reduced.
Fungi have been found to preferentially accumulate polyols while bacteria have a
preference for amino acids and sugars. Other factors that may improve drought
resistance are a robust AMF that will increase N uptake to plants (Baum et al.
2015),
At the end of drought when rains have come, the sudden influx of fresh water
generally results in a flush of microbial activity (Kakumanu et al. 2013). It has still
not been determined whether this is due to the lysis of microbes, a change in
equilibrium of microbes as the concentration gradient changes, or other reasons.
The authors reasoned that they could quantify metabolites that are released when
the concentration gradient changes. It was found that the accumulation of
6 Soil Microbial Metabolomics 183
researchers to determine that glycolysis and the pentose phosphate pathway were
parallel processes and characteristic to the soils studied. Initially, both glucose and
ribose were consumed at similar rates. After 10 days, concentration decreases in the
C-5 labelled ribose showing the preference by microbes to incorporate this
metabolite over glucose, presumably due to the formation of RNA and DNA. Data
for other labelled sugars showed a complex series of reactions occurring in the soil
that include glycolysis, pentose phosphate and gluconeogenesis metabolic path-
ways. PLFA data revealed a preference for the 13C substrates to be taken up by
Gram-negative bacteria, known to be the dominant species around the rhizosphere,
while methanotrophs were found to consume non-plant material, despite its
proximity.
The flux of a series of amino acids was determined using labelled compounds
(Gunina et al. 2014). Compared to fungi, sugars were more efficiently taken up by
bacteria, especially glucose and sucrose, if the concentrations of these metabolites
were low in the soil (Gunina et al. 2014). Ribose uptake was similar to glucose for
Gram-negative bacteria. The pentose phosphate pathway for these bacteria was also
characterised by the uptake of xylose. In contrast, fungi appeared to have a pref-
erence for larger, more complex sugars along with decomposition of glucose to
form triacylglycerols. Acetate, a common metabolite in soils due to it being sourced
in plant litter and cattle slurry, was found to be incorporated preferentially into
Gram-negative bacteria.
Using these labelled metabolites helped the authors to identify that
Gram-negative bacteria were the most efficient at using low molecular weight
metabolites, due to the preference of this type of bacteria to use the anabolic pentose
phosphate pathway. Fungi and filamentous microorganisms were found to be better
utilisers of acidic and complex organic compounds like palmitate and
double-bonded FA. Carbon-13 labelling allowed for comparison between microbial
and soil metabolites in a series of experiments investigating SOM accessibility of
nutrients (Swenson et al. 2015b). Comparing samples of soil that had been fumi-
gated versus untouched soil samples, it was possible to identify the labelled
metabolites of the microbe Pseudomonas stutzeri RCH2. Extracellular metabolites
were found to be more likely to be detected than intracellular ones, indicating that
additional steps such as adding salts to reduce osmosis may not be required. As
minimal processing was a goal of this research, water was used as an extractant, a
solution the authors point out is not ideal for the study of metabolites such as FA or
sterols. Despite this, the labelled metabolites identified included amino acids and
analogues, nucleobases such as uracil, and a series of organic acids.
13 Conclusion
Soil is a rich, complex ground for metabolomics research. The majority of microbes
within soil are still a mystery, but metabolomics is beginning to reveal their secrets.
Even with the advances in understanding how soil microbes interact with plants
6 Soil Microbial Metabolomics 185
References
Abhilash PC, Powell JR, Singh HB, Singh BK (2012) Plant-microbe interactions: novel
applications for exploitation in multipurpose remediation technologies. Trends Biotechnol
30:416–420. doi:10.1016/j.tibtech.2012.04.004
Abu-Nada Y, Kushalappa AC, Marshall WD, Al-Mughrabi K, Murphy A (2007) Temporal
dynamics of pathogenesis-related metabolites and their plausible pathways of induction in
potato leaves following inoculation with Phytophthora infestans. Eur J Plant Pathol 118:375–
391. doi:10.1007/s10658-007-9150-8
Aguilar R, Quesada M, Ashworth L, Herrerias-Diego Y, Lobo J (2008) Genetic consequences of
habitat fragmentation in plant populations: susceptible signals in plant traits and methodolog-
ical approaches. Mol Ecol 17:5177–5188. doi:10.1111/j.1365-294X.2008.03971.x
Ahemad M, Kibret M (2014) Mechanisms and applications of plant growth promoting
rhizobacteria: current perspective. J King Saud Univ Sci 26:1–20. doi:10.1016/j.jksus.2013.
05.001
Ahmed MA, Kroener E, Holz M, Zarebanadkouki M, Carminati A (2014) Mucilage exudation
facilitates root water uptake in dry soils. Funct Plant Biol 41:1129–1137. doi:10.1071/FP13330
Ahuja I, de Vos RCH, Bones AM, Hall RD (2010) Plant molecular stress responses face climate
change. Trends Plant Sci 15:664–674. doi:10.1016/j.tplants.2010.08.002
Alivisatos AP, Blaser MJ, Brodie EL, Chun M, Dangl JL, Donohue TJ, Dorrestein PC, Gilbert JA,
Green JL, Jansson JK, Knight R, Maxon ME, McFall-Ngai MJ, Miller JF, Pollard KS,
Ruby EG, Taha SA (2015) A unified initiative to harness Earth’s microbiomes. Science
350:507–508. doi:10.1126/science.aac8480
Allwood JW, Goodacre R (2010) An introduction to liquid chromatography-mass spectrometry
instrumentation applied in plant metabolomic analyses. Phytochem Anal 21:33–47. doi:10.
1002/pca.1187
186 M.W. Heaven and D. Benheim
Bertram HC, Weisbjerg MR, Jensen CS, Pedersen MG, Didion T, Petersen BO, Duus JØ,
Larsen MK, Nielsen JH (2010) Seasonal changes in the metabolic fingerprint of 21 grass and
legume cultivars studied by nuclear magnetic resonance-based metabolomics. J Agric Food
Chem 58:4336–4341. doi:10.1021/jf904321p
Bhat SV, Booth SC, Vantomme EAN, Afroj S, Yost CK, Dahms TES (2015) Oxidative stress and
metabolic perturbations in Escherichia coli exposed to sublethal levels of
2,4-dichlorophenoxyacetic acid. Chemosphere 135:453–461. doi:10.1016/j.chemosphere.
2014.12.035
Bommarco R, Kleijn D, Potts SG (2013) Ecological intensification: harnessing ecosystem services
for food security. Trends Ecol Evol 28:230–238. doi:10.1016/j.tree.2012.10.012
Bonanomi G, Vinale F, Scala F (2009) The role of natural products in plant-microbe interactions.
In: Osbourn EA, Lanzotti V (eds) Plant-derived natural products: synthesis, function, and
application. Springer, New York
Bonfante P, Genre A (2010) Mechanisms underlying beneficial plant–fungus interactions in
mycorrhizal symbiosis. Nat Commun 1:48
Borisjuk L, Rolletschek H, Neuberger T (2012) Surveying the plant’s world by magnetic
resonance imaging. Plant J 70:129–146. doi:10.1111/j.1365-313X.2012.04927.x
Boyce KC (2005) The evolutionary history of roots and leaves. In: Holbrook NM, Zwieniecki MA
(eds) Vascular transport in plants. Elsevier, San Diego
Braeken K, Daniels R, Ndayizeye M, Vanderleyden J, Michiels J (2008) Quorum sensing in
bacteria-plant interactions. In: Nautiyal CS, Dion P (eds) Molecular mechanisms of plant and
microbe coexistence. Springer, Berlin
Brewin NJ (2010) Root nodules (legume–rhizobium symbiosis). eLS, Nov 2010
Broeckling CD, Broz AK, Bergelson J, Manter DK, Vivanco JM (2008) Root exudates regulate
soil fungal community composition and diversity. Appl Environ Microbiol 74:738–744.
doi:10.1128/aem.02188-07
Bronick CJ, Lal R (2005) Soil structure and management: a review. Geoderma 124:3–22. doi:10.
1016/j.geoderma.2004.03.005
Bundy JG, Davey MP, Viant MR (2009) Environmental metabolomics: a critical review and future
perspectives. Metabolomics 5:3–21. doi:10.1007/s11306-008-0152-0
Burns KN, Kluepfel DA, Strauss SL, Bokulich NA, Cantu D, Steenwerth KL (2015) Vineyard soil
bacterial diversity and composition revealed by 16S rRNA genes: differentiation by geographic
features. Soil Biol Biochem 91:232–247. doi:10.1016/j.soilbio.2015.09.002
Burns RG, DeForest JL, Marxsen J, Sinsabaugh RL, Stromberger ME, Wallenstein MD,
Weintraub MN, Zoppini A (2013) Soil enzymes in a changing environment: current knowledge
and future directions. Soil Biol Biochem 58:216–234. doi:10.1016/j.soilbio.2012.11.009
Canellas LP, Balmori DM, Médici LO, Aguiar NO, Campostrini E, Rosa RCC, Façanha AR,
Olivares FL (2013) A combination of humic substances and Herbaspirillum seropedicae
inoculation enhances the growth of maize (Zea mays L.). Plant Soil 366:119–132
Cappellari LDR, Santoro MV, Nievas F, Giordano W, Banchio E (2013) Increase of secondary
metabolite content in marigold by inoculation with plant growth-promoting rhizobacteria. Appl
Soil Ecol 70:16–22
Carvalhais LC, Dennis PG, Fan B, Fedoseyenko D, Kierul K, Becker A, von Wiren N, Borriss R
(2013) Linking plant nutritional status to plant-microbe interactions. PLoS ONE 8:e68555
Carvalhais LC, Dennis PG, Fedoseyenko D, Hajirezaei MR, Borriss R, von Wirén N (2011) Root
exudation of sugars, amino acids, and organic acids by maize as affected by nitrogen,
phosphorus, potassium, and iron deficiency. J Plant Nutr Soil Sci 174:3–11. doi:10.1002/jpln.
201000085
188 M.W. Heaven and D. Benheim
Cassman KG (1999) Ecological intensification of cereal production systems: yield potential, soil
quality, and precision agriculture. Proc Natl Acad Sci 96:5952–5959. doi:10.1073/pnas.96.11.
5952
Cesco S, Mimmo T, Tonon G, Tomasi N, Pinton R, Terzano R, Neumann G, Weisskopf L,
Renella G, Landi L, Nannipieri P (2012) Plant-borne flavonoids released into the rhizosphere:
impact on soil bio-activities related to plant nutrition. A review. Biol Fertil Soils 48:123–149.
doi:10.1007/s00374-011-0653-2
Chamam A, Sanguin H, Bellvert F, Meiffren G, Comte G, Wisniewski-Dye F, Bertrand C,
Prigent-Combaret C (2013) Plant secondary metabolite profiling evidences strain-dependent
effect in the Azospirillum-Oryza sativa association. Phytochemistry 87:65–77
Chan KG, Atkinson S, Mathee K, Sam CK, Chhabra SR, Cámara M, Koh CL, Williams P (2011)
Characterization of N-acyl-homoserine lactone-degrading bacteria associated with the Zingiber
officinale (ginger) rhizosphere: co-existence of quorum quenching and quorum sensing in
Acinetobacter and Burkholderia. BMC Microbiol 11:1–14. doi:10.1186/1471-2180-11-51
Chaparro JM, Badri DV, Bakker MG, Sugiyama A, Manter DK, Vivanco JM (2013) Root
exudation of phytochemicals in Arabidopsis follows specific patterns that are developmentally
programmed and correlate with soil microbial functions. PLoS ONE 8:e55731
Cheynier V, Comte G, Davies KM, Lattanzio V, Martens S (2013) Plant phenolics: recent
advances on their biosynthesis, genetics, and ecophysiology. Plant Physiol Biochem 72:1–20.
doi:10.1016/j.plaphy.2013.05.009
Clair SB, Lynch JP (2010) The opening of pandora’s box: climate change impacts on soil fertility
and crop nutrition in developing countries. Plant Soil 335:101–115
Coats VC, Rumpho ME (2014) The rhizosphere microbiota of plant invaders: an overview of
recent advances in the microbiomics of invasive plants. Front Microbiol 5:368
Conrad R (1996) Soil microorganisms as controllers of atmospheric trace gases (H2, CO, CH4,
OCS, N2O, and NO). Microbiol Rev 60:609–640
Contreras-Cornejo HA, Macías-Rodríguez L, Beltrán-Peña E, Herrera-Estrella A, López-Bucio J
(2011) Trichoderma-induced plant immunity likely involves both hormonal and camalexin
dependent mechanisms in Arabidopsis thaliana and confers resistance against necrotrophic
fungi Botrytis cinerea. Plant Signaling Behav 6:1554–1563
Contreras-Cornejo HA, Ortiz-Castro R, Lopez-Bucio J (2013) Promotion of plant growth and the
induction of systemic defence by Trichoderma: physiology, genetics and gene expression. In:
Mukherjee PK, Horwitz BA, Singh US, Mukherjee M, Schmoll M (eds) Trichoderma: biology
and applications. CABI, Oxfordshire
Cooper JE (2007) Early interactions between legumes and rhizobia: disclosing complexity in a
molecular dialogue. J Appl Microbiol 103:1355–1365. doi:10.1111/j.1365-2672.2007.03366.x
Coûteaux MM, Bottner P, Berg B (1995) Litter decomposition, climate and liter quality. Trends
Ecol Evol 10:63–66. doi:10.1016/S0169-5347(00)88978-8
Crowther TW, Boddy L, Jones TH (2012) Functional and ecological consequences of saprotrophic
fungus–grazer interactions. ISME J 6:1992–2001
Dakora FD, Phillips DA (2002) Root exudates as mediators of mineral acquisition in low-nutrient
environments. Plant Soil 245:35–47. doi:10.1023/a:1020809400075
Das K, Mukherjee AK (2007) Crude petroleum-oil biodegradation efficiency of Bacillus subtilis
and Pseudomonas aeruginosa strains isolated from a petroleum-oil contaminated soil from
north-east India. Bioresour Technol 98:1339–1345. doi:10.1016/j.biortech.2006.05.032
De-la-Pena C, Loyola-Vergas VM (2014) Biotic interactions in the rhizosphere: a diverse
cooperative enterprise for plant productivity. Plant Physiol 166:701–719
del Mónaco SM, Rodríguez ME, Lopes CA (2016) Pichia kudriavzevii as a representative yeast of
north patagonian winemaking terroir. Int J Food Microbiol 230:31–39. doi:10.1016/j.
ijfoodmicro.2016.04.017
Delort AM, Combourieu B (2001) In situ 1H NMR study of the biodegradation of xenobiotics:
application to heterocyclic compounds. J Ind Microbiol Biotech 26:2–8. doi:10.1038/sj.jim.
7000022
6 Soil Microbial Metabolomics 189
Dennis PG, Miller AJ, Hirsch PR (2010) Are root exudates more important than other sources of
rhizodeposits in structuring rhizosphere bacterial communities? FEMS Microbiol Ecol 72:313–
327. doi:10.1111/j.1574-6941.2010.00860.x
Desai C, Pathak H, Madamwar D (2010) Advances in molecular and “-omics” technologies to
gauge microbial communities and bioremediation at xenobiotic/anthropogen contaminated
sites. Bioresour Technol 101:1558–1569. doi:10.1016/j.biortech.2009.10.080
Dhawi F, Datta R, Ramakrishna W (2015) Mycorrhiza and PGPB modulate maize biomass,
nutrient uptake and metabolic pathways in maize grown in mining-impacted soil. Plant Physiol
Biochem 97:390–399. doi:10.1016/j.plaphy.2015.10.028
Di Marco G, Gismondi A, Canuti L, Scimeca M, Volpe A, Canini A (2014) Tetracycline
accumulates in Iberis sempervirens L. through apoplastic transport inducing oxidative stress
and growth inhibition. Plant Biol 16:792–800. doi:10.1111/plb.12102
Dinesh R, Anandaraj M, Srinivasan V, Hamza S (2012) Engineered nanoparticles in the soil and
their potential implications to microbial activity. Geoderma 173–174:19–27. doi:10.1016/j.
geoderma.2011.12.018
Edenhofer OR, Pichs-Madruga Y, Sokona E, Farahani S, Kadner K, Seyboth A, Adler I, Baum S,
Brunner P, Eickemeier B, Kriemann J, Savolainen S, Schlömer, C von Stechow, Zwickel T
(eds) IPCC, 2014: climate change 2014: mitigation of climate change. Contribution of working
group III to the fifth assessment report of the Intergovernmental Panel on Climate Change,
Cambridge, UK and New York, NY, USA
Ewert F, Rounsevell MDA, Reginster I, Metzger MJ, Leemans R (2005) Future scenarios of
European agricultural land use: I. estimating changes in crop productivity. Agric Ecosyst
Environ 107:101–116. doi:10.1016/j.agee.2004.12.003
Foley JA, DeFries R, Asner GP, Barford C, Bonan G, Carpenter SR, Chapin FS, Coe MT,
Daily GC, Gibbs HK, Helkowski JH, Holloway T, Howard EA, Kucharik CJ, Monfreda C,
Patz JA, Prentice IC, Ramankutty N, Snyder PK (2005) Global consequences of land use.
Science 309:570–574. doi:10.1126/science.1111772
Food and Agriculture Organization (FAO) (2002) World agriculture towards 2015/2030: Summary
report. In: Food and Agriculture Organization of the United Nations (ed). Food and Agriculture
Organization (FAO), Rome
Food and Agriculture Organization of the United Nations (2013) World agriculture towards
2030/2050. In: Alexandratos N, Bruinsma J (ed) ESA Working paper No 12–03. Rome, Italy
Frisvad JC, Smedsgaard J, Larsen TO, Samson RA (2004) Mycotoxins, drugs and other extrolites
produced by species in Penicillium subgenus Penicillium. Stud Mycol 2004:201–241
Gamir J, Pastor V, Kaever A, Cerezo M, Flors V (2014) Targeting novel chemical and constitutive
primed metabolites against Plectosphaerella cucumerina. Plant J 78:227–240. doi:10.1111/tpj.
12465
Garcia-Sánchez M, Garcia-Romera I, Cajthaml T, Tlustoš P, Száková J (2015) Changes in soil
microbial community functionality and structure in a metal-polluted site: the effect of digestate
and fly ash applications. J Environ Manage 162:63–73. doi:10.1016/j.jenvman.2015.07.042
Geras’kin SA, Fesenko SV, Alexakhin RM (2008) Effects of non-human species irradiation after
the chernobyl NPP accident. Environ Int 34:880–897. doi:10.1016/j.envint.2007.12.012
González-Fernández R, Valero-Galván J, Gómez-Gálvez FJ, Jorrín-Novo JV (2015) Unraveling
the in vitro secretome of the phytopathogen Botrytis cinerea to understand the interaction with
its hosts. Front Plant Sci 6. doi:10.3389/fpls.2015.00839
Gunina A, Dippold MA, Glaser B, Kuzyakov Y (2014) Fate of low molecular weight organic
substances in an arable soil: from microbial uptake to utilisation and stabilisation. Soil Biol
Biochem 77:304–313. doi:10.1016/j.soilbio.2014.06.029
Habib MT, Heller T, Polle A (2013) Molecular physiology of tree ectomycorrhizal interactions. In:
Eshel A, Beckman T (eds) Plant roots: the hidden half, 4th edn. CRC Press, Boca Raton
FEZ Haichar, Roncato MA, Achouak W (2012) Stable isotope probing of bacterial community
structure and gene expression in the rhizosphere of Arabidopsis thaliana. FEMS Microbiol
Ecol 81:291–302. doi:10.1111/j.1574-6941.2012.01345.x
190 M.W. Heaven and D. Benheim
Hartmann A, Schmid M, Dv Tuinen, Berg G (2008) Plant-driven selection of microbes. Plant Soil
321:235–257. doi:10.1007/s11104-008-9814-y
Hernandez-Soriano MC, Jimenez-Lopez JC (2014) Metabolomics for soil contamination
assessment
Heuck C, Weig A, Spohn M (2015) Soil microbial biomass C:N: P stoichiometry and microbial
use of organic phosphorus. Soil Biol Biochem 85:119–129. doi:10.1016/j.soilbio.2015.02.029
Hodge A, Fitter AH (2010) Substantial nitrogen acquisition by arbuscular mycorrhizal fungi from
organic material has implications for N cycling. Proc Nat Acad Sci USA 107:13754–13759
Holland JM (2004) The environmental consequences of adopting conservation tillage in Europe:
reviewing the evidence. Agric Ecosyst Environ 103:1–25. doi:10.1016/j.agee.2003.12.018
Horrigan L, Lawrence RS, Walker P (2002) How sustainable agriculture can address the
environmental and human health harms of industrial agriculture. Environ Health Perspect
110:445–456
Huang X-F, Chaparro JM, Reardon KF, Zhang R, Shen Q, Vivanco JM (2014) Rhizosphere
interactions: root exudates, microbes, and microbial communities. Botany 92:267–275
Imfeld G, Vuilleumier S (2012) Measuring the effects of pesticides on bacterial communities in
soil: a critical review. Eur J Soil Biology 49:22–30. doi:10.1016/j.ejsobi.2011.11.010
Itoh Y, Kodama K, Furuya K, Takahashi S, Haneishi T, Takiguchi Y, Arai M (1980) A new
sesquiterpene antibiotic, heptelidic acid producing organisms, fermentation, isolation and
characterization. J Antibiot 33:468–473
Jha P, Panwar J, Jha PN (2015) Secondary plant metabolites and root exudates: guiding tools for
polychlorinated biphenyl biodegradation. Int J Environ Sci Technol 12:789–802. doi:10.1007/
s13762-014-0515-1
Jia X, Liu C, Song H, Ding M, Du J, Ma Q, Yuan Y (2016) Design, analysis and application of
synthetic microbial consortia. Synth Syst Biotechnol 1:109–117. doi:10.1016/j.synbio.2016.
02.001
Jin L, Son Y, DeForest JL, Kang YJ, Kim W, Chung H (2014) Single-walled carbon nanotubes
alter soil microbial community composition. Sci Total Environ 466–467:533–538. doi:10.
1016/j.scitotenv.2013.07.035
Jin L, Son Y, Yoon TK, Kang YJ, Kim W, Chung H (2013) High concentrations of single-walled
carbon nanotubes lower soil enzyme activity and microbial biomass. Ecotoxicol Environ Saf
88:9–15. doi:10.1016/j.ecoenv.2012.10.031
Joanisse GD, Bradley RL, Preston CM, Munson AD (2007) Soil enzyme inhibition by condensed
litter tannins may drive ecosystem structure and processes: the case of Kalmia angustifolia.
New Phytol 175:535–546. doi:10.1111/j.1469-8137.2007.02113.x
Johansen A, Pedersen AL, Jensen KA, Karlson U, Hansen BM, Scott-Fordsmand JJ, Winding A
(2008) Effects of C60 fullerene nanoparticles on soil bacteria and protozoans. Environ Toxicol
Chem 27:1895–1903. doi:10.1897/07-375.1
Jones DL (1998) Organic acids in the rhizosphere—a critical review. Plant Soil 205:25–44. doi:10.
1023/a:1004356007312
Jones OAH, Sdepanian S, Lofts S, Svendsen C, Spurgeon DJ, Maguire ML, Griffin JL (2014)
Metabolomic analysis of soil communities can be used for pollution assessment. Environ
Toxicol Chem 33:61–64. doi:10.1002/etc.2418
Jonkers W, Rodriguez Estrada AE, Lee K, Breakspear A, May G, Kistler HC (2012) Metabolome
and transcriptome of the Interaction between Ustilago maydis and Fusarium verticillioides In
Vitro. Appl Environ Microbiol 78:3656–3667. doi:10.1128/aem.07841-11
Kabouw P, van Dam NM, van der Putten WH, Biere A (2012) How genetic modification of roots
affects rhizosphere processes and plant performance. J Exp Bot 63:3475–3483. doi:10.1093/
jxb/err399
Kakumanu ML, Cantrell CL, Williams MA (2013) Microbial community response to varying
magnitudes of desiccation in soil: a test of the osmolyte accumulation hypothesis. Soil Biol
Biochem 57:644–653. doi:10.1016/j.soilbio.2012.08.014
Karimi A, Khodaverdiloo H, Sepehri M, Sadaghiani MR (2011) Arbuscular mycorrhizal fungi and
heavy metal contaminated soils. Afr J Micro Res 5:1571–1576
6 Soil Microbial Metabolomics 191
Lin CY, Viant MR, Tjeerdema RS (2006) Metabolomics: methodologies and applications in the
environmental sciences. J Pestic Sci 31:245–251. doi:10.1584/jpestics.31.245
Ling LL, Schneider T, Peoples AJ, Spoering AL, Engels I, Conlon BP, Mueller A, Schaberle TF,
Hughes DE, Epstein S, Jones M, Lazarides L, Steadman VA, Cohen DR, Felix CR,
Fetterman KA, Millett WP, Nitti AG, Zullo AM, Chen C, Lewis K (2015) A new antibiotic
kills pathogens without detectable resistance. Nature 517:455–459. doi:10.1038/nature14098
Lloyd JR, Pearce CI, Coker VS, Pattrick RAD, Van Der Laan G, Cutting R, Vaughan DJ,
Paterson-Beedle M, Mikheenko IP, Yong P, Macaskie LE (2008) Biomineralization: linking
the fossil record to the production of high value functional materials. Geobiology 6:285–297.
doi:10.1111/j.1472-4669.2008.00162.x
Loh J, Carlson RW, York WS, Stacey G (2002) Bradyoxetin, a unique chemical signal involved in
symbiotic gene regulation. Proc Nat Aca Sci 99:14446–14451. doi:10.1073/pnas.222336799
López-Bucio J, Pelagio-Flores R, Herrera-Estrella A (2015) Trichoderma as biostimulant:
exploiting the multilevel properties of a plant beneficial fungus. Sci Hortic 196:109–123.
doi:10.1016/j.scienta.2015.08.043
Lorenz MG, Wackernagel W (1994) Bacterial gene transfer by natural genetic transformation in
the environment. Microbiol Rev 58:563–602
Lorito M, Woo SL, Harman Gary E, Monte E (2010) Translational research on trichoderma: from
‘omics to the field. Annu Rev Phytopathol 48:395–417. doi:10.1146/annurev-phyto-073009-
114314
Lozupone CA, Knight R (2007) Global patterns in bacterial diversity. Proc Nat Aca Sci
104:11436–11440. doi:10.1073/pnas.0611525104
Lugan R, Niogret M-F, Leport L, Guégan J-P, Larher FR, Savouré A, Kopka J, Bouchereau A
(2010) Metabolome and water homeostasis analysis of Thellungiella salsuginea suggests that
dehydration tolerance is a key response to osmotic stress in this halophyte. Plant J 64:215–229.
doi:10.1111/j.1365-313X.2010.04323.x
Lumbsch HT, Leavitt SD (2011) Goodbye morphology? a paradigm shift in the delimitation of
species in lichenized fungi. Fungal Divers 50:59–72. doi:10.1007/s13225-011-0123-z
Luo ZB, Janz D, Jiang XN, Gobel C, Wildhagen H, Tan Y, Rennenberg H, Feussner I, Polle A
(2009) Upgrading root physiology for stress tolerance by ectomycorrhizas: insights from
metabolite and transcriptional profiling into reprogramming for stress anticipation. Plant
Physiol 151:1902–1917
Lynch JP (2007) Roots of the second green revolution. Aust J Bot 55:493–512. doi:10.1071/
BT06118
MacCormack TJ, Goss GG (2008) Identifying and predicting biological risks associated with
manufactured nanoparticles in aquatic ecosystems. J Ind Ecol 12:286–296. doi:10.1111/j.1530-
9290.2008.00041.x
Mandal SM, Chakraborty D, Dey S (2010) Phenolic acids act as signaling molecules in
plant-microbe symbioses. Plant Signal Behav 5:359–368. doi:10.4161/psb.5.4.10871
Manlay RJ, Feller C, Swift MJ (2007) Historical evolution of soil organic matter concepts and
their relationships with the fertility and sustainability of cropping systems. Agric Ecosyst
Environ 119:217–233. doi:10.1016/j.agee.2006.07.011
Maron P-A, Mougel C, Ranjard L (2011) Soil microbial diversity: methodological strategy, spatial
overview and functional interest. C R Biol 334:403–411. doi:10.1016/j.crvi.2010.12.003
Marshall TJ, Holmes JW (1980) Soil physics. Cambridge University Press, Cambridge
Mastouri F, Björkman T, Harman GE (2010) Seed treatment with Trichoderma harzianum
alleviates biotic, abiotic, and physiological stresses in germinating seeds and seedlings.
Phytopathology 100:1213–1221
Matson PA, Parton WJ, Power AG, Swift MJ (1997) Agricultural Intensification and Ecosystem
Properties. Science 277:504–509. doi:10.1126/science.277.5325.504
Maurer-Jones MA, Gunsolus IL, Murphy CJ, Haynes CL (2013) Toxicity of engineered
nanoparticles in the environment. Anal Chem 85:3036–3049. doi:10.1021/ac303636s
McBride MB (1994) Environmental chemistry of soils. Oxford university press, Oxford
6 Soil Microbial Metabolomics 193
Pettibone JM, Louie SM (2015) Research highlights: detecting, characterizing and quantifying the
presence and impact of carbon nanomaterials in environmental systems. Environ Sci Nano
2:308–311. doi:10.1039/c5en90014j
Pishchik VN, Vorobyev NI, Chernyaeva II, Timofeeva SV, Kozhemyakov AP, Alexeev YV,
Lukin SM (2002) Experimental and mathematical simulation of plant growth promoting
rhizobacteria and plant interaction under cadmium stress. Plant Soil 243:173–186. doi:10.1023/
a:1019941525758
Ponge J-F (2013) Plant–soil feedbacks mediated by humus forms: a review. Soil Biol Biochem
57:1048–1060. doi:10.1016/j.soilbio.2012.07.019
Ponomarova O, Patil KR (2015) Metabolic interactions in microbial communities: untangling the
Gordian knot. Curr Opin Microbiol 27:37–44. doi:10.1016/j.mib.2015.06.014
Rabie GH (1998) Induction of fungal disease resistance in Vicia faba by dual inoculation with
Rhizobium leguminosarum and vesicular-arbuscular mycorrhizal fungi. Mycopathologia
141:159–166. doi:10.1023/a:1006937821777
Rasmussen S, Parsons AJ, Jones CS (2012) Metabolomics of forage plants: a review. Ann Bot
110:1281–1290. doi:10.1093/aob/mcs023
Rastogi G, Sani R (2011) Molecular techniques to assess microbial community structure, function,
and dynamics in the environment. In: Ahmad I, Ahmad F, Pichtel J (eds) Microbes and
microbial technology. Springer, New York
Rawat L, Singh Y, Shukla N, Kumar J (2011) Alleviation of the adverse effects of salinity stress in
wheat (Triticum aestivum L.) by seed biopriming with salinity tolerant isolates of Trichoderma
harzianum. Plant Soil 347:387–400. doi:10.1007/s11104-011-0858-z
Redmond JW, Batley M, Djordjevic MA, Innes RW, Kuempel PL, Rolfe BG (1986) Flavones
induce expression of nodulation genes in Rhizobium. Nature 323:632–635
Rennenberg H, Dannenmann M, Gessler A, Kreuzwieser J, Simon J, Papen H (2009) Nitrogen
balance in forest soils: nutritional limitation of plants under climate change stresses. Plant Biol
11:4–23. doi:10.1111/j.1438-8677.2009.00241.x
Reynolds HL, Packer A, Bever JD, Clay K (2003) Grassroots ecology: plant-microbe-soil
interactions as drivers of plant community structure and dynamics. Ecology 84:2281–2291
Řezanka T, Sigler K (2009) Odd-numbered very-long-chain fatty acids from the microbial, animal
and plant kingdoms. Prog Lipid Res 48:206–238. doi:10.1016/j.plipres.2009.03.003
Riding MJ, Herbert BMJ, Ricketts L, Dodd I, Ostle N, Semple KT (2015) Harmonising conflicts
between science, regulation, perception and environmental impact: the case of soil conditioners
from bioenergy. Environ Int 75:52–67. doi:10.1016/j.envint.2014.10.025
Rivero J, Gamir J, Aroca R, Pozo MJ, Flors V (2015) Metabolic transition in mycorrhizal tomato
roots. Front Microbiol 6:598
Roberts WK, Selitrennikoff CP (1988) Plant and bacterial chitinases differ in antifungal activity.
Microbiology 134:169–176. doi:10.1099/00221287-134-1-169
Rochfort S, Ezernieks V, Mele P, Kitching M (2015) NMR metabolomics for soil analysis provide
complementary, orthogonal data to MIR and traditional soil chemistry approaches—a land use
study. Magn Reson Chem 53:719–725. doi:10.1002/mrc.4187
Rochfort S, Ezernieks V, Yen A (2009) NMR-based metabolomics using earthworms as potential
indicators for soil health. Metabolomics 5:95–107. doi:10.1007/s11306-008-0140-4
Rockström J, Steffen W, Noone K, Persson A, Chapin Iii FS, Lambin E, Lenton TM, Scheffer M,
Folke C, Schellnhuber HJ, Nykvist B, de Wit CA, Hughes T, van der Leeuw S, Rodhe H,
Sörlin S, Snyder PK, Costanza R, Svedin U, Falkenmark M, Karlberg L, Corell RW, Fabry VJ,
Hansen J, Walker B, Liverman D, Richardson K, Crutzen P, Foley J (2009) Planetary
boundaries: exploring the safe operating space for humanity. Ecol Soc 14
Rudresh DL, Shivaprakash MK, Prasad RD (2005) Tricalcium phosphate solubilizing abilities of
Trichoderma spp. in relation to P uptake and growth and yield parameters of chickpea (Cicer
arietinum L.). Can J Microbiol 51:217–222
Ruzicka D, Hausmann N, Barrios-Masias F, Jackson L, Schachtman D (2012) Transcriptomic and
metabolic responses of mycorrhizal roots to nitrogen patches under field conditions. Plant Soil
350:145–162. doi:10.1007/s11104-011-0890-z
6 Soil Microbial Metabolomics 195
Saharan BS, Nehra V (2011) Plant growth promoting rhizobacteria: a critical review. Life Sci Med
Res 21:1–30
Saia S, Rappa V, Ruisi P, Abenavoli MR, Sunseri F, Giambalvo D, Frenda AS, Martinelli F
(2015a) Soil inoculation with symbiotic microorganisms promotes plant growth and nutrient
transporter genes expression in durum wheat. Front Plant Sci 6:815. doi:10.3389/fpls.2015.
00815
Saia S, Ruisi P, Fileccia V, Di Miceli G, Amato G, Martinelli F (2015b) Metabolomics suggests
that soil inoculation with arbuscular mycorrhizal fungi decreased free amino acid content in
roots of durum wheat grown under n-limited. P-Rich Field Cond. PLoS ONE 10:e0129591.
doi:10.1371/journal.pone.0129591
Samolski I, Rincón AM, Pinzón LM, Viterbo A, Monte E (2012) The qid74 gene from
Trichoderma harzianum has a role in root architecture and plant biofertilization. Microbiology
158:129–138
Sanchez DH, Siahpoosh MR, Roessner U, Udvardi M, Kopka J (2008) Plant metabolomics reveals
conserved and divergent metabolic responses to salinity. Physiol Plant 132:209–219. doi:10.
1111/j.1399-3054.2007.00993.x
Sardans J, Peñuelas J, Rivas-Ubach A (2011) Ecological metabolomics: overview of current
developments and future challenges. Chemoecology 21:191–225. doi:10.1007/s00049-011-
0083-5
Sarry J-E, Kuhn L, Ducruix C, Lafaye A, Junot C, Hugouvieux V, Jourdain A, Bastien O,
Fievet JB, Vailhen D, Amekraz B, Moulin C, Ezan E, Garin J, Bourguignon J (2006) The early
responses of Arabidopsis thaliana cells to cadmium exposure explored by protein and
metabolite profiling analyses. Proteomics 6:2180–2198. doi:10.1002/pmic.200500543
Sasser M (2006) Bacterial identification by gas chromatographic analysis of fatty acids methyl
esters (GC-FAME). Microbial ID Inc
Scandiani MM, Luque AG, Razori MV, Ciancio Casalini L, Aoki T, O’Donnell K, Cervigni GDL,
Spampinato CP (2015) Metabolic profiles of soybean roots during early stages of Fusarium
tucumaniae infection. J Exp Bot 66:391–402. doi:10.1093/jxb/eru432
Schlesinger WH (1991) Biogeochemistry: an analysis of global change. Academic Press, San
Diego
Schmidt MWI, Torn MS, Abiven S, Dittmar T, Guggenberger G, Janssens IA, Kleber M,
Kogel-Knabner I, Lehmann J, Manning DAC, Nannipieri P, Rasse DP, Weiner S, Trumbore SE
(2011) Persistence of soil organic matter as an ecosystem property. Nature 478:49–56
Schützendübel A, Polle A (2002) Plant responses to abiotic stresses: heavy metal-induced
oxidative stress and protection by mycorrhization. J Exp Bot 53:1351–1365
Segarra G, Casanova E, Bellido D, Odena MA, Oliveira E, Trillas I (2007) Proteome, salicylic acid
and jasmonic acid changes in cucumber plants inoculated with Trichoderma asperellum strain
T34. Proteomics 7:3943–3952
Selosse M-A, Strullu-Derrien C (2015) Origins of terrestrial flora: a symbiosis with fungi? BIO
Web Conf 4:1–12
Seo J-S, Keum Y-S, Li Q (2009) Bacterial degradation of aromatic compounds. Int J Environ Res
Pub Health 6:278
Shah V, Belozerova I (2009) Influence of metal nanoparticles on the soil microbial community and
germination of lettuce seeds. Water Air Soil Pollut 197:143–148. doi:10.1007/s11270-008-
9797-6
Simpson AJ, Simpson MJ, Soong R (2012) Nuclear magnetic resonance spectroscopy and its key
role in environmental research. Environ Sci Technol 46:11488–11496. doi:10.1021/es302154w
Simpson M, McKelvie J (2009) Environmental metabolomics: new insights into earthworm
ecotoxicity and contaminant bioavailability in soil. Anal Bioanal Chem 394:137–149. doi:10.
1007/s00216-009-2612-4
Singh OV (2006) Proteomics and metabolomics: the molecular make-up of toxic aromatic
pollutant bioremediation. Proteomics 6:5481–5492. doi:10.1002/pmic.200600200
196 M.W. Heaven and D. Benheim
Singh S, Parihar P, Singh R, Singh VP, Prasad SM (2016) Heavy metal tolerance in plants: role of
transcriptomics, proteomics, metabolomics and ionomics. Front Plant Sci 6. doi:10.3389/fpls.
2015.01143
Sinsabaugh RL, Turner BL, Talbot JM, Waring BG, Powers JS, Kuske CR, Moorhead DL,
Follstad Shah JJ (2016) Stoichiometry of microbial carbon use efficiency in soils. Ecol Monogr
86:172–189. doi:10.1890/15-2110.1
Sláviková E, Vadkertiová R (2003) The diversity of yeasts in the agricultural soil. J Basic
Microbiol 43:430–436. doi:10.1002/jobm.200310277
Smith FA, Smith SE (2015) How harmonious are arbuscular mycorrhizal symbioses? inconsistent
concepts reflect different mindsets as well as results. New Phytol 205:1381–1384
Smith SE, Jakobsen I, Gronlund M, Smith FA (2011) Roles of arbuscular mycorrhizas in plant
phosphorus nutrition: interactions between pathways of phosphorus uptake in arbuscular
mycorrhizal roots have important implications for understanding and manipulating plant
phosphorus acquisition. Plant Physiol 156:1050–1057
Souza LA, Camargos LS, Schiavinato MA, Andrade SAL (2014) Mycorrhization alters foliar
soluble amino acid composition and influences tolerance to Pb in Calopogonium mucunoides.
Theoret Exp Plant Physiol 26:211–216. doi:10.1007/s40626-014-0019-x
Stefaniuk M, Oleszczuk P, Ok YS (2016) Review on nano zerovalent iron (nZVI): from synthesis
to environmental applications. Chem Eng J 287:618–632. doi:10.1016/j.cej.2015.11.046
Stone M (2009) Metal balance helps explain survival of microbial “superhero”. Microbe 4:
164–165
Styger G, Prior B, Bauer FF (2011) Wine flavour and aroma. J Ind Microbiol Biotech 38:
1145–1159
Sutton R, Sposito G (2005) Molecular structure in soil humic substances: the new view. Environ
Sci Technol 39:9009–9015. doi:10.1021/es050778q
Swenson TL, Bowen BP, Nico PS, Northen TR (2015a) Competitive sorption of microbial
metabolites on an iron oxide mineral. Soil Biol Biochem 90:34–41. doi:10.1016/j.soilbio.2015.
07.022
Swenson TL, Jenkins S, Bowen BP, Northen TR (2015b) Untargeted soil metabolomics methods
for analysis of extractable organic matter. Soil Biol Biochem 80:189–198. doi:10.1016/j.
soilbio.2014.10.007
Tate RL (2000) Soil Microbiology. Wiley, New York
Tepper CS, Anderson AJ (1990) Interactions between pectic fragments and extracellular
components from the fungal pathogen Colletotrichum lindemuthianum. Phys Mol Plant Pathol
36:147–158. doi:10.1016/0885-5765(90)90103-5
Tilman D, Cassman KG, Matson PA, Naylor R, Polasky S (2002) Agricultural sustainability and
intensive production practices. Nature 418:671–677
Tittonell P (2014) Ecological intensification of agriculture—sustainable by nature. Curr Opin
Environ Sustain 8:53–61
Torsvik V, Øvreås L (2002) Microbial diversity and function in soil: from genes to ecosystems.
Curr Opin Microbiol 5:240–245. doi:10.1016/S1369-5274(02)00324-7
Tremaroli V, Workentine ML, Weljie AM, Vogel HJ, Ceri H, Viti C, Tatti E, Zhang P, Hynes AP,
Turner RJ, Zannoni D (2009) Metabolomic investigation of the bacterial response to a metal
challenge. Appl Environ Microbiol 75:719–728. doi:10.1128/aem.01771-08
Valentinuzzi F, Pii Y, Vigani G, Lehmann M, Cesco S, Mimmo T (2015) Phosphorus and iron
deficiencies induce a metabolic reprogramming and affect the exudation traits of the woody
plant Fragaria ananassa. J Exp Bot 66:6483–6495. doi:10.1093/jxb/erv364
van Dam NM, Bouwmeester HJ (2016) Metabolomics in the rhizosphere: tapping into
belowground chemical communication. Trends Plant Sci 21:256–265. doi:10.1016/j.tplants.
2016.01.008
van den Berg EM, van Dongen U, Abbas B, van Loosdrecht MCM (2015) Enrichment of DNRA
bacteria in a continuous culture. ISME J 9:2153–2161. doi:10.1038/ismej.2015.26
6 Soil Microbial Metabolomics 197
Van Der Heijden MGA, Bardgett RD, Van Straalen NM (2008) The unseen majority: soil
microbes as drivers of plant diversity and productivity in terrestrial ecosystems. Ecol Lett
11:296–310. doi:10.1111/j.1461-0248.2007.01139.x
Van Der Heijden MGA, Klironomos JN, Ursic M, Moutoglis P, Streitwolf-Engel R, Boller T,
Wiemken A, Sanders IR (1998) Mycorrhizal fungal diversity determines plant biodiversity,
ecosystem variability and productivity. Nature 396:69–72. doi:10.1038/23932
van Leeuwen C, Seguin G (2006) The concept of terroirs in viticulture. Wine Res 17:1–10
Vinale F, Sivasithamparam K, Ghisalberti EL, Marra R, Woo SL, Lorito M (2008) Trichoderma–
plant–pathogen interactions. Soil Biol Biochem 40:1–10. doi:10.1016/j.soilbio.2007.07.002
Walker TS, Bais HP, Grotewold E, Vivanco JM (2003) Root exudation and rhizosphere biology.
Plant Physiol 132:44–51. doi:10.1104/pp.102.019661
Walker V, Bertrand C, Bellvert F, Moënne-Loccoz Y, Bally R, Comte G (2011) Host plant
secondary metabolite profiling shows a complex, strain-dependent response of maize to plant
growth-promoting rhizobacteria of the genus Azospirillum. New Phytol 189:494–506
Wallenstein MD, Haddix ML, Ayres E, Steltzer H, Magrini-Bair KA, Paul EA (2013) Litter
chemistry changes more rapidly when decomposed at home but converges during decompo-
sition–transformation. Soil Biol Biochem 57:311–319. doi:10.1016/j.soilbio.2012.09.027
Wallenstein MD, Hess AM, Lewis MR, Steltzer H, Ayres E (2010) Decomposition of aspen leaf
litter results in unique metabolomes when decomposed under different tree species. Soil Biol
Biochem 42:484–490. doi:10.1016/j.soilbio.2009.12.001
Wallenstein MD, Weintraub MN (2008) Emerging tools for measuring and modeling the in situ
activity of soil extracellular enzymes. Soil Biol Biochem 40:2098–2106. doi:10.1016/j.soilbio.
2008.01.024
Wang W, Li Y, Wang H, Zu Y (2014) Differences in the activities of eight enzymes from ten soil
fungi and their possible influences on the surface structure, functional groups, and element
composition of soil colloids. PLoS ONE 9:e111740. doi:10.1371/journal.pone.0111740
Wania F, Mackay D (1996) Tracking the distribution of persistent organic pollutants. Environ Sci
Technol 30:390A–397A
Warren CR (2013) High diversity of small organic N observed in soil water. Soil Biol Biochem
57:444–450. doi:10.1016/j.soilbio.2012.09.025
Warren CR (2014) Organic N molecules in the soil solution: what is known, what is unknown and
the path forwards. Plant Soil 375:1–19. doi:10.1007/s11104-013-1939-y
Warren CR (2015) Comparison of methods for extraction of organic N monomers from soil
microbial biomass. Soil Biol Biochem 81:67–76. doi:10.1016/j.soilbio.2014.11.005
Watzinger A (2015) Microbial phospholipid biomarkers and stable isotope methods help reveal
soil functions. Soil Biol Biochem 86:98–107. doi:10.1016/j.soilbio.2015.03.019
Wehner J, Antunes PM, Powell JR, Mazukatow J, Rillig MC (2010) Plant pathogen protection by
arbuscular mycorrhizas: a role for fungal diversity? Pedobiologia 53:197–201
Whitfield MÅ, Stephenson GL, Simpson AJ, Simpson MJ (2013) Comparison of earthworm
responses to petroleum hydrocarbon exposure in aged field contaminated soil using traditional
ecotoxicity endpoints and 1H NMR-based metabolomics. Environ Pollut 182:263–268. doi:10.
1016/j.envpol.2013.07.026
Wieland G, Neumann R, Backhaus H (2001) Variation of microbial communities in soil,
rhizosphere, and rhizoplane in response to crop species, soil type, and crop development. Appl
Environ Microbiol 67:5849–5854
Wissuwa M, Mazzola M, Picard C (2008) Novel approaches in plant breeding for
rhizosphere-related traits. Plant Soil 321:409–430. doi:10.1007/s11104-008-9693-2
Woo SL, Scala F, Ruocco M, Lorito M (2006) The molecular biology of the interactions between
trichoderma spp., phytopathogenic fungi, and plants. Phytopathology 96:181–185. doi:10.
1094/phyto-96-0181
Workentine ML, Harrison JJ, Weljie AM, Tran VA, Stenroos PU, Tremaroli V, Vogel HJ, Ceri H,
Turner RJ (2010) Phenotypic and metabolic profiling of colony morphology variants evolved
from Pseudomonas fluorescens biofilms. Environ Microbiol 12:1565–1577. doi:10.1111/j.
1462-2920.2010.02185.x
198 M.W. Heaven and D. Benheim
Yang M, Kim T-Y, Hwang H-C, Yi S-K, Kim D-H (2008) Development of a palm portable mass
spectrometer. J Am Soc Mass Spectrom 19:1442–1448. doi:10.1016/j.jasms.2008.05.011
Zahir AZ, Arshad M, Frankenberger ET Jr (2004) Plant growth promoting rhizobacteria:
application and respectives in agriculture. Adv Agron 81:97–168
Zhang Y, Ruyter-Spira C, Bouwmeester HJ (2015) Engineering the plant rhizosphere. Curr Opin
Biotechnol 32:136–142. doi:10.1016/j.copbio.2014.12.006
Ziegler M, Engel M, Welzl G, Schloter M (2013) Development of a simple root model to study the
effects of single exudates on the development of bacterial community structure. J Microbiol
Methods 94:30–36
Chapter 7
Community Metabolomics
in Environmental Microbiology
1 Introduction
and a proteome entails the complete set of proteins expressed and modified fol-
lowing their expression by the genome (Wilkins 2009). A very basic outline of a
metabolomics experimental setup is shown in Fig. 1.
The metabolome can be used to study the underlying biochemical responses of
an organism/population/community to an environmental stimulus or combination of
stimuli. However, because both populations and communities of microorganisms
are often difficult to culture, metabolomic analysis of such assemblages must take a
different approach from that taken by studies using tissue or biofluid(s) from larger
organisms such as humans. A new and effective way to do this is to borrow
techniques and approaches from the field of metagenomics.
Metagenomics is a rapidly growing area of the genome sciences founded by
Venter et al. (2004) who hit upon the idea of undertaking the analysis of the total
complement of microbial DNA extracted directly from entire microbial commu-
nities in their native habitats without worrying about which species it came from.
This approach allowed them to sequence such communities in their entirety,
regardless of the ability of individual component members to be cultured in the
laboratory (Handelsman 2004). Early studies had great success. For instance, when
the technique was applied to the analysis of samples from the Sargasso sea over 1.2
million previously unknown genes were identified (Venter et al. 2004).
Metagenomics allows one to see, in ever-increasing detail, the vast microbial and
metabolic diversity that exists in the biosphere. It has been utilised in several
high-profile publications in studies ranging from the aforementioned study of the
genetic diversity of microbes in seawater (Venter et al. 2004) to phosphorus and
carbon management in sewage sludge (Garcia et al. 2006; Sales and Lee 2015), to
the effects of warming on carbon sequestration and lignin oxidation in soil (Feng
et al. 2008).
It is of note, however, that the other omic sciences have yet to replicate this
success. The first detailed study on the NMR-based metabolomics of microbial
communities in environmental samples was published by Jones et al. (2014) who
looked at the effects of pollutants on soil metabolic profiles in old mine sites in the
UK. There have since been a few similar studies including Swenson et al. (2015)
and Rochfort et al. (2015) who both looked at metabolites in the extractable organic
matter in soil, and Llewellyn et al. (2015) who used community metabolomics as a
7 Community Metabolomics in Environmental Microbiology 201
2 Soil Science
and cast samples. However, this work has not been followed up in detail and the
amount of diagnostic information that can be obtained from FT-IR is limited.
A more complicated study was that mentioned above by Jones et al. (2014),
which utilised 1H-nuclear magnetic resonance spectroscopy (NMR)-based com-
munity metabolic profiling to assess the changes in biochemical profiles of com-
munities living in contaminated soils from various sites in the UK, each with very
different physicochemical characteristics (levels of metal contamination, underlying
geology and soil type). Each site could be clearly distinguished on the basis of the
metabolic profile of its microbial community. While some of these site differences
may also have been caused by additional abiotic factors (such as soil type or pH),
pattern recognition analysis of the data showed that both site- and
contaminant-specific effects on the metabolic profiles could be discerned. The study
therefore acts as a proof of principle for the use of community metabolomics of
microbial populations from whole soil samples (rather than single isolates) as a
diagnostic tool for pollution assessment. Assigning peaks in NMR spectra of soil
extracts is difficult but software tools such as Chenomx NMR Suite that overlay
library spectra of known compounds over the sample spectra can help (an example
is shown in Fig. 2). Recently developments in NMR software such as Bayesil,
(http://bayesil.ca/) a web based system that automatically identifies and quantifies
metabolites developed at the University of Alberta (Canada) may also be of help in
future.
Rochfort et al. (2015) also used NMR to assess metabolites in soils. They found
similar results to Jones et al. (2014), although in the Rochfort study there were more
Fig. 2 Section of a 1H NMR of soil extract (black) overlain with NMR spectra of lactate (red) and
valine (blue)
7 Community Metabolomics in Environmental Microbiology 203
resonances as a result of lipids and terpenes in the NMR spectra. The authors put
this down to differences between the extraction methodologies employed in each
study, although it seems equally possible that soil type, condition or land use could
have been responsible. Both the Jones and Rochfort studies demonstrated that soil
extracts could be measured by NMR, and there were no obvious interference or line
broadening as a result of paramagnetic materials that might have been present in the
samples. It is interesting to note that Rochfort et al. also used mid-infrared spec-
troscopy (MIR) to analyse their soil samples and found some differences. Whilst the
NMR metabolomic data were more associated with land use rather than location,
the MIR data were correlated more to location and inorganic chemical analysis. The
results may reflect the differences between NMR and MIR data (the latter technique
is far less sensitive) but may also indicate that it may be beneficial to combine two
or more analytical methods in such studies to ensure a comprehensive soil analysis.
It should also be noted that some microbial metabolites rapidly adsorb to soil and
thus one should also keep in mind that soil physical properties such as mineral
composition, surface area, shape and porosity present a notably different set of
challenges in soil analysis compared to the cell and tissue extractions more common
in metabolomics. Swenson et al. (2015) showed that even after a short incubation,
with water, many metabolites, for example amines and carboxylic acids, could be
adsorbed to soil and/or chelated by metals or other ions in the soil or extraction
medium. Cationic and anionic metabolites seem to be most affected, whilst the
majority of neutral metabolites were recovered. This is an important finding since
metabolites not recovered in the extraction procedure could be overlooked in the
subsequent analysis. Other factors that might affect such binding include soil type,
metabolite-soil contact time, temperature, moisture levels, and which extraction
solvents are used. Swenson et al. (2015) point out that there is, however, a silver
lining to this problem as understanding how specific metabolites bind to soil par-
ticles under differing environmental conditions may also increase our understanding
of how such compounds will behave in response to future climatic changes (e.g.
heat or precipitation) and thereby affect substrate availability and the structure of
soil microbial communities in the future.
Analysing small molecule metabolites, in addition to more abundant macro-
molecules, could also help to increase knowledge of biogeochemical cycles. For
example, 13C-labelled carbon sources could be fed to plants or pumped into/through
the soil and the bacterial metabolome studied to assess where the carbon is trans-
ported or to detect subtle shifts in microbial populations interacting with plant litter.
Indeed, the use of NMR techniques has already provided valuable data supporting
the occurrence, diversity and extent of carbon cycling in the carbohydrate meta-
bolism of microorganisms; for a review see Portais and Delort (2002).
Targeted studies have also been undertaken in order to measure flux rates of
specific metabolic pathways, for example through the use of isotope labelling
studies. Scullion et al. (2003) used FT-IR-based metabolic analysis to assess several
potential foods (oat grain, and fresh and aged tobacco) for the earthworm species
Lubricus terrestris and L. rubellus. As casts aged, there was a predictable microbial
succession (from bacteria to fungi) in both earthworm species. Each species was
204 O.A.H. Jones et al.
also found to differ in terms of their cast microbiology in response to food type.
Analysis of ageing casts by FT-IR spectroscopy indicated greater chemical changes
in casts of L. terrestris than L. rubellus irrespective of food type.
Community metabolomics of soils is potentially very useful since the identifi-
cation of biomarkers indicative of a defined response to a pollutant or pollutants,
before major outward changes become apparent, would be very useful in preventing
damage to a variety of sensitive systems (e.g. eutrophication). This could poten-
tially have applications in testing of contaminated land prior to redevelopment (e.g.
house-building on old industrial sites). It could also be useful for the water industry
since the discharge of toxic substances to the sewage network can have negative
effects on wastewater treatment plants, which rely heavily on bacteria and other
microorganisms to function effectively. Similar research could also identify
microorganisms with favourable metabolisms for bioremediation work.
Community metabolomics is not limited to the terrestrial environment.
Llewellyn et al. (2015) used ‘non-targeted’ community metabolomics to determine
patterns in metabolite profiles associated with particulate organic matter (POM) at
four locations from two long-term monitoring stations in the western English
Channel. They identified a range of compounds including amino-acid derivatives,
diacylglyceryltrimethylhomoserine (DGTS) lipids, oxidised fatty acids (oxylipins),
various glycosylated compounds, oligohexoses, phospholipids, triacylglycerides
(TAGs) and oxidised TAGs. Metabolic profiles varied significantly across the four
locations with the largest differences for both the polar and lipid fractions again
being due to geographic location (and/or time). Smaller differences were also
associated with depth.
The work demonstrated that community metabolomics is not only applicable to
the aquatic, as well as the terrestrial, environment but that it has the potential to
contribute significantly to the comprehensive and unbiased characterisation of
marine microbial populations; particularly if linked to metagenomic data. The
authors also speculated that the oxylipins could have a possible link to the for-
mation of oxygenated volatile organic compounds produced by microbial species
that are important in atmospheric chemistry.
was uniquely tolerant to CuSO4 and WS was uniquely tolerant to AgNO3. These
results suggest that the morphological variants are imbued with unique properties.
To further explore the link between the metabolite profiles and metal tolerance,
another set of PLS-DA models were built which related the specific metabolites
responsible to metal tolerance across all three strains (in other words, the basal
metabolite states across all strains was considered together). In this analysis,
tryptophan, glutathione, methionine, adenosine and glucose were elevated in sen-
sitivity for both copper and silver, whereas proline was strongly correlated to
sensitivity for copper only and lactate and NAD+ increased with sensitivity to
silver.
Overall, the results from this study indicate that the biofilms under study had
distinct metabolic states that conferred some advantages to stress, and that mor-
phological variants further allow the bacteria to develop unique profiles. These
laboratory results are particularly important to understanding the environmental
survival and viability of bacterial strains.
One of the proposed mechanisms by which biofilms are known to differ from
planktonic cultures is in sugar metabolism. In particular, exo-polysaccharide
matrices are thought to provide structure for formation of the biofilms (Harrison
et al. 2007). The NMR approach commonly employed for metabolomics analysis is
advantageous in that it is robust, quantitative and highly reproducible. One disad-
vantage, however, is the relatively small number of metabolites (*30) that can be
characterised from extracts, with specific metabolites related to sugar metabolism
being poorly characterised. For this reason, in a study of P. fluorescens response to
copper (Booth et al. 2011) a more sensitive gas chromatography-mass spectrometry
(GC-MS) method was employed in addition to NMR. The two analytical platforms
provided identification of 79 unique metabolites, and hence a much larger coverage
of metabolic space. Pairwise multivariate comparisons indicated that while copper
exposure induces a significant metabolic response in both planktonic and biofilm
cultures, the nature of this response is highly different. Only three metabolites
(NAD+, phosphoric acid and glutathione) were common to both responses.
Overall, the planktonic cultures were found to be far more ‘reactive’ to exposure,
characterised by an oxidative stress response with changes to the tricarboxylic acid
(TCA) cycle, glycolysis, pyruvate and nicotinate and niacinamide metabolism. On
the other hand, the biofilm response was dominated by shifts in exo-polysaccharide
metabolism, suggesting a ‘protective’ response. While alterations in levels of glu-
tathione indicate that there was still oxidative stress in the biofilms, the lack of
involvement of energy pathways suggests that the biofilms have alternate methods
for enhancing protection, which is consistent with previous observations (González
et al. 2010).
7 Community Metabolomics in Environmental Microbiology 209
Given the obvious advantages of being able to tightly control growth and exposure
conditions in a laboratory setting, it is no surprise that this is one of the most
popular methods used to study the effect of exposure to environmental toxicants.
Studies of bacterial metabolism are exquisitely sensitive to a multitude of factors,
including sampling conditions, sample processing, and in the context of metabo-
lomics, data acquisition and analysis. Numerous studies have been conducted on
appropriate methods to quench bacterial metabolism, which has recently been
assessed in the context of bacterial metabolomics (van Gulik 2010). The studies
described above were accomplished using cold methanol quenching, which is the
most common technique, but may suffer from leakage of metabolites during the
process. As the metabolomic scientists interest is often not in absolute quantifica-
tion of metabolites in the cells, but relative quantification between different popu-
lations, an inherent assumption in these experiments is that the quenching and
metabolite extraction processes impact the different populations in the same way.
In spite of these limitations, our understanding of the highly unique and specific
alterations in microbial metabolism in response to metals has been greatly enhanced
by metabolomics methods. The studies described here provide some clues as to how
bacteria can adapt, or exist in a uniquely ‘primed’ state for exposure to toxicants.
There remains much to do in the laboratory, including further testing of mixed
microbial communities, testing of complex toxicity profiles consisting of multiple
toxicants more closely reflective of real-world situations under chronic exposure
conditions. From a technical perspective, further expansion of the metabolome
coverage would also be advantageous, for example using liquid chromatography-
mass spectrometry (LC-MS) methods. In summary, metabolomics experiments in
the laboratory have contributed significantly to our understanding of the mecha-
nisms by which microbial populations are metabolically differentiated and, in fact,
these types of studies could have important industrial applications.
medium, the incomplete oxidation of the substrate and the kinetic limitations in
electron transfer from microorganisms to the anode. This latter process was pre-
viously thought to occur strictly through redox mediators that shuttle the electrons.
However, only a decade ago, microbes capable of direct electron transfer have been
discovered (Min et al. 2005). Although not currently capable of large-scale elec-
tricity generation, MFCs present an attractive technology for self-sufficient
wastewater treatment with modest electricity generation.
Complex microbial communities underpin AD and MFC applications but the
metabolic roles of the individual microbes are still largely undetermined. Moreover,
the species are strongly connected through syntrophies where the waste products of
one provide resources for another. Consequently, to understand, exploit and extend
the application of these systems the ‘ecophysiological’ roles of the individual
populations and species must be determined. Questions, such as how do the
resulting syntrophic interactions structure the system-level behaviour? can then be
addressed to support full-scale engineering and operation. We propose that a uni-
fication of metabolomics and other ‘omics approaches—particularly metagenomics
—could provide a high-throughput solution to link taxonomy with function. This
approach could also enable the construction and validation of ‘ecosystems biology’
models operating at the level of the whole community.
The vast majority of organisms in anaerobic reactor biofilms and MFCs have not
yet been cultured. To study these organisms requires the application of techniques
from the ‘molecular toolbox’ to characterize the complex, mixed microbial con-
sortia present. Much prior research has focused on extracting DNA from these
communities and polymerase chain reaction (PCR)-amplifying 16S rRNA genes.
This gene is present in all prokaryotes and can be used as a marker for the taxa
present (Clarridge 2004). To date, most 16S rRNA studies have used either
gel-based, fingerprinting techniques, such as DGGE, which provides a ‘barcode’ of
community diversity but without direct sequence information, or clone libraries,
which do provide direct sequence reads but through cost and time constraints are
limited in size (Phung et al. 2004). Both methods will fail to resolve rare taxa.
Quantitative PCR assays can be used to obtain absolute abundances of even rare
organisms but only for a predetermined target.
Until recently there was no economical method capable of resolving the com-
plete community structure. Next-generation sequencing technologies have trans-
formed this situation by providing orders-of-magnitude more sequence data at the
same cost (Margulies et al. 2005). This is driving the ‘omics revolution’, allowing
the sequencing of complete genomes from isolated organisms and environmental
DNA.
Application to MFCs and AD systems is still in its infancy. The genomes of
potentially important species have, however, been sequenced; for example,
212 O.A.H. Jones et al.
‘heavy’ and ‘light’ DNA fractions, indicated the autotrophic—or at least, mixo-
trophic—potential of crenarchaeal clones. This is a fascinating example of how
culture-independent techniques, and disparate datasets, can together reveal the
metabolic function of target groups, but it is not a high-throughput and generically
applicable approach while metagenomics is a high-throughput technique, the
connection to function is not direct.
necessary to resolve the pathways in individual samples and to link the components
of those pathways to species. Its success, however, is dependent on coupling it to a
computational pipeline capable of extracting the information and an experimental
strategy that maximises the statistical power of the complete data set to give
meaningful and valid results. The individual components of such a pipeline exist; it
is possible to identify microbial taxa from marker genes using resources such as the
Ribosomal Database Project (Cole et al. 2009); metagenomics reads can be anno-
tated to functional genes using MG-RAST (Meyer et al. 2008); and potential
metabolic pathways can be constructed from the KEGG database (Kanehisa et al.
2004). Similarly, tools exist for converting GC-MS peaks into predictions of
molecule identity (Horai et al. 2010; Atherton et al. 2006). The key will be inte-
grating the information in a single pipeline within a single statistical framework that
allows for errors, mislabelling and potentially contradictory information.
Deriving the optimal or most likely set of possible pathways and their divisions
amongst species will be a highly complex optimisation problem. A Bayesian
probabilistic approach will most likely provide the best method to solve this. This
will have the advantage of predicting not a single solution but a complete distri-
bution of possible results. Finally, any statistical technique is limited by the quality
of the data set. It is vital to link these tools to experiments that maximise the
information available. This should be achieved by running replicate systems to
determine the variability of the responses and, conversely, by using carefully
planned perturbations to expand the range of system states explored. These per-
turbations could be through changing substrate concentrations or operating condi-
tions, such as temperature. These requirements underpin the importance of the
high-throughput nature of ‘omics techniques to obtain, at reasonable cost, multiple
datasets, each containing huge amounts of information.
The final step of an ecosystems biology approach is integrating this information
into mathematical models capable of predicting both community composition and
bioreactor function. These will take the metabolic-taxonomic framework elucidated
above and add dynamics. The actual mathematical structures to do this are well
established: in a well-mixed bioreactor, a system of ordinary differential equations
would be appropriate, possibly with the addition of environmental noise; more
complex models will be necessary if spatial localisation of groups of organisms in
anaerobic granules or on the anodes of MFCs are deemed important. The greater
challenge than the model structure will be parameterisation. Once again a Bayesian
approach will be vital to propagate that uncertainty into predictions that formally
account for it.
The holistic, polyphasic approach outlined above can be viewed as systems biology
at the community level. It is not desirable to reconstruct and model every reaction
and process in every microbial cell, but it is desirable to identify those pathways,
7 Community Metabolomics in Environmental Microbiology 215
syntrophies and ecological interactions that are critical for controlling the com-
munity—and by extension, process—behaviour. The integration of ‘omics’
approaches coupled to process monitoring, and advanced microscopy, can generate
comprehensive, integrated datasets at microorganism, biofilm and bioreactor level.
This will enable the link between processes occurring at microorganism level (scale
c. 1 µm–1 mm) and the processes occurring within bioreactors (scale > 1 m). This
will, in turn, lead to improved reactor monitoring capability, reactor design, per-
formance and operational stability.
The past two decades has seen the rapid expansion of ‘omics’ oriented research,
examining the abundance of the vast array of genes (metagenomics), RNA mole-
cules (meta-transcriptomics), proteins (meta-proteomics) and small molecule
metabolites (meta/community metabolomics) present in a wide range of different
environments. To date, it is genomics that has been the poster child of the omics
era, with the global media avidly following the progress of high-profile research
projects that have enabled us to map the human genome (Human Genome
Sequencing 2004), the genomes of various economically important plants (Goff
et al. 2002), animals (Hillier et al. 2004) and disease causing organisms (Heidelberg
et al. 2000). More recently, we have begun to describe the vast content of
‘metagenomic’ DNA, seemingly aiming to catalogue the entirety of life around us
using the DNA barcode.
As our analysis of the global meta-genome continues to benefit from recent
advances in robotics, DNA sequencing chemistries, analytical and computational
procedures, the costs associated with sequencing such large volumes of DNA
continue to tumble. For example, the consumable costs associated with sequencing
the human genome have decreased from tens of millions of (US) dollars in 2007, to
just a few thousand (US) dollars or under today (Drmanac et al. 2010). With so
much data now being generated, the major challenges associated with most
large-scale metagenomic studies concern data storage and how to make sense of the
vast amounts of DNA sequence data generated.
There are several terms in use for the application of metabolomics in
ecological/environmental studies. For instance, the term “Ecotoxicogenomics” was
proposed by Snape et al. (2004) to describe the integration of genomic-based
science into the field of ecotoxicology. “EcoGenomics” (or ecological genomics)
was used by Chapman (2001) to describe the application of genomics based
techniques to ecology. In both cases the term genomics was taken to encompass all
the ‘omic sciences’ namely genomics (genome sequencing and the annotation of
216 O.A.H. Jones et al.
Modern molecular methods have provided tools for the global scientific community
to produce a continual deluge of DNA sequence data. These data, which are gen-
erated at an exponential rate, perhaps doubling in volume every 18 months (Lathe
et al. 2008), now requires terabyte-scale computation and ever expanding storage
facilities to reduce the widening gap between rates of data collection and inter-
pretation. Whilst it is the aim of researchers to increase the volume of useful DNA
data stored in their own databases and in online repositories such as NCBI’s
GenBank (www.ncbi.nlm.nih.gov/genbank/), much of the data being stored by
researchers are of limited value. Increasingly, it is being recognised that much, or
even most, of the storage space is used to house data that could be thrown away.
For this reason, it is now common practice to delete raw data files (e.g.
218 O.A.H. Jones et al.
electropherogram data) immediately after the data have been processed and con-
densed into far smaller text files containing the DNA sequence data. Due to recent
technical advances, the amount of data produced by metabolomic studies is
beginning to pose similar challenges and new standards of data storage and inter-
pretation are now required to minimise the cataloguing of redundant information.
Even once all of our data have been collected, adequately stored and compared to
appropriate reference and metadata there remain many challenges to maximise the
value of the large datasets characteristic of meta-omic studies. To date, many
large-scale metagenomic and community metabolomic studies have sought only to
address primarily pure, or fundamental, research aims, cataloguing the complexity
of various biological systems and to provide new insights into the functional
capabilities of many different microbial communities. Whilst these are worthy
research aims, it remains crucial that our new understanding of microbial diversity
and function be exploited to its full potential, addressing much broader, applied
research questions of benefit to key industrial partners, healthcare providers and
environmental protection agencies.
The development and application of new, more sensitive, biological indicators of
environmental health will be a key growth area of metagenomics and metabolomics
led research. For example, there is an increasing interest in the monitoring and
restoration of freshwater systems, aimed at maximising their value for ecological,
recreation and economic purposes. Macroinvertebrate communities have been used
around the globe as biological indicators of stream health for decades. Extensive
research has permitted classification of a wide range of macroinvertebrate taxa into
pollution tolerant, sensitive and facultative categories which are used to provide
indices of water quality (e.g., Hilsenhoff 1987) and a well-researched body of
information now supports their use (Feld and Hering 2007; Trigal et al. 2007).
This macroinvertebrate community data provide little or no information as to the
potential causes of any observed declines in freshwater ecological health. However,
recent advances in meta-omics centred research mean that it is now possible to
rapidly characterise (i) the abundance of ‘functional groups’ of bacteria, such as
those conferring resistance to any heavy metal (e.g., cobalt, zinc and cadmium) or
the resistance and ability to degrade organic pollutants (e.g., PCBs), (ii) the
abundance and activity of genes encoding for these ‘key’ functions and (iii) the
presence and abundance of functionally related metabolites. As such, the era of
meta-omics research promises to revolutionise the application of biological indi-
cators such that the rapid analysis of microbial communities may not only be used
to provide an indication of general ecological stress, but also to identify specific
drivers of environmental degradation within any site. This is of particular impor-
tance for the restoration of urban ecosystems in which the primarily drivers of
environmental changes are frequently hard to distinguish amongst the large
7 Community Metabolomics in Environmental Microbiology 219
Acknowledgments OAHJ thanks colleagues from the Australia and New Zealand Metabolomics
Network (ANZMN), Proteomics and Metabolomics Victoria (PMV), RMIT University (Australia),
the University of Cambridge (UK) and the Centre for Ecology and Hydrology Wallingford
(UK) for help and support in metabolomics research over the years.
References
Amann RI, Binder BJ, Olson RJ, Chisholm SW, Devereux R, Stahl DA (1990) Combination of
16S ribosomal-RNA-targeted oligonucleotide probes with flow-cytometry for analyzing mixed
microbial-populations. Appl Environ Microbiol 56:1919–1925
Atherton H, Bailey N, Zhang W, Taylor J, Major H, Shockcor J, Clarke K, Griffin J (2006) A
combined 1H-NMR spectroscopy- and mass spectrometry-based metabolomic study of the
PPAR-alpha null mutant mouse defines profound systemic changes in metabolism linked to the
metabolic syndrome. Physiol Genomics 27:178–186
Beale DJ, Karpe AV, McLeod JD, Gondalia SV, Muster TH, Othman MZ, Palombo EA, Joshi D
(2016) An ‘omics’ approach towards the characterisation of laboratory scale anaerobic
digesters treating municipal sewage sludge. Water Res 88:346–357
Bochner BR, Gadzinski P, Panomitros E (2001) Phenotype microarrays for high-throughput
phenotypic testing and assay of gene function. Genome Res 11:1246–1255
Boles BR, Thoendel M, Singh PK (2004) Self-generated diversity produces “insurance effects” in
biofilm communities. Proc Nat Aca Sci USA 101:16630–16635
220 O.A.H. Jones et al.
Booth SC, Workentine ML, Wen J, Shaykhutdinov R, Vogel HJ, Ceri H, Turner RJ, Weljie AM
(2011) Differences in metabolism between the biofilm and planktonic response to metal stress.
J Proteome Res 10:3190–3199
Brazma A, Hingamp P, Quackenbush J, Sherlock G, Spellman P, Stoeckert C, Aach J, Ansorge W,
Ball CA, Causton HC, Gaasterland T, Glenisson P, Holstege FCP, Kim IF, Markowitz V,
Matese JC, Parkinson H, Robinson A, Sarkans U, Schulze-Kremer S, Stewart J, Taylor RG,
Vilo J, Vingron M (2001) Minimal information about a microarray experiment (MIAME)—
towards standards for microarray data. Nat Genet 29:365–371
Bundy JG, Willey TL, Castell RS, Ellar DJ, Brindle KM (2005) Discrimination of pathogenic
clinical isolates and laboratory strains of Bacillus cereus by NMR-based metabolomic
profiling. FEMS Microbiol Lett 242:127–136
Castillo JA, Staton SJ, Taylor TJ, Herckes P, Hayes MA (2012) Exploring the feasibility of
bioaerosol analysis as a novel fingerprinting technique. Anal Bioanal Chem 403:15–26
Chapman RW (2001) EcoGenomics—a consilience for comparative immunology? Dev Comp
Immunol 25:549–551
Chasteen TG, Bentley R (2002) Biomethylation of Selenium and Tellurium: microorganisms and
plants. Chem Rev 103:1–26
Chaudhuri SK, Lovley DR (2003) Electricity generation by direct oxidation of glucose in
mediatorless microbial fuel cells. Nat Biotechnol 21:1229–1232
Clarridge JE (2004) Impact of 16S rRNA gene sequence analysis for identification of bacteria on
clinical microbiology and infectious diseases. Clin Microbiol Rev 17:840–862
Cole JR, Wang Q, Cardenas E, Fish J, Chai B, Farris RJ, Kulam-Syed-Mohideen AS,
McGarrell DM, Marsh T, Garrity GM, Tiedje JM (2009) The Ribosomal Database Project:
improved alignments and new tools for rRNA analysis. Nucleic Acids Res 37:D141–D145
Collins G, Mahony T, McHugh S, Gieseke A, de Beer D, O’Flaherty V (2005) Distribution,
dynamics and in situ ecophysiology of Crenarchaeota in anaerobic wastewater treatment
granular biofilms. Water Sci Technol 52:233–239
Drmanac R, Sparks AB, Callow MJ, Halpern AL, Burns NL, Kermani BG, Carnevali P,
Nazarenko I, Nilsen GB, Yeung G, Dahl F, Fernandez A, Staker B, Pant KP, Baccash J,
Borcherding AP, Brownley A, Cedeno R, Chen LS, Chernikoff D, Cheung A, Chirita R,
Curson B, Ebert JC, Hacker CR, Hartlage R, Hauser B, Huang S, Jiang Y, Karpinchyk V,
Koenig M, Kong C, Landers T, Le C, Liu J, McBride CE, Morenzoni M, Morey RE, Mutch K,
Perazich H, Perry K, Peters BA, Peterson J, Pethiyagoda CL, Pothuraju K, Richter C,
Rosenbaum AM, Roy S, Shafto J, Sharanhovich U, Shannon KW, Sheppy CG, Sun M,
Thakuria JV, Tran A, Vu D, Zaranek AW, Wu XD, Drmanac S, Oliphant AR, Banyai WC,
Martin B, Ballinger DG, Church GM, Reid CA (2010) Human genome sequencing using
unchained base reads on self-assembling DNA nanoarrays. Science 327:78–81
Eisen JA (2007) Environmental shotgun sequencing: its potential and challenges for studying the
hidden world of microbes. PLoS Biol 5:e82
Feld CK, Hering D (2007) Community structure or function: effects of environmental stress on
benthic macroinvertebrates at different spatial scales. Freshw Biol 52:1380–1399
Feng X, Simpson AJ, Wilson KP, Dudley Williams D, Simpson MJ (2008) Increased cuticular
carbon sequestration and lignin oxidation in response to soil warming. Nat Geosci 1:836–839
Fredrickson JK, Romine MF, Beliaev AS, Auchtung JM, Driscoll ME, Gardner TS, Nealson KH,
Osterman AL, Pinchuk G, Reed JL, Rodionov DA, Rodrigues JLM, Saffarini DA, Serres MH,
Spormann AM, Zhulin IB, Tiedje JM (2008) Towards environmental systems biology of
Shewanella. Nat Rev Microbiol 6:592–603
Garcia MH, Ivanova N, Kunin V, Warnecke F, Barry KW, McHardy AC, Yeates C, He S,
Salamov AA, Szeto E, Dalin E, Putnam NH, Shapiro HJ, Pangilinan JL, Rigoutsos I,
Kyrpides NC, Blackall LL, McMahon KD, Hugenholtz P (2006) Metagenomic analysis of two
enhanced biological phosphorus removal (EBPR) sludge communities. Nat Biotechnol
24:1263–1269
Goff SA, Ricke D, Lan TH, Presting G, Wang RL, Dunn M, Glazebrook J, Sessions A, Oeller P,
Varma H, Hadley D, Hutchinson D, Martin C, Katagiri F, Lange BM, Moughamer T, Xia Y,
7 Community Metabolomics in Environmental Microbiology 221
Budworth P, Zhong JP, Miguel T, Paszkowski U, Zhang SP, Colbert M, Sun WL, Chen LL,
Cooper B, Park S, Wood TC, Mao L, Quail P, Wing R, Dean R, Yu YS, Zharkikh A, Shen R,
Sahasrabudhe S, Thomas A, Cannings R, Gutin A, Pruss D, Reid J, Tavtigian S, Mitchell J,
Eldredge G, Scholl T, Miller RM, Bhatnagar S, Adey N, Rubano T, Tusneem N, Robinson R,
Feldhaus J, Macalma T, Oliphant A, Briggs S (2002) A draft sequence of the rice genome
(Oryza sativa L. ssp japonica). Science 296:92–100
González AG, Shirokova LS, Pokrovsky OS, Emnova EE, Martínez RE, Santana-Casiano JM,
González-Dávila M, Pokrovski GS (2010) Adsorption of copper on Pseudomonas aureofa-
ciens: protective role of surface exopolysaccharides. J Colloid Interface Sci 350:305–314
Handelsman J (2004) Metagenomics: application of genomics to uncultured microorganisms.
Microbiol Mol Biol Rev 68:669–685
Harrison JJ, Ceri H, Turner RJ (2007) Multimetal resistance and tolerance in microbial biofilms.
Nat Rev Microbiol 5:928–938
Heidelberg JF, Eisen JA, Nelson WC, Clayton RA, Gwinn ML, Dodson RJ, Haft DH, Hickey EK,
Peterson JD, Umayam L, Gill SR, Nelson KE, Read TD, Tettelin H, Richardson D,
Ermolaeva MD, Vamathevan J, Bass S, Qin HY, Dragoi I, Sellers P, McDonald L,
Utterback T, Fleishmann RD, Nierman WC, White O, Salzberg SL, Smith HO, Colwell RR,
Mekalanos JJ, Venter JC, Fraser CM (2000) DNA sequence of both chromosomes of the
cholera pathogen Vibrio cholerae. Nature 406:477–483
Heidelberg JF, Paulsen IT, Nelson KE, Gaidos EJ, Nelson WC, Read TD, Eisen JA, Seshadri R,
Ward N, Methe B, Clayton RA, Meyer T, Tsapin A, Scott J, Beanan M, Brinkac L,
Daugherty S, DeBoy RT, Dodson RJ, Durkin AS, Haft DH, Kolonay JF, Madupu R,
Peterson JD, Umayam LA, White O, Wolf AM, Vamathevan J, Weidman J, Impraim M,
Lee K, Berry K, Lee C, Mueller J, Khouri H, Gill J, Utterback TR, McDonald LA,
Feldblyum TV, Smith HO, Venter JC, Nealson KH, Fraser CM (2002) Genome sequence of
the dissimilatory metal ion-reducing bacterium Shewanella oneidensis. Nat Biotechnol
20:1118–1123
Hillier LW, Miller W, Birney E, Warren W, Hardison RC, Ponting CP, Bork P, Burt DW,
Groenen MAM, Delany ME, Dodgson JB, Chinwalla AT, Cliften PF, Clifton SW,
Delehaunty KD, Fronick C, Fulton RS, Graves TA, Kremitzki C, Layman D, Magrini V,
McPherson JD, Miner TL, Minx P, Nash WE, Nhan MN, Nelson JO, Oddy LG, Pohl CS,
Randall-Maher J, Smith SM, Wallis JW, Yang SP, Romanov MN, Rondelli CM, Paton B,
Smith J, Morrice D, Daniels L, Tempest HG, Robertson L, Masabanda JS, Griffin DK,
Vignal A, Fillon V, Jacobbson L, Kerje S, Andersson L, Crooijmans RPM, Aerts J, van der
Poel JJ, Ellegren H, Caldwell RB, Hubbard SJ, Grafham DV, Kierzek AM, McLaren SR,
Overton IM, Arakawa H, Beattie KJ, Bezzubov Y, Boardman PE, Bonfield JK, Croning MDR,
Davies RM, Francis MD, Humphray SJ, Scott CE, Taylor RG, Tickle C, Brown WRA,
Rogers J, Buerstedde JM, Wilson SA, Stubbs L, Ovcharenko I, Gordon L, Lucas S,
Miller MM, Inoko H, Shiina T, Kaufman J, Salomonsen J, Skjoedt K, Wong GKS, Wang J,
Liu B, Yu J, Yang HM, Nefedov M, Koriabine M, deJong PJ, Goodstadt L, Webber C,
Dickens NJ, Letunic I, Suyama M, Torrents D, von Mering C, Zdobnov EM, Makova K,
Nekrutenko A, Elnitski L, Eswara P, King DC, Yang S, Tyekucheva S, Radakrishnan A,
Harris RS, Chiaromonte F, Taylor J, He JB, Rijnkels M, Griffiths-Jones S, Ureta-Vidal A,
Hoffman MM, Severin J, Searle SMJ, Law AS, Speed D, Waddington D, Cheng Z, Tuzun E,
Eichler E, Bao ZR, Flicek P, Shteynberg DD, Brent MR, Bye JM, Huckle EJ, Chatterji S,
Dewey C, Pachter L, Kouranov A, Mourelatos Z, Hatzigeorgiou AG, Paterson AH, Ivarie R,
Brandstrom M, Axelsson E, Backstrom N, Berlin S, Webster MT, Pourquie O, Reymond A,
Ucla C, Antonarakis SE, Long MY, Emerson JJ, Betran E, Dupanloup I, Kaessmann H,
Hinrichs AS, Bejerano G, Furey TS, Harte RA, Raney B, Siepel A, Kent WJ, Haussler D,
Eyras E, Castelo R, Abril JF, Castellano S, Camara F, Parra G, Guigo R, Bourque G, Tesler G,
Pevzner PA, Smit A, Fulton LA, Mardis ER, Wilson RK (2004) Sequence and comparative
analysis of the chicken genome provide unique perspectives on vertebrate evolution. Nature
432:695–716
222 O.A.H. Jones et al.
Hilsenhoff WL (1987) An improved biotic index of organic stream pollution. Great Lakes Entomol
20:31–39
Horai H, Arita M, Kanaya S, Nihei Y, Ikeda T, Suwa K, Ojima Y, Tanaka K, Tanaka S,
Aoshima K, Oda Y, Kakazu Y, Kusano M, Tohge T, Matsuda F, Sawada Y, Hirai MY,
Nakanishi H, Ikeda K, Akimoto N, Maoka T, Takahashi H, Ara T, Sakurai N, Suzuki H,
Shibata D, Neumann S, Iida T, Funatsu K, Matsuura F, Soga T, Taguchi R, Saito K,
Nishioka T (2010) MassBank: a public repository for sharing mass spectral data for life
sciences. J Mass Spectrom 45:703–714
International Human Genome Sequencing Consortium (2004) Finishing the euchromatic sequence
of the human genome. Nature 431:931–945
Lisa T, Casale C, Domenech C (1994) Cholinesterase, acid phosphatase, and phospholipase C of
Pseudomonas aeruginosa under hyperosmotic conditions in a high-phosphate medium. Curr
Microbiol 28:71–76
Jones OAH, Cheung VL (2007) An introduction to metabolomics and its potential application in
veterinary science. Comp Med 57:436–442
Jones OAH, Sdepanian S, Lofts S, Svendsen C, Spurgeon DJ, Maguire ML, Griffin JL (2014)
Metabolomic analysis of soil communities can be used for pollution assessment. Environ Sci
Technol 33:61–64
Kanehisa M, Goto S, Kawashima S, Okuno Y, Hattori M (2004) The KEGG resource for
deciphering the genome. Nucleic Acids Res 32:D277–D280
Lathe W, Williams J, Mangan M, Karolchik D (2008) Genomic data resources: challenges and
promises. Nature Education 1:3
Lester JN, Birkett JW (1999) Microbiology and chemistry for environmental scientists and
engineers, 2nd edn. E and FN Spon Ltd, London
Lettinga G (1995) Anaerobic-digestion and waste-water treatment systems. Antonie Van
Leeuwenhoek 67:3–28
Lettinga G, Vanvelsen AFM, Hobma SW, Dezeeuw W, Klapwijk A (1980) Use of the upflow
sludge blanket (USB) reactor concept for biological wastewater-treatment, especially for
anaerobic treatment. Biotechnol Bioeng 22:699–734
Llewellyn CA, Sommer U, Dupont CL, Allen AE, Viant MR (2015) Using community
metabolomics as a new approach to discriminate marine microbial particulate organic matter in
the western English Channel. Prog Oceanogr 137(Part B):421–433
Lykidis A, Chen C, Tringe SG, McHardy AC, Copeland A, Kyrpides NC, Hugenholtz P,
Macarie H, Olmos A, Monroy O, Liu W (2011) Multiple syntrophic interactions in a
terephthalate-degrading methanogenic consortium. ISME J 5:122–130
Margulies M, Egholm M, Altman WE, Attiya S, Bader JS, Bemben LA, Berka J, Braverman MS,
Chen YJ, Chen ZT, Dewell SB, Du L, Fierro JM, Gomes XV, Godwin BC, He W, Helgesen S,
Ho CH, Irzyk GP, Jando SC, Alenquer MLI, Jarvie TP, Jirage KB, Kim JB, Knight JR,
Lanza JR, Leamon JH, Lefkowitz SM, Lei M, Li J, Lohman KL, Lu H, Makhijani VB,
McDade KE, McKenna MP, Myers EW, Nickerson E, Nobile JR, Plant R, Puc BP, Ronan MT,
Roth GT, Sarkis GJ, Simons JF, Simpson JW, Srinivasan M, Tartaro KR, Tomasz A, Vogt KA,
Volkmer GA, Wang SH, Wang Y, Weiner MP, Yu PG, Begley RF, Rothberg JM (2005)
Genome sequencing in microfabricated high-density picolitre reactors. Nature 437:376–380
McHugh S, O’Reilly C, Mahony T, Colleran E, O’Flaherty V (2003) Anaerobic granular sludge
bioreactor technology. Rev Environ Sci Biotechnol 2:225–245
Methe BA, Nelson KE, Eisen JA, Paulsen IT, Nelson W, Heidelberg JF, Wu D, Wu M, Ward N,
Beanan MJ, Dodson RJ, Madupu R, Brinkac LM, Daugherty SC, DeBoy RT, Durkin AS,
Gwinn M, Kolonay JF, Sullivan SA, Haft DH, Selengut J, Davidsen TM, Zafar N, White O,
Tran B, Romero C, Forberger HA, Weidman J, Khouri H, Feldblyum TV, Utterback TR, Van
Aken SE, Lovley DR, Fraser CM (2003) Genome of Geobacter sulfurreducens: metal
reduction in subsurface environments. Science 302:1967–1969
Meyer F, Paarmann D, D’Souza M, Olson R, Glass EM, Kubal M, Paczian T, Rodriguez A,
Stevens R, Wilke A, Wilkening J, Edwards RA (2008) The metagenomics RAST server—a
7 Community Metabolomics in Environmental Microbiology 223
public resource for the automatic phylogenetic and functional analysis of metagenomes. BMC
Bioinform 9:386
Min BK, Cheng SA, Logan BE (2005) Electricity generation using membrane and salt bridge
microbial fuel cells. Water Res 39:1675–1686
Morrison N, Bearden D, Bundy JG, Collette T, Currie F, Davey MP, Haigh NS, Hancock D,
Jones OAH, Rochfort S, Sansone S-A, Stys D, Teng Q, Field D, Viant MR (2007) Standard
reporting requirements for biological samples in metabolomics experiments: environmental
context. Metabolomics 3:203–210
Nadell CD, Xavier JB, Foster KR (2009) The sociobiology of biofilms. FEMS Microbiol Rev
33:206–224
Nicholson JK, Holmes E, Wilson ID (2005) Gut microorganisms, mammalian metabolism and
personalized health care. Nat Rev Microbiol 3:431–438
Oh SE, Logan BE (2005) Hydrogen and electricity production from a food processing wastewater
using fermentation and microbial fuel cell technologies. Water Res 39:4673–4682
Peršoh D (2015) Plant-associated fungal communities in the light of meta’omics. Fungal Divers
75:1–25
Petrova OE, Sauer K (2009) A novel signalling network essential for regulating Pseudomonas
aeruginosa biofilm development. PLoS Pathog 5:e1000668
Phung NT, Lee J, Kang KH, Chang IS, Gadd GM, Kim BH (2004) Analysis of microbial diversity
in oligotrophic microbial fuel cells using 16S rDNA sequences. FEMS Microbiol Lett 233:77–
82
Portais J-C, Delort A-M (2002) Carbohydrate cycling in micro-organisms: what can 13C-NMR tell
us? FEMS Microbiol Rev 26:375–402
Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F,
Yamada T, Mende DR, Li J, Xu J, Li S, Li D, Cao J, Wang B, Liang H, Zheng H, Xie Y, Tap J,
Lepage P, Bertalan M, Batto JM, Hansen T, Le Paslier D, Linneberg A, Nielsen HB,
Pelletier E, Renault P, Sicheritz-Ponten T, Turner K, Zhu H, Yu C, Li S, Jian M, Zhou Y, Li Y,
Zhang X, Li S, Qin N, Yang H, Wang J, Brunak S, Dore J, Guarner F, Kristiansen K,
Pedersen O, Parkhill J, Weissenbach J, Bork P, Ehrlich SD, Wang J (2010) A human gut
microbial gene catalogue established by metagenomic sequencing. Nature 464:59–65
Rochfort S, Ezernieks V, Mele P, Kitching M (2015) NMR metabolomics for soil analysis provide
complementary, orthogonal data to MIR and traditional soil chemistry approaches—a land use
study. Magn Reson Chem 53:719–725
Sales CM, Lee PKH (2015) Resource recovery from wastewater: application of meta-omics to
phosphorus and carbon management. Curr Opin Biotechnol 33:260–267
Scullion J, Elliott GN, Huang WE, Goodacre R, Worgan H, Darby R, Bailey MJ, Gwynn-Jones D,
Griffith GW, Winson MK, Williams PA, Clegg C, Draper J (2003) Use of earthworm casts to
validate FT-IR spectroscopy as a ‘sentinel’ technology for high-throughput monitoring of
global changes in microbial ecology: the 7th international symposium on earthworm ecology,
Cardiff, Wales, 2002. Pedobiologia 47:440–446
Snape JR, Maund SJ, Pickford DB, Hutchinson TH (2004) Ecotoxicogenomics: the challenge of
integrating genomics into aquatic and terrestrial ecotoxicology. Aquat Toxicol 67:143–154
Sogin ML, Morrison HG, Huber JA, Mark Welch D, Huse SM, Neal PR, Arrieta JM, Herndl GJ
(2006) Microbial diversity in the deep sea and the underexplored “rare biosphere”. Proc Natl
Acad Sci USA 103:12115–12120
Swenson TL, Jenkins S, Bowen BP, Northen TR (2015) Untargeted soil metabolomics methods
for analysis of extractable organic matter. Soil Biol Biochem 80:189–198
Timmins ÉM, Howell SA, Alsberg BK, Noble WC, Goodacre R (1998) Rapid differentiation of
closely related Candida species and strains by pyrolysis mass spectrometry and fourier
transform infrared spectroscopy. J Clin Microbiol 36:367–374
Tremaroli V, Workentine M, Weljie AM, Vogel H, Ceri H, Viti C, Tatti E, Zhang P, Hynes A,
Turner R, Zannoni D (2009) Metabolomic investigation of the bacterial response to a metal
challenge. Appl Environ Microbiol 75:719–728
224 O.A.H. Jones et al.
Farhana R. Pinu
1 Introduction
Food safety is a major concern worldwide, which has gained enormous attention in
the last two decades mainly because of the emergence of new food-borne pathogens
and other chemical hazards. Additionally, there has been an extreme increase in the
food-borne illness incidences along with large-scale outbreaks. Many factors con-
tribute to these growing concerns, including the industrialisation and mass pro-
duction of agricultural products, the increasing number of imported food products,
and changes in food consumption patterns due to consumer lifestyle changes
(Motarjemi et al. 2008). Ready-to-eat and fast foods, especially, have gained great
popularity, and thus also resulted in many food-borne illness incidences in recent
years. Moreover, the tremendous upsurge in global population is also almost
forcing producers towards mass production of agricultural products without giving
much attention to quality and safety issues (Motarjemi and Lelieveld 2014). The
world has already seen an introduction of a massive amount of genetically modified
(GM) crops in the last 40 years, in response to the need to feed over 6.5 billion
people. Consumers are doubtful about the safety of consuming GM food products;
therefore, scientific attention is required to unravel the facts about the safety of
eating such foods (Pinu 2015).
In many third-world countries, food quality and safety are often overlooked. In
addition, many food vendors deliberately contaminate food products with unwanted
materials (e.g. melamine in powdered milk) to increase profits, and others do so
Food safety issues around the world are mainly monitored by the World Health
Organisation (WHO) and The Food and Agriculture Organisation (FAO) of the
United Nations. The Codex Alimentarius Commission (CAC) is an intergovern-
mental body that is operated under WHO and FAO. In 1997, CAC defined food
safety as the assurance that a food or food product will not cause any harm to the
consumers when it is prepared and/or eaten according to its intended use. Therefore,
a food will only be considered as safe when it will not cause any long- or short-term
illness to the consumer. It is the responsibility of food safety management to make
sure that a food is safe before it is sold to its intended consumers. The maintenance
of food hygiene is another important parameter during industrial food production
that can ensure the safety and quality of food products. However, consumers are
also responsible for following the instructions provided on the packaging to ensure
the post-sale safety of food. Therefore, the key responsibilities that allow the proper
maintenance of food safety follow a chain of action from regulators, to producers
and then to consumers (Fig. 1).
8 Metabolomics: Applications to Food Safety and Quality Research 227
Fig. 2 The seven principles of Hazard Analysis Critical Control Points (HACCP)
carried out to determine the food quality by testing the raw material and the end
product, by monitoring the environment before releasing the products. Auditing and
consumer complaints handling are also associated with HACCP implementation in
an industry.
In addition to regulatory bodies and industries, consumers also play a vital role
in maintaining food hygiene and safety. Many consumers prefer to buy inexpensive
food materials that do not have appropriate labels and safety information, and
consumptions of these foods might pose a health risk (Motarjemi et al. 2001).
Moreover, some ignore the instructions provided within the packaging of the food
products and some never report defective foods that may cause public health
concerns. Therefore, the liability of consumers cannot be overlooked and raising
awareness might help to improve current food safety issues. Food safety-related
campaigns organised by government and non-governmental organisations and
social media can help in raising awareness in consumers.
The role of the scientific community is also important in maintaining food safety
and quality. Ongoing research related to food products is always beneficial,
8 Metabolomics: Applications to Food Safety and Quality Research 229
bringing new insight into different food pathogens and chemicals that contaminate
food and food products. Moreover, toxicological and ecological knowledge of
microbial and chemical spoilage provided by scientists allows us to manage the
food safety situation in a better way by undertaking control measurements
(Motarjemi and Lelieveld 2014).
The current mass production and industrialisation of different foods and food
products have been initiated in order to feed the increasing world population. The
‘green revolution’ has also taken place in the last decades, enabling production of
vast amounts of crops mainly using genetically modified (GM) plants. There is still
an insecurity about GM food products, as many believe that long-term consumption
of these food materials may cause some deleterious effects on human health
including allergies and other immunological disorders (Maghari and Ardekani
2011; Krimsky 2015). However, this is still under question as at least two different
groups of scientists exist who either consider GM food as safe or harmful (Krimsky
2015). Due to the lack of long-term and consistent research on the effect of GM
food on human health, it is still not possible to come to a conclusion and this is why
consumers are even more doubtful about the safety of GM products. In addition,
climate change is another key factor that has prompted development of new plants
or crop varieties that will withstand the gradual changes in environmental condi-
tions. For instance, many efforts have already been undertaken to develop new rice
varieties that can withstand considerably unfavourable conditions including heat
stress, drought and high salinity (Nokkoul and Wichitparp 2014; Van Oort et al.
2015; Nguyen et al. 2016). Barley is another crop variety that has been used widely
as a model to study and develop climate-resilient crops (Dawson et al. 2015).
Therefore, the world has observed huge changes in the quality and type of foods
and raw materials, which pose both a significant advantage and sometimes a new
threat to food safety and quality. Chemical contaminants and xenobiotic molecules,
including pesticide residues and organic halogenated compounds, also pose sig-
nificant potential impacts on both human health and the environment, as these
molecules can take a very long time to break down (over 50 years) in the envi-
ronment. The effects of pesticides and other xenobiotics on aquatic and other
environments (e.g. infertility of sea birds) is well documented, and attempts have
already been undertaken to reduce the rate of contamination by banning many
known chemical contaminants (Walker 1990; Falkowska and Reindl 2015;
Gustafson et al. 2015; Pérez et al. 2015). In addition to chemical hazards, food
safety is also threatened by microbial exposure. For instance, overall changes in
lifestyle have steadily forced us to adapt to comparatively different food habits, and
introduced new types of foods (e.g. raw food). As a result, many new food-borne
pathogens have emerged or other pathogens re-emerged because of these newly
found transmission vehicles. Thus, many food-borne outbreaks which occurred in
230 F.R. Pinu
the last 20 years were caused by bacteria, viruses, and protozoa, and many more
pathogens are being introduced via food contamination every year. Therefore, there
is an ongoing need for a proper risk assessment and management system to control
outbreaks or even a pandemic related to any food-borne pathogen.
Major current challenges can be identified as follows:
I. Emergence of new food pathogens and control of outbreaks
II. Potential effects from genetically modified foods
III. Emerging chemical contaminants and xenobiotics
IV. Adulterations of food materials.
Food safety is not only a public health issue, but also has serious social and
economic consequences. Food-borne illness can cause havoc in any country and
sometimes worldwide when it is an epidemic that cause loss of many lives. For
instance, food- and water-borne diseases are the main reasons of death of over 2.2
million people annually (WHO 2006), children, immunocompromised and elderly
populations are often the most affected. This is not only a burden for many poor
countries, but also endangers the international developmental efforts that have been
undertaken to combat poverty (Kuchenmüller et al. 2009). These food-borne ill-
nesses can be caused by various agents including pathogenic microbes, heavy
metals, chemical contaminants and other toxic materials found in different food
sources (e.g. toxins in wild mushrooms) (Anater et al. 2016; Signes-Pastor et al.
2016). Among pathogenic microbes, different strains of Escherichia coli, Listeria
monocytenes, Salmonella spp. and even some food-borne viruses including nor-
ovirus and Hepatitis C have gained particular attention because of outbreaks related
to contaminated food consumption in many countries (Marušic 2011). Moreover,
many incidents were also reported of contamination of foods due to the presence of
heavy metals and chemicals. In particular, the presence of arsenic in rice and other
crops in many developing countries and melamine in milk products have gained
public attention in recent years (Signes-Pastor et al. 2016). Most previous research
on food quality and safety have mainly focused on determining the food compo-
sition and stress response of food pathogens, and have helped to add knowledge on
what should be present in a particular food and how the growth of pathogenic
microbes can be controlled using different techniques.
8 Metabolomics: Applications to Food Safety and Quality Research 231
To know more about the quality of food and food products, it is important to
determine the composition, which also allows us to decide whether a given food is
safe to consume, by providing information about any potential hazard. Much
research has already been undertaken by focusing mainly on food composition of
different crops, fresh food materials, and raw ingredients for other food products.
Determination of food composition is not only focused on differentiating between
different food products to ensure their safety, but is also intended to gain insight
about the originality of some speciality foods and fermented food products, e.g.
fruit juices, balsamic vinegars, and wines. One of the main reasons for determining
originality is to control the adulteration of food material with unwanted ingredients
that should not be present in that specific food. For instance, nuclear magnetic
resonance (NMR) profiling of fruit juices has been carried out to determine the
country of origin and also to discover if those fruit juices were produced using real
juice or concentrate (Spraul et al. 2009a, b; Tomita et al. 2015). In addition, volatile
compound analysis of balsamic vinegars through the use of gas chromatography
and mass spectrometry (GC-MS) allowed determination of the effects of ageing
materials or woods on vinegar quality (Chinnici et al. 2009; Callejón et al. 2010).
Many studies have also focused on regional or country-specific food products
including fresh vegetables, seafood, fish and ready-to-eat foods. This is mainly true
for those countries either with low average incomes or where environmental pol-
lution is a considerably bigger problem than in other developed countries. In
countries that lack proper implementation of laws, regulations and auditing systems
required for maintaining food safety, food stuffs can become deliberately contam-
inated with unwanted materials by vendors in order to gain more profit (Pinu 2015).
In this situation, food composition studies that help to determine the type and
diversity of contaminants are very useful. However, there is still a huge scope for
improvements to technologies for rapid detection and identification of those food
contaminants. Using these technologies, food auditors will be able to manage sit-
uations better by analysing food samples in real time while examining a food
processing unit. Such technologies are still very limited currently, and research
should focus on developing more user-friendly techniques and detectors using
cutting-edge technologies.
One of the most demanding areas of research in food safety is the study of
food-borne pathogens, which is mainly because of the many outbreaks related to the
consumption of food products contaminated with pathogenic microbes. Food is
232 F.R. Pinu
Metabolomics is one of the most recently introduced ‘omics’ that aims to analyse
small molecules (metabolites) in a given biological sample. Although metabolomics
was initially defined as the technical area by which it would be possible to detect
and identify all the metabolites produced by a cell or an organism (Fiehn 2002;
Bino et al. 2004), this has not been realised, mainly due to the diversity of
metabolites. However, metabolomics is already established as a powerful tool for
studying the metabolism and physiology of many living organisms, and thus this
approach has been applied to a diverse range of research areas, including biomarker
and drug discovery, agriculture, nutrition, bioremediation, plant biotechnology and
also food science (Anizan et al. 2012; Badilita et al. 2014; Hall and de Maagd 2014;
Li et al. 2014; Lima et al. 2014; Booth et al. 2015; El Amrani et al. 2015; Melnik
2015). Metabolomics has been used to study food systems including food ingre-
dients, food processing and food pathogens; it has gained popularity in the last
10 years and numerous studies have already been carried out (Vikram et al. 2004;
Badilita et al. 2014; Kusano et al. 2014; Inoue et al. 2015; Le Boucher et al. 2015;
Ragone et al. 2015). Therefore, a distinct research area entitled ‘food metabolomics’
is well established that refers to the application of metabolomics in food system
processes, from farm to consumers (Kim et al. 2016). Recently a term ‘foodomics’
has been introduced within the scientific community that refers to the application of
‘omics approaches including genomics, proteomics, transcriptomics and metabo-
lomics in food science (Herrero et al. 2012; Cifuentes and Rutledge 2013;
D’Alessandro and Zolla 2013; Ibáñez and Cifuentes 2014; Laghi et al. 2014; Inoue
and Toyo’oka 2015). However, food metabolomics deals only with the most
downstream product of cell metabolism, metabolites, present in a given food or
food system.
The food metabolome is very complex in nature, and also widely variable,
depending on the type of food and the raw materials. Therefore, many thousands of
metabolites are present in food are highly variable in terms of polarity and
molecular weight (Shulaev 2006). The differing concentrations of metabolites in
food items pose a major challenge in the development of analytical tools to detect as
many metabolites as possible within a single analysis (Kueger et al. 2012). So far,
there is no such analytical instrument available; therefore, for the better under-
standing of the metabolome by analysing as many metabolites as possible, the use
of multiple analytical technologies is suggested by many scientists (Hall et al. 2002;
Dunn and Ellis 2005; Villas-Bôas et al. 2007; Sumner 2010; Hall and Hardy 2012;
Pinu et al. 2014). In addition, one of the major challenges that a food scientist needs
to overcome while analysing food is the complex sample matrix. The matrix effect
(ME) can create challenges in the detection and quantification of compounds that
are present at very low concentrations in different food. ME is also responsible for
poor and unreliable data that can affect the reproducibility, repeatability, linearity
234 F.R. Pinu
and accuracy of the methods used by various analytical instruments (Trufelli et al.
2011). To avoid and reduce the ME, a sample clean-up step using Solid Phase
Extraction (SPE) or Solid Phase Microextraction (SPME) or liquid extraction is
usually necessary before analysis by other methods (Jiang et al. 2012). More effi-
cient chromatographic separation is also suggested by Trufelli et al. (2011), in
which two-dimensional separation techniques (both GC and LC) can also be
applied (Marriott et al. 2012; Mondello et al. 2012). However, pre-analytical steps
can be time-consuming, arduous and often can cause loss of analytes, which is not
appropriate for an unbiased profiling approach (Villas-Bôas et al. 2007; Cappiello
et al. 2010; Trufelli et al. 2011). Although metabolomics was initially introduced
mainly as an unbiased and non-targeted approach, both targeted and non-targeted
analyses are performed for any biological samples to better answer the research
questions. Therefore, both targeted and non-targeted metabolomics are gaining
popularity for the analysis food products.
A significant improvement has also been achieved in metabolomics workflow,
including sample preparation, quenching, metabolite extraction and acquisition of
data (Fig. 3). In recent years, many sample preparation protocols have been pub-
lished that allow better detection of metabolites with a wide range of chemical
properties (Anizan et al. 2010; Biais et al. 2012; Teo et al. 2013; Brennan 2014;
Chan et al. 2014; Le Gall 2015; Rejczak and Tuzimski 2015). In short, an appro-
priate quenching method is used to stop the ongoing enzymatic activities after the
collection of a food or microbial samples. After quenching, metabolites are
extracted using a suitable extraction solvent (e.g. chloroform/methanol/water).
However, it is noteworthy that metabolite profiles may vary depending on the
metabolite extraction protocols; therefore, it is better to use at least a few extraction
protocols to obtain a global metabolite profile of any biological sample (Duportet
et al. 2012, Jäpelt et al. 2015). Once metabolites are extracted from the sample, they
are ready for analysis by an instrument of choice. The acquired data then needs to
be explored using different statistical and chemometric approaches (Aggio et al.
2011, 2014; Gowda et al. 2014; Pluskal et al. 2010; Robotti and Marengo 2016;
Smith et al. 2006; Xia et al. 2012): including feature detection, alignment,
biomarker identification and chemical structure elucidation (Fig. 3).
Fig. 3 Workflow for the analysis of the food metabolome. Here, MS Mass spectrometry and NMR
Nuclear Magnetic Resonance spectroscopy
unknown metabolites present in a food. It is well known that powerful detectors are
the main factors for the analysis of metabolites. Two technologies, Nuclear
Magnetic Resonance (NMR) and Mass Spectrometry (MS) have been employed
widely in metabolomics (Shulaev 2006; Villas-Bôas et al. 2007; Dieterle et al.
2011; Herrero et al. 2012; Kueger et al. 2012; Zhang et al. 2012; Balan et al. 2013;
Ibáñez et al. 2013; Badilita et al. 2014; Senyuva et al. 2015). However, there are
many other instrumental techniques, including Fourier transform infra-red spec-
troscopy (FTIR), which are available for metabolite profiling of food samples.
NMR has been broadly used for untargeted metabolite profiling of complex
mixtures (i.e. fruit juices, wines, spirits, urine and blood) (Ogrinc et al. 2003). NMR
spectroscopy is increasingly renowned for its efficacy, non-invasiveness
(non-destructive), throughput and linearity (Laghi et al. 2014). Moreover, NMR
spectroscopy also provides structural, chemical-kinetics and other information in
multidimensional applications (Dieterle et al. 2011). Thus, high resolution NMR
spectroscopy along with multivariate data analysis has been used for direct char-
acterisation of fruit juices (Cuny et al. 2008), wine (Lee et al. 2009; Pinu et al.
2014), grape berries (Pereira et al. 2006; Mulas et al. 2011), olive oil (Del Coco
et al. 2012; Piccinonna et al. 2016) and beer (Almeida et al. 2006; Rodrigues et al.
2011). To obtain a global metabolite profile of a complex sample, NMR needs to be
coupled with another non-targeted analytical approach (e.g. MS). But there are
some serious drawbacks to using NMR for metabolome analysis. Different
parameters of the sample, e.g. salinity, pH and concentrations of metal ions, can
affect the sensitivity of NMR spectrometers and can cause difficulty in
bioinformatics-based resonance assignments (Lewis et al. 2012). To avoid these
236 F.R. Pinu
MS ion source, making GC-MS the best combination of separation technique with
mass spectrometry detection. The instrumentation has been developed considerably
during the last 50 years. GC-MS is a highly sensitive analytical platform, which
also provides excellent instrument repeatability, around 5 % or below (Villas-Bôas
et al. 2005). However, an extra step of sample derivatisation is required for the
analysis of semi- and non-volatile metabolites, and GC-MS has been widely used
for the last 50 years for the analysis of a wide range of metabolites present in foods
(Table 1). LC is another powerful separation technique that allows rapid analysis of
small amounts of sample. The main advantage of this separation technique over GC
is that no previous derivatisation of the sample is required to analyse the
non-volatile compounds (Scalbert et al. 2009). This technique is often coupled with
MS and sometimes also with NMR (Shulaev 2006; Zhou et al. 2012). For LC-MS, a
wide range of different detectors is used, ranging from ultra-high resolution MS
such as FTICR or orbitrap to low-resolution MS such as ion traps and triple quads
and hybrid systems. The most recent addition is the ion-mobility TOF-MS system
(Lanucara et al. 2014). The development of methods depends on the nature of the
metabolites to be analysed. LC-MS has been applied widely in metabolite profiling
(both targeted and non-targeted) of complex biological samples (Berg et al. 2012;
Theodoridis et al. 2012). CE is an efficient and rapid technique that can separate a
wide range of charged metabolites within a single analytical run (Villas-Bôas et al.
2005). CE is often coupled to MS and it is considered a promising analytical
instrument in metabolomics (Shulaev 2006). CE-MS has high resolving power and
requires very small volumes of samples (1–20 nL). Thus, it has been used for both
targeted and non-targeted high throughput analysis of metabolites (Nevedomskaya
et al. 2010; Sato and Yanagisawa 2010; Ramautar et al. 2011). The major drawback
for CE-MS is its poor sensitivity; thus the detection limit is several magnitudes
higher than those of chromatographic methods (Table 1). The introduction of a
small volume of sample is inadequate for the detection of many metabolites (Cai
and Henion 1995). Moreover, low recovery and irreversible adsorption of analytes
onto the capillary wall also can occur in CE. For these reasons, CE-MS is mostly
used in metabolomics as a combination of different protocols targeting different
groups of metabolites combined with sample preparation steps to concentrate the
metabolites in the samples, making it a lower throughput technology in metabo-
lomics and certainly more suitable for targeted analysis.
Fourier transform infrared spectroscopy (FTIR) is another analytical technique
that is extensively applied in the food industry because it is rapid, highly automated,
reproducible, non-destructive and cost-effective (Bauer et al. 2008; Versari et al.
2010). Nevertheless, FTIR is insensitive for complex liquid samples and water in
the samples often increases the background noise, increasing the limit of detection
and reducing linearity (Achá et al. 1998). The data obtained from FTIR are also
very complex and there few databases available for assisting with identification of
metabolites (Berthomieu and Hienerwadel 2009).
Table 1 Comparisons of different analytical instruments used in food safety and quality research
238
A large number of chemicals have entered our food chain as a result of the wide
application of different growth-promoting agents (e.g. clenbuterol in pigs, recom-
binant growth hormones in fish and steroids in bovine animals), antibiotics and
pesticides that help us to produce large amounts of agricultural products. Therefore,
both food ingredients and the environment are facing the burden of chemical
exposures, many of which are unwanted and pose a threat to human health.
Moreover, deliberate contamination of food ingredients with unwanted chemicals
(e.g. melamine) also poses a serious risk to consumers. Therefore, these chemicals
should be banned or their use should be limited as set by the regulatory authorities.
To achieve this, development of high throughput methods that would enable study
of these contaminants is urgently needed. Both targeted and non-targeted meta-
bolomics demonstrate enormous potential in detecting and identifying these
chemicals in our food. For instance, targeted analysis using a suitable analytical
platform (e.g. NMR and MS) is beneficial for the study of already-known chemical
contaminants in food. However, it is more problematic when the contaminants are
either not known or are unknown breakdown products of a familiar compound. An
untargeted metabolomics approach could be a better way to study those novel
compounds and also to determine and validate candidate biomarkers that could be
used for tackling illegal practices in food production (Dervilly-Pinel et al. 2012).
Although NMR was initially the method of choice for detecting and identifying
chemical contaminants in food because of its capacity to quantify and elucidate the
8 Metabolomics: Applications to Food Safety and Quality Research 241
structure of molecules (Dieterle et al. 2011), MS is currently more popular due to its
sensitivity, and robustness of coupling with various separation systems (e.g. GC,
LC and CE). Moreover, tandem MS/MS or MSn experiments can now elucidate the
structure of any unknown metabolites, and thus are useful for the development of
multi-residue methods to detect and identify chemical contaminants accurately.
Many LC-MS-based methods have been published recently that have been used to
study chemical contaminants (Table 1) in different crops, including herbal teas,
nutraceuticals, rice and maize, and also in animals (Castro-Puyana and Herrero
2013). For instance, 255 veterinary drug residues and other chemical contaminants
were determined within 10 min using a UHPLC-MS/MS method (Zhan et al.
2012). In addition, high resolution MS/MS has been widely applied for the analysis
of pesticides, toxins and antibiotics. More than 500 pesticides were screened in
fruits and vegetables using an orbitrap tandem MS/MS method (Alder et al. 2011).
De Dominicis et al. (2012) also reported an orbitrap-MS/MS-based protocol for
analysing pesticides and toxins simultaneously in bakery and other foods. In
addition to analysing pesticides and other xenobiotic molecules, attempts have also
been made to develop methods using LC-MS to identify other contaminants in
food, including melamine in infant formula (Inoue et al. 2015). Many other
LC-MS/MS protocols have already been published that analysed a wide range of
food and fermented products including fruits, vegetables, wine, baby foods and
cereals (Lacina et al. 2012; Pérez-Ortega et al. 2012; Fan et al. 2014).
While LC has been the choice of separation technique for the analysis of con-
taminants in food materials, GC still remains the mostly used system coupled with
high resolution MS for the determination of environmental pollutants in food such
as polychlorinated biphenyls (PCBs), polycyclic aromatic hydrocarbons (PAHs) or
polybrominated diphenyl ethers (PBDEs) in various samples including fruits,
vegetables, cereals, teas, fish muscle, dietary supplements and sheep milk
(Mastovska and Wylie 2012; Storelli et al. 2012; Banerjee et al. 2013; Cao et al.
2014; Duedahl-Olesen et al. 2015a, b; Walorczyk et al. 2015). For example,
Sapozhnikova and Lehotay (2013) accurately quantified over 80 pollutants (PCBs,
PAHs and pesticides) in fish within 9 min using a low pressure GC-MS/MS
method. In addition to tandem MS used with a GC system, two-dimensional GC
(GCxGC) analysis is also gaining popularity for the analysis of pollutants, which
allows determination of these compounds in very small amounts (0.1 µg/kg)
(Kalachova et al. 2012, 2013; Giri et al. 2015).
Some applications of food safety in terms of chemical contaminant determina-
tion also make use of direct MS analysis either as direct analysis in real time
(DART) or as desorption electrospray analysis (DESI). Rapid and high throughput
analysis of food samples can be obtained using DART-MS, as it directly analyses
samples from the surface, thus decreasing sample preparation time significantly
compared with those for the other coupled MS systems (Castro-Puyana et al. 2013).
DART-MS combined with high resolution analysers has been applied to analyse
pesticides in fruits and grains (Schurek et al. 2008; Farré et al. 2013). Similarly,
DESI-MS has been used for the determination of pesticides in fruit peels and
vegetables (García-Reyes et al. 2009; Zhang et al. 2009).
242 F.R. Pinu
Food pathogens are one of the major threats for food safety, and large numbers of
people around the world suffer from diseases including diarrhoea, dysentery and
other forms of food poisoning that are directly caused by various food pathogens,
such as Salmonella spp., Shigella spp., Listeria monocytogenes, Campylobacter
jejuni and E. coli. Moreover, food spoilage microorganisms (e.g. Pseudomonas
spp., Acinetobacter spp., Botrytis spp.) are not necessarily pathogenic for people,
but can cause severe economic damage because of the spoilage of a wide range of
food materials. If there was a way to detect these pathogens or spoilage microbes in
the early stages of their growth in food products, it would be possible to reduce
dramatically the numbers of food-borne outbreaks and subsequent significant losses
of food by undertaking appropriate measurements to control their further growth.
Traditional identification and cultural methods for pathogens or food spoilage
microbes are time consuming, and therefore it is necessary to develop techniques
that would enable the rapid detection of microbes soon after the contamination
occurred in a food system (Xu et al. 2010). The metabolomics approach has already
shown huge potential for developing analytical methods to detect food pathogens in
their early stages of growth in a food product (Li et al. 2011; Beale et al. 2014). In
this approach, an experiment is performed using both a contaminated and a
non-contaminated food, and the metabolites from both are analysed. Multivariate or
discriminant analyses are performed to determine a group of potential candidate
biomarkers that can distinguish between the two conditions. Once validated, these
biomarkers can be used for the determination of pathogens in a real food sample, or
a simpler screening technique (e.g. enzymatic or colorimetric) can be developed to
be used routinely by food auditors.
GC-MS has been the choice of analytical instrument for determining pathogenic
growth in a food, as this technique is very efficient in determining the volatile
organic compounds (VOC) produced by microorganisms. Food spoilage microbes
generally produce many VOCs as part of their metabolism, and this causes alter-
ation in the sensory properties of the food (Li et al. 2011). VOC analysis is
attractive for food sampling and is also advantageous because of rapid and
non-invasive sample preparation. Once collected in the headspace using an
appropriate SPME cartridge, VOCs produced by spoilage or pathogenic microor-
ganisms are ready to be analysed by GC-MS. Xu et al. (2010) published a
VOC-based GC-MS metabolite profiling approach to determine the potential
biomarkers of spoilage of pork by S. typhimurium, and identified 16 metabolites
that clearly distinguished the naturally spoiled from S typhimurium-contaminated
pork meats. Moreover, Li et al. (2011) demonstrated the capability of metabolomics
in assigning biomarkers to the spoilage microbes, Botrytis allii and Burkholderia
cepacia, and they identified 16 volatile metabolites related to post-harvest onion
spoilage. In addition to volatile metabolites, other primary metabolites including
sugars and amino acids (dextrose, glycine, tyrosine and histidine) were also
8 Metabolomics: Applications to Food Safety and Quality Research 243
characterised as potential biomarkers that could be used for the early detection of
E. coli O157:H7 and of different strains of Salmonella in ground beef and chicken
(Cevallos-Cevallos et al. 2011).
Among other MS-based techniques, Matrix-assisted laser desorption/ionisation
coupled to time of flight-MS (MALDI-TOF-MS) also has been used to identify
different strains of pathogenic microbes in biological samples, including food and
beverages (Böhme et al. 2010, 2012; Picariello et al. 2012; Ojima-Kato et al. 2014;
Beale et al. 2014; Jadhav et al. 2015). MALDI-TOF-MS is very useful for iden-
tifying microbial strains using either whole cells or cell extracts and has been
applied to epidemiological studies, biological warfare agents, detection of antibiotic
resistance pathogens, detection of water- and food-borne pathogens and entero-
toxins (Tsilia et al. 2012). This technique is rapid, sensitive and inexpensive, and
thus well received by microbiologists (Singhal et al. 2015). Recently, rapid
detection and source tracking of L. monocytogenes was carried out using
MALDI-TOF-MS in Australian dairy products (Jadhav et al. 2015). Staphylococcus
aureus strain characterisation was also performed in Italian dairy products using
MALDI-TOF-MS fingerprinting (Böhme et al. 2012). However, the main drawback
of using this technique is the inability to identify new species, as the identification
process depends on the existing spectral database that contains the peptide mass
fingerprints of the type strains (Singhal et al. 2015).
In contrast to chemical contaminants, food toxins that pose serious health hazards
for consumers are mainly produced naturally by different microorganisms (fungi,
algae and others) growing on food substrates or during the food preparation and
storage. Many of these toxins are produced as by-products of fungal metabolism
(mainly Penicillium, Aspergillus and Fusarium) in contaminated foods including
beer, wine, bread, rice and maize, and are thus known as mycotoxins. The deter-
mination of mycotoxins is very similar to that of chemical contaminants and mainly
carried out by multi-residue analysis (Giacometti et al. 2013; Gottfried and
Herebian 2013; Aniołowska and Steininger 2014; Pizzutti et al. 2014;
Rodríguez-Carrasco et al. 2015). In addition to mycotoxins, toxins produced by
some algae are also of interest for food safety. This type of toxin is usually found in
contaminated seafood and fish and may affect people, such as fish and different
types of shellfish poisoning (e.g. neurotoxic, paralytic, diarrhoeic and amnesic). It is
important to study algal toxins to know about their modes of action and toxic doses
in detail, to help to take appropriate precautions to prevent an outbreak. MS-based
metabolomics approaches have been developed to detect and quantify several
groups of algal toxins, such as ciguatoxins and domoic acid (Yogi et al. 2011;
Beach et al. 2014; Stewart and McLeod 2014).
244 F.R. Pinu
GM or transgenic crops have gained popularity since the first commercial plantings
in 1996. GM technology has allowed us to produce crops and food materials with
enhanced nutritional properties, increased yield; and moreover, these crops can be
resistant to various pests and diseases, and adverse environments, including drought
and salinity (Parrott et al. 2010). A wide range of GM crops are now commercially
cultivated around the world, including soybean, maize, cotton, canola, potatoes and
tomatoes. Although the new traits in GM crops are mainly from the introduction of
small RNAs or some regulatory proteins, it is still unknown if even that small
amount of change could cause an overall alteration in other metabolic pathways,
and thus adverse modification of cellular downstream products (Delaney 2015). The
effects of growing transgenic plants on the environment have also been well doc-
umented (Liu et al. 2012; Brookes and Barfoot 2013; Knight et al. 2013), which has
in turn raised concerns about the long-term consumption of GM food materials on
human and animal health.
As the long-term effects of consuming GM food ingredients are still under
question, it is time to do more research on toxicological aspects that could provide
assurance to consumers about their safety. A few research projects have already
been carried out that addressed the effects of consumption of GM feed ingredients
by animal models. For instance, Sheng et al. (2014) evaluated the toxicity and
allergenicity of GM rice as expressed in human serum albumin in rats, and Qi et al.
(2015) assessed the safety of consumption of similar GM rice by rats using their
urine metabolome. Although both sets of authors found significant differences in
profiles between GM and non-GM rice-consuming rats, they concluded that GM
rice could be considered safe. However, both these studies were performed for a
limited period of time (only 90 days), and therefore it is still too soon to conclude if
long-term consumption (e.g. 5–10 years) of such GM rice by the rats would have
any adverse effects on their health.
Much research has been undertaken in last 10 years to acquire more knowledge
regarding the compositional differences between GM and non-GM crops and food
materials (Barros et al. 2010; Asiago et al. 2012; Cao et al. 2012; Liu et al. 2012;
Clarke et al. 2013; Kusano et al. 2014). This provides valuable insight into the
nutritional properties compared with those of conventional crops, and if any
unintended chemicals or proteins are present in that given crop that might pose a
threat to consumers. Metabolomics (both targeted and non-targeted), especially
metabolite profiling, is very useful in this regard, as it allows the generation of data
on the comprehensive composition of any GM crop (Rischer and
Oksman-Caldentey 2006). For example, Kusano et al. (2014) recently published a
study using metabolomics and ionomics approaches to determine the chemical
diversity of a soybean lineage representing 35 years of breeding. The authors used
different analytical platforms, including CE-TOF-MS, GC-TOF-MS and
LC-qTOF-MS, to determine the global metabolite profiles of soybeans, and found
that newer varieties are completely different from older ones. However, they found
8 Metabolomics: Applications to Food Safety and Quality Research 245
Both targeted and non-targeted metabolomics have been widely applied to determine
food quality by analysing any adulteration during food processing, or to confirm
authenticity of a food or beverage. Adulteration is usually performed deliberately to
gain more profit, using unwanted substances, such as the addition of melamine to
milk powder, synthetic dyes in spices, and the use of horsemeat in other meat
products (Senyuva et al. 2015). Some of these adulterations might cause serious
health issues (Chu and Wang 2013) and some of them are related to economic
adulterations to consumers (Everstine et al. 2013). However, in all cases, it is
essential to know the overall quality of the food materials and also to determine if the
food product is adulterated or not before it can be sold in the market. Authentication
of food is a useful step, and attempts have been made to determine potential markers
in different food matrices that can differentiate between adulterated and normal food.
For example, both targeted and untargeted metabolomics approaches using
LC-MS/MS have been used to authenticate Indian citrus fruits, and several
metabolites (didymin, rhoifolin, isorhoifolin, neohesperidin, hesperidin, naringin,
narirutin, limonin glucoside, and vicenin) were characterised as potential markers
(Jandrić et al. 2015). Arias et al. (2016) recently published a metabolomics study
using UHPLC–QTof-MS (ESI + mode) to distinguish the control (non-medicated
pigs) and pigs treated with ronidazole, dimetridazole and metronidazole where they
have identified at least four ionic features that could be used as potential biomarkers
of illegal 5-nitroimidazole abuse. GC-MS is another popular analytical platform that
has been successfully implemented to determine the quality of various food products.
For instance, GC-MS has been used to monitor the kimchi fermentation process (Park
et al. 2016). Moreover, fatty acid profiling using GC-MS has also been found to be
useful in determining adulteration in flaxseed oil (Sun et al. 2015). Recently,
Isotope-ratio MS (IRMS) along with either GC or LC has become another technique
that can be used for authentication of foods including wines, essential oils in man-
darin, and flavoured strawberry foods (Schipilliti et al. 2010; Guyon et al. 2011;
246 F.R. Pinu
7 Conclusion
Acknowledgements The author would like to thank Dr Damian Martin, Megan Jones and
Science and Publishing Office of Plant and Food Research Ltd for their support during the writing
of this manuscript.
References
Achá V, Naveau H, Meurens M (1998) Extractive sampling methods to improve the sensitivity of
FTIR spectroscopy in analysis of aqueous liquids. Analusis 26:157–163
Ackerman LK, Noonan GO, Begley TH (2009) Assessing direct analysis in real-time-mass
spectrometry (DART-MS) for the rapid identification of additives in food packaging. Food
Addit Contam Part A 26:1611–1618
Aggio R, Villas-Bôas SG, Ruggiero K (2011) Metab: an R package for high-throughput analysis
of metabolomics data generated by GC-MS. Bioinformatics 27:2316–2318
Aggio RBM, Mayor A, Reade S, Probert CSJ, Ruggiero K (2014) Identifying and quantifying
metabolites by scoring peaks of GC-MS data. BMC Bioinformatics 15:374
Alder L, Steinborn A, Bergelt S (2011) Suitability of an orbitrap mass spectrometer for the
screening of pesticide residues in extracts of fruits and vegetables. J AOAC Int 94:1661–1673
Almeida C, Duarte IF, Barros A, Rodrigues J, Spraul M, Gil AM (2006) Composition of beer by
1H NMR spectroscopy: effects of brewing site and date of production. J Agric Food Chem
54:700–706
Alvarez-Ordóñez A, Broussolle V, Colin P, Nguyen-The C, Prieto M (2015) The adaptive
response of bacterial food-borne pathogens in the environment, host and food: implications for
food safety. Int J Food Microbiol 213:99–109
Anater A, Manyes L, Meca G, Ferrer E, Luciano FB, Pimpão CT, Font G (2016) Mycotoxins and
their consequences in aquaculture: a review. Aquaculture 451:1–10
Aniołowska M, Steininger M (2014) Determination of trichothecenes and zearalenone in different
corn (Zea mays) cultivars for human consumption in Poland. J Food Compos Anal 33:14–19
Anizan S, Bichon E, Monteau F, Cesbron N, Antignac JP, le Bizec B (2010) A new reliable
sample preparation for high throughput focused steroid profiling by gas chromatography-mass
spectrometry. J Chromatogr A 1217:6652–6660
Anizan S, Bichon E, Duval T, Monteau F, Cesbron N, Antignac JP, Le Bizec B (2012) Gas
chromatography coupled to mass spectrometry-based metabolomic to screen for anabolic
practices in cattle: identification of 5a-androst-2-en-17-one as new biomarker of
4-androstenedione misuse. J Mass Spectrom 47:131–140
248 F.R. Pinu
Arias M, Chevallier OP, Graham SF, Gasull-Gimenez A, Fodey T, Cooper KM, Crooks SRH,
Danaher M, Elliott CT (2016) Metabolomics reveals novel biomarkers of illegal
5-nitromimidazole treatment in pigs. Further evidence of drug toxicity uncovered. Food
Chem 199:876–884
Arroyo-Manzanares N, Diana Di Mavungu J, Uka V, Malysheva SV, Cary JW, Ehrlich KC,
Vanhaecke L, Bhatnagar D, de Saeger S (2015) Use of UHPLC high-resolution Orbitrap mass
spectrometry to investigate the genes involved in the production of secondary metabolites in
Aspergillus flavus. Food Addit Contam Part A 32:1656–1673
Aru V, Pisano MB, Savorani F, Engelsen SB, Cosentino S, Cesare Marincola F (2016)
Metabolomics analysis of shucked mussels’ freshness. Food Chem 205:58–65
Asiago VM, Hazebroek J, Harp T, Zhong C (2012) Effects of genetics and environment on the
metabolome of commercial maize hybrids: a multisite study. J Agric Food Chem 60:11498–
11508
Badilita V, Adhikari SS, MacKinnon N, Wallrabe U, Korvink JG (2014) Disposable 1H NMR
detectors for food quality monitoring: application to butter and citrus juice discrimination.
Chem Biol Microsyst Soc 1710–1712
Balan M, Nicolescu A, Stavarache C, Ciobanu M, Deleanu C (2013) Fast NMR juice identification
based on sugars and other plant metabolites from fruits. Rev Roum Chim 58:175–182
Banerjee K, Mujawar S, Utture SC, Dasgupta S, Adsule PG (2013) Optimization of gas
chromatography-single quadrupole mass spectrometry conditions for multiresidue analysis of
pesticides in grapes in compliance to EU-MRLs. Food Chem 138:600–607
Barros E, Lezar S, Anttonen MJ, van Dijk JP, Röhlig RM, Kok EJ, Engel KH (2010) Comparison
of two GM maize varieties with a near-isogenic non-GM variety using transcriptomics,
proteomics and metabolomics. Plant Biotechnol J 8:436–451
Bauer R, Nieuwoudt H, Bauer FF, Kossmann J, Koch KR, Esbensen KH (2008) FTIR
spectroscopy for grape and wine analysis. Anal Chem 80:1371–1379
Beach DG, Walsh CM, McCarron P (2014) High-throughput quantitative analysis of domoic acid
directly from mussel tissue using Laser Ablation Electrospray Ionization—tandem mass
spectrometry. Toxicon 92:75–80
Beale DJ, Morrison PD, Palombo EA (2014) Detection of Listeria in milk using non-targeted
metabolic profiling of Listeria monocytogenes: a proof-of-concept application. Food Control
42:343–346
Begley M, Hill C (2015) Stress adaptation in food-borne pathogens. Ann Rev Food Sci Technol
6:191–210
Berg M, Vanaerschot M, Jankevics A, Cuypers B, Breitling R, Dujardin JC (2012) LC-MS
metabolomics from study design to data-analysis—using a versatile pathogen as a test case.
Comput Struct Biotechnol J 4:e201301002
Berthomieu C, Hienerwadel R (2009) Fourier transform infrared (FTIR) spectroscopy. Photosynth
Res 101:157–170
Biais B, Bernillon S, Deborde C, Cabasson C, Rolin D, Tadmor Y, Burger J, Schaffer AA,
Moing A (2012) Precautions for harvest, sampling, storage, and transport of crop plant
metabolomics samples. Methods Mol Biol 860:51–63
Bino RJ, Hall RD, Fiehn O et al (2004) Potential of metabolomics as a functional genomics tool.
Trends Plant Sci 9:418–425
Böhme K, Fernández-No IC, Barros-Velázquez J, Gallardo JM, Calo-Mata P, Cañas B (2010)
Species differentiation of seafood spoilage and pathogenic gram-negative bacteria by
MALDI-TOF mass fingerprinting. J Proteome Res 9:3169–3183
Böhme K, Morandi S, Cremonesi P, Fernández No IC, Barros-Velázquez J, Castiglioni B,
Brasca M, Cañas B, Calo-Mata P (2012) Characterization of Staphylococcus aureus strains
isolated from Italian dairy products by MALDI-TOF mass fingerprinting. Electrophoresis
33:2355–2364
Booth SC, Weljie AM, Turner RJ (2015) Metabolomics reveals differences of metal toxicity in
cultures of Pseudomonas pseudoalcaligenes KF707 grown on different carbon. Front
Microbiol 6:827
8 Metabolomics: Applications to Food Safety and Quality Research 249
Cirlini M, Caligiani A, Palla L, Palla G (2011) HS-SPME/GC-MS and chemometrics for the
classification of Balsamic Vinegars of Modena of different maturation and ageing. Food Chem
124:1678–1683
Clarke JD, Alexander DC, Ward DP, Ryals JA, Mitchell MW, Wulff JE, Guo L (2013)
Assessment of genetically modified soybean in relation to natural variation in the soybean seed
metabolome. Sci Rep 3:3083
Cuny M, Vigneau E, Le Gall G, Colquhoun I, Lees M, Rutledge DN (2008) Fruit juice
authentication by 1H NMR spectroscopy in combination with different chemometric tools.
Anal Bioanal Chem 390:419–427
D’Alessandro A, Zolla L (2013) Foodomics to investigate meat tenderness. TrAC Trend Anal
Chem 52:47–53
Da Rosa JS, Freitas-Silva O, Pacheco S, de Oliveira Godoy RL, de Rezende CM (2012) LC-MS
based screening and targeted profiling methods for complex plant: coffee a case study. Curr
Drug Metab 13:1244–1250
Da Silva EP, De Martinis ECP (2013) Current knowledge and perspectives on biofilm formation:
The case of Listeria monocytogenes. Appl Microbiol Biotechnol 97:957–968
Dawson IK, Russell J, Powell W, Steffenson B, Thomas WTB, Waugh R (2015) Barley: a
translational model for adaptation to climate change. New Phytol 206:913–931
De Dominicis E, Commissati I, Suman M (2012) Targeted screening of pesticides, veterinary
drugs and mycotoxins in bakery ingredients and food commodities by liquid
chromatography-high-resolution single-stage Orbitrap mass spectrometry. J Mass Spectrom
47:1232–1241
Delaney B (2015) Safety assessment of foods from genetically modified crops in countries with
developing economies. Food Chem Toxicol 86:132–143
Del Coco L, Schena FP, Fanizzi FP (2012) 1H Nuclear magnetic resonance study of olive oils
commercially available as Italian products in the United States of America. Nutrients 4:343–
355
Dervilly-Pinel G, Courant F, Chéreau S, Royer AL, Boyard-Kieken F, Antignac JP, Monteau F, Le
Bizec B (2012) Metabolomics in food analysis: application to the control of forbidden
substances. Drug Test Anal 4:59–69
Dais P, Hatzakis E (2013) Quality assessment and authentication of virgin olive oil by NMR
spectroscopy: a critical review. Anal Chim Acta 765:1–27
Dieterle F, Riefke B, Schlotterbeck G, Ross A, Senn H, Amberg A (2011) NMR and MS methods
for metabonomics. Methods Mol Biol 691:385–415
Doué M, Dervilly-Pinel G, Pouponneau K, Monteau F, le Bizec B (2015) Direct analysis in real
time—high resolution mass spectrometry (DART-HRMS): a high throughput strategy for
identification and quantification of anabolic steroid esters. Drug Test Anal 7:603–608
Duedahl-Olesen L, Navaratnam MA, Jewula J, Jensen AH (2015a) PAH in some brands of tea and
coffee. Polycyclic Aromat Compd 35:74–90
Duedahl-Olesen L, Aaslyng M, Meinert L, Christensen T, Jensen AH, Binderup ML (2015b)
Polycyclic aromatic hydrocarbons (PAH) in Danish barbecued meat. Food Control 57:169–176
Dunn WB (2011) Mass spectrometry in systems biology: an introduction. Methods Enzymol
500:15–35
Dunn WB, Ellis DI (2005) Metabolomics: current analytical platforms and methodologies. TrAC
Trend Anal Chem 24:285–294
Duportet X, Aggio RBM, Carneiro S, Villas-Bôas SG (2012) The biological interpretation of
metabolomic data can be misled by the extraction method used. Metabolomics 8:410–421
El-Aneed A, Cohen A, Banoub J (2009) Mass spectrometry, review of the basics: electrospray,
MALDI, and commonly used mass analyzers. Appl Spectrosc Rev 44:210–230
El Amrani A, Dumas AS, Wick LY, Yergeau E, Berthomé R (2015) “Omics” Insights into PAH
degradation toward improved green remediation biotechnologies. Environ Sci Technol
49:11281–11291
Everstine K, Spink J, Kennedy S (2013) Economically motivated adulteration (EMA) of food:
common characteristics of EMA incidents. J Food Prot 76:723–735
8 Metabolomics: Applications to Food Safety and Quality Research 251
Falkowska L, Reindl AR (2015) Dietary exposure to, and internal organ transfer of, selected
halogenated organic compounds in birds eating fish from the Southern Baltic. J Environ Sci
Health Part A Toxiic/Hazard Subst Environ Eng 50:1029–1039
Fan S, Zhao P, Yu C, Pan C, Li X (2014) Simultaneous determination of 36 pesticide residues in
spinach and cauliflower by LC-MS/MS using multi-walled carbon nanotubes-based dispersive
solid-phase clean-up. Food Addit Contam Part A 31:73–82
Farré M, Picó Y, Barceló D (2013) Direct peel monitoring of xenobiotics in fruit by direct analysis
in real time coupled to a linear quadrupole ion trap-orbitrap mass spectrometer. Anal Chem
85:2638–2644
Fiehn O (2002) Metabolomics—the link between genotypes and phenotypes. Plant Mol Biol
48:155–171
Font G, Ruiz MJ, Fernández M, Picó Y (2008) Application of capillary electrophoresis-mass
spectrometry for determining organic food contaminants and residues. Electrophoresis
29:2059–2078
Forsberg ND, Wilson GR, Anderson KA (2011) Determination of parent and substituted
polycyclic aromatic hydrocarbons in high-fat salmon using a modified quechers extraction,
dispersive SPE and GC-MS. J Agric Food Chem 59:8108–8116
García-Gómez D, García-Hernández M, Rodríguez-Gonzalo E, Carabias-Martínez R (2012) A fast
and reliable method for the quantitative determination of benzimidazoles and metabolites in
milk by LC-MS/MS with on-line sample treatment. Anal Bioanal Chem 404:2909–2914
García-Reyes JF, Jackson AU, Molina-Díaz A, Cooks RG (2009) Desorption electrospray
ionization mass spectrometry for trace analysis of agrochemicals in food. Anal Chem 81:820–
829
Gelting RJ, Baloch MA, Zarate-Bermudez M, Hajmeer MN, Yee JC, Brown T, Yee BJ (2015) A
systems analysis of irrigation water quality in an environmental assessment of an E. coli O157:
H7 outbreak in the United States linked to iceberg lettuce. Agric Water Manag 150:111–118
Giacometti J, Tomljanović AB, Josić D (2013) Application of proteomics and metabolomics for
investigation of food toxins. Food Res Int 54:1042–1051
Gilbert-López B, Robles-Molina J, García-Reyes JF, Molina-Díaz A (2010) Rapid determination
of BTEXS in olives and olive oil by headspace-gas chromatography/mass spectrometry
(HS-GC-MS). Talanta 83:391–399
Giri A, Khummueng W, Mercier F, Kondjoyan N, Tournayre P, Meurillon M, Ratel J, Engel E
(2015) Relevance of two-dimensional gas chromatography and high resolution olfactometry
for the parallel determination of heat-induced toxicants and odorants in cooked food.
J Chromatogr A 1388:217–226
Glinski M, Weckwerth W (2006) The role of mass spectrometry in plant systems biology. Mass
Spectrom Rev 25:173–214
Gottfried S, Herebian D (2013) Determination of mycotoxins in food matrices using LC-MS/MS
compared with high-resolution orbitrap™ MS technology. Curr Anal Chem 9:99–107
Gowda H, Ivanisevic J, Johnson CH, Kurczy ME, Benton HP, Rinehart D, Nguyen T, Ray J,
Kuehl J, Arevalo B, Westenskow PD, Wang J, Arkin AP, Deutschbauer AM, Patti GJ,
Siuzdak G (2014) Interactive XCMS online: simplifying advanced metabolomic data
processing and subsequent statistical analyses. Anal Chem 86:6931–6939
Grimalt S, Harbeck S, Shegunova P, Seghers J, Sejerøe-Olsen B, Emteborg H, Dabrio M (2015)
Development of a new cucumber reference material for pesticide residue analysis: feasibility
study for material processing, homogeneity and stability assessment. Anal Bioanal Chem
407:3083–3091
Guo C, Huang XY, Yang MJ, Wang S, Ren ST, Li H, Peng XX (2014) GC/MS-based
metabolomics approach to identify biomarkers differentiating survivals from death in crucian
carps infected by Edwardsiella tarda. Fish Shellfish Immunol 39:215–222
Gustafson KD, Belden JB, Bolek MG (2015) The effects of the herbicide atrazine on freshwater
snails. Ecotoxicology 24:1183–1197
252 F.R. Pinu
Guyon F, Gaillard L, Salagoïty MH, Médina B (2011) Intrinsic ratios of glucose, fructose, glycerol
and ethanol 13C/12C isotopic ratio determined by HPLC-co-IRMS: toward determining
constants for wine authentication. Anal Bioanal Chem 401:1551–1558
Hall R, Beale M, Fiehn O, Hardy N, Sumner L, Bino R (2002) Plant metabolomics: the missing
link in functional genomics strategies. Plant Cell 14:1437–1440
Hall RD, Hardy NW (2012) Practical applications of metabolomics in plant biology. Methods Mol
Biol 860:1–10
Hall RD, de Maagd RA (2014) Plant metabolomics is not ripe for environmental risk assessment.
Trend Biotechnol 32:391–392
Han J, Datla R, Chan S, Borchers CH (2009) Mass spectrometry-based technologies for
high-throughput metabolomics. Bioanalysis 1:1665–1684
Han Y, Song L, Zhao P, Li Y, Zou N, Qin Y, Li X, Pan C (2016) Residue determination of
glufosinate in plant origin foods using modified Quick Polar Pesticides (QuPPe) method and
liquid chromatography coupled with tandem mass spectrometry. Food Chem 197:730–736
Harrigan GG, Stork LG, Riordan SG et al (2007) Impact of genetics and environment on
nutritional and metabolite components of maize grain. J Agric Food Chem 55:6177–6185
Herman KM, Hall AJ, Gould LH (2015) Outbreaks attributed to fresh leafy vegetables, United
States, 1973–2012. Epidemiol Infect 143:3011–3021
Herrero M, Simõ C, García-Cañas V, Ibáñez E, Cifuentes A (2012) Foodomics: MS-based
strategies in modern food science and nutrition. Mass Spectrom Rev 31:49–69
Humphrey T (2004) Salmonella, stress responses and food safety. Nat Rev Microbiol 2:504–509
Ibáñez C, García-Cañas V, Valdés A, Simó C (2013) Novel MS-based approaches and
applications in food metabolomics. TrAC Trend Anal Chem 52:100–111
Ibáñez E, Cifuentes A (2014) Foodomics: food science and nutrition in the postgenomic era, vol
64. Elsevier, Amsterdam, pp 395–440
Inoue K, Toyo’oka T (2015) Foodomics, vol 68. Elsevier, Amsterdam, pp 653–684
Inoue K, Tanada C, Sakamoto T, Tsutsui H, Akiba T, Min JZ, Todoroki K, Yamano Y, Toyo’Oka
T (2015) Metabolomics approach of infant formula for the evaluation of contamination and
degradation using hydrophilic interaction liquid chromatography coupled with mass
spectrometry. Food Chem 181:318–324
Ison SA, Delannoy S, Bugarel M, Nightingale KK, Webb HE, Renter DG, Nagaraja TG,
Loneragan GH, Fach P (2015) Genetic diversity and pathogenic potential of attaching and
effacing Escherichia coli O26: H11 strains recovered from bovine feces in the United States.
Appl Environ Microbiol 81:3671–3678
Jadhav S, Gulati V, Fox EM, Karpe A, Beale DJ, Sevior D, Bhave M, Palombo EA (2015) Rapid
identification and source-tracking of Listeria monocytogenes using MALDI-TOF mass
spectrometry. Int J Food Microbiol 202:1–9
Jandrić Z, Islam M, Singh DK, Cannavan A (2015) Authentication of Indian citrus fruit/fruit juices
by untargeted and targeted metabolomics. Food Control (in press)
Janssens G, Mangelinckx S, Courtheyn D, De Kimpe N, Matthijs B, Le Bizec B (2015)
Simultaneous detection of androgen and estrogen abuse in breeding animals by gas
chromatography-mass spectrometry/combustion/isotope ratio mass spectrometry
(GC-MS/C/IRMS) evaluated against alternative methods. J Agric Food Chem 63:7574–7581
Jäpelt KB, Nielsen NJ, Wiese S, Christensen JH (2015) Metabolic fingerprinting of Lactobacillus
paracasei: a multi-criteria evaluation of methods for extraction of intracellular metabolites.
Anal Bioanal Chem 407:6095
Jiang H, Cao H, Zhang Y, Fast DM (2012) Systematic evaluation of supported liquid extraction in
reducing matrix effect and improving extraction efficiency in LC-MS/MS based bioanalysis for
10 model pharmaceutical compounds. J Chromatogr B Anal Technol Biomed Life Sci 891–
892:71–80
Juan-Borrás M, Periche A, Domenech E, Escriche I (2015) Routine quality control in honey
packaging companies as a key to guarantee consumer safety. The case of the presence of
sulfonamides analyzed with LC-MS-MS. Food Control 50:243–249
8 Metabolomics: Applications to Food Safety and Quality Research 253
Ogrinc N, Košir IJ, Spangenberg JE, Kidrič J (2003) The application of NMR and MS methods for
detection of adulteration of wine, fruit juices, and olive oil. A review Anal Bioanal Chem
376:424–430
Ojima-Kato T, Yamamoto N, Suzuki M, Fukunaga T, Tamura H (2014) Discrimination of
Escherichia coli O157, O26 and O111 from other serovars by MALDI-TOF MS based on the
S10-germs method. PLoS One 9:e113458
O’Leary D, McCabe EM, McCusker MP, Martins M, Fanning S, Duffy G (2015) Acid
environments affect biofilm formation and gene expression in isolates of Salmonella enterica
Typhimurium DT104. Int J Food Microbiol 206:7–16
Ordoudi SA, Cagliani LR, Lalou S, Naziri E, Tsimidou MZ, Consonni R (2015) 1H NMR-based
metabolomics of saffron reveals markers for its quality deterioration. Food Res Int 70:1–6
Osselaere A, Devreese M, Goossens J, Vandenbroucke V, De Baere S, De Backer P, Croubels S
(2013) Toxicokinetic study and absolute oral bioavailability of deoxynivalenol, T-2 toxin and
zearalenone in broiler chickens. Food Chem Toxicol 51:350–355
OzFoodNet Working Group (2014) OzFoodNet quarterly report, 1 April to 30 June 2013.
Commun Dis Intell Q Rep 38:E376–E382
Pacilli M, Cortese MM, Smith S et al (2015) Outbreak of gastroenteritis in adults due to Rotavirus
genotype G12P[8]. Clin Infect Dis 61:e20–e25
Park SE, Yoo SA, Seo SH, Lee KI, Na CS, Son HS (2016) GC–MS based metabolomics approach
of Kimchi for the understanding of Lactobacillus plantarum fermentation characteristics. LWT
—Food Sci Technol 68:313–321
Parrott W, Chassy B, Ligon J, Meyer L, Petrick J, Zhou J, Herman R, Delaney B, Levine M (2010)
Application of food and feed safety assessment principles to evaluate transgenic approaches to
gene modulation in crops. Food Chem Toxicol 48:1773–1790
Pereira GE, Gaudillere JP, Pieri P, Hilbert G, Maucourt M, Deborde C, Moing A, Rolin D (2006)
Microclimate influence on mineral and metabolic profiles of grape berries. J Agric Food Chem
54:6765–6775
Pérez-Ortega P, Gilbert-López B, García-Reyes JF, Ramos-Martos N, Molina-Díaz A (2012)
Generic sample treatment method for simultaneous determination of multiclass pesticides and
mycotoxins in wines by liquid chromatography-mass spectrometry. J Chromatogr A 1249:32–
40
Pérez S, Rial D, Beiras R (2015) Acute toxicity of selected organic pollutants to saltwater (mysid
Siriella armata) and freshwater (cladoceran Daphnia magna) ecotoxicological models.
Ecotoxicology 24:1229–1238
Picariello G, Mamone G, Addeo F, Ferranti P (2012) Novel mass spectrometry-based applications
of the ‘omic’: sciences in food technology and biotechnology. Food Technol Biotechnol
50:286–305
Piccinonna S, Ragone R, Stocchero M, del Coco L, de Pascali SA, Schena FP, Fanizzi FP (2016)
Robustness of NMR-based metabolomics to generate comparable data sets for olive oil cultivar
classification. An inter-laboratory study on Apulian olive oils. Food Chem 199:675–683
Pinu FR (2015) Metabolomics-The new frontier in food safety and quality research. Food Res Int
72:80–81
Pinu FR, Edwards PJB, Jouanneau S, Kilmartin PA, Gardner RC, Villas-Boas SG (2014)
Sauvignon blanc metabolomics: grape juice metabolites affecting the development of varietal
thiols and other aroma compounds in wines. Metabolomics 10:556–573
Pisano MB, Scano P, Murgia A, Cosentino S, Caboni P (2016) Metabolomics and microbiological
profile of Italian mozzarella cheese produced with buffalo and cow milk. Food Chem 192:618–
624
Pizarro C, Rodríguez-Tecedor S, Pérez-del-Notario N, González-Sáiz JM (2011) Recognition of
volatile compounds as markers in geographical discrimination of Spanish extra virgin olive oils
by chemometric analysis of non-specific chromatography volatile profiles. J Chromatogr A
1218:518–523
Pizzutti IR, De Kok A, Scholten J, Righi LW, Cardoso CD, Necchi Rohers G, Da Silva RC (2014)
Development, optimization and validation of a multimethod for the determination of 36
256 F.R. Pinu
Wan Q, Wu G, He Q, Tang H, Wang Y (2015) The toxicity of acute exposure to T-2 toxin
evaluated by the metabonomics technique. Mol Biosyst 11:882–891
World Health Organization (2006) WHO consultation to develop a strategy to estimate the global
burden of foodborne diseases. World Health Organization, Geneva, p vii. Available from http://
www.who.int/foodsafety/publications/foodborne_disease/fbd_2006.pdf
Xia J, Mandal R, Sinelnikov IV, Broadhurst D, Wishart DS (2012) MetaboAnalyst 2.0-a
comprehensive server for metabolomic data analysis. Nucleic Acids Res 40:W127–W133
Xu Y, Cheung W, Winder CL, Goodacre R (2010) VOC-based metabolic profiling for food
spoilage detection with the application to detecting Salmonella typhimurium-contaminated
pork. Anal Bioanal Chem 397:2439–2449
Yang Y, Zheng N, Zhao X, Zhang Y, Han R, Yang J, Zhao S, Li S, Guo T, Zang C, Wang J (2016)
Metabolomic biomarkers identify differences in milk produced by Holstein cows and other
minor dairy animals. J Proteomics 136:174–182
Yogi K, Oshiro N, Inafuku Y, Hirama M, Yasumoto T (2011) Detailed LC-MS/MS analysis of
ciguatoxins revealing distinct regional and species characteristics in fish and causative alga
from the pacific. Anal Chem 83:8886–8891
Zeng W, Hazebroek J, Beatty M, Hayes K, Ponte C, Maxwell C, Zhong CX (2014) Analytical
method evaluation and discovery of variation within maize varieties in the context of food
safety: transcript profiling and metabolomics. J Agric Food Chem 62:2997–3009
Zhan J, Yu XJ, Zhong YY, Zhang ZT, Cui XM, Peng JF, Feng R, Liu XT, Zhu Y (2012) Generic
and rapid determination of veterinary drug residues and other contaminants in raw milk by ultra
performance liquid chromatography-tandem mass spectrometry. J Chromatogr B Anal Technol
Biomed Life Sci 906:48–57
Zhang A, Sun H, Wang P, Han Y, Wang X (2012) Modern analytical techniques in metabolomics
analysis. Analyst 137:293–300
Zhang D, Park JA, Kim DS, Kim SK, Shin SJ, Shim JH, Shin SC, Kim JS, Abd El-Aty AM,
Shin HC (2015) A simple extraction method for the simultaneous detection of tetramisole and
diethylcarbamazine in milk, eggs, and porcine muscle using gradient liquid
chromatography-tandem mass spectrometry. Food Chem 192:299–305
Zhang XZ, Li CJ, Chen SS, Li XJ, Han H, Ma XD (2009) Direct determination of atrazine residue
on Chinese cabbage leaf using desorption electrospray ionization-tandem mass spectrometry
and its application for diagnosing atrazine drift phytotoxicity. J AOAC Int 92:1587–1592
Zhou B, Xiao JF, Tuli L, Ressom HW (2012) LC-MS-based metabolomics. Mol Bio Syst 8:470–
481
Chapter 9
Microbial Metabolomics in Biomass Waste
Management
1 Introduction
Plant biomass has a complex structure that consists of cellulose and hemicellulose
surrounded by lignin (Fig. 9.1a) together with trace amounts of protein and fatty
acids (Eriksson et al. 1990; Hori et al. 1985; Sánchez 2009). Cellulose is the most
abundant and most studied polysaccharide and it forms the basic structure of cell
walls of all plants and algae. Cellulose is predominantly found within the primary
layer of the plant cell wall and its presence decreases in the secondary and tertiary
cell wall layers. It occurs as an aggregated, lateral, microfibril network that forms
highly complex mesh-like structures. To date, cellulose has only been partially
understood using advanced technologies such as nuclear magnetic resonance
(NMR) spectroscopy, X-ray crystallography and electron microscopy (O’Sullivan
1997; Fernandes et al. 2011; Harris et al. 2012).
Cellulose is an almost linear molecule composed of β-D-glucopyranose, linked
by β-1, 4-polyanhydroglucose, with cellobiose as the smallest repetitive unit
(Fig. 9.1b), and is thus a β-glucan (Kumar et al. 2008; O’Neill et al. 2004; Stone
1958). Hemicelluloses form the secondary structural component of the lignocel-
lulose complex (Fig. 9.1c). They are composed of hetero-structures of acelyted and
other derived monosaccharides, mostly pentoses. Various hemicelluloses have been
reported to be directly or indirectly associated with cellulose. They include sugars
such as xylans, xyloglucans, mannans, pectins, homogalacturonans, rhamnogalac-
turonans, arabinans and galactans.
Fig. 9.1 a–d Complex structure of Plant biomass. a Schematic characterization of plant biomass
structure comprising lignocellulose complex. b Fragment of cellulose with the reducing,
non-reducing and cellobiose component of cellulose. c Representative example of hemicellulose
(β-glucans). d Structures of (i) Hydroxycinnamyl alcohols-p-coumaryl alcohol, coniferyl alcohol
and sinapyl alcohol and (ii) derived structural units H, G and S
9 Microbial Metabolomics in Biomass Waste Management 263
More recently, SSF has been developed for a number of industrial bioprocesses
such as biomass conversion (Bak et al. 2009; Brethauer and Studer 2014; Cheng
and Liu 2012). Degradation and bioconversion of the lignocellulose complex from
various biomass sources has increased the production of industrial metabolites,
biofuels and secondary metabolites which can be used, for example, as medicinal
compounds. The use of biomass addresses issues related to managing the consid-
erable amounts of waste material generated during numerous agricultural and allied
processes. SSF employs a mixture of different organisms and/or enzymes in a single
step to generate considerably higher bioconversion of biomass as compared to SmF
methodologies (Brijwani et al. 2010; Kausar et al. 2010; Lee 1997; Sarkar et al.
2012).
Recently, Cheng and Liu (2012) have shown the effects of pre-treatment in
hydrogen production from milled cornstalk. SSF meditated by T. reesei Rut-30 was
applied to this substrate, followed by sludge seeding at 35 and 55 °C. It was
observed that within 4 days of seeding, the 6-day pre-treated substrate generated
about 200 mL H2 when incubated at 55 °C. Additionally, considerable amounts of
low molecular weight fatty acids and ethanol were produced. In a similar fashion,
rice straw pre-treated with Phanerochaete chrysosporium displayed considerable
lignin peroxidase (LiP) and manganese peroxidase (MnP) activities. It was
observed that after 30 days of pre-treatment, these enzymes had degraded about
33 % of the lignin. Follow-up enzymatic degradation resulted in the degradation of
about 17 % of the glucan and 3 % of the xylan (Bak et al. 2009).
Consolidated bioprocessing (CBP) (Fig. 9.2) is a relatively new methodology
for improving biomass conversion to products of commercial interest (Brethauer
Fig. 9.2 A conceptual design of a multi-species biofilm membrane (MBM) reactor for CBP to
generate ethanol from pre-treated wheat straw. Diagram taken from Brethauer and Studer 2014;
published by The Royal Society of Chemistry (Wiethölter et al. 2003) on behalf of the Centre
National de la Recherche Scientifique (CNRS) and the RSC
9 Microbial Metabolomics in Biomass Waste Management 267
and Studer 2014; Lynd et al. 2005). CBP combines the individual processes of
biomass hydrolysis and subsequent fermentation to generate products such as
ethanol in a single step. It eliminates the time-consuming separate biological
pre-treatment process. It also eliminates the need for separate fermentations of
different sugars, such as tetroses or pentoses, which cannot be fermented by general
industrial fermenters such as Saccharomyces cerevisiae (Lynd et al. 2005).
CBP relies on the development of cellulolytic organisms via strategies such as
metabolic engineering or genetic engineering of native species. The first strategy
has been reported to generate about 0.47 g ethanol/g hexose using the thermophilic
bacterium, Geobacillus thermoglucosidasius. Clostridium thermocellum and C.
cellulolyticum, two thermophilic biomass degrading bacteria, have been reported to
generate about 50 g/L ethanol on pre-treated cellulose. The second strategy
involves recombinant DNA technology to mediate heterologous expression of
cellulolytic enzymes in naturally occurring biomass degrading microorganisms.
These strategies have been successful at the laboratory scale; however, success
under industrial conditions has not yet been reported (Olson et al. 2012). In a fungal
(mushroom)-based CBP experiment, Kamei et al. (2014) described the use of spent
sawdust waste from Lentinula edodes cultivation as a substrate for white-rot fungus,
Phlebia spp. MG-60. They observed about 45 % ethanol production from the
mushroom sawdust waste in about 400 h due to increased saccharification.
Symbiotic fermentation may be carried out as part of CBP. The strategy is to
develop a symbiotic consortium of different microorganisms, or their lignocellu-
lolytic enzymes, in either a single batch or continuous batch fermentation. It has
been shown previously that fungi such as Trichoderma spp. and Aspergillus
spp. have the ability to co-culture during cellulose degradation (Brijwani et al.
2010; Gupte and Madamwar 1997). A similar approach has been investigated in
lignocellulose degradation by termites (Brune 2014) and lignin degradation by
co-culture of wood-rot fungi (Qi-he et al. 2011). Mixed microbiota taken from
biogas digested sludge inoculated onto wheat stalk was able to produce 37 mL/g of
H2 after 200 h of incubation. In addition, the concentration of volatile fatty acids,
such as acetate, propionate and butyrate, also increased considerably. Furthermore,
about 75 % of the cellulose was solubilized during this process (Chu et al. 2011).
Additionally, Cheng and Liu (2012) reported that corn stalk substrate pre-treated
with T. reesei RUT C30 by SSF and then seeded with winery sludge was able to
generate H2 gas up to 200 mL within 6 days.
An experiment displaying the synergistic properties of cellulolytic enzymes was
recently reported. Crude, filtered and dialysed fungal enzyme samples obtained
from SSF were mixed to an already operating SmF biodegradation sample. The
resulting symbiosis of Trichoderma and Aspergillus enzymes caused degradation of
cellulose and hemicellulose, as evidenced from a conversion of up to 64 % biomass
to reducing sugars. Under anaerobic conditions, this enzyme mix in combination
with S. cerevisiae was able to generate up to 43 g/L ethanol (about 84 % of the
theoretical maximum) within 7 h (Pirota et al. 2014).
268 A.V. Karpe et al.
Various fungi such as those from the divisions of Ascomycota and Basidiomycota
have been reported as effective biomass degraders. They generate an array of
enzymes including endo- and exo-glucanases, β-glucosidase, xylanases, arabino-
furanosidases and pectinases (Brink and Vries 2011; González-Centeno et al.
2010). The resultant degradation caused by these enzymes generates useful
9 Microbial Metabolomics in Biomass Waste Management 269
in agriculture. While the Dixon et al. report considered issues such as data acqui-
sition, data accuracy and a number of gaps in metabolic pathway information, it is
apparent that the same issues have relevance in biomass related metabolomics. For
example, in biomass metabolomics, the analyst has to deal with differential con-
centrations of larger substrates such as oligo- and polysaccharides, peptones and
smaller, but varied, metabolites such as sugars, amino acids, fatty acids and aro-
matics. One of the reported studies considering an untargeted metabolomic
approach in biomass degradation was the rumen bacterial mediated degradation of
birch wood xylan and apple pectin (Villas-Bôas et al. 2006). The bacterial species
Clostridium proteoclasticum, Butyrivibrio fibrisolvens and Streptococcus bovis
were used. The biomass degradation quantified by GC-MS indicated the presence
of about 40 different sugars, sugar acids, fatty acids and acids of lignin and tannin
degradation including gallic acid, coumaric acid and quinic acid (among others). In
our research, winery biomass was degraded using ascomycete fungi such as T.
harzianum, A. niger, P. chrysogenum, P. citrinum and S. cerevisiae (as a negative
control). An untargeted approach using GC-MS based metabolomic analysis was
used which yielded 233 metabolite peaks. These consisted of biomass degraded
mono- and oligosaccharides, sugar alcohols, sugar acids, fatty acids, amino acids,
organic acids and aromatic acids, although the major metabolites present in the
biomass degrading fungal profile were pentoses, hexoses and to a lesser degree fatty
acids and amino acids. The results were in agreement with considerable enzyme
activities of cellulases, xylanases and β-glucosidase in the biomass degrading fungi
(Karpe et al. 2015c). In further research, the winery biomass was degraded by a
symbiotic mixture of all the above mentioned fungi, except S. cerevisiae; and the
degradation level increased considerably, indicated primarily by the higher enzyme
activities, especially xylanase. The enhanced enzyme activity affected overall
metabolic output where the higher metabolite concentration consisted of fatty acids
such as oleanolic acid, glyceric acid, heptadecanoic acid and tetradecanoic acid.
Additionally, the mixed fungal degradation was also able to yield some industrially
useful secondary metabolites such as gallic acid, inositol, β-sitosterol and mannitol
in higher concentrations (Karpe et al. 2015a).
By comparison to the above untargeted approach, targeted metabolomics deals
with quantitative analysis of a limited number of the metabolites which represent
the complete metabolome output of an organism. On a superficial level, targeted
metabolomics resembles metabolic fingerprinting, however they are significantly
different. While, the fingerprinting approach classifies different metabolic (func-
tional) groups and identifies signature metabolic patterns to each organism (quali-
tatively or quantitatively), the targeted approach selectively screens any
non-targeted metabolites and quantifies only targeted metabolites. The presence
of targeted metabolites or metabolic groups in an organism’s metabolome is
therefore a prerequisite for targeted metabolomics (Shulaev 2006). The quantitative
approach combined with selectivity increases accuracy and sensitivity of the
analysis (for those specific metabolites). The main issues associated with wider use
of a targeted approach are the current incomplete knowledge of organism specific
metabolic pathways, the identification of intermediate metabolites, and their
9 Microbial Metabolomics in Biomass Waste Management 271
structural elucidation and availability of metabolites in pure form that can be used
as a reference standard and, in general, an increased level of complexity. A targeted
approach, therefore, has been used less compared to both non-targeted and fin-
gerprinting metabolomic approaches. It has been argued that targeted approaches
are largely limited to either genetic behavioural studies or clinical applications, as
indicated in a recent review by Putri et al. (2013).
In spite of these limitations, targeted metabolomics methods are becoming more
prominent in studies that encompass multiple ‘omics’ approaches. One recent study
on targeted metabolomics involved the quantification of sugars, sugar phosphates,
organic and amino acids within the S. cerevisiae metabolome (Kato et al. 2012). On
a GC-MS system, around 99 metabolites were targeted using mass-to-charge (m/z)
channel based screening of a mass spectrum range of 89–299 m/z units. Based on
the data collected, the samples were then quantitatively re-analysed on an
LC-QQQ-MS. The combined approach was able to detect metabolites concentra-
tions of as low as to 1.4 femtomole in the yeast metabolome to a very high accuracy
(R2 = 99.94 %) (Kato et al. 2012). Another reported work involved the quantifi-
cation of the Clostridium thermocellum metabolic profile. The thermophilic bac-
terium was grown on cellobiose and its metabolic output was analysed by
LC-QQQ-MS/MS (using 13C-isotopes). About 45 metabolites including amino
acids and organic acids were quantified, some present in concentrations as low as
0.8 ng/mL of detection limit and 2.7 ng/mL of quantification level (Cui et al. 2013).
The targeted approach has also been applied in the assessment of metabolites
that may be responsible for development of fungal tolerance in horticulture crops.
A good example is the polyphenol assessment of wine-grape varieties displaying
tolerance towards powdery and downy mildew; the fungal infections are caused by
Uncinula necator and Plasmopara viticola, respectively. The analysis of secondary
metabolites belonging to phenolic acids, dihydrochalcones, stilbenes, flavanols and
flavanols was performed by the combination of two different UPLC-QQQ-MS
systems. About 55 metabolites including gallic and coumaric acids, resveratrol,
procyanidine, cathechins and naringenin were quantified with results ranging from
30 ng/mL to 203 μg/mL (Ehrhardt et al. 2014).
An interesting application of combined (targeted and non-targeted) metabolomic
approaches was recently demonstrated by Godelmann et al. (2013). This group
performed untargeted and targeted metabolic profiling using GC-TOF-MS and
1
H-NMR, respectively, to differentiate the grape variety, its vintage and geo-
graphical distribution. Specifically, 16 metabolites were quantified which included
organic acids such as lactate, citrate, malate, succinate, acetate, fumarate and tar-
trate; alcohols such as methanol, ethanol, 3-methylbutanedioland glycerol; and
secondary metabolites such as shikimate, caftarate and acetone. The concentrations
ranged from 79.3 mg/L for shikimate to 120.2 g/L for ethanol. Chemometric sta-
tistical analysis was able to classify the geographic regions and vintage year to
about 90 and 97 % accuracy, respectively. The experiment provides not only
quality parameters for wine grapes, but also indicates the potential of applying
specific fungal/bacterial species to extract targeted metabolites on a commercial
scale from those substrates.
272 A.V. Karpe et al.
Fig. 9.3 The general structure of chief antibacterial metabolites secreted by P. communes fungus.
The structures represent a Lovastatin and b 3-isobutyl hexahydropyrrolo[1, 2-a]pyrazine-1,
4-dione (α-Ketoisovaleryl-Leu-Pro lactam)
Although the FTIR approach was unable to evaluate individual metabolites, it could
separate the overall metabolite spectra into several groups providing a defined
analysis pathway. The authors suggested using a more specific metabolic flux
analysis to obtain a specific fingerprint related to fermentation of rye grass biomass
by rumen microbiota. The study also indicated the likelihood of increasing protein
utilisation by increasing the availability of soluble carbohydrates to rumen micro-
biota. The authors also indicated the use of FTIR as a preliminary
non-discriminatory technique to establish a rapid understanding of metabolite
interactions, which can then be studied in detail by a secondary flux analysis
(Kingston-Smith et al. 2013). The study, albeit a preliminary investigation, indi-
cated that the metabolic fingerprint and flux pattern in the fermentation of plant
biomass by rumen microbiota were specific to the type of plant source.
Another widely used method for metabolite fingerprinting is liquid chro-
matography with electrospray ionisation-mass spectrometry (LC-MS). Due to
protonation/de-protonation during ionic adduct formation, the analyte is observed
as a molecular ion. The mass-to-charge ratio (m/z) calculation by ESI is then used
to predict the putative metabolite in numerous types of samples (Draper et al. 2013).
Secretome studies of Trichoderma hamatum GD12 in relation with Arabidopsis in
the presence of other fungi showed the presence of cellulose binding-motif (CBM).
Further analysis by combined 1H-NMR and direct infusion-ESI-MS (DI-ESI-MS)
showed the presence of 30 metabolites of at least fivefold change, based on
unbiased positive and negative ionisation mode discrimination (Studholme et al.
2013). Similarly, in a recent study of Aspergillus nidulans mediated degradation of
Quercus suber cork, the secretome analysis showed ca. 28 active protein species, of
which ca. 12 % comprised plant cell wall degradation proteins. These were further
classified as β-glucosidases, arabinosidase, alcohol oxidase and oxidoreductases.
The metabolic output of this secretome was analysed by ultra-high-performance
liquid chromatography coupled to electrospray ionisation-high resolution mass
spectrometry (UHPLC-ESI-HRMS). The organic extracts that resulted from A.
nidulans mediated degradation displayed the presence of signature metabolites such
as low molecular weight aromatics (e.g. ferrulates, veratraldehyde and cinnamate),
about five β-aryl ether linked metabolites and a pinoresinol group compound
(Martins et al. 2014). Secretome analysis of Fusarium spp. Q7-31T by ESI coupled
with tandem mass spectrometry (LC-MS/MS) indicated the presence of ca.
10 glycoside hydrolase enzymes out of a total of 28, most of them responsible for
producing smaller glucan metabolites by hydrolysing biomass substrates (Tian et al.
2015). A further downstream metabolic effect on the sugar release due to Fusarium
spp. based biomass degradation was analysed by FTIR metabolite fingerprinting.
The results showed 13 % decrease in glucose and about 11 % increase in uronic
acids for Fusarium circinatuum mediated rotting of Pinus pinaster biomass.
Additionally, the content of methyl esterified polysaccharide in pectins also
increased following post-Fusarium culturing (Vivas et al. 2014). It has been
observed that fingerprinting of biomass degradation has been mostly focused
towards proteome/secretome analysis, as indicated from some recent research
(Kosmides et al. 2013; Martins et al. 2014; Studholme et al. 2013; Tian et al. 2015;
9 Microbial Metabolomics in Biomass Waste Management 275
Vivas et al. 2014; Cattaneo et al. 2014). Additional elucidation of the metabolic
output of these secretomes still needs to be applied in the context of biomass
conversion, as currently is applied in plant–fungal parasitic or symbiotic and
microbiome relations (Draper et al. 2011; Kingston-Smith et al. 2013).
Some of the earlier metabolism flux related research was performed on the
galactoglycome of T. reesei during its growth on lactose. The work showed that
lactose, when used as a carbon source, acted an initial inducer for galactokinase and
aldose reductase XYL1. These enzymes further form the oligosaccharides which
serve as inducers for cellulase formation (Hartl et al. 2007; Seiboth et al. 2007).
A more downstream quantitative metabolic flux analysis of this fungus used
sophorose as an initial inducer for cellulase formation and activity. The induction
did not affect any balance between intracellular and extracellular proteins, the latter
of which consist of cellulases and hemicellulases, a critical factor for industrial
application of this fungus (Rautio et al. 2006). A similar, but a more amino acid
focussed experiment, was conducted to optimize the growth medium composition
for the basidiomycete Pleurotus sapidus by using 13C-glucose based metabolic flux.
It was observed that the isotopic glucose contributed to fungal amino acid synthesis,
ranging from 22 (asparagine/aspartate) to 92 % (alanine). In addition, the threonine
aldolase enzyme based mechanism to generate glycine from threonine was
observed to be inactive or absent in this fungus. Additionally, glucose was utilised
at a rate of 0.24 mmol g/hour, with about 35 % of its output going to the pentose
phosphate pathway (Fraatz et al. 2014).
Similar approaches have been reviewed by Kubicek (2013) and asserts the
necessity of multi ‘Omics’ approach for understanding cellulolytic properties of T.
reesei for industrial applications. It also indicated a limited number of approaches
used in metabolome studies of T. reesei and other fungal enzyme outputs during
biomass degradation as compared to genomics, transcriptomics and proteomics.
engineering can provide novel in silico design and fabrication materials, the con-
struction of new biological parts, devices, systems and machines, as well as the
re-design of existing, natural biological systems for useful purposes (Wagner and
Alper 2016; Maynard 2015).
The application of metabolomics to biological engineering will become
increasingly important for the production of chemicals of value from agricultural
wastes, which are either high-value fine chemicals (e.g. medicinal products) or bulk
low-value commodities (e.g. fuels) (Ellis and Goodacre 2012). The integration of
metabolomics and fluxomics at each stage of biological engineering projects will
enable metabolic pathways with a desired outcome to become the focus of bio-
engineering design, where the genetic/metabolic optimisation of those pathways
will result in more efficient conversion of low-cost starting materials into highly
desirable products (Ellis and Goodacre 2012; Chen 2015). For example, this
emerging field that has already been identified as a sustainable solution for pro-
ducing an alternative source of Omega-3 fatty acids that is not reliant on wild fish
harvests or aquaculture; work has already been performed to create recombinant
sources of Omega-3 fatty acids in a canola and yeast (Adarme-Vega et al. 2012).
Bioprocessing of polyols using lactic acid bacteria has been investigated as an
alternative to current industrial food production approaches (Ortiz et al. 2013). The
major challenge for all bioengineering studies is to balance the flux in the pathway
of interest in order to obtain high yields and maintain productivity of the target
microorganism (Yuan et al. 2013).
Fig. 9.4 Design strategy of rational and evolutionary direct and inverse metabolic engineering
experiments. Adapted from Hasunuma and Kondo (2012)
9 Microbial Metabolomics in Biomass Waste Management 279
Papini et al. (2012) described a rational metabolic engineering strategy that was
used to express the fungal genes of the phosphoketolase pathway in S. cerevisiae
and the effects of the expression of this recombinant route in yeast using intra-
cellular flux analysis based on 13C labelling. Several bacterial species and fila-
mentous fungi utilise the phosphoketolase pathway for glucose dissimilation as an
alternative to the Embden-Meyerhof-Parnas pathway. However, through the use of
fluxomics, Papini et al. (2012) observed yeast using the recombinant pathway and it
was observed that the utilisation of the phosphoketolase pathway did not interfere
with glucose assimilation through the Embden-Meyerhof-Parnas pathway.
Furthermore, the expression of the recombinant pathway contributed to an increase
in the acetyl coenzyme supply, therefore enabling future potential metabolic
engineering strategies to be considered that utilise acetyl CoA as precursor for the
biosynthesis of industrially relevant and value-added compounds (Papini et al.
2012). Huang et al. (2015) applied biological evolutionary engineering to activate
the glycerol utilisation pathway for fumaric acid production. In this study, an
evolved strain G80 was selected that was tolerant to high concentrations of crude
glycerol to produce fumaric acid.
While A. niger is an effective organism in the biodegradation of biomass and has
been widely applied for the production of high value-added products (Karpe et al.
2015b; Krull et al. 2010; Meijer et al. 2009), the morphology of this fungus, the
transport phenomena and the related metabolic activity result in unreliable yield
within industrial bioprocessing facilities (Krull et al. 2010). As such, further
characterization of the molecular and cell biology of A. niger, in particular char-
acterising interactions between the growth conditions, cell morphology,
spore-hyphae-interactions and value-added product formation, is of great impor-
tance for ensuring a sustainable bioprocessing industry (Krull et al. 2010).
Conversely, Rumbold et al. (2009) used a substrate-oriented approach as opposed
to a product-oriented approach towards the selection of a microbial production host
that avoids the need for extensive metabolic engineering. In this work, six
microorganisms were evaluated and the performance of two bacteria (Escherichia
coli and Corynebacterium glutamicum), two yeasts (S. cerevisiae and Pichia
stipitis) and two fungi (A. niger and T. reesei) were assessed for their ability to
degrade lignocellulosic hydrolysates. The study was particularly focused on their
ability to utilise monosaccharides, characterising the resistance against lignocellu-
losic hydrolysates inhibitors and assess growth and utilisation rates on different
feedstock (with feedstocks thermally pre-treated under mild acidic conditions fol-
lowed by enzymatic hydrolysis and non-enzymatic treatments). Rumbold et al.
(2009) concluded that carbon source versatility and inhibitor resistance were the
major discriminators of the microorganisms tested, with P. stipitis and A. niger
found to the moist effective. C. glutamicum and S. cerevisiae were shown to be the
least adapted to renewable feedstock. These results highlight the complexity of
substrate-orientated approaches that remove the over engineering of biological
systems and utilising natural metabolic pathways present in substrate matched
microorganisms.
280 A.V. Karpe et al.
References
O’Neill MA, Ishii T, Albersheim P, Darvill AG (2004) Rhamnogalacturanan II: structure and
function of a borate cross-linked cell wall pectic polysaccharide. Annu Rev Plant Biol 55:109–
139
O’Sullivan A (1997) Cellulose: the structure slowly unravels. Cellulose 4:173–207
Ogura T, Date Y, Kikuchi J (2013) Differences in cellulosic supramolecular structure of
compositionally similar rice straw affect biomass metabolism by paddy soil microbiota.
PLoS ONE 8:e66919
Olson DG, McBride JE, Joe Shaw A, Lynd LR (2012) Recent progress in consolidated
bioprocessing. Curr Opin Biotechnol 23:396–405
Ortiz ME, Bleckwedel J, Raya RR, Mozzi F (2013) Biotechnological and in situ food production
of polyols by lactic acid bacteria. Appl Microbiol Biotechnol 97:4713–4726
Papini M, Nookaew I, Siewers V, Nielsen J (2012) Physiological characterization of recombinant
Saccharomyces cerevisiae expressing the Aspergillus nidulans phosphoketolase pathway:
validation of activity through 13C-based metabolic flux analysis. Appl Microbiol Biotechnol
95:1001–1010
Pedersen H, Carlsen M, Nielsen J (1999) Identification of enzymes and quantification of metabolic
fluxes in the wild type and in a recombinant Aspergillus oryzae strain. Appl Environ Microb
65:11–19
Pirota RPB, Delabona P, Farinas C (2014) Simplification of the biomass to ethanol conversion
process by using the whole medium of filamentous fungi cultivated under solid-state
fermentation. BioEnerg Res 7:744–752
Pribowo A, Arantes V, Saddler JN (2012) The adsorption and enzyme activity profiles of specific
Trichoderma reesei cellulase/xylanase components when hydrolyzing steam pretreated corn
stover. Enzyme Microb Technol 50:195–203
Putri SP, Nakayama Y, Matsuda F, Uchikata T, Kobayashi S, Matsubara A, Fukusaki E (2013)
Current metabolomics: practical applications. J Biosci Bioeng 115:579–589
Qi-He C, Krügener S, Hirth T, Rupp S, Zibek S (2011) Co-cultured production of
lignin-modifying enzymes with white-rot fungi. Appl Biochem Biotechnol 165:700–718
Ralph J, Lundquist K, Brunow G, Lu F, Kim H, Schatz PF, Marita JM, Hatfield RD, Ralph SA,
Christensen JH, Boerjan W (2004) Lignins: natural polymers from oxidative coupling of
4-hydroxyphenyl- propanoids. Phytochem Rev 3:29–60
Rani KS, Seenayya G (1999) High ethanol tolerance of new isolates of Clostridium thermocellum
strains SS21 and SS22. World J Microbiol Biotechnol 15:173–178
Rautio J, Smit B, Wiebe M, Penttila M, Saloheimo M (2006) Transcriptional monitoring of steady
state and effects of anaerobic phases in chemostat cultures of the filamentous fungus
Trichoderma reesei. BMC Genom 7:247
Rehman MSU, Kim I, Chisti Y, Han J-I (2013) Use of ultrasound in the production of bioethanol
from lignocellulosic biomass. EEST Part A: Energy Sci Res 30:1391–1410
Rodríguez A, Carnicero A, Perestelo F, De La Fuente G, Milstein O, Falcón MA (1994) Effect of
Penicillium chrysogenum on lignin transformation. Appl Environ Microbiol 60:2971–2976
Roessner U, Bowne J (2009) What is metabolomics all about? Biotechniques 46:363
Rumbold K, van Buijsen HJJ, Overkamp KM, van Groenestijn JW, Punt PJ, van der Werf MJ
(2009) Microbial production host selection for converting second-generation feedstocks into
bioproducts. Microb Cell Fact 8:64
Sánchez C (2009) Lignocellulosic residues: biodegradation and bioconversion by fungi.
Biotechnol Adv 27:185–194
Sarkar N, Ghosh SK, Bannerjee S, Aikat K (2012) Bioethanol production from agricultural wastes:
an overview. Renew Energy 37:19–27
Saunders EC, Souza DP, Chambers JM, Ng M, Pyke J, McConville MJ (2015) Use of 13C stable
isotope labelling for pathway and metabolic flux analysis in Leishmania parasites. In:
Peacock C (ed) Parasite genomics protocols. Springer, New York
Seiboth B, Gamauf C, Pail M, Hartl L, Kubicek CP (2007) The D-xylose reductase of Hypocrea
jecorina is the major aldose reductase in pentose and D-galactose catabolism and necessary for
β-galactosidase and cellulase induction by lactose. Mol Microbiol 66:890–900
9 Microbial Metabolomics in Biomass Waste Management 287
Zha Y, Westerhuis JA, Muilwijk B, Overkamp KM, Nijmeijer BM, Coulier L, Smilde AK, Punt PJ
(2014) Identifying inhibitory compounds in lignocellulosic biomass hydrolysates using an
exometabolomics approach. BMC Biotechnol 14:22
Zuroff T, Curtis W (2012) Developing symbiotic consortia for lignocellulosic biofuel production.
Appl Microbiol Biotechnol 93:1–13
Chapter 10
Beyond Metabolomics: A Review
of Multi-Omics-Based Approaches
1 Introduction
‘Omics’-based techniques comprise a suite of tools and approaches, each with their
own specific protocols and frameworks for setting minimum data and reporting
standards (Field et al. 2008; Morrison et al. 2006; Sansone et al. 2007; Sumner et al.
2007; Orchard and Kerrien 2010). This suite of techniques primarily comprises
metagenomics, transcriptomics, proteomics and metabolomics. There are also a
number of other specialized ‘omic’-based approaches that are included under the
broader ‘omics’ banner, such as lipidomics, fluxomics (metabolic flux analysis),
toxicogenomics, nutrigenomics and foodomics. However, these additional ‘omics’
approaches are not considered within the context of this chapter, as they are subcat-
egories of the aforementioned suite of techniques. In addition, for completeness, it
should be noted that within the context of this chapter, (meta)genomics has been
defined as the analysis of genetic material recovered from an organism or environ-
mental samples (Handelsman 2004); (meta)transcriptomics is the analysis of RNA
molecules, including messenger RNA (mRNA), Ribosomal RNA (rRNA), transfer
RNA (tRNA) and other non-coding RNA produced by an organism or a population of
organisms (Pascault et al. 2015); (meta)proteomics is the analysis of proteins produced
by an organism or population of organisms, and their function (Douterelo et al. 2014);
and lastly, metabolomics is the analysis of the small chemical compounds produced
and consumed by an organism or a population of organisms (Beale et al. 2016a).
Microbial Potential
1 2 3 4
Fig. 1 Chronical order of ‘omics’ based techniques commonly used in multi-omics studies.
Adapted from Abram (2015)
10 Beyond Metabolomics: A Review of Multi-Omics-Based Approaches 291
(Barh et al. 2013; Blanchet and Smolinska 2016; Fondi and Liò 2015; Kohl et al.
2014; Schneider and Orchard 2011; Zhang et al. 2010). Instead, the aim of this
chapter is to provide an overview of the different approaches used in multi-omics-
based research and the tools used to integrate multi-omics data, using examples from
the literature from a range of environmental, industrial and biomedical applications
to highlight the value of extending beyond metabolomics only research.
Multi-omics based techniques are inherently data-rich studies. For example, the
human genome comprises ca. 20,000–25,000 protein coding genes (Pertea and
Salzberg 2010) and the human metabolome is estimated to comprise over 40,000
metabolites (Forsythe and Wishart 2009). As such, the challenge in all ‘omic-based
investigations’, which is only compounded further when applying a multi-omics
approach, is the handling of these large and complex datasets (Kohl et al. 2014;
Röling et al. 2010). Röling et al. (2010) characterized the data processing and
quantitative comprehension of multi-omic information as a bottleneck in the overall
workflow, requiring input and interpretation of ‘systems biologists’ and microbi-
ologists. In addition, the authors of this review propose that it needs further input
from the bioanalytical chemist/biochemist/biostatistician to first evaluate the quality
and validity of the study (experimental) design, as well as the quality of the data
acquired from the instrument, before even attempting to integrate and synthesize
findings (indeed the old adage of poor data in equals poor data out applies). In any
case, assuming the data obtained are of high quality and are valid [following each
‘omics’ specific protocol and frameworks for minimum data and reporting stan-
dards as highlighted by the various societies (Field et al. 2008; Sansone et al. 2007;
Sumner et al. 2007)], there are a number of approaches to analyzing and inter-
preting multi-omics data, namely: post-data analysis integration and integrated data
analysis techniques.
In a post-data analysis approach, datasets are analyzed in isolation of each other
and key features are networked in a post analysis exercise through the synthesis of
significant features at joint nodes in the overall model metabolic pathway. This
approach has been used in previous studies that focused on characterizing and
assessing biological wastewater treatment systems (Beale et al. 2016b), the
microbial resistance of marine sediments after an oil spill (Kimes et al. 2013) and
characterizing permafrost (Hultman et al. 2015). In contrast, an integrated
multi-omics approach employs specialized tools to merge datasets prior to under-
taking any data analysis and interpretation (Kuo et al. 2013), thus enabling simi-
larities of each omic approach to be statistically derived, as opposed to relying on
human interpretation. The principal differences between the post-data analysis and
integrated analysis approaches are graphically presented in Fig. 2.
In addition to post-data analysis integration and integrated data analysis tech-
niques, a third model-based integration approach has been identified. However, a
292 D.J. Beale et al.
Fig. 2 Principal differences between the post-data analysis and integrated analysis approaches
3 Application of Multi-Omics
There are many examples in the literature of multi-omic studies, with various levels
of integration. Some studies comprise simple levels of integrations (i.e. combing
two different -omics datasets) through to more comprehensive and computationally
demanding studies (i.e. integration of multiple omics datasets). Typically, a
two-omics integration study combines either metagenomics or transcriptomics data
with proteomics (Sunagar et al. 2016; Wildburger et al. 2015) or metabolomics
datasets (Tokimatsu et al. 2005b; Garcia-Alcalde et al. 2011; Kamburov et al. 2011)
or combines proteomics and metabolomics datasets (Xu et al. 2015). These studies
demonstrate how predicted functional metabolism (metagenomics) or gene
expression (transcriptomics) relate to actual protein and metabolite expression and,
by extension, provide a means to self-validate findings through cross-referencing
experimental findings (Fondi and Liò 2015). The following section provides some
examples of multi-omics studies applied to various fields of research, all of which
provided an extension beyond a metabolomics only study (or other singular
omics-based approaches) and enabled further depth of analysis that would other-
wise not be achieved.
Cytoscape with MODAM and – Transcriptomics Unspecified – Multi-Omic Data Miner and OmicsAnalyzer Open Enjalbert et al. (2011),
Cytoscape with OmicsAnalyzer – Proteomics were designed as an accessible and handy Xia et al. (2010)
– Metabolomics Cytoscape plugin that facilitates omics
– Fluxomics analysis
– Compile all biologically-relevant information
regarding the model system through web link
association
– Map the network components with
multi-omics data
– Model omics data
295
(continued)
Table 1 (continued)
296
of crucial microbial populations within water sources, pipes and treatment process.
In addition, it is anticipated that through multi-omics approaches, new insights into
microbial populations in terms of diversity, resilience and activity when exposed to
shocks and stresses (such as illegal discharges within sewer networks and opera-
tional shock during treatment) will be obtained. Furthermore, multi-omics may
provide insight into driving higher biogas and methane production during treatment
and collection systems for reuse.
The safety and quality of potable water is often assumed and taken for granted by
consumers in most developed countries. However, our understanding of potable
water biofilms is limited, partly as they are difficult to access and traditional
microbiology approaches fail to provide sufficient information on their composition
and activity (Douterelo et al. 2014). To date, numerous researchers have applied
omics-based techniques to characterize and assess aquatic biofilms, in a range of
environments. For example, Shaw et al. (2014), Chao et al. (2013) assessed the
community structure of potable water biofilms using metagenomics when exposed
to different water treatment strategies. Metagenomics was also used to investigate
intertidal marine biofilm communities (Zhang et al. 2013) and black fungal biofilms
growth in domestic water fixtures (Heinrichs et al. 2013). Metabolomics has also
been used to investigate biofilms found on the surface of copper pipes (Beale et al.
2010; Beale et al. 2012) and within potable water distribution networks (Beale et al.
2013). However, the application of multi-omics approaches has been limited. Leary
et al. (2014) used a metagenomic and metaproteomic approach to analyze the
composition and function of marine biofilms; others have investigated microbial
communities in extreme environments using metagenomics and proteomics (Singer
2012; Schneider and Riedel 2010). Nevertheless, it has been identified that there is a
need for a multi-omics approaches to assess biofilms. This has resulted in the
recently proposed term “Biofomics” and web-based interface for biofilm data
(Lourenço et al. 2012). Biofomics provides a framework, database depository and
selection of statistical tools for researchers to follow, merge and examine data from
different approaches such as metagenomics, transcriptomics, proteomics and key
metabolites (metabolomics) (Nozhevnikova et al. 2015). The biofilm data results
from experiments involving several types of bacterial genera such as Salmonella
spp., Escherichia coli and Candida spp., and is supported by the minimum infor-
mation about a biofilm experiment (MIABiE) initiative (Lourenço et al. 2012). To
date, the majority of biofomics work has been related to the biomedical and clinical
fields.
An issue of particular concern to infrastructure asset managers is biofilms that
cause biocorrosion/biodeterioration (also known as MIC). Due to their potential
financial and economic impact to infrastructure, there has been a considerable
amount of research published on the role of microorganisms in promoting
302 D.J. Beale et al.
corrosion. The majority of this work has been undertaken to address the problem of
MIC in offshore oil and gas pipelines, and concrete structures with some prelimi-
nary research on microbial/metal surface interactions in water pipes. As such, a
number of extensive reviews have been compiled on MIC mechanisms over the
past 20 years (Videla and Herrera 2009; Edyvean and Videla 1991; Videla 2003;
Beech and Gaylarde 1999; Flemming 1994; Beech et al. 2014).
Corrosion is the result of a series of chemical, physical and (micro) biological
processes leading to the deterioration of materials. The mechanisms of MIC and
MIC inhibition are not completely understood, because they cannot be linked to a
single biochemical reaction or specific microbial species or group (Kip and van
Veen 2015). As such, MIC biofilm communities can be studied at both their
compositional and functional levels through the use of multi-omics. A number of
traditional techniques, such as clone libraries and genetic fingerprinting, along with
more recent metagenomics and transcriptomics, are being used to characterize and
understand MIC biofilms (Chakraborty et al. 2014).
Relatively few metabolomic-MIC studies have been reported. The role of cor-
rosion products on MIC of carbon steel has been investigated by gas
chromatography-mass spectrometry (GC-MS) (Liu et al. 2000), where S2− and
organic acids were found to destroy the protective layer and promote hydrogen
permeation. Furthermore, GC-MS-based metabolomics has also been explored as a
potential tool in monitoring MIC in copper pipes in water distribution systems
(Beale et al. 2010, 2012). It was found that the biofilm metabolites of bacteria
causing copper pipe MIC comprised a combination of organic acids, amino acids
and lipids. These are common in biological metabolic processes, specifically those
relating to soluble monomers and sulphite reducing substrates. In addition, the
range of carboxylic acids released from the isolates (Bosea, Methylobacterium,
Paenibacillus, Sphingomonas and Variovorax) suggests that the corrosion potential
of these biofilms varies, which would account for localized pitting corrosion
commonly observed in metallic pipes (Beale et al. 2014a). Kouremenos et al.
(2014) investigated the metabolic profile of Pseudomonas putida in potable water
exposed to high and low doses of soluble and insoluble iron using LC-TOFMS and
identified metabolites that included nucleobases, nucleosides, dipeptides, amino
acids, fatty acids, sugars and phospholipids as a response to exposure. While not
directly related to MIC, the approach by Kouremenos et al. (2014) and the pre-
liminary work of Beale et al. (2010, 2012) demonstrate the feasibility of GC and
LC-based metabolomics techniques to assess microbial populations exposed to
metals or isolated from biofilms known to cause MIC in potable water networks. In
a study by Booth et al. (2011), the difference in response to metal stress between
sessile and planktonic bacterial populations was characterized. The planktonic
population had an oxidative stress response to copper ion exposure, whereas the
same species within a biofilm environment responded by shifts in
exo-polysaccharide-related metabolism. This finding suggests that microbial
responses pertinent to corrosion are different between sessile and planktonic pop-
ulations, and through more research using metabolomic-based techniques, linkages
between the metabolite activity of both sessile and planktonic populations and their
10 Beyond Metabolomics: A Review of Multi-Omics-Based Approaches 303
Metabolomics studies have been used to investigate the gut microbial population
structure associated with a wide range of human diseases (Alam et al. 2014; Garrett
2015; Gevers et al. 2014; Goldman et al. 2014; Ley et al. 2006; Smith et al. 2013).
Expression profiling studies have contributed significantly to the understanding of
underlying molecular mechanisms of several diseases. In the context of cancer
research, this has resulted in the improved classification of tumour subtypes
(Karnovsky et al. 2012). However, the lack of early diagnostic markers still remains
a problem (Diamandis 2010). Proteomics and metabolomics have the potential to
provide additional biological insight for solving this problem (Enjalbert et al. 2011).
The multi-omics approach has been applied to identify markers related to
retinoblastoma which is caused by the RB1 gene inactivation. The miRNA pathway
analysis, coupled with miRNA microarray indicated a presence of 18 novel
miRNAs responsible for the onset of this type of retinal cancer (Guha et al. 2015).
In addition to cancer studies, multi-omics approach has been applied to other
studies such as asthma-COPD overlap syndrome (Trentacoste et al.), autism dis-
orders, sickle cell anemia and kidney disorders among many (Megan et al. 2016;
Zeidan-Chulia et al. 2014; Goodman et al. 2016; Cisek et al. 2015). A very recent
research conducting ACOS pattern among asthma patients recorded a trend of
increased Immunoglobulin E (IgE) antibody. A differential gene expression of
Toll-like receptor 10 (TLR10), an asthma related gene was observed. Also, a further
study of single nucleotide polymorphisms (SNPs) indicated a role of another gene
IL21R in ACOS. Based on pathway analysis, the pattern was also observed to affect
the activities related to cytochrome P450 system (Megan et al. 2016).
A recent review by Higdon et al. (2015) indicated numerous domains (genes,
RNA and proteins) related to autism spectrum disorders (ASD) such as fragile-X
304 D.J. Beale et al.
mental retardation protein (FMRP) and chromatin modifying genes and postsy-
naptic and embryonic development proteins. Expression analysis models such as
linear models for microarray analysis (LIMMA) and significance analysis of
microarrays (SAM) have been used to identify differential expressions based on
several databases and networks such as KEGG, HMDB, BioCyc, Panther among
many others. The former modelling approach (LIMMA) was used to identify
Alzheimer’s related genes and signal transduction pathways such as NOTCH and
Wnt in mitochondrial expressional system of ASD patients (Zeidan-Chulia et al.
2014).
Overall, it has been observed that multi-omics research has been at fairly
advanced levels in cancer research, and has made inroads into other clinical
researches. It is expected that a rapid growth in those studies will be observed in
near future, with a further expansion in other related fields, thereby leading to
‘personalized medicine’ development.
3.6 Biofuels
4 Conclusion
Reference
Beale DJ, Dunn MS, Marney D (2010) Application of GC–MS metabolic profiling to ‘blue-green
water’ from microbial influenced corrosion in copper pipes. Corros Sci 52:3140–3145
Beale DJ, Dunn MS, Morrison PD, Porter NA, Marlow DR (2012) Characterisation of bulk water
samples from copper pipes undergoing microbially influenced corrosion by diagnostic
metabolomic profiling. Corros Sci 55:272–279
Beale DJ, Karpe AV, Mcleod JD, Gondalia SV, Muster TH, Othman MZ, Palombo EA, Joshi D
(2016b) An ‘omics’ approach towards the characterisation of laboratory scale anaerobic
digesters treating municipal sewage sludge. Water Res 88:346–357
Beale DJ, Morrison PD, Key C, Palombo EA (2014a) Metabolic profiling of biofilm bacteria
known to cause microbial influenced corrosion. Water Sci Technol 69:1–8
Beale DJ, Morrison PD, Palombo EA (2014b) Detection of Listeria in milk using non-targeted
metabolic profiling of Listeria monocytogenes: a proof-of-concept application. Food Control
42:343–346
Beech IB, Gaylarde CC (1999) Recent advances in the study of biocorrosion—an overview. Rev
De Microbiol 30:177–190
Beech IB, Sztyler M, Gaylarde CC, Smith WL, Sunner J (2014) 2—Biofilms and biocorrosion. In:
Liengen T, Féron D, Basséguy R, Beech IB (eds) Understanding biocorrosion. Woodhead
Publishing, Cambridge
Beer LL, Boyd ES, Peters JW, Posewitz MC (2009) Engineering algae for biohydrogen and
biofuel production. Curr Opin Biotechnol 20:264–271
Blanchet L, Smolinska A (2016) Data fusion in metabolomics and proteomics for biomarker
Discovery. In: Jung K (ed) Statistical analysis in proteomics. Springer, New York
Booth SC, Workentine ML, WEN J, Shaykhutdinov R, Vogel HJ, Ceri H, Turner RJ, Weljie AM
(2011) Differences in metabolism between the biofilm and planktonic response to metal stress.
J Proteome Res 10:3190–3199
Chakraborty A, Dasgupta CK, Bhadury P (2014) 4—application of molecular techniques for the
assessment of microbial communities in contaminated sites. In: Das S (ed) Microbial
Biodegradation and bioremediation. Elsevier, Amsterdam
Chao Y, Ma L, Yang Y, JU F, Zhang XX, Wu WM, Zhang T (2013) Metagenomic analysis
reveals significant changes of microbial compositions and protective functions during drinking
water treatment. Scientific Reports, vol 3
Chariton AA, Ho KT, Proestou D, Bik H, simpson SL, Portis LM, Cantwell MG, Baguley JG,
Burgess RM, Pelletier MM, Perron M, Gunsch C, Matthews RA (2014) A molecular-based
approach for examining responses of eukaryotes in microcosms to contaminant-spiked
estuarine sediments. Environ Toxicol Chem 33:359–369
Cisek K, Krochmal M, Klein J, Mischak H (2015) The application of multi-omics and systems
biology to identify therapeutic targets in chronic kidney disease. Nephrol Dial Transplant
gfv364
Cohen A, Bont L, Engelhard D, Moore E, Fernandez D, Kreisberg-Greenblatt R, Oved K, Eden E,
Hays JP (2015) A multifaceted ‘omics’ approach for addressing the challenge of antimicrobial
resistance. Future Microbiol 10:365–376
Dassey AJ, Hall SG, Theegala CS (2014) An analysis of energy consumption for algal biodiesel
production: comparing the literature with current estimates. Algal Res 4:89–95
Diamandis EP (2010) Cancer biomarkers: can we turn recent failures into success? JNCI J Natl
Cancer Inst 102:1462–1467
Douterelo I, Boxall JB, Deines P, Sekar R, Fish KE, Biggs CA (2014) Methodological approaches
for studying the microbial ecology of drinking water distribution systems. Water Res 65:134–
156
Edyvean RGJ, Videla HA (1991) Biological corrosion. Interdis Sci Rev 16:267–282
El-Serehy HA, AL-Rasheid KA, AL-Misned FA, AL-Talasat AAR, Gewik MM (2016)
Microbial–meiofaunal interrelationships in coastal sediments of the Red Sea. Saudi J Biol
Sci 23:327–334
Enjalbert B, Jourdan F, Portais JC (2011) Intuitive visualization and analysis of multi-omics data
and application to Escherichia coli carbon metabolism. PLoS ONE 6:e21318
10 Beyond Metabolomics: A Review of Multi-Omics-Based Approaches 307
Erickson AR, Cantarel BL, Lamendella R, Darzi Y, Mongodin EF, Pan C, Shah M, Halfvarson J,
Tysk C, Henrissat B, Raes J, Verberkmoes NC, Fraser CM, Hettich RL, Jansson JK (2012)
Integrated metagenomics/metaproteomics reveals human host-microbiota signatures of crohn’s
disease. PLoS ONE 7:e49138
Field D, Garrity G, Gray T, Morrison N, Selengut J, Sterk P, Tatusova T, Thomson N, Allen MJ,
Angiuoli SV, Ashburner M, Axelrod N, Baldauf S, Ballard S, Boore J, Cochrane G, COLE J,
Dawyndt P, De Vos P, Depamphilis C, Edwards R, Faruque N, Feldman R, Gilbert J, Gilna P,
Glockner FO, Goldstein P, Guralnick R, Haft D, Hancock D, Hermjakob H, Hertz-Fowler C,
Hugenholtz P, Joint I, Kagan L, Kane M, Kennedy J, Kowalchuk G, Kottmann R, Kolker E,
Kravitz S, Kyrpides N, Leebens-Mack J, Lewis SE, Li K, Lister AL, Lord P, Maltsev N,
Markowitz V, Martiny J, Methe B, Mizrachi I, Moxon R, Nelson K, Parkhill J, Proctor L,
White O, Sansone SA, Spiers A, Stevens R, Swift P, Taylor C, Tateno Y, Tett A, Turner S,
Ussery D, Vaughan B, Ward N, Whetzel T, San Gil I, Wilson G, Wipat A (2008) The
minimum information about a genome sequence (MIGS) specification. Nat Biotechnol 26:
541–547
Flemming HC (1994) Microbial deterioration of materials—fundamentals—economical and
technical overview. Werkst Korros-Mater Corros 45:5–9
Fondi M, Liò P (2015) Multi -omics and metabolic modelling pipelines: challenges and tools for
systems microbiology. Microbiol Res 171:52–64
Forsythe IJ, Wishart DS (2009) Exploring human metabolites using the human metabolome
database. In: Current protocols in bioinformatics, Chap. 14, Unit 14.8
Garcia-Alcalde F, Garcia-Lopez F, Dopazo J, Conesa A (2011) Paintomics: a web based tool for
the joint visualization of transcriptomics and metabolomics data. Bioinformatics 27:137–139
Garrett WS (2015) Cancer and the microbiota. Science 348:80–86
Gevers D, Kugathasan S, Denson LA, Vazquez-Baeza Y, Van Treuren W, Ren B, Schwager E,
Knights D, Song SJ, Yassour M, Morgan XC, Kostic AD, Luo C, Gonzalez A, Mcdonald D,
Haberman Y, Walters T, Baker S, Rosh J, Stephens M, Heyman M, Markowitz J,
Baldassano R, Griffiths A, Sylvester F, Mack D, Kim S, Crandall W, Hyams J,
Huttenhower C, Knight R, Xavier RJ (2014) The treatment-naive microbiome in new-onset
Crohn’s disease. Cell Host Microbe 15:382–392
Goldman SM, Kamel F, Ross GW, Jewell SA, Marras C, Hoppin JA, Umbach DM,
Bhudhikanok GS, Meng C, Korell M, Comyns K, Hauser RA, Jankovic J, Factor SA,
Bressman S, Lyons KE, Sandler DP, Langston JW, Tanner CM (2014) Peptidoglycan
recognition protein genes and risk of parkinson’s disease. Mov Disord 29:1171–1180
Gomez-Alvarez V, Revetta R, Domingo JW (2012) Metagenome analyses of corroded concrete
wastewater pipe biofilms reveal a complex microbial system. BMC Microbiol 12:122
Goodman SR, Pace BS, Hansen KC, D’alessandro A, Xia Y, Daescu O, Glatt SJ (2016)
Minireview: multiomic candidate biomarkers for clinical manifestations of sickle cell severity:
early steps to precision medicine. Exp Biol Med 241:772–781
Graeber M, Boehm S, Kuever J (2014) 3—molecular methods for studying biocorrosion. In:
Liengen T, Féron D, Basséguy R, Beech IB (eds) Understanding biocorrosion. Woodhead
Publishing, Cambridge
Grimplet J, Cramer GR, Dickerson JA, Mathiason K, Van Hemert J, Fennell AY (2009) VitisNet:
“Omics” integration through grapevine molecular networks. PLoS ONE 4:e8365
Guha N, Deepak SA, Lateef S, Padmanabhan A, Gundimeda S, Ghosh A, Mallipatna A, Suresh
Babu V (2015) A multi-omics approach to identify biomarkers of clinically advanced
retinoblastoma for diagnostics and therapeutic applications. FASEB J 29:417.7
Günther OP, Shin H, Ng RT, Mcmaster WR, Mcmanus BM, Keown PA, Tebbutt SJ, lê Cao KA
(2014) Novel multivariate methods for integration of genomics and proteomics data:
applications in a kidney transplant rejection study. OMICS: J Integr Biol 18:682–695
Handelsman J (2004) Metagenomics: application of genomics to uncultured microorganisms.
Microbiol Mol Biol Rev 68:669–685
308 D.J. Beale et al.
Heinrichs G, Hübner I, Schmidt CK, De hoog GS, Haase G (2013) Analysis of black fungal
biofilms occurring at domestic water taps (I): compositional analysis using tag-encoded flx
amplicon pyrosequencing. Mycopathologia 175:387–397
Higdon R, Earl RK, Stanberry L, Hudac CM, Montague E, Stewart E, Janko I, Choiniere J,
Broomall W, Kolker N, Bernier RA, Kolker E (2015) The promise of multi-omics and clinical
data integration to identify and target personalized healthcare approaches in autism spectrum
disorders. OMICS 19:197–208
Hook SE, Osborn HL, Spadaro DA, Simpson SL (2014) Assessing mechanisms of toxicant
response in the amphipod Melita plumulosa through transcriptomic profiling. Aquat Toxicol
146:247–257
Hossain A, Salleh A, Boyce AN, Chowdhury P, Naqiuddin M (2008) Biodiesel fuel production
from algae as renewable energy. Am J Biochem Biotechnol 4:250–254
Hultman J, Waldrop MP, Mackelprang R, David MM, Mcfarland J, Blazewicz SJ, Harden J,
Turetsky MR, Mcguire AD, Shah MB, Verberkmoes NC, Lee LH, Mavrommatis K,
Jansson JK (2015) Multi-omics of permafrost, active layer and thermokarst bog soil
microbiomes. Nature 521:208–212
Jadhav S, Gulati V, Fox EM, Karpe A, Beale DJ, Sevior D, Bhave M, Palombo EA (2015) Rapid
identification and source-tracking of Listeria monocytogenes using MALDI-TOF mass
spectrometry. Int J Food Microbiol 202:1–9
Jadhav S, Sevior D, Bhave M, Palombo EA (2014) Detection of Listeria monocytogenes from
selective enrichment broth using MALDI–TOF Mass Spectrometry. J Proteomics 97:100–106
Jansson J, Willing B, Lucio M, Fekete A, Dicksved J, Halfvarson J, Tysk C, Schmitt-Kopplin P
(2009) Metabolomics reveals metabolic biomarkers of crohn’s disease. PLoS ONE 4:e6386
Jones OAH, Sdepanian S, Lofts S, Svendsen C, Spurgeon DJ, Maguire ML, Griffin JL (2014)
Metabolomic analysis of soil communities can be used for pollution assessment. Environ
Toxicol Chem 33:61–64
Jozefczuk S, Klie S, Catchpole G, Szymanski J, Cuadros-Inostroza A, Steinhauser D, Selbig J,
Willmitzer L (2010) Metabolomic and transcriptomic stress response of Escherichia coli. Mol
Syst Biol 6:364
Junker BH, Klukas C, Schreiber F (2006) Vanted: a system for advanced data analysis and
visualization in the context of biological networks. BMC Bioinf 7:1–13
Kaever A, Landesfeind M, Feussner K, Mosblech A, Heilmann I, Morgenstern B, Feussner I,
Meinicke P (2015) MarVis-pathway: integrative and exploratory pathway analysis of
non-targeted metabolomics data. Metabolomics 11:764–777
Kamburov A, Cavill R, Ebbels TM, Herwig R, Keun HC (2011) Integrated pathway-level analysis
of transcriptomics and metabolomics data with IMPaLA. Bioinformatics 27:2917–2918
Karnovsky A, Weymouth T, Hull T, Tarcea VG, Scardoni G, Laudanna C, Sartor MA,
Stringer KA, Jagadish HV, Burant C, Athey B, Omenn GS (2012) Metscape 2 bioinformatics
tool for the analysis and visualization of metabolomics and gene expression data.
Bioinformatics 28:373–380
Karsenti E, Acinas SG, Bork P, Bowler C, Vargas CD, Raes J, Sullivan M, Arendt D, Benzoni F,
Claverie JM, Follows M, Gorsky G, Hingamp P, Iudicone D, Jaillon O, Kandels-Lewis S,
Krzic U, Not F, Ogata H, Pesant S, Reynaud EG, Sardet C, Sieracki ME, Speich S,
Velayoudon D, Weissenbach J, Wincker PW, Consortium TT (2011) A holistic approach to
marine eco-systems biology. PLoS Biol 9:e1001177
Kato H, Takahashi S, Saito K (2011) Omics and integrated omics for the promotion of food and
nutrition science. J Tradit Complement Med 1:25–30
Kimes NE, Callaghan AV, Aktas DF, Smith WL, Sunner J, Golding BT, Drozdowska M,
Hazen TC, Suflita JM, Morris PJ (2013) Metagenomic analysis and metabolite profiling of
deep-sea sediments from the gulf of mexico following the deepwater horizon oil spill. Front
Microbiol 4:50
King ZA, Dräger A, Ebrahim A, Sonnenschein N, Lewis NE, Palsson BO (2015) Escher: a web
application for building, sharing, and embedding data-rich visualizations of biological
pathways. PLoS Comput Biol 11:e1004321
10 Beyond Metabolomics: A Review of Multi-Omics-Based Approaches 309
Kip N, Van Veen JA (2015) The dual role of microbes in corrosion. ISME J 9:542–551
Kohl M, Megger DA, Trippler M, Meckel H, Ahrens M, Bracht T, Weber F, Hoffmann AC,
Baba HA, Sitek B, Schlaak JF, Meyer HE, Stephan C, Eisenacher M (2014) A practical data
processing workflow for multi-OMICS projects. Biochim Biophys Acta 1844:52–62
Kouremenos KA, Beale DJ, Antti H, Palombo EA (2014) Liquid chromatography time of flight
mass spectrometry based environmental metabolomics for the analysis of Pseudomonas putida
bacteria in potable water. J Chromatogr B 966:179–186
Kovács E, Wirth R, Maróti G, Bagi Z, Nagy K, Minárovits J, Rákhely G, Kovács KL (2015)
Augmented biogas production from protein-rich substrates and associated metagenomic
changes. Bioresour Technol 178:254–261
Krämer A, Green J, Pollard J, Tugendreich S (2013) Causal analysis approaches in ingenuity
pathway analysis. Bioinform 30:523–530
Kuo TC, Tian TF, Tseng YJ (2013) 3Omics: a web-based systems biology tool for analysis,
integration and visualization of human transcriptomic, proteomic and metabolomic data. BMC
Syst Biol 7:64
Kutmon M, Van Iersel MP, Bohler A, Kelder T, Nunes N, Pico AR, Evelo CT (2015) PathVisio 3:
an extendable pathway analysis toolbox. PLoS Comput Biol 11:e1004085
Leary DH, Li RW, Hamdan LJ, Hervey WJ, Lebedev N, Wang Z, Deschamps JR,
Kusterbeck AW, Vora GJ (2014) Integrated metagenomic and metaproteomic analyses of
marine biofilm communities. Biofouling 30:1211–1223
Ley RE, Turnbaugh PJ, Klein S, Gordon JI (2006) Microbial ecology—human gut microbes
associated with obesity. Nature 444:1022–1023
Lin K, Kools H, De Groot PJ, Gavai AK, Basnet RK, Cheng F, Wu J, Wang X, Lommen A,
Hooiveld GJ, Bonnema G, Visser RG, Muller MR, Leunissen JA (2011) Madmax—
MANAGEMENT and analysis database for multiple *omics experiments. J Integr Bioinform
8:160
Liu H, Xu L, Zeng J (2000) Role of corrosion products in biofilms in microbiologically induced
corrosion of carbon steel. Br Corros J 35:131–135
Llewellyn CA, Sommer U, Dupont CL, Allen AE, Viant MR (2015) Using community
metabolomics as a new approach to discriminate marine microbial particulate organic matter in
the western English Channel. Prog Oceanogr 137 Part B:421–433
Lourenço A, Ferreira A, Veiga N, Machado I, Pereira MO, Azevedo NF (2012) BiofOmics: a web
platform for the systematic and standardized collection of high-throughput biofilm data. PLoS
One 7:e39960
Ma X, Zheng X, Zhang M, Yang X, Chen L, Huang L, Cao S (2014) Electron beam irradiation of
bamboo chips: degradation of cellulose and hemicelluloses. Cellulose 21:3865–3870
Machado D, Herrgård M (2014) Systematic evaluation of methods for integration of transcriptomic
data into constraint-based models of metabolism. PLoS Comput Biol 10:e1003580
Megan EH, Michael HC, Merry-Lynn M, Peter JC, Ruth TS, Diego JM, Barry JM, James DC,
Russell PB, Edwin KS, Craig PH (2016) A multi-Omics Approach Identifies Inflammatory
Patterns In The asthma-COPD overlap syndrome. D91. Genetics and Genomics of COPD.
American Thoracic Society
Mohan AM, Bibby KJ, Lipus D, Hammack RW, Gregory KB (2014) The functional potential of
microbial communities in hydraulic fracturing source water and produced water from natural
gas extraction characterized by metagenomic sequencing. PLoS ONE 9:e107682
Morrison N, Wood AJ, Hancock D, Shah S, Hakes L, Gray T, Tiwari B, Kille P, Cossins A,
Hegarty M, Allen MJ, Wilson WH, Olive P, Last K, Kramer C, Bailhache T, Reeves J,
Pallett D, Warne J, Nashar K, Parkinson H, Sansone SA, Rocca-Serra P, Stevens R, Snape J,
Brass A, Field D (2006) Annotation of environmental OMICS data: application to the
transcriptomics domain. OMICS 10:172–178
Neria-González I, Wang ET, Ramírez F, Romero JM, Hernández-Rodríguez C (2006)
Characterization of bacterial community associated to biofilms of corroded oil pipelines from
the southeast of Mexico. Anaerobe 12:122–133
310 D.J. Beale et al.
Neuweger H, Persicke M, Albaum SP, Bekel T, Dondrup M, Huser AT, Winnebald J, Schneider J,
Kalinowski J, Goesmann A (2009) Visualizing post genomics data-sets on customized pathway
maps by ProMeTra-aeration-dependent gene expression and metabolism of Corynebacterium
glutamicum as an example. BMC Syst Biol 3:82
Noecker C, Eng A, Srinivasan S, Theriot CM, Young VB, Jansson JK, Fredricks DN, Borenstein E
(2016) Metabolic model-based integration of microbiome taxonomic and metabolomic profiles
elucidates mechanistic links between ecological and metabolic variation. mSystems 1:
e00013-15
Nozhevnikova AN, Botchkova EA, Plakunov VK (2015) Multi-species biofilms in ecology,
medicine, and biotechnology. Microbiology 84:731–750
Orchard S, Kerrien S (2010) Molecular interactions and data standardisation. Methods Mol Biol
604:309–318
Osborn HL, Hook SE (2013) Using transcriptomic profiles in the diatom Phaeodactylum
tricornutum to identify and prioritize stressors. Aquat Toxicol 138–139:12–25
Pap B, Györkei Á, Boboescu IZ, Nagy IK, Bíró T, Kondorosi É, Maróti G (2015)
Temperature-dependent transformation of biogas-producing microbial communities points to
the increased importance of hydrogenotrophic methanogenesis under thermophilic operation.
Bioresour Technol 177:375–380
Park JJ, Wang H, Gargouri M, Deshpande RR, Skepper JN, Holguin FO, Juergens MT,
Shachar-Hill Y, Hicks LM, Gang DR (2015) The response of Chlamydomonas reinhardtii to
nitrogen deprivation: a systems biology analysis. Plant J 81:611–624
Pascault N, Loux V, Derozier S, Martin V, Debroas D, Maloufi S, Humbert J-F, Leloup J (2015)
Technical challenges in metatranscriptomic studies applied to the bacterial communities of
freshwater ecosystems. Genetica 143:157–167
Pertea M, Salzberg SL (2010) Between a chicken and a grape: estimating the number of human
genes. Genome Biol 11:206
Pomraning KR, Kim Y-M, Nicora CD, Chu RK, Bredeweg EL, Purvine SO, Hu D, Metz TO,
Baker SE (2016) Multi-omics analysis reveals regulators of the response to nitrogen limitation
in Yarrowia lipolytica. BMC Genom 17:1–18
Röling WFM, Ferrer M, Golyshin PN (2010) Systems approaches to microbial communities and
their functioning. Curr Opin Biotechnol 21:532–538
Saito K, Arai S, Kato H (2005) A nutrigenomics database–integrated repository for publications
and associated microarray data in nutrigenomics research. Br J Nutr 94:493–495
Sansone SA, Fan T, Goodacre R, Griffin JL, Hardy NW, Kaddurah-Daouk R, Kristal BS,
Lindon J, Mendes P, Morrison N, Nikolau B, Robertson D, Sumner LW, Taylor C, Van Der
Werf M, Van Ommen B, Fiehn O (2007) The metabolomics standards initiative. Nat
Biotechnol 25:846–848
Santo Domingo JW, Revetta RP, Iker B, Gomez-Alvarez V, Garcia J, Sullivan J, Weast J (2011)
Molecular survey of concrete sewer biofilm microbial communities. Biofouling 27:993–1001
Schneider MV, Orchard S (2011) Omics technologies, data and bioinformatics principles. In:
Mayer B (ed) Bioinformatics for omics data: methods and protocols. Humana Press, New York
Schneider T, Riedel K (2010) Environmental proteomics: analysis of structure and function of
microbial communities. Proteomics 10:785–798
Shaw JLA, Monis P, Fabris R, Ho L, Braun K, Drikas M, Cooper A (2014) Assessing the impact
of water treatment on bacterial biofilms in drinking water distribution systems using
high-throughput DNA sequencing. Chemosphere 117:185–192
Singer SW (2012) Targeted isolation of proteins from natural microbial communities living in an
extreme environment. Methods Mol Biol 881:63–72
Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly Y, Glickman JN,
Garrett WS (2013) The microbial metabolites, short-chain fatty acids, regulate colonic treg cell
homeostasis. Science 341:569–573
Srinivasan S, Morgan MT, Fiedler TL, Djukovic D, Hoffman NG, Raftery D, Marrazzo JM,
Fredricks DN (2015) Metabolic signatures of bacterial vaginosis. mBio 6:e00204–e00215
10 Beyond Metabolomics: A Review of Multi-Omics-Based Approaches 311
Stasiewicz MJ, Den Bakker HC, Wiedmann M (2015) Genomics tools in microbial food safety.
Curr Opin Food Sci 4:105–110
Sumner LW, Amberg A, Barrett D, Beale MH, Beger R, Daykin CA, Fan TWM, Fiehn O,
Goodacre R, Griffin JL, Hankemeier T, Hardy N, Harnly J, Higashi R, Kopka J, Lane AN,
Lindon JC, Marriott P, Nicholls AW, Reily MD, Thaden JJ, Viant MR (2007) Proposed
minimum reporting standards for chemical analysis. Metabolomics 3:211–221
Sunagar K, Morgenstern D, Reitzel AM, Moran Y (2016) Ecological venomics: how genomics,
transcriptomics and proteomics can shed new light on the ecology and evolution of venom.
J Proteomics 135:62–72
Takahashi S, Saito K, Jia H, Kato H (2014) An integrated multi-omics study revealed metabolic
alterations underlying the effects of coffee consumption. PLoS One 9:e91134
Talbot G, Topp E, Palin MF, Massé DI (2008) Evaluation of molecular methods used for
establishing the interactions and functions of microorganisms in anaerobic bioreactors. Water
Res 42:513–537
Thakur NL, Jain R, Natalio F, Hamer B, Thakur AN, Müller WEG (2008) Marine molecular
biology: an emerging field of biological sciences. Biotechnol Adv 26:233–245
Theriot CM, Koenigsknecht MJ, Carlson JRPE, Hatton GE, Nelson AM, Li B, Huffnagle GBZ,
Li J, Young VB (2014) Antibiotic-induced shifts in the mouse gut microbiome and
metabolome increase susceptibility to Clostridium difficile infection. Nat Commun 5
Thimm O, Blasing O, Gibon Y, Nagel A, Meyer S, Kruger P, Selbig J, Muller LA, Rhee SY,
Stitt M (2004) Mapman: a user-driven tool to display genomics data sets onto diagrams of
metabolic pathways and other biological processes. Plant J 37:914–939
Tokimatsu T, Sakurai N, Suzuki H, Ohta H, Nishitani K, Koyama T, Umezawa T, Misawa N,
Saito K, Shibata D (2005a) KaPPA-view: a web-based analysis tool for integration of transcript
and metabolite data on plant metabolic pathway maps. Plant Physiol 138:1289–1300
Tokimatsu T, Sakurai N, Suzuki H, Ohta H, Nishitani K, Koyama T, Umezawa T, Misawa N,
Saito K, Shibatanenell D (2005b) KaPPA-view. a web-based analysis tool for integration of
transcript and metabolite data on plant metabolic pathway maps. Plant Physiol 138:1289–1300
Trentacoste EM, Shrestha RP, Smith SR, glé C, Hartmann AC, Hildebrand M, Gerwick WH
(2013) Metabolic engineering of lipid catabolism increases microalgal lipid accumulation
without compromising growth. Proc Natl Acad Sci USA 110:19748–19753
Turnbaugh PJ, Gordon JI (2008) An invitation to the marriage of metagenomics and
metabolomics. Cell 134:708–713
Usadel B, Nagel A, Thimm O, Redestig H, Blaesing OE, Palacios-Rojas N, Selbig J,
Hannemann J, Piques MC, Steinhauser D, Scheible WR, Gibon Y, Morcuende R,
Weicht D, Meyer S, Stitt M (2005) Extension of the visualization tool mapman to allow
statistical analysis of arrays, display of corresponding genes, and comparison with known
responses. Plant Physiol 138:1195–1204
Valdés A, Ibáñez C, Simó C, García-Cañas V (2013) Recent transcriptomics advances and
emerging applications in food science. TrAC Trends Anal Chem 52:142–154
Vanwonterghem I, Jensen PD, Ho DP, Batstone DJ, Tyson GW (2014) Linking microbial
community structure, interactions and function in anaerobic digesters using new molecular
techniques. Curr Opin Biotechnol 27:55–64
Videla HA (2003) Biocorrosion and biofouling of metals and alloys of industrial usage. Present
state of the art at the beginning of the new millennium. Rev Metal 34:256–264
Videla HA, Herrera LK (2009) Understanding microbial inhibition of corrosion. a comprehensive
overview. Int Biodeterior Biodegradation 63:896–900
Vincke E, Boon N, Verstraete W (2001) Analysis of the microbial communities on corroded
concrete sewer pipes—a case study. Appl Microbiol Biotechnol 57:776–785
Wagele B, Witting M, Schmitt-Kopplin P, Suhre K (2012) Mass TRIX reloaded: combined
analysis and visualization of transcriptome and metabolome data. PLoS One 7:e39860
Wheelock AM, Wheelock CE (2013) Trials and tribulations of ‘omics data analysis: assessing
quality of SIMCA-based multivariate models using examples from pulmonary medicine. Mol
Bio Syst 9:2589–2596
312 D.J. Beale et al.
Wildburger NC, Lichti CF, Leduc RD, Schmidt M, Kroes RA, Moskal JR, Nilsson CL (2015)
Quantitative proteomics and transcriptomics reveals metabolic differences in attracting and
non-attracting human-in-mouse glioma stem cell xenografts and stromal cells. EuPA Open
Proteomics 8:94–103
Xia J, Fjell CD, Mayer ML, Pena OM, Wishart DS, Hancock RE (2013) Inmex–a web-based tool
for integrative meta-analysis of expression data. Nucleic Acids Res 41:W63–W70
Xia T, Hemert JV, Dickerson JA (2010) OmicsAnalyzer: a Cytoscape plug-in suite for modeling
omics data. Bioinformatics 26:2995–2996
Xu Y, Dou D, Ran X, Liu C, Chen J (2015) Integrative analysis of proteomics and metabolomics
of anaphylactoid reaction induced by Xuesaitong injection. J Chromatogr A 1416:103–111
Yanagawa K, kouduka M, Nakamura Y, Hachikubo A, Tomaru H, Suzuki Y (2014) Distinct
microbial communities thriving in gas hydrate-associated sediments from the eastern Japan
Sea. J Asian Earth Sci 90:243–249
Yizhak K, Benyamini T, Liebermeister W, Ruppin E, Shlomi T (2010) Integrating quantitative
proteomics and metabolomics with a genome-scale metabolic network model. Bioinformatics
26:i255–i260
Zeidan-Chulia F, De Oliveira B-H, Salmina A, Casanova M, Gelain D, Noda M, Verkhratsky A,
Moreira JC (2014) Altered expression of Alzheimer’s disease-related genes in the cerebellum
of autistic patients: a model for disrupted brain connectome and therapy. Cell Death Dis 5:
e1250
Zhang W, Li F, Nie L (2010) Integrating multiple ‘omics’ analysis for microbial biology:
application and methodologies. Microbiology 156:287–301
Zhang W, Wang Y, Lee OO, Tian R, Cao H, Gao Z, Li Y, Yu L, Xu Y, Qian PY (2013)
Adaptation of intertidal biofilm communities is driven by metal ion and oxidative stresses.
Scientific reports, vol 3
Index
Note: Page numbers followed by f and t indicate figures and tables respectively
A Bacteria, 149–150
ABC transmembrane proteins (ATP-binding bacterial soil inoculants, 166–167
cassette), 6 and metal toxicity, 204
Abiotic stress adaptation, 167 biofilm versus planktonic response to
Adipose tissue form (ATF), 92 copper, 208
Adulteration of food material, 231, 245–246 investigations of biofilms and
After TAG synthesis (ATS), 304 morphological variants, 207–208
Akkermansia muciniphila, 24, 28, 58 methodological considerations and
Alkylphosphocoline, 95 conclusions, 209
Aminosugars, 93 tellurite resistance, 205–206, 205f
Ammonia fibre explosion (AFEX), 264 Bacteroidetes, 14
Anaerobic digesters, 300 Bayesian probabilistic approach, 214
Anaerobic digestion, 209, 210 Before TAG synthesis (BTS), 304
Anaerobic reactors, 210 β-oxidation inhibitors, 97
Analytical instruments in metabolomics, Bifidobacteria, 16
advancements of, 234–239 Bifidobacterium, 24, 28
Anthropogenic effects on soil microbes, Biodefense and Emerging Infections Research
174–175 Resources Repository (BEI Resources), 61
climatic change effects, 181–183 Biofertilizers, 166
engineered nanomaterials (ENM), 175–177 Biofilms, 162, 178, 301–303
heavy metal contamination, 177–179 and morphological variants, 207–28
organic contaminants, 179–181 versus planktonic response to copper, 208
Antibiotics, 27, 67, 162, 167, 173, 180–181, Biofomics, 294t, 301
241, 243 Biofuels, 304
Antimonials, pentavalent and trivalent, 94 Biomarkers, 233, 240, 242–243, 245
Arabidopsis–Pseudomonas rhizosphere model, Biomass waste management, microbial
164 metabolomics in, 261, 263–265
Arabidopsis thaliana, 93, 164–165, 167, 169 biomass degradation/conversion methods,
Arbuscular mycorrhizal fungi (AMF), 161, 265–267
163, 170–171, 178, 180, 181–182 future research, 280–281
Arginine deprivation response (ADR), 91 metabolic engineering approaches, 276
Aspergillus nidulans, 274 to develop fungal bioprocessing
Aspergillus oryzae, 265 abilities, 277–280
Aspergillus spp., 265, 267 microbial process metabolomics and its
Atmospheric pressure ionisation (API), 236 application to biomass, 268
Autism spectrum disorders (ASD), 14, 28, 303 classification of metabolomic
approaches, 268–269
B metabolic fingerprinting, 272–275
Bacillus subtilis, 134, 174, 181 metabolic flux (fluxomics), 275–276
Single nucleotide polymorphisms (SNPs), 303 Synechococcus sp. PCC 7002, 128
16S rRNA gene, 53–54, 59–61, 211–212 Systems biology, 214, 278
Small-colony variants (SCVs), 207
Sodium fluoroacetate (NaFAc), 101 T
Soil, complexity of, 147–157 T5 cells, 206
Soil community, 128–131 Targeted metabolomics methods, 271
Soil exometabolomics studies, 122, 127 Tellurite resistance, 205–206, 205f
Soil inoculants, 165–168 Tellurium oxide, 205
Soil matrix, 148 Terroir, 169
Soil microbial metabolomics, 147 Thermoanaerobacter ethanolicus, 265
anthropogenic effects on soil microbes, Thermophilic bacteria, 265, 267
174–175 Thin layer chromatography (TLC), 272
climatic change effects, 181–182 3Omics, 294t
engineered nanomaterials (ENM), Time of flight (ToF), 236
175–177 Time-of-flight secondary ion mass
heavy metal contamination, 171–173 spectrometry (TOF-SIMS), 136
organic contaminants, 179–181 Toll-like receptor 10 (TLR10), 303
complexity of soil, 147–157 Toll-like receptors (TLRs), 62, 63
extracellular enzymes, 173–174 Total dietary fibre (TDF), 17
exudates, 162 Toxoplasma gondii, 87
rhizoengineering, 163–164 Trametes spp., 265
humus, 168–169 Transmembrane protein family PulD, 6
LC–MS analyses, 158t Tree litter, 172
metabolite coverage over the lifespan of Triacylglycerol (TAG) synthesis, 304
plants, 165 Trichoderma, 167, 168
nascent field, 157–159 Trichoderma hamatum, 274
rhizosphere, 159 Trichoderma reesei, 276
map of, 159–161 Trichoderma spp., 265, 267
metabolomics, 161–162 “Trojan Horse” strategy, 86
nutrients in, 170–172 “Trojan rabbit” model, 86
soil inoculants, 165–168 Trypanosoma, 83
terroir, 169 Trypanosoma brucei, 83, 92
tools for, 183–184 Trypanosoma cruzi, 83, 87
Soil microorganisms, 119, 133 Trypanosomatidae, 83
metabolomics (see Soil microbial Type IV secretion systems (T4SS), 6
metabolomics)
Soil organic matter (SOM), 120, 122 U
exometabolomics for analysis of, 120–126 Ulcerative colitis (UC), 27
Soil science, 201–204 Ultra-high-performance liquid chromatography
Solid phase extraction (SPE), 234 coupled to electrospray ionisation-high
Solid phase microextraction (SPME), 234 resolution mass spectrometry
Solid state fermentation (SSF), 265–266 (UHPLC-ESI-HRMS), 274
Stable isotope labeling approaches, 99, 103 Ultrasensitive analytical methods, 8
Stable isotope probing (SIP) techniques, Uncinula necator, 271
131–133, 134 Upflow anaerobic sludge blanket (UASB)
Steam explosion (SE) experiment, 264 reactor, 210
Streptococcus bovis, 270
Streptomyces coelicolor, 134 V
Submerged fermentation/shake flask VANTED (visualization and analysis of
fermentation (SmF), 265 networks with related experimental data),
SWCNT (single-walled carbon nanotubes), 176 296t
Symbiotic fermentation, 267 Visceral leishmaniasis (VL), 83
Index 321