Irit Sagi - Nikolaos A Afratis - Collagen - Methods and Protocols-Humana Press (2019)
Irit Sagi - Nikolaos A Afratis - Collagen - Methods and Protocols-Humana Press (2019)
Irit Sagi - Nikolaos A Afratis - Collagen - Methods and Protocols-Humana Press (2019)
Collagen
Methods and Protocols
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Edited by
This Humana Press imprint is published by the registered company Springer Science+Business Media, LLC, part of
Springer Nature.
The registered company address is: 233 Spring Street, New York, NY 10013, U.S.A.
Preface
Collagen is the most abundant protein in mammals, making up about 25% of the whole-
body protein content. Collagen is involved in many physiological and pathological situa-
tions. Specifically, collagen has been shown to be involved either directly or indirectly in the
promotion of cell adhesion and differentiation, cell functions that are related to the control
of embryonic development and regeneration. However, the uncontrolled synthesis and
accumulation of collagen from fibroblasts lead to pathological conditions in fibrotic-related
diseases and tumorigenesis.
Collagen is the main structural protein in extracellular matrix and connective tissue. The
basic fundamental structure consists of three coiled subunits: two α1(I) chains and one α2
(I). There are at least 26 different types of collagen, and all of them contain three-stranded
helical segments of similar structure. The differentiation between each type of collagen is
due to segments that interrupt the triple helix and that fold into other kinds of three-
dimensional structures. Collagen types I, II, and III consist the 80% of the collagen in
the body.
Collagen is widely used in many applications like food, cosmetics, pharmaceuticals,
cosmetic surgery, artificial skin, and glue (the name collagen comes from the
Greek κóλλα (kolla), meaning “glue,” and suffix -γεν, -gen, denoting “producing”). The
importance of collagen in health and disease and the interest of the researchers to further
investigate the in vitro and in vivo mechanisms of collagen were the objective reasons that
motivated us to create this Collagen: Methods and Protocols book. This volume compiles a
collection of state-of-the-art protocols that will serve not only as recipes for bench scientists
but also as accepted methods for the collagen field. Each chapter describes a specific topic
and technique by an expert in the field and provides a detailed protocol, which includes
some most helpful tips in the Notes section.
The content of the book is separated into four different parts. Specifically, Part I focuses
on in vitro models for the characterization of collagen formation. In detail, it describes a
mimetic model for mineralization of type-I collagen fibrils, cross-linked collagen scaffolds,
and methods to clone, mutate, and isolate collagen. Part II highlights a large-scale analysis
of collagen with mass spectrometry in order to elucidate the proteomics, degradomics,
interactomes, and cross-linking of collagen. It also presents the triple-helix hydroxylation
of collagen by solid-state NMR spectroscopy.
Furthermore, Part III describes high-resolution imaging approaches for collagen by the
use of scanning electron microscopy and multiphoton imaging. In addition, a new tool for
degraded and denatured collagen histopathology and collagen-binding integrin analysis is
presented. Finally, Part IV introduces the role of collagen during physiological and patho-
logical conditions and provides an overview of collagenous matrix in tumor microenviron-
ment. The reader will find protocols to analyze the collagen levels from primary fibroblasts,
to produce collagen-based peptides for inhibition of matrix metalloproteinases, and to assess
collagen deposition during aging and liver fibrosis.
v
vi Preface
We hope that this volume on collagen will be a useful tool for the scientists working in
the field and will inspire them to develop new techniques and models that will create the next
generation of collagen protocols. We would also like to thank all the authors and co-authors
that participated on this book for their tremendous contributions. It was a great pleasure to
work with all of them, and we believe that their effort was critical for the high quality and
repeatability of the research protocols.
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
vii
viii Contents
ix
x Contributors
Abstract
The size and relatively high GC content of cDNAs are challenges for efficient targeted engineering of large
collagens. There are both basic biological and therapeutic interests in the ability to modify collagens, as this
would allow for studies precisely describing interactions of collagens with specific interaction partners,
addressing consequences of individual disease-causing mutations, and assessing therapeutic applicability of
precision medicine approaches. Using collagen VII as an example, we will here describe a strategy for rapid
and simple modification of cDNAs encoding large collagens. The method is flexible and can be used for the
creation of point mutations, small or large deletions, and insertion of DNA.
Key words Collagen VII, Gibson assembly, Dystrophic epidermolysis bullosa, Collagenopathy, Exon
skipping
1 Introduction
Irit Sagi and Nikolaos A. Afratis (eds.), Collagen: Methods and Protocols, Methods in Molecular Biology, vol. 1944,
https://doi.org/10.1007/978-1-4939-9095-5_1, © Springer Science+Business Media, LLC, part of Springer Nature 2019
3
4 Olivier Bornert and Alexander Nyström
2 Materials
3. Lipofectamine® 2000.
4. Opti-MEM Reduced Serum Medium.
5. 20 mg/ml phleomycin
6. 10 cm cell culture dish
7. Blue sample buffer (BSB) 4: 4 M Urea, 0.5 M Trizma® base,
0.05% bromophenol blue, 10% SDS, 50% glycerol, 0.25 M
DTT, pH 6.8.
8. 7.5% Mini-PROTEAN® TGX precast gels (see Note 1)
9. Mini-PROTEAN® Tetra Vertical Electrophoresis cell.
10. TGS electrophoresis buffer: 25 mM Trizma® base, 192 mM
glycine, 0.1% SDS, pH 8.3.
11. Criterion™ blotter.
12. Transfer buffer: 1 Tris-borate (TB) buffer, 50 mM Trizma®
base, and 50 mM boric acid (see Note 2).
13. Amersham Protran nitrocellulose membrane.
14. Antibodies: Rabbit polyclonal antibody directed against the
epitope of the mouse monoclonal LH7.2 antibody (home-
made and verified to be specific for collagen VII [27], the
epitope is in the NC1 domain of collagen VII) and rabbit
polyclonal ProC7 antibody (homemade, raised against the
end of the NC2 domain of collagen VII; specific for the
pro-form of collagen VII) (see Note 3).
15. Tris-buffered saline-Tween 20 (TBS-T): 50 mM Trizma®
base, 150 mM NaCl, 0.05% Tween 20, pH 7.4.
16. Blocking buffer: TBS-T + 5% nonfat dry milk.
17. Enhanced chemiluminescence detection reagent, e.g., ECL™
Prime.
18. Developing system, e.g., Amersham™ imager 600.
19. 1 Phospho-buffered saline (PBS).
20. 50 μg/ml L-Ascorbic acid.
21. 50 ml Falcon Conical Centrifuge Tube.
22. 1 mM Pefabloc.
23. 500 mM EDTA pH 8.0.
24. Collagen IV.
25. 100 ng laminin-332.
Cloning and Mutagenesis Strategies for Large Collagens 7
3 Methods
3.1 Strategy Design 1. First, a strategy that will be used to modify the DNA sequence
and Generation of DNA of the target cDNA has to be designed. As an example, we will
Fragments with describe a procedure to remove a specific DNA region (from a
Overlapping Ends few amino acids to deletion of large sequences) from COL7A1
cDNA cloned into the vector pcDNA3.1 (Fig. 1).
2. The strategy is based on the use of PCR dividing the target
cDNA and vector, in this case COL7A1 and pcDNA3.1, into
fragments. In order to allow efficient amplification of the frag-
ments by PCR, dividing the mother sequence into fragments of
2.5 to 3 kb maximum is recommended.
3. As shown in Fig. 1, the strategy described here can be used to
delete a specific region of DNA. Not described here, by using
Fig. 1 Mutagenesis of COL7A1 using Gibson cloning. This figure presents an overview of the strategy used to
modify the COL7A1 cDNA sequence in the pcDNA3.1 vector by Gibson assembly. In step 1, specific pairs of
primers with overlapping sequence are designed to ensure reconstitution of the mother DNA construct, as well
as excision of a specific DNA sequence in COL7A1. These primers are then used to amplify selected regions of
the mother DNA construct (step 2). Finally, through Gibson reaction (step 3), the fragments are assembled
together in order to create a new DNA construct lacking the target DNA sequence
8 Olivier Bornert and Alexander Nyström
Table 1
Primers used for amplification of COL7A1 and deletion of exon73. Primers used to delete exon73 are
shown in red. Overlap is indicated with lower case
See Note 4
a
3.2 Gibson Assembly 1. Mix 50 ng of each PCR product together in a PCR tube. For
and Bacterial optimal reaction, the total volume should not exceed 20 μl
Transformation (see Note 6). Add a 1:1 volume of NEBuilder® HiFi DNA
assembly 2 Master Mix to the mixture. A control reaction
lacking one of the fragments is highly recommended to assess
the efficacy and specificity of the reaction.
2. Incubate reactions in a thermocycler at 50 C for 60 min.
Figure 2 provides a detailed step-by-step illustration of the
Gibson assembly® strategy [9]. Following incubation, samples
can be used directly or stored at 20 C for subsequent
transformation.
3. Thaw on ice NEB 10-alpha (see Note 7) Competent E. coli
bacteria, and incubate 2 μl of the assembled product with 50 μl
the bacteria for 30 min on ice. Heat shock the samples at 42 C
during 45 s, and cool down on ice for 2 min. Resuspend the
transformed bacteria in 200 μl Super Optimal broth with
Catabolite repression (SOC) medium, and plate the mixture
on a LB-agar-ampicillin petri dish. Incubate the plate at 37 C
overnight.
4. Select ten clones for analysis, and resuspend them individually
in 3 ml of LB medium with 100 μg/ml ampicillin in a 15 ml
10 Olivier Bornert and Alexander Nyström
Fig. 2 Gibson cloning in detail. In order to assemble two pieces of DNA together using Gibson assembly, it is
necessary to design primers containing a sequence that binds to the mother sequence and an overlapping
end, which correspond to the second DNA fragment. After amplification by PCR, the 50 end will be digested
with a 50 exonuclease. Then, complementary DNA sequences will anneal together, and a DNA polymerase will
be used to extend the fragment. After sealing of the gaps with a DNA ligase, the construct can be used to
transform bacteria. Finally, the newly generated product can be analyzed by sequencing
3.3 Collagen VII 1. HEK293 cells in DMEM/F12 medium are grown until a
Expression in HEK293 confluence of 70% and then transfected with Lipofectamine®
Cells 2000 with purified plasmids in a 10 cm cell culture dish. First,
Cloning and Mutagenesis Strategies for Large Collagens 11
3.4 First Assessment 1. Seed selected HEK293 cells carrying the construct of interest
of Functionality on poly-L-lysine-coated 10 cm tissue culture plates, and grow
of Engineered Collagen the cells to confluence.
VII Variants 2. Wash the plates three times with 3 ml PBS.
3. Add 10 ml serum-free DMEM and 50 μg/ml L-ascorbic acid
(see Note 11).
4. Next day add an additional 50 μg/ml L-ascorbic acid.
5. After 2 days harvest the medium, and add it to a Falcon 50 ml
Conical Centrifuge Tube, and centrifuge at 1000 g for 5 min
12 Olivier Bornert and Alexander Nyström
Fig. 3 Analyses of mutated collagen VII. Western blot for collagen VII on
conditioned medium from HEK293 cells expressing wild-type (WT) or collagen
VII with an internal deletion (the nucleotides corresponding to exon73 have been
deleted (Δ73)). The modified collagen VII has a deletion of 201 bp in the central
region of the COL7A1 cDNA (bp 6015 to 6216). As the deletion is small in
comparison to the large size of collagen VII, no significant difference in size can
be seen by Western blot. Usage of polyclonal LH7.2 and ProColVII antibodies
allows ensuring that the collagen VII is produced in full, as these antibodies,
respectively, bind the NC1 and the NC2 domains of collagen VII
to remove cells and debris (see Note 12). Add the medium to a
fresh 50 ml Falcon tube.
6. Cool the medium to 4 C, and add ammonium sulfate to a
saturation of 20%.
7. Precipitate the medium for 1 h at 4 C by rotating the tube
head over head.
8. Centrifuge 6000 g for at 4 C for 20 min.
9. Discard the supernatant and dissolve the pellet in 200 μl TBS.
Optional, if the protein is not to be used for limited trypsin
digestion, add 1 mM Pefabloc and 5 mM EDTA.
10. Limited trypsin digestion can be performed to give quick
information of correct folding of collagen VII (for a detailed
protocol please see [6]). The functionality of the protein can
further be assessed by determining binding to 100 ng immo-
bilized laminin-332 or collagen IV, as described in detail in
ref. 2.
4 Notes
Acknowledgments
References
1. Phillips CL, Lever LW, Pinnell SR et al (1991) 7. Fritsch A, Spassov S, Elfert S et al (2009)
Construction of a full-length murine pro alpha Dominant-negative effects of COL7A1 muta-
2(I) collagen cDNA by the polymerase chain tions can be rescued by controlled overexpres-
reaction. J Invest Dermatol 97:980–984 sion of normal collagen VII. J Biol Chem
2. Bornert O, Kühl T, Bremer J et al (2016) 284:30248–30256. https://doi.org/10.
Analysis of the functional consequences of tar- 1074/jbc.M109.045294
geted exon deletion in COL7A1 reveals pro- 8. Gibson DG (2011) Enzymatic assembly of
spects for dystrophic epidermolysis bullosa overlapping DNA fragments. Methods Enzy-
therapy. Mol Ther J Am Soc Gene Ther mol 498:349–361. https://doi.org/10.1016/
24:1302–1311. https://doi.org/10.1038/ B978-0-12-385120-8.00015-2
mt.2016.92 9. Gibson DG, Young L, Chuang R-Y et al (2009)
3. Chmel N, Bornert O, Hausser I et al (2018) Enzymatic assembly of DNA molecules up to
Large deletions targeting the triple-helical several hundred kilobases. Nat Methods
domain of collagen VII Lead to mild Acral 6:343–345. https://doi.org/10.1038/
dominant dystrophic Epidermolysis Bullosa. J nmeth.1318
Invest Dermatol 138:987–991. https://doi. 10. Ricard-Blum S (2011) The collagen family.
org/10.1016/j.jid.2017.11.014 Cold Spring Harb Perspect Biol 3:a004978.
4. Nyström A, Bornert O, Kühl T et al (2018) https://doi.org/10.1101/cshperspect.
Impaired lymphoid extracellular matrix a004978
impedes antibacterial immunity in epidermoly- 11. Varki R, Sadowski S, Uitto J, Pfendner E
sis bullosa. Proc Natl Acad Sci U S A 115: (2007) Epidermolysis bullosa II. Type VII col-
E705–E714. https://doi.org/10.1073/pnas. lagen mutations and phenotype-genotype cor-
1709111115 relations in the dystrophic subtypes. J Med
5. Chen M, Costa FK, Lindvay CR et al (2002) Genet 44:181–192. https://doi.org/10.
The recombinant expression of full-length type 1136/jmg.2006.045302
VII collagen and characterization of molecular 12. Chen M, Keene DR, Costa FK et al (2001) The
mechanisms underlying dystrophic epidermo- carboxyl terminus of type VII collagen med-
lysis bullosa. J Biol Chem 277:2118–2124. iates antiparallel dimer formation and constitu-
https://doi.org/10.1074/jbc.M108779200 tes a new antigenic epitope for epidermolysis
6. Nyström A, Bruckner-Tuderman L, Kern JS Bullosa acquisita autoantibodies. J Biol Chem
(2013) Cell- and protein-based therapy 276:21649–21655. https://doi.org/10.
approaches for epidermolysis bullosa. Methods 1074/jbc.M100180200
Mol Biol Clifton NJ 961:425–440. https:// 13. Rattenholl A, Pappano WN, Koch M et al
doi.org/10.1007/978-1-62703-227-8_29 (2002) Proteinases of the bone morphogenetic
Cloning and Mutagenesis Strategies for Large Collagens 15
Abstract
Collagen is the most widely used substratum in cell culture and biomaterials applications. In this chapter, we
describe a simple procedure to isolate collagen, which can be employed to a wide range of tissue sources,
and subsequently use it to study the collagen crosslinking and stabilization abilities of various compounds.
The protocol is designed for a multi-well format assay and thus can be used for simultaneous assessment of
multiple number of compounds and can be easily adapted to a high-throughput screening setup.
Key words Collagen, Crosslinking, Extracellular matrix, Fibrillation, Quarter staggered structure
1 Introduction
Collagen is the most abundant protein in the human body and plays
a vital role in several physiological and pathological conditions
[1–3]. It is used in several biomedical applications and holds a
central role in treating a myriad of abnormalities such as abnormal
wound healing, cancer, etc. [4]. Because of its wide presence and
diverse functions, isolation and characterization of collagen are
important aspects in biomaterials preparation and understanding
the molecular mechanisms of various diseases. One of the
intriguing properties of the isolated collagen fibrils, in vitro, is its
ability to arrange into an quarter-staggered structure in physiologi-
cal conditions resembling the in vivo fibril formation [5]. This
property can be utilized to comprehend the influence of molecules
on the three-dimensional spatial organization of collagens, which
are crucial in dictating the cellular behavior.
Irit Sagi and Nikolaos A. Afratis (eds.), Collagen: Methods and Protocols, Methods in Molecular Biology, vol. 1944,
https://doi.org/10.1007/978-1-4939-9095-5_2, © Springer Science+Business Media, LLC, part of Springer Nature 2019
17
18 Venkat Raghavan Krishnaswamy
2 Materials
2.1 Collagen This protocol can be employed to isolate collagen from a variety of
Isolation sources including skin and tendon. The most commonly used
source is rat tail tendon which is almost entirely made up of colla-
gen type I. The isolation of tendon from rat tail is explained by
Rajan et al. [6].
1. Wash buffer 1: Mix three volumes of methanol with one vol-
ume of chloroform. Store the solution in an airtight bottle at
4 C.
2. Wash buffer 2: Prepare 0.5 M sodium acetate by dissolving
41.02 g of sodium acetate in 1 L of deionized water.
3. Extraction reagent: Make up 28.73 mL of pure glacial acetic
acid to 1 L with deionized water to prepare 0.5 M acetic acid.
Store at 4 C.
4. Precipitating solution: To prepare 2.8 M sodium chloride,
dissolve 163.6 g of sodium chloride in 800 mL, and make up
the volume to 1 L using extraction reagent, store at 4 C.
5. Dialysate: Make up 1.14 mL of pure glacial acetic acid to 1 L
with deionized water to prepare 0.02 M acetic acid. Store
at 4 C.
3 Methods
3.1 Collagen 1. After harvesting the tissue, remove the unwanted tissues like
Isolation soft tissue, clotted blood, etc. that are present. Wash the sam-
ples twice thoroughly with cold deionized water.
2. Wash the tissue samples with wash buffer 1. Repeat two more
times, and ensure all the fat particles are removed.
3. Wash the samples once with wash buffer 2.
4. Wash the samples extensively with extraction reagent, and
incubate the tissue in at least 10 volumes of extraction solution
for 48 to 72 h (see Note 1).
5. Homogenize the samples in a blender with extraction reagent.
Centrifuge the samples at 14,000 g for 30 min at 4 C. Save
the supernatant, and re-extract the collagen once from the
pellet by repeating the homogenization. Pool both the super-
natants (see Note 2).
6. For 750 mL of collagen supernatant, gradually add 250 mL of
precipitating solution with mild stirring overnight at 4 C.
7. Centrifuge the solution at 6000 g for 10 min at 4 C. Dis-
card the supernatant, and collect the pellet.
8. Dissolve the pellet in a suitable volume of cold extraction
reagent.
9. Dialyze the collagen solution extensively against dialysate at
least for 24–48 h (see Note 3).
10. Freeze the samples as a thin layer on a lyophilized flask by
rotating the flask with the sample on an ice-salt slurry. Freeze
the collagen coated flasks at 80 C for a minimum of
12–16 h.
11. Freeze dry the samples thoroughly. Store the samples at
80 C in an airtight container.
12. Dissolve a portion of the lyophilized sample in the dialysate,
and estimate the collagen content using standard procedures.
The purity of the collagen can be assessed by a standard sodium
dodecyl sulfate-polyacrylamide gel electrophoresis (see Note 4)
(Fig. 1).
4 Notes
References
1. Prockop DJ, Kivirikko KI (1995) Collagens: new target to limit fibrosis. J Biol Chem 283
molecular biology, diseases, and potentials for (38):25879–25886. https://doi.org/10.1074/
therapy. Annu Rev Biochem 64:403–434. jbc.M804272200
https://doi.org/10.1146/annurev.bi.64. 6. Rajan N, Habermehl J, Cote MF, Doillon CJ,
070195.002155 Mantovani D (2006) Preparation of ready-to-
2. Mienaltowski MJ, Birk DE (2014) Structure, use, storable and reconstituted type I collagen
physiology, and biochemistry of collagens. Adv from rat tail tendon for tissue engineering appli-
Exp Med Biol 802:5–29. https://doi.org/10. cations. Nat Protoc 1(6):2753–2758. https://
1007/978-94-007-7893-1_2 doi.org/10.1038/nprot.2006.430
3. Myllyharju J, Kivirikko KI (2001) Collagens and 7. Krishnaswamy VR, Lakra R, Korrapati PS
collagen-related diseases. Ann Med 33(1):7–21 (2014) Keloid collagen–cell interactions: struc-
4. Lee CH, Singla A, Lee Y (2001) Biomedical tural and functional perspective. RSC Adv 4
applications of collagen. Int J Pharm 221 (45):23642–23648. https://doi.org/10.1039/
(1–2):1–22 C4RA01995D
5. Chung HJ, Steplewski A, Chung KY, Uitto J,
Fertala A (2008) Collagen fibril formation. A
Chapter 3
Abstract
Chemical cross-linking of collagen-based devices is used as a means of increasing the mechanical stability
and control the degradation rate upon implantation. Herein, we describe techniques to produce cross-
linked with glutaraldehyde (GTA; amine terminal cross-linker), 4-arm polyethylene glycol succinimidyl
glutarate (4SP; amine terminal cross-linker), diphenyl phosphoryl azide (DPPA; carboxyl terminal cross-
linker), and 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC; carboxyl terminal cross-linker) colla-
gen films. In addition, we provide protocols to characterize the biophysical (swelling), biomechanical
(tensile), and biological (metabolic activity, proliferation and viability using human dermal fibroblasts and
THP-1 macrophages) properties of the cross-linked collagen scaffolds.
1 Introduction
Irit Sagi and Nikolaos A. Afratis (eds.), Collagen: Methods and Protocols, Methods in Molecular Biology, vol. 1944,
https://doi.org/10.1007/978-1-4939-9095-5_3, © Springer Science+Business Media, LLC, part of Springer Nature 2019
23
24 Ignacio Sallent et al.
2 Materials
3. 4% paraformaldehyde (PFA).
4. 1 Phosphate-buffered saline (PBS).
5. Triton-X 100 solution in 0.2% PBS.
3 Methods
3.1 Collagen Cross- All steps detailed in this paragraph need to be performed on ice to
Linking and Film avoid denaturation of collagen and prevent its premature gelation.
Preparation The temperature of the centrifuge, if possible, should be set to 4 C
before starting the procedure. The following instructions describe
the production of a film of 3 4 cm dimensions, for other mold
dimensions the volume of the reagents need to be scaled up.
1. Mix 0.42 ml of PBS 10 with 0.2 ml of 1 M NaOH in a 15 ml
screw-cap tube using a benchtop vortex.
2. Add 4.2 ml of the collagen stock solution to the mixture, and
mix thoroughly with the vortex.
3. Use the pH meter and 1 M NaOH to neutralize the solution
(pH 7.2–7.4).
4. Add the 0.25 ml of the chosen cross-linker, and mix with the
benchtop vortex (see Note 2).
5. Centrifuge the solution at 2000 rpm for 10 s to remove the
bubbles.
6. Pour 4 ml of the solution on the nonadherent mold (3 4 cm).
7. Incubate the sample (mold and collagen) at 37 C for 1 h.
8. Dry the sample in the flow hood at room temperature (RT) for
at least 24 h.
9. Use tweezers to peel off the collagen film from the nonadher-
ent surface.
10. Store the films in a dark and dry environment until use.
3.2 Swelling Assay 1. Use a blade and a scalpel to cut five pieces (n ¼ 5) from the
collagen film, and record their dry weight using a laboratory
scale (see Note 3).
Chemically Cross-Linked Collagen Scaffolds 27
3.3 Tensile Test 1. Use a scalpel with a sharp blade and a ruler to cut the collagen
film in three stripes of 1 4 cm (see Note 4).
2. Hydrate the samples overnight in PBS 1 at 37 C (see
Note 5).
3. Blot the samples with filter paper to remove the excess surface
water.
4. Using the calipers, take five measurements of the thickness of
the sample in the middle region (see Note 6).
5. Insert the sample into the upper grip of the tensile tester, and
tighten (see Note 7).
6. Bring the crosshead into an appropriate range for the sample
and affix the sample to the lower grip (see Note 8).
7. Set up the software from the mechanical tester to the following
tensile test parameters: Preload, 0.01 N; preload speed, 5 mm/
min; test speed, 10 mm/min.
8. Initiate the test, and export the results to excel.
9. Calculate the following parameters: stress at break (MPa),
strain at break (%) and Young’s modulus (MPa), using the
width and the thickness of the samples. Stress at break ¼ force
at break/original cross-sectional area of the sample, Young’s
modulus ¼ slope of the linear region of the stress (MPa)/strain
(no units) curve (see Note 8) (Table 1).
Table 1
Swelling ratio, thickness, and mechanical data of the produced collagen films
Cross-linking Swelling ratio Thickness Stress at break Strain at break Young’s modulus
agent (%) (μm) (MPa) (%) (MPa)
NC 100 16 178 4 0.03 0.01 8.33 3.00 0.20 0.04
GTA 21 9 # 67 6 # 0.19 0.04 # 4.19 2.82 1.95 0.33 #
4SP 80 9 232 33 0.07 0.03 # 13.28 3.13 0.38 0.04
EDC 125 7 185 39 0.03 0.01 15.78 3.08 0.10 0.03
DPPA 91 23 83 12 # 0.05 0.01 18.22 1.89 # 0.22 0.01
GTA significantly decreased water absorption. GTA and DPPA significantly reduced thickness. GTA induced the highest
stress at break. DPPA induced the highest strain at break. GTA induced the highest modulus. # indicates statistically
significant difference ( p < 0.05)
28 Ignacio Sallent et al.
Fig. 1 Metabolic activity (a), proliferation (b), viability (c), and rhodamine phalloidin (red) and DAPI (blue)
images (d) of DF seeded onto the collagen scaffolds. DF on GTA cross-linked scaffolds presented reduced
Chemically Cross-Linked Collagen Scaffolds 29
3.4 Cell Culture 1. Using a scalpel with a blade cut the collagen films into 1 1 cm
squares.
3.4.1 Scaffold
Preparation 2. In the laminar flow hood, place the films in 24-well plates (see
Note 9).
3. Use the UV from the laminar flow hood for 1 h to partially
sterilize the collagen scaffolds in the plates. Keep the lid of the
plates open during the process.
4. Fix the collagen films to the bottom of the plates with the help
of autoclaved tweezers and silicone rings.
5. Remove the excess of cross-linker by washing the films three
times with sterile PBS 1 for 20 min.
6. Sterilize the films with 0.5 ml of 70% ethanol for 30 min.
7. Wash the films three times in sterile PBS 1.
3.4.3 THP-1 Seeding 1. Incubate the sterilized films at 37 C and 5% CO2 with 1 ml
cell-specific media for THP-1 cells for 3 days.
2. Collect the RPMI supplemented media incubated with the
scaffolds (preconditioned media), and filter it through 0.2 μm
sterile filters. Keep it at 37 C in the water bath until
further use.
3. Seed THP-1 into new 24-well plates and onto the scaffolds at
26 103 cells/cm2 with THP-1 complete media supplemen-
ted with 100 ng/ml of PMA to induce adherent mature
macrophage-like state for 6 h.
4. Wash off nonattached cells with sterile PBS 1.
5. Add 0.5 ml of the filtered preconditioned media to the macro-
phages attached to the 24-well plates TCP and 0.5 ml of fresh
RPMI supplemented media to the macrophages attached to
the scaffolds. Create a positive control for macrophage
ä
Fig. 1 (continued) metabolic activity at day 3 and 7 and reduced proliferation at all three time points in
comparison to the NC samples. None of the cross-linking treatments significantly affected DF viability (c). DF
adopted a spindle-shaped morphology in all the groups except for GTA, on which cells spread in all directions
(d). Treatments: non-cross-linked collagen films (NC), collagen films cross-linked with glutaraldehyde (GTA),
4-arm PEG succinimidyl glutarate (4SP), 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC), and diphe-
nylphosphoryl azide (DPPA). Tissue culture plastic (TCP) was used as control. Significance compared to *NC
group ( p < 0.05)
30 Ignacio Sallent et al.
Fig. 2 Metabolic activity (a), proliferation (b), viability (c), elongated cells proportion (d), and rhodamine
phalloidin (red) and DAPI (blue) images (e) of THP-1 seeded onto the collagen scaffolds. By day 1, LPS
significantly increased THP-1 metabolic activity (a); and 4SP significantly reduced THP-1 proliferation (b) in
Chemically Cross-Linked Collagen Scaffolds 31
3.5 Cell Proliferation 1. At the appropriate time point remove media from cells.
and Morphology Assay 2. Gently wash scaffolds with PBS 1.
3. Add 0.5 ml of 4% PFA, and allow the sample to fix for 30 min at
RT (see Note 10).
4. Wash the sample with 1 PBS.
5. Incubate the samples with 0.2 ml of 0.2% Triton-X solution for
5 min.
6. Wash the sample with PBS 1.
7. Add rhodamine-phalloidin solution enough to cover the bot-
tom of the well and/or scaffolds (around 150 μl).
8. Incubate 1 h at RT. Protect the samples from the light during
this procedure and in subsequent manipulations.
9. Wash the sample with PBS 1.
10. Add 150 μl of DAPI solution, and incubate for 5 min at RT.
11. Wash the sample three times with PBS 1 (see Note 11).
12. Image the samples in a fluorescence or confocal microscope
using a 10 objective and an excitation/emission wavelength
of 358/461 nm for DAPI and 540/565 nm for rhodamine
phalloidin.
13. Take 4–5 representative images of every sample.
14. Use ImageJ or other software for an automated counting of the
nuclei per image (see Note 12) and for morphology assessment
(see Note 13).
15. Graph the number of cells per area to evaluate cell
proliferation.
Fig. 2 (continued) comparison to the NC samples. By day 2, GTA, EDC, and 4SP significantly reduced THP-1
metabolic activity (a); and all the cross-linking treatments significantly reduced THP-1 proliferation (b) in
comparison to the NC. None of the cross-linking treatments significantly affected macrophage viability (c). All
treatments elicited a significant lower proportion of elongated cells than LPS, whereas THP-1 in GTA, EDC and
DPPA presented a significant higher number of elongated cells than TCP at both time points, likewise NC at
day 2 (d). THP-1 adopted a rounded morphology, independently of the treatment and time in culture. Some
elongated macrophages (white arrows) were observed predominantly in LPS- and GTA-treated samples,
whereas cell clusters (black arrows) were mainly observed in LPS condition (e). Treatments: non-cross-linked
collagen films (NC), collagen films cross-linked with glutaraldehyde (GTA), 4-arm PEG succinimidyl glutarate
(4SP), 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC), and diphenylphosphoryl azide (DPPA). Tissue
culture plastic (TCP) and lipopolysaccharides in TCP (LPS) were used as controls. Significance compared to
*NC group, compared to #TCP and compared to +LPS group ( p < 0.05)
32 Ignacio Sallent et al.
Fig. 3 Metabolic activity (a), proliferation (b), viability (c), elongated cells proportion (d), and rhodamine
phalloidin (red) and DAPI (blue) images (e) of THP-1 cultured with preconditioned media from the cross-linked
Chemically Cross-Linked Collagen Scaffolds 33
3.6 Cell Metabolic 1. Cell metabolic activity can be assessed by the incubation of the
Activity Assay cells with 10% alamarBlue® for 2 h on the day of the time
(alamarBlue®) point, as per manufacturer’s protocol (see Note 14).
2. Cell metabolic activity is expressed as % of reduced alamar-
Blue® and normalized to the activity of cells in TCP.
3.7 Cell Viability 1. Prepare staining solution by diluting 1:1000 the calcein AM
Assay and the ethidium homodimer-1 stock solutions with PBS 1.
2. Add 150 μl of DMSO to a well with cells alone (no scaffold) to
create a negative control with dead cells. Place the plate in the
incubator for 20 min.
3. Remove culture medium from the wells, and wash cells
with PBS.
4. Add 150 μl of staining solution to each scaffold.
5. Incubate at 37 C, 5% CO2 for 30 min.
6. Wash the sample with PBS 1 (see Note 11).
7. Image the samples in a fluorescence or confocal microscope
using a 10 objective and an excitation/emission wavelength
of 495/515 nm and 528/617 nm for Calcein AM and ethi-
dium homodimer-1, respectively.
8. Take 3–5 images of every sample.
9. Count viable (green) and dead (red) cells with ImageJ or
another software.
10. Calculate cell viability (%) by dividing the number of alive cells
by the total number of cells.
Fig. 3 (continued) collagen films. None of the cross-linking treatments significantly affected THP-1 metabolic
activity (a). Only cells treated with LPS showed a decreased proliferation in comparison with the NC samples
(b). None of the cross-linking treatments significantly affected macrophage viability (c). LPS showed a higher
number of elongated cells than TCP at both times points. All treatments showed values higher than TCP and
lower than LPS. Significant differences were found between LPS and TCP at both time points, whereas only
4SP showed a significant difference with LPS at day 1 (d). THP-1 adopted a rounded morphology, indepen-
dently of the treatment and time in culture. Some elongated macrophages were observed (white arrows)
predominantly in LPS-, NC-, and GTA-treated samples. Some cell clusters were observed (black arrows) in all
the conditions except for TCP (e). Treatments: non-cross-linked collagen films (NC), collagen films cross-
linked with glutaraldehyde (GTA), 4-arm PEG succinimidyl glutarate (4SP), 1-ethyl-3-(3-dimethylaminopropyl)
carbodiimide (EDC), and diphenylphosphoryl azide (DPPA). Tissue culture plastic (TCP) and lipopolysacchar-
ides (LPS) in TCP were used as controls. Significance compared to *NC group, compared to #TCP and
compared to +LPS group ( p < 0.05)
34 Ignacio Sallent et al.
4 Notes
Acknowledgments
References
1. Gelse K, Poschl E, Aigner T (2003) Col- 14. Zeugolis DI, Paul GR, Attenburrow G (2009)
lagens–structure, function, and biosynthesis. Cross-linking of extruded collagen fibers -- a
Adv Drug Deliv Rev 55(12):1531–1546 biomimetic three-dimensional scaffold for tis-
2. Brodsky B, Ramshaw JA (1997) The collagen sue engineering applications. J Biomed Mater
triple-helix structure. Matrix Biol 15 Res A 89(4):895–908. https://doi.org/10.
(8–9):545–554 1002/jbm.a.32031
3. Smith-Mungo LI, Kagan HM (1998) Lysyl 15. Weadock K, Olson RM, Silver FH (1983) Eval-
oxidase: properties, regulation and multiple uation of collagen crosslinking techniques.
functions in biology. Matrix Biol 16 Biomater Med Devices Artif Organs 11
(7):387–398 (4):293–318
4. Yamauchi M, Taga Y, Hattori S, Shiiba M, Ter- 16. Jorge-Herrero E, Fernandez P, Turnay J,
ajima M (2018) Analysis of collagen and elastin Olmo N, Calero P, Garcia R, Freile I,
cross-links. Methods Cell Biol 143:115–132. Castillo-Olivares JL (1999) Influence of differ-
https://doi.org/10.1016/bs.mcb.2017.08. ent chemical cross-linking treatments on the
006 properties of bovine pericardium and collagen.
5. Fu MX, Wells-Knecht KJ, Blackledge JA, Biomaterials 20(6):539–545
Lyons TJ, Thorpe SR, Baynes JW (1994) Gly- 17. van Wachem PB, Zeeman R, Dijkstra PJ,
cation, glycoxidation, and cross-linking of col- Feijen J, Hendriks M, Cahalan PT, van Luyn
lagen by glucose. Kinetics, mechanisms, and MJ (1999) Characterization and biocompati-
inhibition of late stages of the Maillard reac- bility of epoxy-crosslinked dermal sheep col-
tion. Diabetes 43(5):676–683 lagens. J Biomed Mater Res 47(2):270–277
6. Reddy GK (2004) Cross-linking in collagen by 18. Haugh MG, Jaasma MJ, O’Brien FJ (2009)
nonenzymatic glycation increases the matrix The effect of dehydrothermal treatment on
stiffness in rabbit achilles tendon. Exp Diabe- the mechanical and structural properties of
sity Res 5(2):143–153. https://doi.org/10. collagen-GAG scaffolds. J Biomed Mater Res
1080/15438600490277860 A 89(2):363–369. https://doi.org/10.1002/
7. Greenberg CS, Birckbichler PJ, Rice RH jbm.a.31955
(1991) Transglutaminases: multifunctional 19. Wess TJ, Orgel JP (2000) Changes in collagen
cross-linking enzymes that stabilize tissues. structure: drying, dehydrothermal treatment
FASEB J 5(15):3071–3077 and relation to long term deterioration. Ther-
8. Bailey AJ, Light ND, Atkins ED (1980) Chem- mochim Acta 365(1-2):119–128. https://doi.
ical cross-linking restrictions on models for the org/10.1016/S0040-6031(00)00619-5
molecular organization of the collagen fibre. 20. Weadock KS, Miller EJ, Bellincampi LD,
Nature 288(5789):408–410 Zawadsky JP, Dunn MG (1995) Physical cross-
9. Orgel JP, Irving TC, Miller A, Wess TJ (2006) linking of collagen fibers: comparison of ultra-
Microfibrillar structure of type I collagen in violet irradiation and dehydrothermal
situ. Proc Natl Acad Sci U S A 103 treatment. J Biomed Mater Res 29
(24):9001–9005. https://doi.org/10.1073/ (11):1373–1379. https://doi.org/10.1002/
pnas.0502718103 jbm.820291108
10. Glowacki J, Mizuno S (2008) Collagen scaf- 21. Torres DS, Freyman TM, Yannas IV, Spector M
folds for tissue engineering. Biopolymers 89 (2000) Tendon cell contraction of collagen-
(5):338–344. https://doi.org/10.1002/bip. GAG matrices in vitro: effect of cross-linking.
20871 Biomaterials 21(15):1607–1619
11. Cliche S, Amiot J, Avezard C, Gariepy C 22. Chen RN, Ho HO, Sheu MT (2005) Charac-
(2003) Extraction and characterization of col- terization of collagen matrices crosslinked
lagen with or without telopeptides from using microbial transglutaminase. Biomaterials
chicken skin. Poult Sci 82(3):503–509. 26(20):4229–4235. https://doi.org/10.
https://doi.org/10.1093/ps/82.3.503 1016/j.biomaterials.2004.11.012
12. Friess W (1998) Collagen-biomaterial for drug 23. Stachel I, Schwarzenbolz U, Henle T, Meyer M
delivery. Eur J Pharm Biopharm 45 (2010) Cross-linking of type I collagen with
(2):113–136 microbial transglutaminase: identification of
cross-linking sites. Biomacromolecules 11
13. Charulatha V, Rajaram A (2003) Influence of (3):698–705. https://doi.org/10.1021/
different crosslinking treatments on the physi- bm901284x
cal properties of collagen membranes. Bioma-
terials 24(5):759–767
Chemically Cross-Linked Collagen Scaffolds 37
24. Zeugolis DI, Panengad PP, Yew ES, Biomater 9(7):7191–7199. https://doi.org/
Sheppard C, Phan TT, Raghunath M (2010) 10.1016/j.actbio.2013.02.021
An in situ and in vitro investigation for the 33. Orenstein SB, Qiao Y, Klueh U, Kreutzer DL,
transglutaminase potential in tissue engineer- Novitsky YW (2010) Activation of human
ing. J Biomed Mater Res A 92(4):1310–1320. mononuclear cells by porcine biologic meshes
https://doi.org/10.1002/jbm.a.32383 in vitro. Hernia 14(4):401–407. https://doi.
25. Orban JM, Wilson LB, Kofroth JA, El-Kurdi org/10.1007/s10029-010-0634-7
MS, Maul TM, Vorp DA (2004) Crosslinking 34. Witherel CE, Graney PL, Freytes DO, Wein-
of collagen gels by transglutaminase. J Biomed garten MS, Spiller KL (2016) Response of
Mater Res A 68(4):756–762. https://doi.org/ human macrophages to wound matrices
10.1002/jbm.a.20110 in vitro. Wound Repair Regen 24
26. Moshnikova AB, Afanasyev VN, Proussakova (3):514–524. https://doi.org/10.1111/wrr.
OV, Chernyshov S, Gogvadze V, Beletsky IP 12423
(2006) Cytotoxic activity of 1-ethyl-3- 35. Delgado LM, Fuller K, Zeugolis DI (2017)
(3-dimethylaminopropyl)-carbodiimide is Collagen cross-linking: biophysical, biochemi-
underlain by DNA interchain cross-linking. cal, and biological response analysis. Tissue
Cell Mol Life Sci 63(2):229–234. https://doi. Eng Part A 23(19–20):1064–1077. https://
org/10.1007/s00018-005-5383-x doi.org/10.1089/ten.TEA.2016.0415
27. Speer DP, Chvapil M, Eskelson CD, Ulreich J 36. Lotz C, Schmid FF, Oechsle E, Monaghan
(1980) Biological effects of residual glutaralde- MG, Walles H, Groeber-Becker F (2017)
hyde in glutaraldehyde-tanned collagen bioma- Cross-linked collagen hydrogel matrix resisting
terials. J Biomed Mater Res 14(6):753–764. contraction to facilitate full-thickness skin
https://doi.org/10.1002/Jbm.820140607 equivalents. ACS Appl Mater Interfaces 9
28. Gough JE, Scotchford CA, Downes S (2002) (24):20417–20425. https://doi.org/10.
Cytotoxicity of glutaraldehyde crosslinked col- 1021/acsami.7b04017
lagen/poly(vinyl alcohol) films is by the mech- 37. Monaghan M, Browne S, Schenke-Layland K,
anism of apoptosis. J Biomed Mater Res 61 Pandit A (2014) A collagen-based scaffold
(1):121–130. https://doi.org/10.1002/jbm. delivering exogenous microRNA-29B to mod-
10145 ulate extracellular matrix remodeling. Mol
29. Hass V, Luque-Martinez IV, Gutierrez MF, Ther 22(4):786–796. https://doi.org/10.
Moreira CG, Gotti VB, Feitosa VP, Koller G, 1038/mt.2013.288
Otuki MF, Loguercio AD, Reis A (2016) Col- 38. Sanami M, Sweeney I, Shtein Z, Meirovich S,
lagen cross-linkers on dentin bonding: stability Sorushanova A, Mullen A, Miraftab M,
of the adhesive interfaces, degree of conversion Shoseyov O, O’Dowd C, Pandit A, Zeugolis
of the adhesive, cytotoxicity and in situ MMP D (2016) The influence of poly(ethylene gly-
inhibition. Dent Mater 32(6):732–741. col) ether tetrasuccinimidyl glutarate on the
https://doi.org/10.1016/j.dental.2016.03. structural, physical, and biological properties
008 of collagen fibers. J Biomed Mater Res B 104
30. Delgado LM, Bayon Y, Pandit A, Zeugolis DI (5):914–922
(2015) To cross-link or not to cross-link? 39. Marinucci L, Lilli C, Guerra M, Belcastro S,
Cross-linking associated foreign body response Becchetti E, Stabellini G, Calvi EM, Locci P
of collagen-based devices. Tissue Eng Part B (2003) Biocompatibility of collagen mem-
21(3):298–313. https://doi.org/10.1089/ branes crosslinked with glutaraldehyde or
ten.TEB.2014.0290 diphenylphosphoryl azide: an in vitro study. J
31. Brown BN, Londono R, Tottey S, Zhang L, Biomed Mater Res A 67A(2):504–509.
Kukla KA, Wolf MT, Daly KA, Reing JE, Bady- https://doi.org/10.1002/jbm.a.10082
lak SF (2012) Macrophage phenotype as a pre- 40. Petite H, Frei V, Huc A, Herbage D (1994)
dictor of constructive remodeling following Use of diphenylphosphorylazide for cross-
the implantation of biologically derived surgi- linking collagen-based biomaterials. J Biomed
cal mesh materials. Acta Biomater 8 Mater Res 28(2):159–165. https://doi.org/
(3):978–987. https://doi.org/10.1016/j. 10.1002/Jbm.820280204
actbio.2011.11.031 41. Capella-Monsonı́s H, Coentro J, Graceffa V,
32. McDade JK, Brennan-Pierce EP, Ariganello Wu Z, Zeugolis D (2018) An experimental
MB, Labow RS, Michael Lee J (2013) Interac- toolbox for characterization of mammalian col-
tions of U937 macrophage-like cells with lagen type I in biological specimens. Nat Pro-
decellularized pericardial matrix materials: toc 13(3):507–529
influence of crosslinking treatment. Acta
38 Ignacio Sallent et al.
Abstract
The bone and dentin mainly consist of type-I collagen fibrils mineralized by hydroxyapatite (HAP)
nanocrystals. In vitro biomimetic models based on self-assembled collagen fibrils have been widely used
in studying the mineralization mechanism of type-I collagen. In this chapter, the protocol we used to build a
biomimetic model for the mechanistic study of type-I collagen mineralization is described. Type-I collagen
extracted from rat tail tendon or horse tendon is self-assembled into fibrils and mineralized by HAP in vitro.
The mineralization process is monitored by cryoTEM in combination with two-dimensional (2D) and
three-dimensional (3D) stochastic optical reconstruction microscopy (STORM), which enables in situ and
high-resolution visualization of the process.
1 Introduction
Irit Sagi and Nikolaos A. Afratis (eds.), Collagen: Methods and Protocols, Methods in Molecular Biology, vol. 1944,
https://doi.org/10.1007/978-1-4939-9095-5_4, © Springer Science+Business Media, LLC, part of Springer Nature 2019
39
40 Shasha Yao et al.
2 Materials
3 Methods
Fig. 1 Photos showing the effect of mixing speed on preparing the mineralization
solution. (a) The colorless and transparent mineralization solution obtained by
fast mixing. (b) The light blue-colored, cloudy dispersion obtained by slow mixing
Biomimetic Model for Mineralization of Type-I Collagen 43
3.1.3 TEM Imaging The assembled (Fig. 2), stained (Fig. 3), and mineralized (Figs. 4
and Mineralization Degree and 5) type-I collagen fibrils were visualized by TEM operating at
Calculation 100 kV or 200 kV. The mineralization degree increased with
increasing the pAsp concentration from 0 to 240 μg ml1 (Fig. 4)
and increasing the mineralization time from 6 h to 24 h (Fig. 5).
The mineralization degree (m.d.) was calculated by first
splitting the pixel intensity histogram of the TEM images into
three Gaussian distributions (Fig. 6), which correspond to the
Fig. 3 Conventional TEM image of the assembled type-I collagen fibrils after
uranyl acetate positive staining, displaying the ~67 nm periodical structures
together with fine band structures. Reproduced with permission [14]. Copyright
2018, Wiley-VCH
44 Shasha Yao et al.
Fig. 4 Conventional TEM images of the mineralized collagen-I fibrils in the presence of 0 (a), 10 (b), 15 (c),
50 (d), 120 (e), and 240 (f) μg ml1 pAsp. Insets: selected area electron diffraction (SAED) patterns of the
samples, which match with the HAP diffractions and indicate the oriented crystallization. Reproduced with
permission [14]. Copyright 2018, Wiley-VCH
Fig. 5 Conventional TEM images of the collagen-I fibrils mineralized at 6 h (a) and 24 h (b). Insets: SAED
patterns indicating HAP mineralization. Reproduced with permission [14]. Copyright 2018, Wiley-VCH
Fig. 6 Mineralization degree calculation. (a) Segmentation thresholds determined by analyzing the pixel
intensity histogram of the TEM image and splitting the histogram into three Gaussian distributions. Determi-
nation of the regions of the mineralized collagen-I fibrils (b), unmineralized collagen-I fibrils (c), and the
background (d). Reproduced with permission [14]. Copyright 2018, Wiley-VCH
46 Shasha Yao et al.
part of the collagen fibrils (S2) could then be segmented from the
image, and m.d. could then be given by comparing S1 and S2
S1
m:d: ¼
S2
3.2 CryoTEM 1. 3 mg ml1 of type-I collagen stock solution from rat tail was
Experiments mixed with the assembling buffer for cryoTEM study in a 1:29
ratio. 300 μl of as-prepared incubation solution was applied on
3.2.1 Self-Assembly
a parafilm.
of Type-I Collagen
Extracted from Rat Tail 2. Gold Quantifoil grids were floated upside down on the incuba-
Tendon tion solution droplet (Fig. 7). The parafilm was then covered
by a glass petri dish and incubated for 200 min.
3. After incubation, the grids were transferred to the Vitrobot
(FEI, Mark III) in 100% humidity (see Note 6), blotted for 5 s
by filter papers, and then vitrified in liquid ethane to visualize
the collagen fibrils or transferred to the mineralization solution
(see Subheading 3.2.3).
3.2.2 Self-Assembly 1. 1 g of type-I collagen extracted from horse tendon was mixed
of Type-I Collagen overnight with 10 ml of 50 mM aqueous acetic acid, pH 2.5,
Extracted from Horse centrifuged at 3500 g for 10 min, and the supernatant was
Tendon collected and stored at 4 C.
2. CryoTEM grids were laid on a 15 μl drop of collagen solution
for 10 s. The excess of collagen solution was manually blotted,
and the grids were transferred to a 15 μl drop of Hepes buffer,
for 30 min. This procedure was performed inside a glove box,
where temperature and humidity are controlled at 22 C and
100% relative humidity.
3. The grids were then vitrified as described in Subheading 3.2.1
or transferred to the mineralization solution (see Subheading
3.2.3).
Fig. 7 Photo of the gold Quantifoil TEM grids floating on a droplet of buffer
solution applied on a parafilm
Biomimetic Model for Mineralization of Type-I Collagen 47
Fig. 8 CryoTEM image of assembled type-I collagen fibrils. (a) Rat tail type-I collagen fibrils, which are
60–80 nm thick. (b) Zoom-in image of the fibrils showing no periodical structure. (c) A horse tendon type-I
collagen fibril, which is ~300 nm thick and clearly shows the ~67 nm periodical structure. The 10 nm particles
highlighted by yellow circle are gold fiducial markers used for tomography
3.2.4 CryoTEM Imaging 1. CryoTEM imaging of the assembled (Fig. 8 and see Note 7)
and then mineralized (Fig. 9) collagen was performed under
~3 μm defocus on a FEI Titan cryoTEM equipped with a field
emission gun (FEG) and operating at 300 kV or a FEI Tecnai
20 (Type Sphera) TEM equipped with a LaB6 filament
operating at 200 kV and a Gatan cryo-holder operating at
170 C.
2. Images were recorded in FEI Titan using a 2 k 2 k Gatan
CCD camera equipped with a post-column Gatan energy filter
(GIF) or in FEI Tecnai using a 1 k 1 k Gatan CCD camera.
The electron dose used is 10 e Å2 per image (see Note 8).
48 Shasha Yao et al.
Fig. 9 CryoTEM image of mineralized type-I collagen fibrils. (a) Image showing a mineralized rat tail type-I
collagen fibril, displaying stacks of HAP platelets. (b) SAED pattern of the mineralized rat tail collagen fibril,
which shows a narrow but very weak HAP (002) diffraction arc in the collagen fibril axis direction as
highlighted by the yellow arrow. (c) Image showing a mineralized horse tendon type-I collagen fibril. The
fibril is completely impregnated with HAP platelets, and therefore the ~67 nm periodical structures are not
distinguishable. (d) SAED pattern of the mineralized horse tendon type-I collagen fibril, which clearly shows
the HAP (002) diffraction arc in the collagen axis as highlighted by the yellow arrow. The collagen fibril axis
directions in (c) and (d) are highlighted by the blue arrows
3.3 STORM 1. Type-I collagen fibrils (rat tail) were immobilized on the
Experiments amino-silanized LCCD. In the experiments, 200 μl APTES
was dripped onto a LCCD substrate avoiding the light. The
3.3.1 Modification
APTES solution was then removed and dried in an oven at
of the Laser Confocal
100 C prior to use (see Note 9).
Culture Dish (LCCD)
and Self-Assembly 2. 100 μl of 50 μg ml1 type-I collagen (rat tail) solution was
of Type-I Collagen Fibrils dripped on the modified LCCD substrate and incubated at
37 C for 12 h and then gently rinsed with deionized water.
3. The type-I collagen fibrils were further cross-linked with
0.05 wt% glutaraldehyde for 2 h and then gently rinsed with
deionized water (see Note 10).
Biomimetic Model for Mineralization of Type-I Collagen 49
3.3.3 STORM Imaging The type-I collagen (rat tail) fibrils were immersed in the imaging
buffer before STORM imaging.
1. 2D STORM imaging experiments were performed on inverted
optical microscope. For z-stack images of the mineralized type-
I collagen fibrils, the images were recorded, processed, and
analyzed using the Imaris software.
2. 3D STORM imaging experiments were performed on a Nikon
N-STORM microscope equipped with a 100 oil immersion
objective and an Andor camera [24].
3. To identify the zone of interest, a low-magnification fluores-
cence image was acquired prior to STORM imaging.
4. After switching to higher magnification (100 objective), con-
ventional fluorescence images were first acquired. STORM data
acquisition was then started using imaging cycles at one frame
of activation laser illumination (405 nm laser) followed by five
frames of imaging laser illumination (561 nm laser). The inte-
gration time of the camera was set to the one frame mode with
an EM gain of 305,000–10,000 cycles per channel, which was
50 Shasha Yao et al.
4 Notes
Fig. 11 The z-slices of the STORM image of the mineralized collagen-I fibrils,
indicating that the mineralization of HAP inside and on the surface of the
collagen-I fibrils. Reproduced with permission [14]. Copyright 2018, Wiley-VCH
Fig. 13 CryoTEM image of collagen fibrils after exposure of 200 e/Å2, showing
the beam damage (white spots, as highlighted by the yellow circle)
9. The LCCD must be fresh and clean prior to use. After the
removal of the APTES solution, the modified LCCD was ultra-
sonicated with anhydrous ethanol and deionized water for
20 min, respectively, to remove the physical absorbed chemi-
cals. 50 μg ml1 of type-I collagen solution was obtained by
mixing 5 μl of type-I collagen stock solution with 295 μl of
assembling solution and then incubating the mixed solution at
room temperature for 20 min. After 12 h, the LCCD was
gently rinsed with deionized water for at least three times to
remove unassembled type-I collagen.
10. Incubation times of antibodies less than 2 h or calcein labeling
time less than 20 min would lead to inhomogeneous stained
type-I collagen samples (Fig. 14). The labeling process should
avoid the light.
References
1. Weiner S, Traub W, Wagner HD (1999) 4. Hodge AJ (1963) Recent studies with the elec-
Lamellar bone: structure-function relations. J tron microscope on ordered aggregates of the
Struct Biol 126:241–255 tropocollagen macromolecule. In: Ramachan-
2. Beniash E, Traub W, Veis A, Weiner S (2000) A dran GN (ed) Aspects of protein structure.
transmission electron microscope study using Academic Press, New York, pp 289–300
vitrified ice sections of predentin: structural 5. Orgel JPRO, Irving TC, Miller A, Wess TJ
changes in the dentin collagenous matrix (2006) Microfibrillar structure of type I colla-
prior to mineralization. J Struct Biol gen in situ. Proc Natl Acad Sci U S A
132:212–225 103:9001–9005
3. Goldberg M, Kulkarni AB, Young M, Boskey A 6. Quan BD, Sone ED (2013) Cryo-TEM analy-
(2011) Dentin: structure, composition and sis of collagen fibrillar structure. Methods
mineralization. Front Biosci (Elite Ed) Enzymol 532:189–205
3:711–735
54 Shasha Yao et al.
7. Weiner S, Traub W (1986) Organization of 17. Wang Y et al (2012) The predominant role of
hydroxyapatite crystals within collagen fibrils. collagen in the nucleation, growth, structure
FEBS Lett 206:262–266 and orientation of bone apatite. Nat Mater
8. Olszta MJ et al (2007) Bone structure and 11:724
formation: A new perspective. Mater Sci Eng 18. Jiang F, Hörber H, Howard J, Müller DJ
R Rep 58:77–116 (2004) Assembly of collagen into microrib-
9. Gao HJ, Ji BH, Jager IL, Arzt E, Fratzl P bons: effects of pH and electrolytes. J Struct
(2003) Materials become insensitive to flaws Biol 148:268–278
at nanoscale: Lessons from nature. Proc Natl 19. Williams BR, Gelman RA, Poppke DC, Piez
Acad Sci U S A 100:5597–5600 KA (1978) Collagen fibril formation. Optimal
10. Nair AK, Gautieri A, Chang S-W, Buehler MJ in vitro conditions and preliminary kinetic
(2013) Molecular mechanics of mineralized results. J Biol Chem 253:6578–6585
collagen fibrils in bone. Nat Commun 4:1724 20. George A, Veis A (2008) Phosphorylated pro-
11. Chen J, Burger C, Krishnan CV, Chu B, Hsiao teins and control over apatite nucleation, crys-
BS, Glimcher MJ (2005) In vitro mineraliza- tal growth, and inhibition. Chem Rev
tion of collagen in demineralized fish bone. 108:4670–4693
Macromol Chem Phys 206:43–51 21. Price PA, Toroian D, Lim JE (2009) Minerali-
12. Wang J et al (2013) Remineralization of dentin zation by inhibitor exclusion the calcification of
collagen by meta-stabilized amorphous calcium collagen with fetuin. J Biol Chem
phosphate. CrystEngComm 15:6151–6158 284:17092–17101
13. Olszta M, Douglas E, Gower L (2003) Scan- 22. Nudelman F, Lausch AJ, Sommerdijk NAJM,
ning electron microscopic analysis of the min- Sone ED (2013) In vitro models of collagen
eralization of type I collagen via a polymer- biomineralization. J Struct Biol 183:258–269
induced liquid-precursor (PILP) process. Cal- 23. Smeets PJM et al (2017) A classical view on
cif Tissue Int 72:583–591 nonclassical nucleation. Proc Natl Acad Sci U S
14. Shao C et al (2018) Citrate improves collagen A 114:E7882–E7890
mineralization via interface wetting: a physico- 24. Bates M, Jones SA, Zhuang X (2013) Stochas-
chemical understanding of biomineralization tic optical reconstruction microscopy
control. Adv Mater 30:1704876 (STORM): a method for superresolution fluo-
15. Nudelman F et al (2010) The role of collagen rescence imaging. Cold Spring Harb Protoc
in bone apatite formation in the presence of 2013(6):498–520 pdb.top075143
hydroxyapatite nucleation inhibitors. Nat 25. Nudelman F, Bomans PH, George A, Sommer-
Mater 9:1004–1009 dijk NA (2012) The role of the amorphous
16. Niu LN et al (2017) Collagen intrafibrillar phase on the biomimetic mineralization of col-
mineralization as a result of the balance lagen. Faraday Discuss 159:357–370
between osmotic equilibrium and electroneu-
trality. Nat Mater 16:370–378
Part II
Abstract
Solid-state nuclear magnetic resonance spectroscopy (ssNMR) is an emerging technique in structural
methods of studying collagen proteins, capable of identifying features on an atomic length scale in tissues
and protein samples without extensive extraction or purification. Hydroxylation is a key posttranslational
modification of collagen that gives rise to distinctive signals in the ssNMR spectrum of collagen proteins.
Here we outline the type of information that ssNMR can provide and describe the procedures involved in a
ssNMR structural study, with particular focus on using dynamic nuclear polarization to enhance sensitivity
for detecting hydroxylysine residues by ssNMR.
Key words Solid-state nuclear magnetic resonance spectroscopy, NMR, Proline, Lysine, Hydroxyl-
ation, Hydroxyproline, Hydroxylysine, Isotopic enrichment/labelling, Dynamic nuclear polarization
1 Introduction
Irit Sagi and Nikolaos A. Afratis (eds.), Collagen: Methods and Protocols, Methods in Molecular Biology, vol. 1944,
https://doi.org/10.1007/978-1-4939-9095-5_5, © Springer Science+Business Media, LLC, part of Springer Nature 2019
57
58 Wing Ying Chow
1.1 NMR Overview Nuclear magnetic resonance spectroscopy [3–5] relies on the fact
that nuclei have a quantum property called spin. Spin arises from
the number of protons and neutrons in the nuclei, taking values of
0, 1/2, 1, 3/2, and so on; certain nuclei (e.g., 1H, 13C) have a value
of spin >0, and these nuclei can be detected by NMR, i.e., they are
NMR-active. For elements with different isotopes, the nuclei of
these isotopes often have different values of spin. For example, of
the isotopes of carbon, the most abundant (12C) has zero spin and
hence cannot be detected by NMR. On the other hand, 1H and 31P,
both highly abundant, have spin and can be detected by NMR.
Almost all of the nuclei that are used for detection in NMR are
stable (i.e., nonradioactive) isotopes.
In most biomolecular NMR, the focus is on experiments that
involve spin-1/2 nuclei, commonly 1H, 13C, 15N, and 31P. These
nuclei have the property of having two quantum states which have
different energies when placed in a magnetic field. There are appli-
cations detecting NMR of higher-spin nuclei such as 2H (spin-1),
14
N (spin-1), 17O (spin-5/2), 23Na (spin-3/2), and 43Ca (spin-7/
2); however, these often give rise to more complicated NMR
spectra and therefore are beyond the scope of this chapter.
Transitions between the two states of a spin-1/2 nuclei in a
magnetic field occur at a frequency that is determined by an intrin-
sic property of the nuclei and by the strength of the magnetic field.
It is therefore necessary to note which nuclei is being detected, as at
9.4 T, the 1H (proton) frequency is 400 MHz, while 13C would
have a frequency of 100 MHz and 15N would have a frequency of
40 MHz. It is common to refer to NMR spectrometers by the 1H
frequency, so a 9.4 T magnet is commonly called a 400 MHz
spectrometer and an 18.8 T magnet, an 800 MHz spectrometer.
A key part of NMR equipment is the probe head, which sits in
the core of the magnet and plays several important roles: firstly, it
places the sample in the region of the magnet with the most
homogeneous magnetic field; secondly, it regulates the sample
temperature; thirdly, for solid-state NMR, the probe head contains
the stator, which can be adjusted to the magic angle and regulates
the spinning of the rotor, necessary for producing resolved spectra
(see below); fourthly and perhaps most importantly, a coil in the
stator surrounds the sample and converts magnetic behavior of the
spins to an electric current, which is amplified and can be detected
by the resonant circuits. The circuits can be tuned to the specific
characteristic frequency for 1H and other nuclei to deliver radio-
frequency (RF) pulses to manipulate the spin transitions; the com-
bination of the number, timing, spacing, length, and power of the
pulses is what makes up different NMR experiments by selecting for
specific spin interactions.
Since the magnetic behavior of the spins occur at specific fre-
quencies, the electric current induced and measured also shows an
oscillating behavior over time, which has an overall decrease in
Solid-state NMR Investigations of Collagen Hydroxylation 59
signal due to relaxation and is called the free induction decay (FID).
Generally, the FID is not analyzed directly but instead subjected to
Fourier transform which provides a spectrum with a frequency axis.
Fourier transform procedures are routine on modern spectrometers
and computers and are therefore not further described in this
chapter.
1.2 Chemical Shift One of the key advantages of NMR is its sensitivity to small varia-
tions in chemical structure. Differences in the number and types of
bonds lead to relatively small (parts per million level) changes in the
transition frequency. The simplest NMR spectra, so-called 1D
spectra, are plots of the intensity of transition frequencies of one
particular type of nuclei, giving insight into the types of different
chemical environments for these nuclei in the sample.
The transition frequency scales with the external applied mag-
netic field. Therefore, the spectrum is commonly presented with a
frequency scale called the chemical shift (δ), which is relative to parts
per million (ppm) of the transition frequency of the nuclei in a
reference sample, rather than the directly measured frequencies in
MHz. The chemical shift scale has the useful property of enabling
direct comparison of spectra obtained on spectrometers with dif-
ferent magnetic field strengths.
While the method of referencing the chemical shift scale can
seem like a technical detail, it is important for accurate reporting
and correct comparisons to literature-reported chemical shift
values. For 1H and 13C NMR, the chemical shift ppm scale is
referenced by setting the signal of TMS (tetramethylsilane) or
DSS (2,2-dimethyl-2-silapentane-5-sulfonic acid) to zero ppm.
TMS is used by publications in synthetic chemistry as per IUPAC
recommendations [6], while DSS is more commonly used in bio-
molecular NMR spectroscopy due to its better water solubility
[7]. For 13C, the DSS-referenced chemical shift values are greater
than the TMS-referenced values by approximately 2.7 ppm, so it is
important to take this conversion factor into account when com-
paring to literature values.
In solution-state NMR, referencing is often carried out using
the solvent signal. However, this is not the recommended approach
in solid-state NMR due to the highly variable amount of solvent,
the wider temperature range, and the potential inhomogeneity in
solvent distribution, all of which can lead to shifting or broadening
of the solvent signal, complicating attempts to reference and/or
reproduce the spectra. Instead, DSS is sometimes added to wet
pellet protein samples as an internal reference or external referen-
cing is used where DSS or TMS is not added to the sample but
measured separately. In this chapter, we use an external referencing
scheme based on TMS, as we would prefer to facilitate comparisons
with synthetic compounds that model collagen glycosylation [8].
60 Wing Ying Chow
1.3 Solid-State NMR The lack of tumbling motion of the sample in solid-state NMR
and Magic Angle complicates the spectra compared to those of solution-state NMR.
Spinning Since the interaction of the nuclei and the magnetic field is aniso-
tropic and depends on the orientation of surrounding atoms and
bonds, the signals in solid-state NMR spectra of static noncrystal-
line samples are greatly broadened. Moreover, through-space
Solid-state NMR Investigations of Collagen Hydroxylation 61
Fig. 1 Typical 13C and 15N chemical shifts of proline, hydroxyproline, lysine, and
hydroxylysine. For proline, hydroxyproline, and lysine, variations are usually less
than 2 ppm. For hydroxylysine, variations of up to 5 ppm are possible due to
structural changes induced by glycosylation, glycation, and cross-linking.
Changes due to hydroxylation are shown in bold text. Chemical shift values
are referenced to TMS to enable easy comparison to the synthetic chemistry
literature on various hydroxylysine modifications
1.4.2 Dynamic Nuclear The detectable signal in NMR phenomena is based on population
Polarization in Solid- differences between the spin quantum states, which is termed the
State NMR polarization. The energy differences between spin quantum states
are much smaller than thermal energy at room temperatures, which
leads to a very small population difference, a very small polariza-
tion, and consequently a very small detectable signal. One of the
techniques to improve polarization in solid-state NMR is dynamic
nuclear polarization (DNP) [14], which has undergone commer-
cialization and rapid development in the recent years. DNP
enhancement relies on the transfer of polarization from electrons
to nuclei, either directly or through a spin diffusion process,
thereby achieving signal enhancement up to a theoretical limit of
660 for 1H (and even larger for other nuclei). Assuming no loss of
sensitivity via depolarization effects, the time saving is equal to the
signal enhancement squared. In other words, when an enhance-
ment factor of 10 is obtained relative to some reference spectrum,
an experiment with the same signal-to-noise ratio as the reference
spectrum can be conducted in 1/100th of the time. Signals that
would take weeks of signal averaging before accumulating sufficient
intensity above the noise level under routine ssNMR conditions can
be observed in hours with sufficient DNP enhancement.
64 Wing Ying Chow
2 Materials
2.2 Solid Biological While general procedures of preparing samples for DNP NMR
Samples experiments on water-soluble proteins, assemblies, and precipitates
have been described [21], since most collagen samples are poorly
soluble in water, we provide the procedures described below, which
were inspired by prior applications of DNP signal enhancement on
porous materials [22] and can be applied on a wide range of solid
samples. For example, ssNMR experiments have been successfully
performed on tissues dissected from patients or model organisms,
on protein pellets such as those obtained from cell culture-derived
ECM, and on freeze-dried triple-helical peptides obtained from
solid-phase peptide synthesis [11, 12]. 13C-enriched material is
necessary for studies involving hydroxylysine and recommended
for studies involving hydroxyprolines. While this chapter will not
provide a full description of isotope enrichment procedures, which
can vary greatly depending on the scientific question and the type of
samples involved, some general requirements of samples suitable
for solid-state NMR experiments are provided below.
1. NMR experiments can be successfully carried out with slightly
less than 10 mg of isotope-enriched sample, but more is better
for both detection and spinning stability. Up to 30 mg of sample
can usually be packed into a 3.2 mm diameter rotor (see Note 5).
2. Although a high level of isotope enrichment is desirable, a level
of 20–50% can be sufficient (see Note 6).
3. Avoid freeze-drying tissue samples (see Note 7).
4. Avoid mechanical milling of tissue samples (see Note 8).
5. The use of fixatives or preservatives are generally not recom-
mended, as these substances can give rise to irrelevant but
strong signals in the NMR spectra, especially in the case of
natural abundance samples (see Note 9).
6. If a cell lysis step is carried out, it is recommended to rinse the
ECM with phosphate buffered saline (1) or distilled water to
remove any cell debris.
66 Wing Ying Chow
2.3 Stable Radicals 1. Water-soluble binitroxide radicals such as bcTol-M [17] and
for DNP Enhancement AMUPol [18]. Store away from light.
2. Heavy water (D2O).
3. Distilled water (H2O) (for freeze-dried samples only).
3 Methods
3.1 Preparing Carry out this procedure at room temperature. Try to avoid work-
the Radical Stock ing at a bench which is under direct sunlight, as this can lead to
Solution for DNP quenching of the radicals.
Enhancement 1. If the sample is hydrated, use D2O as the solvent. Otherwise, if
the sample is freeze-dried, use a mix of D2O and H2O in a ratio
of 9:1 (v/v) as the solvent. Allow for 4–10 μL of radical
solution for each ssNMR sample to estimate the volume of
radical stock solution required.
2. If using AMUPol, calculate the mass required for a 30 mM
solution. If using bcTol-M, calculate the mass required for a
40 mM solution. Weigh out the mass required using a balance
with sufficient accuracy (see Note 10).
3. Dissolve the solid radical thoroughly with the required amount
of water (pure D2O or mixed D2O/H2O). Check carefully for
any undissolved crystals and sonicate if necessary. Label, wrap
the radical stock solution container with tinfoil to protect from
light, and store in the refrigerator until use.
3.2 Packing For most collagen-containing samples, this procedure can be car-
the ssNMR Rotor ried out at room temperature. In the case of highly temperature-
sensitive biological samples, the procedure can be carried out at low
temperature. The procedures below are specifically for 3.2 mm
diameter rotors.
1. Weigh the empty ssNMR rotor with the cap prior to sample
packing.
2. Weigh the sample. If there is more than 30 mg of sample,
return the excess to storage.
3. If the sample is solid tissue, cut or break up the sample into
pieces that are no larger than 1.5 mm 1.5 mm 4 mm,
keeping the cut pieces in relatively similar sizes where possible.
4. Fill the sample into the rotor with occasional compaction to
ensure an even weight distribution, which is important for
successful MAS. Stop filling when the sample height in the
rotor is about 3 mm below the top edge of the rotor. When
this happens, or when the sample is all filled into the rotor,
weigh the packed rotor to determine the mass of sample in the
rotor.
Solid-state NMR Investigations of Collagen Hydroxylation 67
3.3 Inserting Rotor 1. Set the temperature of the NMR probe head to the desired
into Magnet and Magic value (see Note 14).
Angle Spinning 2. Press “insert” on the MAS control panel.
3.3.1 Insertion 3. Insert the sample by placing the rotor into the sample transfer
on a Probe Head Without line or directly into the opening of the stator within the
the Option of DNP probe head.
Enhancement
3.3.2 Insertion 1. Cool the DNP NMR probe head down to 100 K using the
on a Probe Head MAS and variable temperature gas flow.
with the Option of DNP 2. Press “eject” on the MAS control panel (see Note 15).
Enhancement
3. Place the rotor in the sample holder of the sample transfer line.
4. Press “insert” on the MAS control panel (see Note 16).
68 Wing Ying Chow
3.4 Solid-State NMR Here we present three experiments that are particularly useful for
Experiments studying collagen hydroxylation. We assume that the solid-state
NMR spectrometer had already been used previously for other
13
C/15N experiments and therefore does not cover basic spectrom-
eter setup such as setting the magic angle and initial calibration of
the RF pulse powers, and we assume that a functional pulse
sequence has been installed. For each experiment, some represen-
tative spectra are included to aid interpretation and analysis. Please
check with the local facility manager to ensure that maximum
power levels of the NMR spectrometer are not exceeded.
3.4.1 1D Cross This experiment yields a 1D 13C or 15N NMR spectrum and is the
Polarization (CP) basis of many other ssNMR experiments. Below, the 13C/15N
nucleus is denoted with X (see Note 19).
1. Create a new dataset from a previous CP experiment.
2. Optimize the following parameters for maximum signal-to-
noise ratio (see Note 20):
(a) 1
H 90 pulse length
1
(b) H contact pulse power
1
(c) H-X contact pulse length
1
(d) H decoupling sequence pulse power during signal
acquisition
3. Signal average the optimized CP experiment for at least
4–10 min. Usually this would equate to a minimum of
128 transients.
4. For measuring DNP signal enhancement, the same CP experi-
ment (with same number of transients and parameters) should
be run twice: once with the microwave irradiation switched off
and once again with the microwave irradiation switched
on. The relative ratio of intensity of the two 1D CP spectra is
commonly referred to as the enhancement value (ε). An accept-
able value of ε is 10—many experiments are already much more
feasible at this level of enhancement. On collagen samples, we
have thus far obtained values of ε up to 57.
5. For all other DNP NMR experiments, the microwave irradia-
tion should be switched on prior to the start and kept at the
same level throughout the experiment.
If the referenced spectrum contains a 13C signal at 70 ppm, it
strongly indicates that the sample contains hydroxylated carbon
Solid-state NMR Investigations of Collagen Hydroxylation 69
Fig. 2 1D 13C NMR spectra of adult human bone (natural abundance), 297 K,
400 MHz spectrometer, 10 Hz MAS. Residues that feature prominently in
collagen-containing samples are labelled in bold text
Fig. 3 2D 13C–13C DARR spectra of (a) lysine 13C-labelled mouse skin, no DNP enhancement, 285 K, 20 ms
mixing time, 600 MHz spectrometer, 13,333 Hz MAS; (b) lysine 13C-labelled mouse skin, 110 K, with DNP
enhancement (AMUPol, 30 mA microwave current, ε ¼ 27), 5 ms mixing time, 400 MHz spectrometer,
8889 Hz MAS; (c) proline and glycine 13C, 15N-labelled fetal sheep osteoblast ECM, 110 K, no DNP
enhancement, 20 ms mixing time, 400 MHz spectrometer, 8889 Hz MAS. Annotations indicate signals for
unmodified lysine (orange/light gray) and proline (light gray/light gray) residues, and bold-type labels indicate
signals for hydroxylysine (dark brown/dark gray) and hydroxyproline (dark green/dark gray). We can see from
(b) and (c) that hydroxylysine and hydroxyproline signals do not overlap with each other; thus, we can use the
2D spectral pattern to distinguish between these two hydroxylated residues
Fig. 4 2D 15N–13C spectra of lysine 13C, 15N-labelled adult bovine vascular smooth muscle cell ECM, 110 K,
with DNP enhancement (AMUPol, 30 mA microwave current, ε ¼ 57), 10 ms DARR mixing time, 400 MHz
spectrometer, 8889 Hz MAS. The signals specific to hydroxylysine (a, b; in red/dark gray) and hydroxylysine
(c, d; in teal/light gray) are indicated on the figure. Arrows on the chemical structure indicate the magnetiza-
tion transfer pathways, omitting the initial 1H to 15N CP step, but starting on 15N and ending on 13C, that give
rise to the signals observed. Due to the low number of hydroxylysine residues in the ECM relative to lysine
residues, the sidechain signals from the hydroxylysine pathway (a) are much weaker than those from the
lysine pathway (c)
4 Notes
1. For tissue samples, rotor diameters of 3.2 mm, 4.0 mm, and
7.0 mm have all been successfully used to achieve stable
spinning. Smaller rotor diameters such as 2.5 mm and
1.9 mm may also be used, but more care is needed to balance
the sample.
2. From our experience, the commercial DNP-ssNMR 3.2 mm
diameter MAS probe head produced by Bruker requires cool-
ing with approximately 250 L of liquid nitrogen every 24 h
when running at a set temperature of 100 K.
3. In the cases where the sample mass is limited, restricted depth
rotors should be used to center the sample in the center of the
rotor to improve RF pulse homogeneity and experiment sensi-
tivity. Inert spacers made of materials such as Teflon can be
used for restricting the sample height to the center of the rotor.
Materials low in proton and carbon (where possible) are pre-
ferred, as otherwise the spacer material could be observed in
the NMR spectra; care should also be taken to avoid materials
that show large volume change with temperature variations.
4. For experiments at the low temperature conditions for DNP
enhancement (<200 K), zirconia caps are recommended,
though Vespel caps have also been successfully used. It is
recommended to test shrinkage of the rotor cap by placing
the packed rotor into liquid nitrogen and observing whether
there is any movement or slippage of the cap before inserting
the rotor into the probe head. Use rotors with regular wall
Solid-state NMR Investigations of Collagen Hydroxylation 73
Acknowledgments
The author thanks Dr. Jonathan Clark from the Babraham Institute
for providing the lysine 13C-labelled mouse skin sample,
Mr. Rakesh Rajan and Professor Melinda Duer from the University
of Cambridge for providing the proline and glycine 13C, 15N-
labelled fetal sheep osteoblast ECM sample, Mr. Robert Hayward
and Professor Cathy Shanahan at King’s College London for
providing the lysine 13C, 15N-labelled adult bovine vascular smooth
muscle cell ECM sample, and Dr. Kelsey Collier at FMP Berlin for
useful discussions and comments.
References
1. Shoulders MD, Raines RT (2009) Collagen 4. Cavanagh J, Fairbrother WJ, Palmer AG, Skel-
structure and stability. Annu Rev Biochem ton NJ, Rance M (2010) Protein NMR spec-
78:929–958. https://doi.org/10.1146/ troscopy: principles and practice. Elsevier,
annurev.biochem.77.032207.120833 Amsterdam
2. Yamauchi M, Sricholpech M (2012) Lysine 5. Keeler J (2011) Understanding NMR spectros-
post-translational modifications of collagen. copy. John Wiley & Sons, New Jersey
Essays Biochem 52:113–133. https://doi. 6. Harris RK, Becker ED, De Menezes SMC,
org/10.1042/bse0520113 Granger P, Hoffman RE, Zilm KW (2008)
3. Levitt MH (2013) Spin dynamics: basics of International Union of Pure and Applied
nuclear magnetic resonance. John Wiley & Chemistry physical and biophysical chemistry
Sons, New Jersey division (2008) further conventions for NMR
shielding and chemical shifts IUPAC
Solid-state NMR Investigations of Collagen Hydroxylation 77
Abstract
All tissues contain an extracellular matrix (ECM) which is constantly and dynamically remodeled, either in
physiological or pathological processes, such as fibrosis or cancer. One of the key contributors in the
establishment of a fibrotic state is the abnormal deposition of extracellular matrix and cross-linked proteins,
in particular collagen, leading to tissue stiffening and disruption of organ function. The precise and sensitive
measurement of these cross-links by LC-MS/MS is a very powerful tool for providing a quantitative and
qualitative analysis of fibrosis and is a key requirement in the study of this state, as well as in the development
of drugs for this unmet clinical need.
Key words Collagen, Tissue cross-linking, Immature cross-links, Mature cross-links, Solid phase
extraction (SPE), Mass spectrometry
1 Introduction
Irit Sagi and Nikolaos A. Afratis (eds.), Collagen: Methods and Protocols, Methods in Molecular Biology, vol. 1944,
https://doi.org/10.1007/978-1-4939-9095-5_6, © Springer Science+Business Media, LLC, part of Springer Nature 2019
79
80 Amar Joshi et al.
2 Materials
2.1.2 Reduction Step 1. 12 mg/mL sodium borohydride (NaBH4) buffer: the buffer is
prepared in 20 mM sodium hydroxide (NaOH) solution. The
NaOH stock solution can be prepared at 2 M in water and then
diluted to 20 mM with water for NaBH4 buffer preparation (see
Note 1).
2. Neat acetic acid: >99.7% purity.
3. Benchtop centrifuge.
4. Benchtop heat block.
5. 6 M hydrochloric acid (HCL): 12 M HCL sourced commer-
cially is diluted 1:2 to obtain 6 M HCL (see Note 2).
2.1.3 Sample Cleanup 1. Automated SPE machine (e.g., GX-271 ASPEC™ Liquid
Using Solid Phase Handler).
Extraction (SPE) 2. Benchtop quick spin.
3. Benchtop shaker.
4. Capless graduated microtubes (1.7 mL).
5. C18 columns (e.g., GracePure™ C18).
6. Solid phase extraction (SPE) strong cation exchange (SCX)
columns (e.g., GracePure™ SPE).
7. 5 mL round bottom tubes.
8. Solution 1: 10 mM ammonium formate, 0.1% formic acid,
0.1% heptafluorobutyric acid (HFBA) in water. For a 500 mL
solution preparation, weigh 315.3 mg of ammonium formate
and transfer to a 500 mL glass bottle. Add 500 μL formic acid,
500 μL HFBA and make up to 500 mL with water. Mix (see
Note 2).
9. Solution 2: 10 mM ammonium formate, 0.1% formic acid,
0.1% HFBA, 40% methanol in water. For a 500 mL solution
preparation, weigh 315.3 mg ammonium formate and transfer
to a 500 mL Schott glass bottle. Add 500 μL formic acid,
500 μL HFBA, 200 mL methanol and make up to 500 mL
with water. Mix (see Note 2).
10. Solution 3: 25% ammonium hydroxide solution in water (see
Note 2).
11. Solution 4: 0.1% formic acid in water. For a 500 mL solution
preparation, add 500 μL formic acid and make up to 500 mL
with water. Mix (see Note 2).
12. HFBA solution: 5% HFBA in water (see Note 2).
13. SnapStrip 8-strip PCR tubes.
14. High-performance liquid chromatography (HPLC) screw vials
(with fixed inserts) and lids.
82 Amar Joshi et al.
3 Methods
3.1 Extraction For immature and mature collagen cross-links and hydroxyproline.
and Purification
of Cross-Links
3.1.1 Tissue Sample Note that this tissue preparation is generic and applies for most soft
Preparation tissues, except skin (see Subheading “Skin Preparation”).
1. Weigh out relevant amount of tissue and carry out freeze-
drying. Cut the tissue into smaller pieces to increase the surface
area for freeze-drying (see Note 4).
2. Grind the freeze-dried tissue manually using a spatula and
then collect 20 mg in a 2 mL metal bead lysing matrix tube
(see Note 5).
3. Homogenize the dry tissue twice. The sample becomes a dry
powder.
4. Add 1 mL of PBS and homogenize two more times (see
Note 6).
5. Transfer 500 μL of the homogenate (equivalent to 10 mg dry
tissue) into a 2 mL heat/acid resistant screw cap tube (see
Note 7). Tissue preparation is complete for mature collagen
and mature elastin cross-links samples.
6. For immature collagen cross-links extraction and hydroxypro-
line extraction samples, top up with 370 μL of PBS, to gain a
total volume of 870 μL.
7. Reduction step (see Subheading 3.1.2).
Skin Preparation 1. Weigh out each sample and insert into a 2 mL metal bead lysing
matrix tube (see Note 8).
2. For homogenization of skin samples, add 1 mL of collagenase
solution per 10 mg of tissue.
3. Incubate for 24 h at room temperature. During the incuba-
tion, the samples should be homogenized intermittently (see
Note 9).
84 Amar Joshi et al.
3.1.2 Reduction Step For immature collagen cross-links extraction and hydroxyproline.
For mature collagen and elastin cross-links, go straight to step 5.
1. Add 30 μL of 12 mg/mL NaBH4 buffer into the 870 μL tissue
homogenate (equivalent to a 30 dilution of the buffer). After
addition, shake the tubes manually by hand to ensure the
reduction reaction can occur.
2. Incubate at room temperature for 30 min.
3. Stop the reaction by adding 50 μL of neat acetic acid, which
will bring the pH below 2.5 (see Note 10).
4. Quick spin the samples to remove liquid contents from the lid.
5. Dry the samples using the Genevac at 60 C with the aqueous
program settings.
6. Add 1600 μL of 6 M HCL to the pellet. Heat at 105 C to
hydrolyse for 24 h (see Note 11).
7. Dry the HCL using the Genevac at 60 C with the H2O + NH3
program settings (see Note 12).
8. Reconstitute the samples in 200 μL of water, and place on
shaker for 10 min at high speed.
9. Spin dow the samples at 20,000 g for 1 min.
3.1.3 Sample Cleanup 1. Prepare C18 and SCX columns by inserting adaptors onto the
Using SPE (Automated) columns (see Note 13).
2. Take 50 μL of the sample hydrolysate (i.e., from step 8 of the
reduction procedure) into 400 μL of solution 1.
3. Load onto a SPE C18 column.
4. Perform steps 5–9 in a fume hood (see Note 2).
5. Add 600 μL of solution 1 to the C18 column and elute into a
C18-labeled 5 mL round bottom tube.
6. Add 500 μL of solution 2 to the C18 column and elute into the
abovementioned 5 mL round bottom tube.
7. Add the C18 eluate (1550 μL total) directly onto a SCX
column (which has been pre-rinsed with 2 mL of water and
500 μL of solution 4).
8. Wash the loaded SCX column with 1 mL of water.
9. Elute the SCX cartridge with 3 mL of solution 3 and collect
into a SCX-labeled 5 mL round bottom tube.
Measurement of Collagen Cross-Linking by Mass Spectrometry 85
10. Dry the SCX eluate using the Genevac at 60 C with the
H2O + NH3 program settings.
11. Add 500 μL of 5% HFBA solution.
12. Dry the sample using the Genevac at 60 C with the aqueous
program settings.
13. Reconstitute the dried samples in 50 μL of solution 1 and
transfer to SnapStrip PCR tubes (see Note 14).
14. Spin the samples using a benchtop spin at high speed for 5 min.
15. Transfer 40 μL of the supernatant into HPLC vials; take extra
care not to disturb the pellet.
3.1.4 Protein Assay Perform a protein assay on the samples as per instructions detailed
in the total protein assay kit (see Note 15).
3.2 Ultrahigh- 1. Flow rate: 0.3 mL/min using a multistep gradient method.
Performance Liquid 2. Gradient method: Hold mobile phase A at 96.2% for 1.0 min
Chromatography- and then decrease linearly to 80% within 5 min. Hold mobile
Triple Quadrupole phase A at 80% from 5 min to 7 min. Switch the divert valve
Mass Spectrometer from ion source to the waste at 7 min. Decrease mobile phase A
(UHPLC-MS/MS) linearly to 0% from 7 min to 9 min. Bring back mobile phase A
concentration from 0% to 96.2% linearly at 9.5 min and hold
3.2.1 Chromatographic
for the next 1.5 min, i.e., until 12.0 min. At the end of the
Conditions
gradient program, for the duration of 0.2 min from 11.8 to
12.0 min, decrease the flow rate from 0.3 mL/min to
0.05 mL/min.
3. Total run time: 12.0 min.
4. Column oven temperature: 40 C.
5. Autosampler temperature: Room temperature.
6. Injection volume: 10 μL for the skin, liver, and kidney tissues.
3.2.3 Calibration Carry out all procedures at room temperature unless otherwise
Standards Preparation specified.
1. Pipette out 650 μL of the PYD/DPD mixture from the vial out
of the total volume of 750 μL and transfer it into a plastic test
Measurement of Collagen Cross-Linking by Mass Spectrometry 87
3.2.4 Sample Acquisition 1. Start up the LC-MS/MS system (see Note 16). Perform at least
and Data Analysis five injections of water onto the column using the cross-links
method. This is to equilibrate the column and the mass spec-
trometer ion source with the cross-link method conditions.
2. Following equilibration, inject a few Cross-links Calibration stan-
dard for sensitivity and chromatography check (see Note 17).
3. Inject all the Cross-links Calibration standards (see Note 18),
followed by injecting the processed tissue samples.
Measurement of Collagen Cross-Linking by Mass Spectrometry 89
10. Once the chromatographic data are finalized, apply the correct
conversion factor to the tissue samples. This would give the
actual amount of biomarker present in the total volume of
hydrolysate sample. Conversion factor is the ratio of the vol-
ume of the hydrolysate sample to the volume of the injected
sample (e.g., for a 200 μL hydrolysate sample and an injected
10 μL sample onto the column, the conversion factor would be
200/10 ¼ 20).
11. Generate the final LC-MS/MS results summary file into an
Excel file for further calculations.
12. The amount generated are in terms of pmols/volume of sam-
ple hydrolysate. Convert the amount in terms of pmols/mL.
13. Normalize the amount (pmols/hydrolysate sample) of
DHLNL, HLNL, PYD, and DPD against HYP amount
(pmols/hydrolysate sample). Normalize the amount (pmols/
hydrolysate sample) of all biomarkers against dry tissue weight
(mg) and normalize the amount (pmols/mL) of all biomarkers
against protein content (mg/mL).
14. Conduct the statistical data analysis on Graph Pad Prism soft-
ware, version 6 or latest. Use unpaired t-test for the compari-
son of healthy and diseased animal groups. Use one-way
ANOVA for all treated groups against the untreated group to
compare the effect of one variable at the time (dose or different
drugs). Conduct all the Prism analysis on cleaned data by
excluding the outliers, using the Rout test, with the Q value
set at 1%.
4 Notes
tissue for each sample must be done accordingly (i.e., for liver
samples, 300 mg wet weight is a good starting point). After
each sample has been weighed out, it must be snap frozen in
dry ice (all tubes can be stored at 80 C for 30 min before
being placed on the freeze dryer). It can take up to 2 days for
the samples to freeze-dry completely; at the end of the proce-
dure, perform a qualitative (tissue should appear pale and
lighter in color) and quantitative (check to see approximately
70% of the weight has been deducted).
5. For the cross-links procedure, 10 mg has been determined to
be the ideal amount for the assay (although in some instances
5 mg is used). We usually weigh out 20 mg, add 1 mL PBS, and
take 500 μL of the homogenate across to further steps (which is
equivalent to 10 mg).
6. For certain tissue types, homogenizing two more times is not
sufficient. The tube must be observed thoroughly to ensure an
evenly reconstituted homogenate. The homogenization can be
performed a number of times (when dissolved evenly, the
homogenate usually appears pink). If after 5–6 cycles the pow-
der is not dissolved into the PBS, use a pipette tip to poke at the
tissue that may be stuck to the walls of the tube and then
homogenize again. Occasionally, the metal beads lead to
break of the integrity of the tube, which may in turn lead to
leakage of the homogenate. If this occurs midway, stop the
machine and reweigh the tissue. A good safety catch is to
check the tubes after each homogenization cycle and change
the lids if scratches are observed.
7. It is critical the homogenate is evenly reconstituted before
transferring. Therefore, pipette up and down carefully before
transferring the 500 μL. If any large tissue bits are observed,
repeat the homogenization.
8. If the skin sample is in excess of 14 mg or so, then the sample
should be divided into two, and both subsections should be
taken across in individual metal bead lysing matrix tubes.
9. The homogenization should be performed every 2–3 h (5–6
at every time point) when feasible. Before the end of the 24-h
period, the tissue should be entirely homogenized and no skin
tissue should be visible.
10. Concentrated acetic acid is corrosive to the skin and must
therefore be handled with appropriate care. This step should
be performed in a fume hood.
11. If after the 24-h hydrolysis, the procedure cannot be continued
straight away, the samples containing HCl can be stored a
20 C.
92 Amar Joshi et al.
References
1. Trackman PC (2016) Enzymatic and Zuurmond AM, Bank RA (2005) The type of
non-enzymatic functions of the lysyl oxidase collagen cross-link determines the reversibility of
family in bone. Matrix Biol 52-54:7–18 experimental skin fibrosis. Biochim Biophys Acta
2. van der Slot-Verhoeven AJ, van Dura EA, 1740:60–67
Attema J, Blauw B, De Groot J, Huizinga TW,
Chapter 7
Abstract
We present the development, optimization, and application of constructs, cell lines, covalent cross-linking
methods, and immunoprecipitation strategies that enable robust and accurate determination of collagen
interactomes via mass spectrometry-based proteomics. Using collagen type-I as an example, protocols for
working with large, repetitive, and GC-rich collagen genes are described, followed by strategies for
engineering cells that stably and inducibly express antibody epitope-tagged collagen-I. Detailed steps to
optimize collagen interactome cross-linking and perform immunoprecipitations are then presented. We
conclude with a discussion of methods to elute collagen interactomes and prepare samples for mass
spectrometry-mediated identification of interactors. Throughout, caveats and potential problems research-
ers may encounter when working with collagen are discussed. We note that the protocols presented herein
may be readily adapted to define interactomes of other collagen types, as well as to determine comparative
interactomes of normal and disease-causing collagen variants using quantitative isotopic labeling (SILAC)-
or isobaric mass tags (iTRAQ or TMT)-based mass spectrometry analysis.
1 Introduction
Collagen is the molecular scaffold for animal life [1, 2]. In addition
to forming the primary proteinaceous component of bone, skin,
cartilage, basement membranes, and other tissues, collagen also
facilitates diverse phenomena such as cell–cell signaling, wound
healing, and cell migration [3]. Proper execution of the folding,
modification, and quality control processes required for biogenesis
of this complex protein is, therefore, critical [4, 5]. Collagen pro-
teostasis, encompassing all of these factors, can be disrupted by
mutations in collagen molecules themselves or by dysfunction of
the proteostasis network [6–9], which is the integrated system of
intracellular chaperones, quality control factors, and trafficking
Irit Sagi and Nikolaos A. Afratis (eds.), Collagen: Methods and Protocols, Methods in Molecular Biology, vol. 1944,
https://doi.org/10.1007/978-1-4939-9095-5_7, © Springer Science+Business Media, LLC, part of Springer Nature 2019
95
96 Ngoc-Duc Doan et al.
Table 1
Selected wild-type collagen-I interactors reproducibly identified by mass spectrometry [15]
(continued)
98 Ngoc-Duc Doan et al.
Table 1
(continued)
2 Materials
2.1 Molecular 1. Genes, vectors, and DNA oligomers: Human Col1A1 and
Biology and Cell Col1A2 genes (Origene), pENTR1A vector (Thermo Fisher
Culture Scientific), pTRE-Tight vector (Clontech), pLenti.CMV/TO.
DEST vectors with assorted resistance cassettes (Addgene)
[17], puromycin and hygromycin linear selection markers
(Clontech), DNA primers (Sigma), and RRE, REV, and
VSVG vectors for lentivirus production (Addgene).
2. Enzymes: Q5 Polymerase, BamHI, EcoRV, NotI, T4 ligase,
Antarctic phosphatase (New England BioLabs; NEB), and LR
Clonase (Thermo Fisher Scientific).
Mass Spectrometry-Based Proteomics to Define Intracellular Collagen Interactomes 99
2.2 Preparation All chemicals and supplies used in these protocol steps must be mass
of Protein Samples, spectrometry-grade. RNase-/DNase-free microcentrifuge tubes
Immunoprecipitation, employed can be from a variety of vendors, but should not be
Protein Elution, autoclaved prior to use.
and Precipitation 1. Radioimmunoprecipitation assay (RIPA) buffer: 50 mM Tris
(VWR), 0.5% sodium deoxycholate (Alfa Aesar), 1% Triton
(INTEGRA Chemical), 0.1% sodium dodecyl sulfate (SDS;
Alfa Aesar), 50 mM NaF (Alfa Aesar), 5 mM β-glycerol phos-
phate (Sigma), 1 mM sodium metavanadate (Sigma), 5 mM
ethylene glycol-bis(β-aminoethyl ether)-N,N,N0 ,N0 -tetraacetic
acid (EGTA; STREM Chemicals), 5 mM ethylenediaminete-
traacetic acid (EDTA; Aqua Solutions), 100 μM sodium pyru-
vate (Sigma), 1 mM phenylmethylsulfonyl fluoride (PMSF;
Thermo Fisher Scientific), and protease inhibitor mixture
(Pierce) at pH 7.4 in dd-H2O.
100 Ngoc-Duc Doan et al.
2.3 Preparation All chemicals and supplies used in these protocol steps must be mass
of Tryptic Peptides spectrometry-grade. RNase-/DNase-free microcentrifuge tubes
and Mass employed can be from a variety of vendors, but should not be
Spectrometry autoclaved prior to use.
1. Chemicals: Iodoacetamide (IAA; Sigma), 1,4-dithiothreitol
(DTT; Sigma), urea and formic acid (Fluka).
2. NH4HCO3 buffer: 100 mM NH4HCO3(aq) at pH 8.0.
3. TFA buffer: 0.1% aqueous trifluoroacetic acid (TFA; Sigma).
4. Formic acid buffer: 0.1% aqueous formic acid.
5. TFA/MeCN buffer: 0.1% TFA in 9:1 MeCN:H2O (MeCN
from Sigma).
6. Formic acid/MeCN buffer: 0.1% formic acid in 4:1 MeCN:
H2O.
7. Pierce C18 Peptide Desalting Spin Column (Thermo Fisher
Scientific).
8. Aluminum foil (Reynolds).
9. Water bath at 56 C.
10. EASY-nLC 1000 or similar HPLC with autosampler
connected to a Thermo Fisher Q Exactive Hybrid
Quadrupole-Orbitrap Mass Spectrometer or any similar high-
resolution, accurate-mass mass spectrometer.
11. Proteome Discoverer 2.2 with Mascot search engine.
3 Methods
3.1 Creating Cellular antibodies, to permit the necessary IPs, we use cloning strategies to
Platforms introduce short, well-defined HA or FLAG antibody epitopes
for Biochemical between an ER-targeting signal sequence and collagen-I’s
Studies of Collagen N-propeptide. More recently, Cas9-based approaches may make it
possible to introduce such epitopes directly in endogenous gene
loci [18, 19]. With necessary genes encoding antibody epitope-
tagged collagens in hand, we next engineer cell lines that stably and
inducibly express the tagged collagen proteins. As described below,
in HT-1080 cells, we used a transfection-based strategy to deliver
genes [15]. In Saos-2 cells, we used a lentivirus-based strategy
[15, 20]. Work in both cell types is described below. Either may
be used to introduce genes for other collagens or collagen disease-
causing variants into essentially any cell system of interest. An
advantage of HT-1080 cells for collagen-I studies is that, although
they are capable of synthesizing and secreting various collagen
types, they do not express collagen-I, and therefore the collagen-I
interactome can be readily enriched. On the other hand, Saos-
2 cells do endogenously express collagen-I. In such a system, it is
more straightforward to mimic autosomal dominant collagen-I
pathologies by simply introducing the disease-causing collagen-I
variant in a background of endogenous wild-type collagen-I expres-
sion. Note that we have found that adenoviruses may also be used
for robust transient expression of collagen (not described below, as
the focus is on stable cell line development). An adenovirus-based
strategy can be especially valuable for transient collagen variant
expression in primary cells.
3.1.1 Transfection Col1A1 and Col1A2 genes were obtained prior to transfer into an
Approach to Prepare Stable appropriate expression vector (Fig. 2a), as described below (see
HT-1080 Cells Inducibly Note 1).
Expressing Collagen Genes
1. Using appropriate restriction sites (e.g., BamHI and EcoRV),
insert a DNA sequence (IDT) encoding the preprotrypsin signal
sequence (PPT; to target collagen strands to the secretory path-
way), the desired epitope tag (e.g., HA or FLAG), and a NotI
restriction digestion site (i.e., PPT.HA.NotI or PPT.FLAG.
NotI) into a pTRE-Tight vector (see Notes 2 and 3).
2. PCR-amplify (using the primers shown below which remove
the native signal sequence) collagen-I genes over 35–40 PCR
cycles. For amplification of collagen-I genes, we recommend
the use of Q5 Polymerase with the addition of the High GC
Enhancer (protocol from NEB) and a 2 min extension cycle (see
Note 4).
Col1A1: For. 50 -ACATCAGCGGCCGCACAAGAGGAAGG
CCAAGTCGAG-30 .
Rev. 50 - AAAAAAGTCGACTTACAGGAAGCAGACAGGG-30 .
102 Ngoc-Duc Doan et al.
Fig. 2 (a) Generalized schematic for collagen-I expression constructs. Either the
endogenous collagen signal sequence or the preprotrypsin signal sequence can
be employed. The tag can be HA, FLAG, c-MYC, or any of a number of other short
antibody epitope tags. (b) Structure of dithiobis(succinimidyl propionate) (DSP). (c)
Optimizing DSP cross-linker concentration by evaluating the co-immunoprecipitation
of known collagen-I interactors in the HT-1080 cell system. The control sample
represents HT-1080 cells that do not express HA-tagged collagen-I upon induction.
This figure was adapted from DiChiara et al. [15]
3.1.2 Lentiviral Approach 1. Construct pENTR1A vectors encoding the Col1A1 and
to Prepare Stable Saos- Col1A2 genes by ligating Col1A1 or Col1A2 extracted from
2 Cells Inducibly pTRE.Tight vectors (described above) using the NotI and
Expressing Collagen Genes EcoRV restriction enzymes into a pENTR1A vector already
encoding an ER-targeting PPT signal sequence and an HA
antibody epitope tag (or other desired tag; see Note 7). All
pENTR1A vectors must be sequence-confirmed before
proceeding.
2. Introduce the resulting construct into the appropriate pLenti.
CMV/TO.DEST Gateway destination vector via LR clonase-
mediated recombination (protocol from Thermo Fisher Scien-
tific). The resulting pLenti vectors must be sequence-
confirmed before proceeding to lentiviral production.
3. Produce lentivirus using the third-generation system by
co-transfecting HEK293FT cells with the lentiviral plasmids
and packaging vectors encoding RRE, REV, and VSVG using
Lipofectamine 2000. Briefly, (1) incubate the plasmid mixture
(pLenti vector, 15 μg; RRE, 15 μg; REV, 6 μg; and VSVG,
3 μg) with 60 μL of Lipofectamine 2000 in 3 mL of Opti-MEM
media for 45 min at room temperature (RT); (2) add the
resulting mixture dropwise to a 10 cm dish of HEK293FT
cells (~8 106 cells plated on poly-D-lysine-coated plates
~12 h pre-transfection) and incubate at 37 C for 12 h;
(3) remove and replace media with DMEM (6 mL) and incu-
bate the plates for another 36 h, monitoring for successful
production of lentivirus using Lenti-X GoStix (protocol from
Takara); (4) collect viral supernatant 48 h post-transfection;
and (5) use supernatant immediately for transductions of Saos-
2-TREx cells.
4. Transduce Saos-2TREx (see Note 8) cells with a range of titers
of crude viral supernatants of Tet-responsive HA-collagen-α1
(I). Add polybrene to the viral supernatant (final concentration
of 4 μg/mL during transductions) to increase transduction
efficiency.
104 Ngoc-Duc Doan et al.
3.2 Immunoisolation The protocol below uses Saos-2TREx cells inducibly expressing
of the Collagen-I PPT.HA.Colα1(I) (Saos-2Colα1(I) cells) as an example but can be
Interactome adapted for essentially any cell type of interest. Note that a control
for Proteomic Analysis IP should also always be performed on cells not expressing the
antibody epitope-tagged collagen.
1. Plate Saos-2Colα1(I) cells (~6 106) in 10 cm dishes with 10 mL
of DMEM supplemented with 15% heat-inactivated FBS,
100 IU/mL penicillin, 100 μg/mL streptomycin, 150 μg/
mL hygromycin, and 2 mM L-glutamine. Maintain cells as a
monolayer overnight at 37 C in a humidified atmosphere of
5% CO2(g).
2. Induce HA-tagged wild-type collagen-I expression by over-
night incubation with media containing 4 μg/mL doxycycline
(see Note 10) in the presence of 50 μM ascorbate, a required
cofactor for proper collagen hydroxylation and for induction of
synthesis and secretion of the endogenous, untagged collagen-I
(see Note 11).
3. After 48 h, remove media, wash with PBS, and add 1 mL of
trypsin-EDTA for 5 min. Neutralize trypsin by adding 7 mL of
complete DMEM. Pellet cells by spinning at 1000 g for
5 min.
4. Remove media and wash cells twice with PBS at (RT).
5. Resuspend cell pellet in a 15 mL Falcon tube containing 10 mL
of PBS and an experimentally optimized concentration of DSP
(75 μM for Saos-2Colα1(I) cells; Fig. 2b). Rotate tubes for 30 min
at RT to allow the cross-linking to proceed (see Note 12).
6. Quench excess DSP by adding 1 mL of 1 M aqueous Tris,
pH 8, and rotating for 10–15 min at RT.
7. Pellet cells by centrifuging at 1000 g for 5 min at 4 C,
followed by removal of the supernatant.
8. Lyse cells by resuspending them in 1 mL of RIPA buffer at
4 C. Vortex vigorously for 30 s, and then immediately sonicate
(130 Watt, 20 kHz, 70% amplitude) for 6 cycles of one 15 s
pulse each, returning tube to ice for 15 s between pulse cycles.
Lysates should be clear and fluid after sonication (see Note 13).
9. Post-sonicating, place the lysate solution on ice for 30 min to
ensure complete lysis.
Mass Spectrometry-Based Proteomics to Define Intracellular Collagen Interactomes 105
3.4 Protein Digestion All procedures should be carried out at RT unless otherwise
and Mass specified.
Spectrometry-Based
1. Add 22.5 μL of 8 M aqueous urea to the precipitated protein
Proteomic Studies pellet from Subheading 3.3, step 10. Vortex vigorously
3.4.1 Trypsin Digestion 3 30 s. Centrifuge at 3000 g for 1 min.
of Protein Pellets 2. Add 2.5 μL of 100 mM DTT dissolved in NH4HCO3 buffer to
and Preparation yield a final concentration of 10 mM DTT.
of Desalted Peptides
3. Incubate at 56 C for 1 h.
for Mass Spectrometry
Analysis 4. Cool samples to RT, and centrifuge at 3000 g for 1 min to
collect any H2O condensed on the internal surface of the tube.
5. Add 2.75 μL of 550 mM iodoacetamide to yield a final con-
centration of 55 mM iodoacetamide. Wrap tubes in foil to
protect from light, and mix end-over-end for 45 min (see
Note 20).
6. Add 169.75 μL of NH4HCO3 buffer. Vortex briefly, and then
add 2.5 μL of sequencing grade modified trypsin (0.4 μg/μL)
to yield a final volume of 200 μL (see Note 21).
7. Mix end-over-end at RT for 12 h or for 3 h at 37 C with
agitation on a thermomixer with hot plate.
8. Acidify the sample by adding 11 μL of 98% formic acid to
achieve a final concentration of 5% formic acid.
9. Remove white tip from the bottom of the peptide desalting
spin column. Place column into a 2 mL microcentrifuge tube.
Centrifuge at 3000 g for 1 min and discard flow through.
10. Wash the column with 300 μL of TFA/MeCN buffer, centri-
fuge at 3000 g for 1 min, and discard flow through (see Note
22). Repeat once.
11. Wash the column with 300 μL TFA buffer, centrifuge at
3000 g for 1 min, and discard flow through. Repeat once.
12. Add sample to the column, centrifuge at 3000 g for 3 min,
and discard flow through. Repeat centrifugation if not all of the
sample passed through the column (see Note 23).
13. Wash the sample on the column by adding 50 μL of formic acid
buffer. Centrifuge at 500 g for 3 min. Repeat twice.
14. Place column into a fresh microcentrifuge tube, and elute the
peptides from the column by adding 50 μL of formic acid/
MeCN buffer. Centrifuge at 3000 g for 3 min. Repeat this
elution step once.
15. Evaporate solvent in a vacuum centrifuge until near dryness.
Store at 20 C until mass spectrometry analysis.
Mass Spectrometry-Based Proteomics to Define Intracellular Collagen Interactomes 107
3.4.2 Mass Spectrometry 1. Solubilize digested peptide mixture into a working volume
and Data Analysis (we typically use 30 μL) of HPLC-grade 0.1% aqueous formic
acid by vortexing 3 for 30 s each.
2. Inject samples (typically 1–5 μL volumes are reasonable) into a
C18 reversed-phase resin (3 μM resin; 120 Å pore size) column
(15 cm in length; 50 μM in diameter) on HPLC with auto-
sampler connected to high-resolution, accurate mass/mass
spectrometer. Elute peptides into the mass spectrometer for
analysis over a 140-min gradient of formic acid buffer ranging
to a 95:5 MeCN:H2O in 0.1% formic acid solution (see Notes
24 and 25).
3. Operate the mass spectrometer in data-dependent mode with a
full scan MS spectrum followed by MS/MS for the top ten
precursor ions in each cycle. Depending on instrumentation,
the number of precursor ions selected in each cycle may be
increased.
4. Analyze raw data using Proteome Discoverer 2.2, performing a
database search using Mascot for protein identification
[21]. Depending on instrumentation and study objectives,
search parameters and data analysis used may vary. For qualita-
tive mapping of the collagen-I interactome in these example
cellular systems, Mascot search parameters typically used were
as follows: mass tolerance for precursor ions, 10 ppm; fragment
ion mass tolerance, 15 mmu; missed cleavages of trypsin, 2;
fixed modifications were carbamidomethylation of cysteine;
and variable modifications were methionine oxidation and
hydroxylation of proline, lysine, aspartate, and/or asparagine.
Peptides with unambiguous peptide spectrum match, Mascot
scores 25, and isolation inference 30 were considered iden-
tified, resulting in an average false discovery rate of 0.0077 (see
Notes 26 and 27).
4 Notes
Acknowledgments
References
Abstract
Global characterization of protein N termini provides valuable information on proteome dynamics and
diversity in health and disease. Driven by the progress in mass spectrometry-based proteomics, novel
approaches for the dedicated investigation of protein N termini and protease substrates have been recently
developed. Terminal amine isotopic labeling of substrates (TAILS) is a quantitative proteomics approach
suitable for high-throughput and system-wide profiling of protein N termini in complex biological matri-
ces. TAILS employs isotopic labeling of primary amines of intact proteins in combination with an amine-
reactive high molecular weight polymer (HPG-ALD) for depletion of internal tryptic peptides and high
enrichment of protein N termini by negative selection. Thereby, TAILS allows simultaneous identification
of the natural N termini, protease-generated neo-N termini, and endogenously modified (e.g., acetylated)
N termini. In this chapter, we provide a protocol for tandem mass tag (TMT)-TAILS analysis and further
discuss specific considerations regarding N-terminome data interpretation using Proteome Discoverer™
software.
1 Introduction
Irit Sagi and Nikolaos A. Afratis (eds.), Collagen: Methods and Protocols, Methods in Molecular Biology, vol. 1944,
https://doi.org/10.1007/978-1-4939-9095-5_8, © Springer Science+Business Media, LLC, part of Springer Nature 2019
115
116 Elizabeta Madzharova et al.
Fig. 1 Standard TAILS workflow. The TAILS experiment starts with the exposure of a complex proteome to a
specific test protease. Next, all primary amines (α-amines on the N-terminal and ε-amines of lysine side
chains) are labeled with isobaric tags (TMT or iTRAQ, indicated by multicolored diamonds), and samples are
TMT-TAILS N-Terminomics 117
Fig. 1 (continued) pooled and trypsinized. The newly generated internal tryptic and C-terminal peptides are
depleted using a high-molecular aldehyde-derivatized polymer (HPG-ALD) that binds to the newly generated
free amino group at the N terminus. The labeled N termini and neo-N termini, as well as naturally blocked N
termini (acetylation, cyclization, and methylation, indicated by black square), do not react with the polymer
and are recovered by ultrafiltration. The enriched N-terminome is analyzed by tandem mass spectrometry,
which discriminates between protease-generated neo–N termini and mature protein N termini with help of
quantitative information from TMT/iTRAQ reporter ions
118 Elizabeta Madzharova et al.
2 Materials
2.5 Data Analysis 1. Thermo Scientific™ Proteome Discoverer software v2.2 avail-
able at https://www.thermofisher.com/order/catalog/prod
uct/OPTON-30795.
2. SEQUEST database search engine.
3 Methods
3.1 Proteolysis by The following steps are only required if the proteome is to be
Test Protease exposed to a protease of interest in vitro. For other studies, which
(Optional) focus only on the nature of the N-terminome or using samples that
120 Elizabeta Madzharova et al.
3.3 StageTip 1. Stamp out two pieces of a C18 Empore disk using a blunt-
Desalting ended syringe needle, and press the cored pieces close to the
end of a P200 pipette tip.
122 Elizabeta Madzharova et al.
3.4 LC-MS/MS Analyze preTAILS and TAILS samples with a tandem mass spec-
Analysis trometer coupled to a C18 chromatography system. Due to the
large variation of instrumental setups, details of the mass spectrom-
etry analysis are specific to the instrument used. As an example,
peptides from our dataset were analyzed on a Thermo Scientific Q
Exactive Mass Spectrometer coupled to a Thermo Scientific EASY-
nLC™ 1000 Liquid Chromatography system and a Thermo Scien-
tific PepMap™ RSLC C18 column (2 μm, 100 Å, 75 μm 50 cm).
For each analysis, 2 μg of peptide mixture was loaded at a constant
flow rate of 250 nL/min in Buffer A and eluted using a 2-h
gradient from 6% to 95% Buffer B. A data-dependent acquisition
(DDA) mode was applied to record the spectra with the following
MS1 parameters, scan range 300–1750 m/z, resolution of 70,000,
AGC target 3e6; MS2 parameters, resolution of 17,500, AGC
target 1e6; and higher energy collision dissociation (HCD), nor-
malized collision energy of 32%.
3.5 N-Terminome A variety of tools are available for proteomic assessment of TAILS
Data Analysis N-terminomics data with specific search settings, which vary
depending on the algorithm and the instrument used for data
acquisition [11–15]. Here we describe in detail a workflow for
the analysis of a TMT-TAILS dataset collected after treatment of
secretomes from murine embryonic fibroblasts with GluC endo-
peptidase (test protease) using Proteome Discoverer™ 2.2
software.
1. Open Proteome Discoverer™ 2.2 application and select New
Study/Analysis on the start page.
2. Choose a name for the study and a root directory. Click OK.
3. Select the TMTe 6plex as a quantification method to be used in
the study.
TMT-TAILS N-Terminomics 123
4. Click Add Files and select the mass spectrometry raw file.
Important: Proteome Discoverer accepts .raw, .mgf, .mzData,
.mzXML, and .mzML file formats as an input. Optional: As a
test run, use our MS raw file (available upon request from the
corresponding author).
5. Specify the quantification method used in the input file on the
Input Files page.
6. On the Samples tab, specify samples as “control” and/or
“sample” type.
7. Open New Analysis, select and drag the input file from the
input file page to the Analysis window.
8. Select Workflows tab to create workflows to use for the pro-
cessing and consensus step. Optional: Use our processing and
consensus workflow files (available upon request from the
corresponding author).
9. Create Processing Step Workflow by dragging and connect-
ing the following nodes into the Workflow tree window
(Fig. 2a):
l Spectrum File.
l Spectrum Selector.
l Sequest HT.
l Percolator.
l Reporter Ions Quantifier.
10. For N termini profiling, set the parameters for Sequest HT
node (see Note 5) (Fig. 2b):
l Upload an appropriate database.
l Choose enzyme semi-specific ArgC with one missed cleav-
age (see Note 6).
l Set minimum peptide length of 6 amino acids and maxi-
mum peptide length of 30 amino acids.
l Set precursor mass tolerance at 10 ppm and fragment mass
tolerance at 0.02 Da.
l Set TMT6plex lysine (+229.163 Da) and carboxymethyl of
cysteine (+58.005 Da) as static modifications.
l Set TMT-labeled N-termini (+229.163 Da), oxidation of
methionine (+15.995 Da), N-terminal acetylation
(+42.010565 Da), N-terminal pyro-Glu ( 17.026549
Da), or asparagine deamidation (+ 0.984 Da) as dynamic
modifications.
11. Create Consensus Step Workflow by dragging and connect-
ing the following nodes into the Workflow tree window
(Fig. 2c):
124 Elizabeta Madzharova et al.
Fig. 2 Proteome Discoverer settings. (A) Processing Step Workflow nodes. (B) Sequest HT parameters for
TAILS analysis of mouse secretome. (C) Consensus Step Workflow nodes
TMT-TAILS N-Terminomics 125
l MSF Files.
l PSM Grouper.
l Peptide Validator.
l Peptide and Protein Filter.
l Protein Scorer.
l Protein Grouping.
l Peptide in Protein Annotation.
l Reporter Ions Quantifier.
l Protein FDR Validator.
l As for post-processing nodes, Data Distributions, and Dis-
play Settings.
12. In Grouping and Quantification tab, specify the ratios to
report for quantification and how to group the samples with
respect to the specified factor values.
13. Select Sample type as study variable.
14. Choose Sample type: Control denominators to be used for
bulk ratio generator and press Add Ratio.
15. Press Run in the Analysis window to start the search.
Output of analysis results should be inspected on the “Peptide
Group” level for interpretation of N-terminal peptides and can be
exported to appropriate formats for further analysis, e.g., using
spreadsheet applications. To annotate protein N termini for their
position in the mature protein, several tools are available including
ImproViser [16] and TAILS-ANNOTATOR [4].
4 Notes
References
1. Hartmann EM, Armengaud J (2014) analysis of MMP-2 and MMP-9 substrate
N-terminomics and proteogenomics, getting degradomes by iTRAQ-TAILS quantitative
off to a good start. Proteomics 14:2637–2646 proteomics. Mol Cell Proteomics 9:894–911
2. Lai ZW, Petrera A, Schilling O (2015) Protein 10. Kleifeld O, Doucet A, auf dem Keller U,
amino-terminal modifications and proteomic Prudova A, Schilling O, Kainthan RK et al
approaches for N-terminal profiling. Curr (2010) Isotopic labeling of terminal amines in
Opin Chem Biol 24:71–79 complex samples identifies protein N-termini
3. Yeom J, Ju S, Choi Y, Paek E, Lee C (2017) and protease cleavage products. Nat Biotech-
Comprehensive analysis of human protein nol 28:281–288
N-termini enables assessment of various pro- 11. Cox J, Mann M (2008) MaxQuant enables
tein forms. Sci Rep 7:6599 high peptide identification rates, individualized
4. Kleifeld O, Doucet A, Prudova A, auf dem p.p.b.-range mass accuracies and proteome-
Keller U, Gioia M, Kizhakkedathu JN, Overall wide protein quantification. Nat Biotechnol
CM (2011) Identifying and quantifying pro- 26:1367–1372
teolytic events and the natural N terminome by 12. Deutsch EW, Mendoza L, Shteynberg D,
terminal amine isotopic labeling of substrates. Farrah T, Lam H, Tasman N et al (2010) A
Nat Protoc 6:1578–1611 guided tour of the trans-proteomic pipeline.
5. Marino G, Eckhard U, Overall CM (2015) Proteomics 10:1150–1159
Protein termini and their modifications 13. Craig R, Beavis RC (2004) TANDEM: match-
revealed by positional proteomics. ACS Chem ing proteins with tandem mass spectra. Bioin-
Biol 10:1754–1764 formatics 20:1466–1467
6. van den Berg BHJ, Tholey A (2012) Mass 14. auf dem Keller U, Prudova A, Gioia M, Butler
spectrometry-based proteomics strategies for GS, Overall CM (2010) A statistics-based plat-
protease cleavage site identification. Proteo- form for quantitative N-terminome analysis
mics 12:516–529 and identification of protease cleavage pro-
7. Rogers LD, Overall CM (2013) Proteolytic ducts. Mol Cell Proteomics 9:912–927
post-translational modification of proteins: 15. auf dem Keller U, Overall CM (2012) CLIP-
proteomic tools and methodology. Mol Cell PER: an add-on to the trans-proteomic pipe-
Proteomics 12:3532–3542 line for the automated analysis of TAILS
8. Schlage P, Egli FE, Nanni P, Wang LW, Kiz- N-terminomics data. Biol Chem
hakkedathu JN, Apte SS, auf dem Keller U 393:1477–1483
(2014) Time-resolved analysis of the matrix 16. Videm P, Gunasekaran D, Schröder B, Mayer B,
metalloproteinase 10 substrate degradome. Biniossek ML, Schilling O (2014) Automated
Mol Cell Proteomics 13:580–593 peptide mapping and protein-topographical
9. Prudova A, auf dem Keller U, Butler GS, Over- annotation of proteomics data. BMC Bioinfor-
all CM (2010) Multiplex N-terminome matics 15:207
Part III
Abstract
Scanning electron microscopy is a useful tool for high-resolution morphological characterization of the
extracellular matrix. In certain tissues and surfaces, imaging of the extracellular matrix requires cell removal
before sample preparation. In this protocol, we will describe a method for preparing extracellular matrices
derived from murine colon for imaging under a scanning electron microscope.
Key words Tissue, Colon, Scanning electron microscope, Extracellular matrix, Decellularization
1 Introduction
Irit Sagi and Nikolaos A. Afratis (eds.), Collagen: Methods and Protocols, Methods in Molecular Biology, vol. 1944,
https://doi.org/10.1007/978-1-4939-9095-5_9, © Springer Science+Business Media, LLC, part of Springer Nature 2019
129
130 Elee Shimshoni and Irit Sagi
Fig. 1 Scanning electron micrographs of a murine colon with or without cells colon samples were either
prepared as described in this protocol (image on the right), or were immediately fixed after harvest and stained
with an additional dye that binds to cell membranes (OsO4). Both images were taken in a 2500 magnifica-
tion. Images demonstrate the mucosal surface with and without cells, displaying the colonic crypts. As evident
by the images, the extracellular matrix scaffold underlying the gut epithelium that can be observed in the
decellularized sample is covered by cells in the sample presented on the left
derived from murine colon for imaging of the under SEM using the
following processes – decellularization, fixation, staining, dehydra-
tion, and mounting.
2 Materials
3 Methods
3.1 Decellularization 1. Cut 5–10 mm pieces (in length) of fresh or snap-frozen colon.
2. Wash samples twice with PBS, clearing out any residual feces.
3. Wash samples twice with ddH2O.
4. Place samples in an Eppendorf tube with ddH2O.
5. Freeze samples on dry ice (see Note 1) in ddH2O.
6. Thaw at room temperature and replace ddH2O.
7. Repeat steps 4 and 5 five more times (six times in total).
8. Immerse samples in the ammonium hydroxide solution for
20 min in a rotator.
9. Wash six times in ddH2O (see Note 2).
Fig. 2 Sample mounting after critical point drying Dehydrated colon samples should be mounted as illustrated.
First the colon tube is placed gently on the carbon sticker stuck to the mount and allowed to adhere. Then the
colon is cut open using scissors while trying not to touch the bottom of the sample to not create dents in the
sample. Following, take forceps and gently pull the edges, and stick them to the sticker as well. Finally, stick
small pieces of carbon tape on the edges of the sample to ground the charges that will be interacting with
sample in the microscope
4 Notes
References
1. Hayat MA (2000) Principles and techniques of protocols in microbiology p. Unit 2B.2. John
electron microscopy: biological applications. Wiley & Sons, Hoboken, NJ
Cambridge University Press, Cambridge, UK 3. Brown BN, Barnes CA, Kasick RT et al (2010)
2. Fischer ER, Hansen BT, Nair V et al (2012) Surface characterization of extracellular matrix
Scanning electron microscopy. In: Current scaffolds. Biomaterials 31:428–437
SEM Sample Preparation of ECM 133
Abstract
Degraded and denatured collagens are useful markers for physiological events (e.g., bone formation and
aging) and pathologic conditions (e.g., cancer, arthritis, and fibrosis). Here we describe histological staining
of such collagens using fluorescent collagen hybridizing peptide that can specifically bind to collagen
strands by folding into triple helix. The method can report the amount of denatured collagen and/or
collagen remodeling activity in tissues via localized fluorescence intensity and can be used in conjunction
with conventional staining agents. The collagen hybridizing peptide probes can be used across species and
collagen types, providing a versatile tool not only for pathology and developmental biology but also
histology-based disease diagnosis, staging, and therapeutic screening.
Key words Matrix metalloproteinase, Collagenase, Collagen fiber, Collagen remodeling, Bone
resorption, Immunostaining
1 Introduction
Irit Sagi and Nikolaos A. Afratis (eds.), Collagen: Methods and Protocols, Methods in Molecular Biology, vol. 1944,
https://doi.org/10.1007/978-1-4939-9095-5_10, © Springer Science+Business Media, LLC, part of Springer Nature 2019
135
136 Yang Li and S. Michael Yu
Fig. 1 (a) Schematic of a CHP strand (labeled with X) hybridizing to denatured collagen chains and forming a
collagen triple helix. During disease progression, tissue development, or aging, collagen can be extensively
degraded by collagen-cleaving proteases, causing its triple helix to unfold at the physiological temperature
due to thermal instability. X represents the biotin or fluorescent tag. (b) The thermal activation step used to
generate monomeric CHP strands for collagen hybridization. A dilute CHP solution (15–30 μM) was heated in a
water bath at 80 C for 5 min to dissociate the folded CHP homotrimers. The hot solution was then quenched
in an ice-water bath to room temperature. The solution of monomeric CHP was then ready for immediate use
to hybridize with denatured collagen in tissue samples. The self-trimerizing of CHP takes several hours to
complete in this concentration range, and the effect of the dead time due to quenching (< 3 min) is negligible.
The figure is reproduced from ref. 4 with permission from American Chemical Society
during a polymerase chain reaction (Fig. 1a) [4–6]. Due to the lack
of hybridization sites, the CHP has no affinity toward native, fully
folded collagen molecules [5]. Furthermore, the neutral and
hydrophilic amino acid composition of CHP makes it virtually
nonadherent to other biomolecules [10].
In this chapter, we present detailed procedures for detecting
degraded and denatured collagen in histology slides using biotin-
labeled CHP. The key difference between this method and a stan-
dard immunostaining protocol is that the CHP probe requires a
special heating-and-quenching step before application to samples
(see Fig. 1b). Because of the high triple helix propensity, the CHP
peptides tend to self-assemble into homotrimers in solution over
time (e.g., during storage at 4 C) and lose the driving force for
collagen hybridization. Therefore, the trimeric CHP needs to be
thermally dissociated to single strands by heating briefly at 80 C
before it can be used to bind to unfolded collagen [4]. The hot
CHP solution is quenched quickly to room temperature immedi-
ately prior to addition to tissue samples to prevent undesired ther-
mal denaturation of the tissue. Since the CHP trimerization has a
third-order folding rate with a halftime on the order of hours at low
μM concentrations [11, 12], the heating-and-quenching method
(Fig. 1b) described in this protocol ensures that the active mono-
meric form of CHP is delivered to the denatured collagen allowing
efficient hybridization.
2 Materials
3 Methods
3.1 Slide Preparation 1. Fill two tissue-slide-staining jars with each of the following
and Prestaining Steps solvents: xylene, 200 proof ethanol, 190 proof ethanol, 50%
ethanol, and deionized water. One hundred milliliter of a sol-
vent is added to each jar. Label the jars with their contents.
2. Place the paraffin-embedded slides onto a staining rack, and
immerse the rack in a xylene-containing jar.
3. Incubate the slides for two 5 min cycles in each solvent, follow-
ing a consecutive order of xylene, 200 proof ethanol, 190 proof
ethanol, 50% ethanol, and deionized water. After the final cycle
of washing in water, these deparaffinized slides can be directly
stained by CHP without any antigen-retrieval process. For
OCT-embedded frozen slides (see Note 5).
4. Fill the bottom of the tissue-staining chamber with water.
5. Using the PAP pen, draw a circle around the specimen(s) on
each slide as a hydrophobic barrier to keep staining reagents
localized on the tissue sections. Align the slides on the rack
inside the staining chamber.
6. Add an enough volume of blocking solution to each tissue
sections to fill the space within the barrier circle. To estimate
the volume of solution needed to cover all slides (see Note 4).
Incubate the slides with the blocking solution in the staining
chamber for 20 min to block the non-specific binding sites.
7. Remove the blocking solution by blotting the slides on a paper
towel.
8. Rinse the slides in 100 mL of 1 PBS in a slide-staining jar for
5 min. Rinse three times. (See Note 6, for the optional biotin-
blocking steps.)
9. Blot the slides on a paper towel, and align the slides on the rack
inside the staining chamber. The slides are now ready to be
stained by CHP.
3.2 Collagen 1. Turn on the temperature-controlled water bath, and set the
Hybridizing Peptide temperature to 80 C (see Note 7).
Staining 2. Fill the ice bucket or a glass beaker with ice and water to make
an ice-water bath.
3. Ensure the microtube containing the diluted B-CHP staining
solution is capped tightly. Add the tube onto a floating rack,
and place the rack into the 80 C water bath.
4. Allow the B-CHP staining solution to be heated for 5 min (see
Note 8) to dissociate the trimeric peptides into single strands
(see Fig. 1b).
5. Following heating, immediately vortex and centrifuge the tube
for 10 s to collect the condensation inside the tube.
140 Yang Li and S. Michael Yu
4 Notes
Fig. 2 The optimization tests for CHP staining. (a) Fluorescence images of neighboring cryosections of a
porcine ligament stained with 100 μL of F-CHP solutions at varying concentrations. Before staining, all slides
were denatured in 80 C water to purposefully denature the collagen molecules for CHP hybridization. The
staining time was set at 2 h for all samples. (b) Fluorescence images of a set of heated ligament tissue slides
stained with F-CHP (100 μL, 15 μM) for different incubation time periods. Scale bars: 2 mm. The figure is
reproduced from ref. 4 with permission from American Chemical Society
Fig. 3 Histological visualization of collagen degradation during endochondral ossification using CHP. (a)
Localization of CHP binding in a sagittal section of an 18 d.p.c. mouse embryo double stained with B-CHP
(detected by AlexaFluor647-streptavidin, orange) and an anti-collagen I antibody (detected by AlexaFluor555-
labeled donkey anti-rabbit IgG H&L, cyan). mx maxilla, md mandibular bone, bp basisphenoid bone, bo
basioccipital bone, vc vertebral column, rb rib, h hipbone, d digital bones. (b) Magnified views of the
basioccipital bone (bo) beside the C1 vertebra (v) (top images) and the manubrium sterni (ms, bottom images)
in the sagittal section of a 17 d.p.c. mouse embryo stained in the same fashion. High level of CHP binding is
found in the hypertrophic zone surrounding the newly deposited collagen I bone matrix (yellow arrow heads),
which is visualized by the anti-collagen I antibody. Scale bars: 3 mm (a), 0.5 mm (b). The figure is reproduced
from ref. 4 with permission from American Chemical Society
Acknowledgments
References
1. Bonnans C, Chou J, Werb Z (2014) Remodel- collagen prior to peptide bond hydrolysis.
ling the extracellular matrix in development EMBO J 23:3020–3030
and disease. Nat Rev Mol Cell Biol 8. Danielsen CC (1987) Thermal stability of
15:786–801 human-fibroblast-collagenase-cleavage pro-
2. Page-McCaw A, Ewald AJ, Werb Z (2007) ducts of type-I and type-III collagens. Biochem
Matrix metalloproteinases and the regulation J 247:725–729
of tissue remodelling. Nat Rev Mol Cell Biol 9. Fisher GJ, Kang S, Varani J et al (2002)
8:221–233 Mechanisms of photoaging and chronological
3. Wahyudi H, Reynolds AA, Li Y et al (2016) skin aging. Arch Dermatol 138:1462–1470
Targeting collagen for diagnostic imaging and 10. Li Y, Ho D, Meng H et al (2013) Direct detec-
therapeutic delivery. J Control Release tion of collagenous proteins by fluorescently
240:323–331 labeled collagen mimetic peptides. Bioconjug
4. Hwang J, Huang Y, Burwell TJ et al (2017) In Chem 24:9–16
situ imaging of tissue remodeling with collagen 11. Ackerman MS, Bhate M, Shenoy N et al (1999)
hybridizing peptides. ACS Nano Sequence dependence of the folding of
11:9825–9835 collagen-like peptides. Single amino acids
5. Li Y, Foss CA, Summerfield DD et al (2012) affect the rate of triple-helix nucleation. J Biol
Targeting collagen strands by photo-triggered Chem 274:7668–7673
triple-helix hybridization. Proc Natl Acad Sci U 12. Boudko S, Frank S, Kammerer RA et al (2002)
S A 109:14767–14772 Nucleation and propagation of the collagen
6. Li Y, Yu SM (2013) Targeting and mimicking triple helix in single-chain and trimerized pep-
collagens via triple helical peptide assembly. tides: transition from third to first order kinet-
Curr Opin Chem Biol 17:968–975 ics. J Mol Biol 317:459–470
7. Chung L, Dinakarpandian D, Yoshida N et al
(2004) Collagenase unwinds triple-helical
Chapter 11
Abstract
An important issue in tissue biomechanics is to decipher the relationship between the mechanical behavior
at macroscopic scale and the organization of the collagen fiber network at microscopic scale. Here, we
present a protocol to combine traction assays with multiphoton microscopy in ex vivo murine skin. This
multiscale approach provides simultaneously the stress/stretch response of a skin biopsy and the collagen
reorganization in the dermis by use of second harmonic generation (SHG) signals and appropriate image
processing.
1 Introduction
Irit Sagi and Nikolaos A. Afratis (eds.), Collagen: Methods and Protocols, Methods in Molecular Biology, vol. 1944,
https://doi.org/10.1007/978-1-4939-9095-5_11, © Springer Science+Business Media, LLC, part of Springer Nature 2019
145
146 Stéphane Bancelin et al.
2 Materials
2.1 Traction Device 1. Symmetrical uniaxial traction machine using two identical
motors and sensors, one of each side of the skin sample.
2. Motors: Linear actuators. Resolution below 5 μm, accuracy
below 5 μm, strokes above 40 mm, maximal holding force
above 10 N. Speed between 1 and 100 μm/s (or above).
3. Sensors: Capacity at least 8 N. Resolution below 0.01 N. Over-
load should be important (more than 80 N) so as to avoid
breaking the sensors when screwing the sample in the grips.
Fig. 1 Experimental setup combining multiphoton microscopy and uniaxial traction assay. (a) Schematic
representation and (b) picture. Adapted from Bancelin et al., Sci Rep 2015, doi:https://doi.org/10.1038/
srep17635, published under license CC BY 4.0
3 Methods
3.2 Skin Cutting 1. The skin sample is cut into a dog-bone shape using curved
and Fastening dissection scissors, taking care to hold the sample on the out-
for Traction Assays skirt part (not used in mechanical test) so as to damage the
sample as little as possible (Fig. 2) (see Note 6).
148 Stéphane Bancelin et al.
Fig. 2 Preparation of the depilated and de-epidermized skin sample for mechan-
ical test. (a) Schematic representation and dimensions of the dog-bone-shaped
skin sample. (b) Picture before cutting. The orange arrow shows the black spot
indicating the right foreleg. The red dots show the dog-bone shape the skin will
be cut into. Typical size is 30 20 mm2. Adapted from Bancelin et al., Sci Rep
2015, doi:https://doi.org/10.1038/srep17635, published under license CC BY 4.0
3.3 Traction Assays 1. The motor displacement and the force are measured continu-
Under the Multiphoton ously (every second) during the whole traction assay (Fig. 3).
Microscope 2. SHG imaging is performed continuously while adjusting the
axial position of the objective lens to find the surface of the skin
sample and focus a few μm below.
3. The jaws are moved apart slowly (usually around 0.01 mm s1)
while continuously recording low-resolution SHG images until
Multiscale Skin Biomechanics 149
Fig. 3 Mechanical data from wild-type skin sample. (a) Experimental timetable showing stepwise stretching of
the skin sample (green dotted line) with continuous force measurement (blue continuous line) and time-lapse
imaging of immobile sample after each displacement step (red arrows). (b) Nominal stress/stretch curve for
the same sample, showing the processed mechanical parameters. Adapted from Bancelin et al., Sci Rep
2015, doi:https://doi.org/10.1038/srep17635, published under license CC BY 4.0
3.4 Mechanical Data 1. The global stretch ratio is obtained as the ratio of the length
Processing between the rubber joints of the jaws to the reference length that
is the one measured at the reference position (see Note 12).
150 Stéphane Bancelin et al.
Fig. 4 SHG imaging and image processing of ex vivo stretched skin sample. (a) Raw SHG images at 1.0, 1.2,
and 1.4 stretch ratio. The deformation of the hair follicle network reveals the local skin deformation. (b)
Processed SHG images at the same stretch ratios highlighting fiber orientation for each pixel with a color code.
Adapted from Bancelin et al., Sci Rep 2015, doi:https://doi.org/10.1038/srep17635, published under license
CC BY 4.0
3.5 Image 1. SHG images at approximately the same depth are cropped to
Processing for Local obtain a ROI common to all stretching steps showing the same
Deformation Analysis hair follicles (round or elliptical regions with no SHG, inter-
rupting the collagen fiber network, Fig. 4). The size of this
ROI increases in the loading direction at increasing stretch
ratio.
2. The hair follicles are segmented using the following steps
(Fig. 5): application of a median filter to reduce noise (2 pixel
radius), inversion of contrast, application of circular opening
(8 pixel radius) [19] to smooth the image while preserving
follicle edges, thresholding resulting in a binary image, and
shape detection to identify and label every follicle. If needed,
Multiscale Skin Biomechanics 151
Fig. 5 Hair follicle segmentation in the SHG image for local deformation measurement. (a) Raw and (b)
enhanced SHG image of mice skin papillary dermis; (c) binary SHG image after thresholding; (d) position
detection and numbering of hair follicles; mapping of (e) λxx, (f) λyy, and (g) ω tensorial components calculated
from the deformation of the hair follicle network—see green triangle in (d)—compared to the initial network
before stretching. Scale bar: 50 μm. Reproduced from Bancelin et al., Sci Rep 2015, doi:https://doi.org/10.
1038/srep17635, published under license CC BY 4.0
3.6 Image 1. A fixed number of SHG images (for instance, 6) with maximum
Processing average intensity are selected in every SHG stack for all stretch-
for Collagen ing ratios.
Reorganization 2. Morphological filtering is applied to extract the fiber orienta-
tion in every pixel by means of a mean filter (3 pixel diameter)
and of a morphological opening [19] using a linear structuring
element (Strel; see Note 15). A stack of images is obtained by
152 Stéphane Bancelin et al.
and (3) the entropy, defined as the usual statistical entropy, which
measures the degree of organization in the skin sample indepen-
dently of any main orientation:
90∘
X
S¼ pðθÞ ln ½pðθÞ
θ¼90∘
I ðθÞ
where pðθÞ ¼ P90 .
θ¼90 I ðθÞ
4 Notes
Acknowledgments
References
1. Goulam Houssen Y, Gusachenko I, Schanne- 3. Zipfel WR, Williams RM, Christie R, Nikitin
Klein M-C, Allain J-M (2011) Monitoring AY, Hyman BT, Webb WW (2003) Live tissue
micrometer-scale collagen organization in intrinsic emission microscopy using
rat-tail tendon upon mechanical strain using multiphoton-excited native fluorescence and
second harmonic generation microscopy. J Bio- second harmonic generation. Proc Natl Acad
mech 44:2047–2052 Sci U S A 100:7075–7080
2. Bancelin S, Lynch B, Bonod-Bidaud C, 4. Strupler M, Pena A-M, Hernest M, Tharaux
Ducourthial G, Psilodimitrakopoulos S, P-L, Martin J-L, Beaurepaire E, Schanne-
Dokladal P, Allain J-M, Schanne-Klein M-C, Klein M-C (2007) Second harmonic imaging
Ruggiero F (2015) Ex vivo multiscale quanti- and scoring of collagen in fibrotic tissues. Opt
tation of skin biomechanics in wild-type and Express 15(7):4054–4065
genetically-modified mice using multiphoton
microscopy. Sci Rep 5:17635
Multiscale Skin Biomechanics 155
5. Chen XY, Nadiarynkh O, Plotnikov S, Cam- 13. Mauri A, Ehret AE, Perrini M, Maake C,
pagnola PJ (2012) Second harmonic genera- Ochsenbein-Kolble N, Ehrbar M, Oyen ML,
tion microscopy for quantitative analysis of Mazza E (2015) Deformation mechanisms of
collagen fibrillar structure. Nat Protoc 7 human amnion: quantitative studies based on
(4):654–669 second harmonic generation microscopy. J Bio-
6. Lynch B, Bancelin S, Bonod-Bidaud C, Gues- mech 48(9):1606–1613
quin J-B, Ruggiero F, Schanne-Klein M-C, 14. Alavi SH, Sinha A, Steward E, Milliken JC,
Allain J-M (2017) A novel microstructural Kheradvar A (2015) Load-dependent extracel-
interpretation for the biomechanics of mouse lular matrix organization in atrioventricular
skin derived from multiscale characterization. heart valves: differences and similarities. Am J
Acta Biomater 50:302–311 Physiol Heart Circ Physiol 309(2):
7. Jayyosi C, Affagard J-S, Ducourthial G, H276–H284
Bonod-Bidaud C, Lynch B, Bancelin S, 15. Nesbitt S, Scott W, Macione J, Kotha S (2015)
Ruggiero F, Schanne-Klein M-C, Allain J-M, Collagen fibrils in skin orient in the direction of
Bruyère-Garnier K, Coret M (2017) Affine applied uniaxial load in proportion to stress
kinematics in planar fibrous connective tissues: while exhibiting differential strains around
an experimental investigation. Biomech Model hair follicles. Materials 8(4):1841–1857
Mechanobiol 16(4):1459–1473 16. Caulk AW, Nepiyushchikh ZV, Shaw R, Dixon
8. Lynch B, Bonod-Bidaud C, Ducourthial G, JB, Gleason RL (2015) Quantification of the
Affagard J-S, Bancelin S, passive and active biaxial mechanical behaviour
Psilodimitrakopoulos S, Ruggiero F, Allain and microstructural organization of rat tho-
J-M, Schanne-Klein M-C (2017) How aging racic ducts. J R Soc Interface 12(108). ARTN
impacts skin biomechanics: a multiscale study 20150280
in mice. Sci Rep 7:13750 17. Benoit A, Latour G, Schanne-Klein M-C,
9. Sinclair EB, Andarawis-Puri N, Ros SJ, Laudier Allain J-M (2016) Simultaneous microstruc-
DM, Jepsen KJ, Hausman MR (2012) Relating tural and mechanical characterization of
applied strain to the type and severity of struc- human corneas at increasing pressure. J Mech
tural damage in the rat median nerve using Behav Biomed Mater 60:93–105
second harmonic generation microscopy. Mus- 18. Krasny W, Morin C, Magoariec H, Avril S
cle Nerve 46(6):899–907 (2017) A comprehensive study of layer-specific
10. Wentzell S, Nesbitt RS, Macione J, Kotha S morphological changes in the microstructure
(2013) Measuring strain using digital image of carotid arteries under uniaxial load. Acta
correlation of second harmonic generation Biomater 57:342–351
images. J Biomech 46(12):2032–2038 19. Serra J (1982) Analysis and mathematical mor-
11. Chow MJ, Turcotte R, Lin CP, Zhang YH phology. Academic Press
(2014) Arterial extracellular matrix: a Mechan- 20. Bayan C, Levitt JM, Miller E, Kaplan D, Geor-
obiological study of the contributions and gakoudi I (2009) Fully automated, quantita-
interactions of elastin and collagen. Biophys J tive, noninvasive assessment of collagen fiber
106(12):2684–2692 content and organization in thick collagen
12. Sigal IA, Grimm JL, Jan NJ, Reid K, Minckler gels. J Appl Phys 105(10):102042
DS, Brown DJ (2014) Eye-specific IOP-in- 21. Zipfel WR, Williams RM, Webb WW (2003)
duced displacements and deformations of Nonlinear magic: multiphoton microscopy in
human Lamina Cribrosa. Invest Ophth Vis Sci the biosciences. Nat Biotech 21
55(1):1–15 (11):1369–1377
Chapter 12
Abstract
Integrin-mediated interactions of cells with the extracellular matrix (ECM) are important for their activ-
ities. The ECM itself is a complex network of macromolecules forming aggregates or suprastructures.
Moreover, the molecular composition is important for the macromolecular organization and, thereby, the
functional properties of the ECM. In addition, collagen molecules lose their integrin-binding capabilities
after incorporation into fibrils. Therefore, we have established detailed protocols for the analysis of integrin-
matrix interactions at the supramolecular level.
Key words Cell adhesion assay, Integrins, Matrix superstructures, Collagens, Chondrocytes, Trans-
mission electron microscopy
1 Introduction
Irit Sagi and Nikolaos A. Afratis (eds.), Collagen: Methods and Protocols, Methods in Molecular Biology, vol. 1944,
https://doi.org/10.1007/978-1-4939-9095-5_12, © Springer Science+Business Media, LLC, part of Springer Nature 2019
157
158 Uwe Hansen
2 Materials
10. Collagens:
Collagens II, IX, and XI in their native molar proportions of
ƒII,IX,XI ¼ 8:1:1.
Purified monomeric cartilage collagens II, IX, or XI.
Authentic fibrils fragments from cartilage of 17-day-old
chicken embryos or adult chicken, juvenile or adult bovine
knee joint cartilage or human osteoarthritic hip cartilage.
11. Tris buffer:
50 mM Tris–HCl, 150 mM NaCl, pH 7.4 (for collagen
purification).
50 mM Tris–HCl, 400 mM NaCl, pH 7.4 (for in-vitro
fibrillogenesis).
12. HEPES buffer: 0.1 M HEPES, 0.4 M NaCl, pH 7.4.
13. Staining wash buffer: 150 mM NaCl, 2 mM sodium phos-
phate, pH 7.4.
14. Adhesion blocking buffer: 2 mM Tris–HCl buffer, 150 mM
NaCl, pH 7.4.
15. Collagen reconstitute buffer: 50 mM HEPES, 200 mM NaCl,
pH 7.4.
3 Methods
3.2 Collagen Mixtures of cartilage collagens II, IX, and XI in their native molar
Purification proportions ƒII,IX,XI ¼ 8:1:1 were isolated from chicken embryo
chondrocytes cultured in suspension of agarose as described in
160 Uwe Hansen
3.5 Binding 1. Coat grids at room temperature for 15 min with purified
Experiments on Grids monomeric cartilage collagens II, IX, or XI in HEPES buffer
for Transmission in order to prevent fibril formation.
Electron Microscopy 2. Wash grids with PBS.
3.5.1 Purified Monomeric 3. Block for 60 min with 2% dried skimmed milk in PBS.
Cartilage Collagens II, IX, 4. Incubate with 375 μg/ml of soluble recombinant α1β1-, α2β1-,
or XI α10β1-integrins in the presence of the integrin-activating anti-
body 9EG7 and 2 mM MgCl2 or 10 mM EDTA (negative
control).
5. Wash with PBS.
6. Expose the grids to immuno-gold particles coated with anti-
IgG antibodies.
7. Stain samples with 2% uranyl acetate for 15 min.
8. Take electron micrographs from five randomly selected fields.
9. Count gold particles/field.
Integrin Interaction with Supramolecular Aggregates 163
3.5.2 Reconstituted 1. Reconstitute collagen fibrils from monomeric collagens II, IX,
Collagen Fibrils Containing and XI (300 μg/ml total concentration) as adhesion substrates
Collagen Types II, IX, and XI by adjusting the buffer salinity to collagen reconstitute buffer
and warm to 37 C as described above (see Note 6).
2. Apply 10 μl of 150 μg/ml total concentration of reconstituted
collagen fibril suspension onto the surface of 400 square mesh
uncoated nickel grids.
3. Wash the attached collagen fibrils five times with PBS in order
to remove remaining collagen monomers (see Note 7).
4. Block unspecific binding sites with 2% dried skimmed milk
in PBS.
5. Incubate the reconstituted collagen fibrils with integrins and
primary and secondary antibodies as described under 3.4.
6. Coat the grids with a thin layer of carbon generated on mica
plates and float on distilled water. Put the grid under the
carbon layer floating on water surface and take out both the
grid and the carbon layer.
7. Dry the grid carefully on filter paper.
8. Stain the samples with 2% (v/w) uranyl acetate.
9. Analyze samples by transmission electron microscopy.
4 Notes
Acknowledgments
References
1. Lu P, Weaver VM, Werb Z (2012) The extra- 4. Mercier I, Lechaire JP, Desmouliere A, Gaill F,
cellular matrix: a dynamic niche in cancer pro- Aumailley M (1996) Interactions of human
gression. J Cell Biol 196:395–406 skin fibroblasts with monomeric or fibrillar col-
2. Nelson CM, Bissell MJ (2006) Of extracellular lagens induce different organization of the
matrix, scaffolds, and signaling: tissue architec- cytoskeleton. Exp Cell Res 225:245–256
ture regulates development, homeostasis, and 5. Woltersdorf C, Bonk M, Leitinger B,
cancer. Annu Rev Cell Dev Biol 22:287–309 Huhtala M, K€apyl€a J, Heino J et al (2017)
3. Blaschke UK, Eikenberry EF, Hulmes DJS, The binding capacity of α1β1-, α2β1- and
Galla HJ, Bruckner P (2000) Collagen XI α10β1-integrins depends on noncollagenous
nucleates assembly and limit lateral growth of surface macromolecules rather than the col-
cartilage fibrils. J Biol Chem lagens in cartilage fibrils. Matrix Biol
275:10370–10378 63:91–105
166 Uwe Hansen
6. Eble JA, Beermann B, Hinz HJ, Schmidt- 11. Xu H, Raynal N, Stathopoulos S, Myllyharju J,
Hederich A (2001) α2β1 integrin is not recog- Farndale RW, Leitinger B (2011) Collagen
nized by rhodocytin but is the specific, high binding specificity of the discoidin domain
affinity target of rhodocetin, an receptors: binding sites on collagens II and III
RGD-independent disintegrin and potent and molecular determinants for collagen IV
inhibitor of cell adhesion to collagen. J Biol recognition by DDR1. Matrix Biol 30:16–26
Chem 276:12274–12284 12. Böhme K, Winterhalter KH, Bruckner P
7. de Santana Evangelista K, Andrich F, Figueir- (1995) Terminal differentiation of chondro-
edo de Rezende F, Niland S, Cordeiro MN, cytes in culture is a spontaneous process and is
Horlacher T et al (2009) Plumieribetin, a fish arrested by transforming growth factor-beta2
lectin homologous to mannose-binding B-type and basic fibroblast growth factor in synergy.
lectins, inhibits the collagen-binding α1β1 Exp Cell Res 216:191–198
integrin. J Biol Chem 284:34747–34759 13. Bruckner P, Hörler I, Mendler M, Houze Y,
8. Tulla M, Pentik€ainen OT, Viitasalo T, K€apyl€a J, Winterhalter KH, Eich-Bender SG, Spycher
Impola U, Nykvist P et al (2001) Selective MA (1989) Induction and prevention of chon-
binding of collagen subtypes by integrin α1I, drocyte hypertrophy in culture. J Cell Biol
α2I, and α10I domains. J Biol Chem 109:2537–2545
276:48206–48212 14. Mendler M, Eich-Bender SG, Vaughan L, Win-
9. K€apyl€a J, Ivaska J, Riikonen R, Nykvist P, terhalter KH, Bruckner P (1989) Cartilage
Pentikainen O, Johnson M, Heino J (2000) contains mixed fibrils of collagen types II, IX,
Integrin alpha2 I-domain recognizes type I and XI. J Cell Biol 108:191–197
and type IV collagens by different mechanisms. 15. Lenter M, Uhlig H, Hamann A, Jenö P,
J Biol Chem 275:3348–3354 Imhof B, Vestweber D (1993) A monoclonal
10. Leitinger B (2003) Molecular analysis of colla- antibody against an activation epitope on
gen binding by the human discoidin domain mouse integrin chain β1 blocks adhesion of
receptors, DDR1 and DDR2: identification of lymphocytes to the endothelial integrin α6β1.
collagen binding sites in DDR2. J Biol Chem Proc Natl Acad Sci U S A 90:9051–9055
278:16761–16769
Part IV
Abstract
Proper collagen homeostasis is essential for development and aging of any multicellular organism. During
aging, two extreme scenarios are commonly occurring: a local excess in collagen deposition, for instance
during fibrosis, or a gradual overall reduction of collagen mass. Here, we describe a histological and a
colorimetric method to assess collagen levels in mammalian tissues and in the nematode Caenorhabditis
elegans. The first method is the polychrome Herovici staining to distinguish between young and mature
collagen ratios. The second method is based on hydroxyproline measurements to estimate collagen protein
levels. In addition, we show how to decellularize the multicellular organism C. elegans in order to harvest its
cuticle, one of the two major extracellular matrices, mainly composed of collagen. These methods allow
assessing collagen deposition during aging either in tissues or in whole organisms.
Key words Collagen, Aging, Tissue, Herovici staining, Hydroxyproline, Cuticle, Isolation, Freeze-
cracking, Age-synchronizing, Extracellular matrix, C. elegans
1 Introduction
Irit Sagi and Nikolaos A. Afratis (eds.), Collagen: Methods and Protocols, Methods in Molecular Biology, vol. 1944,
https://doi.org/10.1007/978-1-4939-9095-5_13, © Springer Science+Business Media, LLC, part of Springer Nature 2019
169
170 Alina C. Teuscher et al.
Fig. 1 Herovici staining for mammalian tissue and C. elegans. (a–c) A schematic of the individual steps of
the Herovici staining described in Subheading 3 (see Subheading 3.1). (a) Freeze-cracking of C. elegans
Collagen Deposition During Aging 173
2 Materials
Fig. 1 (continued) (see Subheading 3.1.1). (b) Mammalian tissue harvesting and deparaffinization (see
Subheading 3.1.2). (c) Shared part of the Herovici staining protocol for both C. elegans and mammalian
tissue (see Subheading 3.1.3). (d) Herovici-stained mouse skin specimen. The young collagen (collagen type
III) is stained in blue, whereas the mature collagen (collagen type I) is stained in red. Cytoplasm is counter-
stained in yellow and cell nuclei dark blue to black. Three layers forming the skin (epidermis, dermis,
hypodermis) can be identified. (e) Herovici-stained C. elegans at day 1 of adulthood
174 Alina C. Teuscher et al.
3 Methods
3.1 Herovici Staining All procedures are carried out at room temperature, if not indicated
for Mammalian Tissue otherwise. Glass slides with tissue sections fixed in 4% paraformal-
and C. elegans dehyde (PFA in PBS) are used for the Herovici staining protocol.
Reusable Herovici solutions can be poured back in their original
container. At the end of this procedure, tissue sections will be ready
to be imaged under the microscope. In this section, we first
describe the harvesting and preparation for mammalian tissue (see
Subheading 3.1.1) and then for C. elegans (see Subheading 3.1.2)
followed by the shared methodical part (see Subheading 3.1.4).
3.1.2 Preparation of Poly- 1. Clean microscope slides with Kimwipes and 70%–100% EtOH.
L-Slides for Freeze-
2. Pipette 50 μl Poly-L-Solution on the non-frosted part of the
Cracking of C. elegans microscope slide. Place another microscope slide on top, so
that the transparent parts completely overlay and the frosted
parts are hanging over on the opposite sides.
3. Slide the two slides apart: one side per slide should now be
covered with a thin layer of Poly-L-Solution.
4. Turn the slides around, so that the Poly-L-Solution sides are
facing upward, and place the slides in a 55 C incubator for 1 h
to dry. Slides can now be used directly or stored for several
months in a clean slide box at 4 C.
3.1.3 Harvesting The described C. elegans freeze-cracking method was adopted from
and Preparing C. elegans Duerr 2006 [33]. For freeze-cracking use a minimum of 4000
Samples Using a Freeze- C. elegans or better about 8000–12,000 C. elegans (see Note 15).
Cracking Method (Fig. 1b) For a protocol to age-synchronize and culture C. elegans (see Sub-
heading 3.2.2):
1. Place the fixation solution (4% paraformaldehyde (PFA in
PBS)) and several empty 10 cm petri dishes on ice in order to
pre-chill for later use.
2. Wash C. elegans off culturing plates using M9 buffer, and
collect them in a 15 ml conical centrifuge tube.
3. Centrifuge for 1 min at 1000 rpm (standard table centrifuge is
ca. 200 g; see Note 16), and remove supernatant. Wash the
C. elegans until the supernatant is clean and no bacteria are
visible (see Note 17).
4. Rinse C. elegans by filling up the 15 ml conical centrifuge tube
with 14 ml ddH2O, centrifuge for 1 min at 1000 rpm
(200 g), remove supernatant, and add ddH2O to the pelleted
C. elegans to a total volume of 100 μl.
5. Using a glass Pasteur pipette, place a small drop of animals
(ca.10–20 μl) on a Poly-L-Solution covered slide. Wait shortly
to let the C. elegans settle on the slide. Use the pipette to spread
the C. elegans out so that they do not overlap.
6. Carefully place another Poly-L-Solution covered slide on top of
the slide with the C. elegans so that the Poly-L-covered sides
face each other, while the frosted parts are not overlapping.
7. Put on safety goggles and gloves. Hold the two slides slightly
pressed together into liquid nitrogen until the bubbling stops.
Take the slides out. Wait for ca. 20 s. Then start to snip with
your finger carefully against one of the slides until they rip
apart.
Collagen Deposition During Aging 177
8. Check which slide has most of the C. elegans attached to it, and
place either only this slide or both slides separately into indi-
vidual empty 10 cm petri dishes, which are standing on ice.
9. Pipette 100–200 μl 4% PFA pre-chilled onto the C. elegans
covered part of the slides. Fix the C. elegans by incubating the
slides at 4 C for at least 30 min or up to 24 h.
10. After fixation, rinse the slides gently at least three times with
PBS. Proceed immediately with staining, or store the slides in a
Coplin jar or petri dish containing PBS at 4 C for several days.
3.1.4 The Herovici 1. Immerse slides in Solution 1 for 5 min (see Note 1).
Staining for Both 2. Rinse slides under running tap water for 2 min (see Note 18).
Mammalian Tissue
3. Immerse slides in Solution 2 for 5–6 min (see Notes 5, 6,
and C. elegans (Fig. 1c)
and 19).
4. Rinse slides under running tap water for 2 min (see Note 18).
5. Immerse slides in Solution 3 for 2 min (see Note 8).
6. Transfer slides directly to Solution 4 for 2 min (see Note 9).
7. Rinse slides under running tap water for 2 min (see Note 18).
8. Immerse slides in Solution 5 for 2 min (see Note 12).
9. Transfer slides directly to Solution 6 for 2 min (see Note 13).
10. Transfer slides directly to Solution 7 for 2 min (see Note 14).
11. Immerse the slides in 100% ethanol for 1 min. Repeat this
dehydration step once for a total of two.
12. Immerse the slides in xylol for 2 min. Repeat this step once for
a total of two.
13. Let the slides dry for a minimum of 10 min (see Note 20).
14. Mount the slides with a mounting media (e.g., Eukitt®), and
let them dry under a fume hood (see Note 21).
15. After adding the cover slip, the specimen is ready for imaging.
Examples of a Herovici-stained mammalian tissue section and a
C. elegans are shown in Fig. 1d, e, respectively.
3.2 Total Collagen The collagen content in a tissue or whole-organism sample can be
Quantification determined in multiple ways (Fig. 2). It is generally advised to select
for Mammalian Tissue the normalization entity to be as conclusive as possible. Here, total
and C. elegans collagen quantification is combined with either absolute animal
number (C. elegans) or with total protein determination for both
mammalian tissue and C. elegans. Here, we focus mainly on how to
establish aged C. elegans cultures and on the assessment of collagen
levels in C. elegans, since the use of hydroxyproline measurement in
mammalian tissue is well-established.
178 Alina C. Teuscher et al.
a b c
harvest
(in well)
(in well)
formula (1)
store at-80°C
0.1
a) harvest 0.5
b) wash
95°C, 20 h c) count
in acid L4 L4
split sonicate
1 2 3 4 5 6 7 8 9 10 11 12
35 µl 15 µl 1 2 3 4 5 6 7 8 9 10 11 12
A A
B B
C C
store at -80°C
D D
E E
F F
G
H
G
H
0
split
1 2 3 4 5 6 7 8 9 10 11 12
A 1 2 3 4 5 6 7 8 9 10 11 12
B
C
D
95°C, 20 h A
B
C
D
E
E
F
G
H in acid F
G
H
Fig. 2 Total collagen quantification using hydroxyproline assay for mammalian tissue and C. elegans. (a, b)
Schematic of the collagen assay that can be performed using mammalian tissue (a) as well as C. elegans
samples (b). Please see Subheading 3 (see Subheading 3.2) for detailed protocol steps. (c) An example of how
to quantify the collagen-to-protein ratio for larval L4 stage spe-9(hc88) C. elegans samples is shown. (d)
Formula (1) shows how to calculate the ratio of collagen content to total protein levels. The dilution term
referring to the digestion is omitted when the total protein content is determined using the hydrolyzed sample.
Formula (2) shows how to normalize collagen content per individual C. elegans
3.2.1 Mammalian Tissue There are several ways to assess hydroxyproline levels in mammalian
(Fig. 2a) tissues. Here, we briefly outline the use of a colorimetric method
using the QuickZyme Kit for both hydroxyproline and total protein
measurements. The advantage of this protocol is that each HCl
hydrolyzed tissue sample can be split to measure (1) hydroxyproline
levels to estimate collagen protein content and (2) to measure total
protein levels from the same sample in order to normalize the
collagen content to total protein levels. Equivalent kits or protocols
Collagen Deposition During Aging 179
3.2.2 Synchronizing C. elegans samples are prepared in one batch and harvested in user-
C. elegans for an defined intervals over the animal’s lifespan. To age-synchronize the
Age-Dependent Time C. elegans population, we describe a bleach preparation protocol
Course Analysis (Fig. 2b) to harvest eggs from gravid C. elegans adults. For each time point
and condition, 2000–5000 animals are required; therefore
ca. 10,000–100,000 eggs should be harvested in the initial lysis step.
1. Use multiple culturing plates containing a large amount of
gravid C. elegans adults to perform a population lysis. Wash
the culturing plates with sterile H2O. Pipette the H2O across
the plate several times to loosen C. elegans that are stuck onto
the bacterial lawn.
2. Collect the liquid containing the C. elegans in a sterile 15 ml
conical centrifuge tube. Centrifuge for 1 min at 1000 rpm
(standard table centrifuge is ca. 200 g; see Note 24). Discard
supernatant. Wash residual bacteria away from C. elegans by
filling up the 15 ml conical centrifuge tube with sterile H2O,
spin down, and discard the supernatant. Repeat this step until
the supernatant is clear. Spin down to pellet gravid C. elegans,
and add H2O to a total volume of 3.5 ml.
3. Put on gloves and eye protection. Add 1 ml of 5% sodium
hypochlorite solution (or household bleach), and add 0.5 ml
of 5 N NaOH. Close 15 ml conical centrifuge tube, and shake
well. In 1-min intervals, hold the 15 ml conical centrifuge tube
under a dissecting scope to observe lysis of gravid C. elegans,
and shake well after observation to optimize lysis. Once almost
all C. elegans are lysed, add M9 buffer to a total volume of 15 ml,
and centrifuge at 3000 rpm (ca. 1800 g) for 1 min to pellet the
eggs (see Note 25). Discard supernatant, and wash the eggs at
least three times by filling up to 15 ml with M9 buffer and
re-pelleting the eggs by centrifugation at 3000 rpm (ca. 1800 g)
for 1 min. Discard the supernatant (see Note 26).
180 Alina C. Teuscher et al.
4. After lysis and subsequent washing steps, the samples are incu-
bated while rotating for 12 h in M9 buffer supplemented with
5 μg/ml cholesterol.
5. 500–1000 larval L1 C. elegans larvae are placed on each 10 cm
culturing plate. If a temperature-sensitive sterile strain is used,
the sterility mechanism has to be activated at the needed time
point (see Note 27). If floxuridine (FUDR) is used to avoid
offsprings, the animals are transferred at the larval L4 stage to
50 μg/ml FUDR culturing plates (see Notes 28 and 29).
6. 2000–5000 C. elegans (2–10 plates) are harvested for each
condition and time point. Transfer all animals from the plates
to a 15 ml conical centrifuge tube. The animals are centrifuged
briefly at low speed (200 rpm, ca. 8 g) to pellet aged C. ele-
gans adults. The suspension containing unhatched eggs or
larvae (if FUDR is used) is aspirated and discarded. Repeat
this washing step three times or until only the desired
C. elegans adults are present and supernatant is clear (see Note
30).
7. From the last wash that was filled up with M9 buffer to a total
volume of 14 ml, take five times 20 μl aliquots, and place these
aliquots on a petri dish lid to count the number of animals in
each 20 μl drop using a dissecting scope (see Notes 31 and 32).
To estimate total number of C. elegans, average the number of
C. elegans from each 20 μl drop and multiply by 700 to get an
estimate of the total number of C. elegans present in 14 ml
sample.
8. Centrifuge for 1 min at 1000 rpm (ca. 200 g) to softly pellet
C. elegans. Discard supernatant. Use a glass Pasteur pipette to
transfer the C. elegans pellet to a labelled 1.5 ml Eppendorf
collection tube, fill up with sterile H2O to a total volume of
200 μl, and store at 80 C (see Notes 16, 33, and 34).
9. At the end of the time course, once all samples have been
collected, proceed by thawing all tubes. At least 3–5 freeze-
thaw cycles should be performed by thawing at room tempera-
ture and freezing in liquid nitrogen (or dry ice with EtOH or
by placing back into the 80 C).
10. Then sonicate all samples on ice until all animals are disrupted.
The C. elegans cuticle is the hardest to break. With a standard
sonication device, it will take several repeats to break down all
C. elegans (see Note 35).
3.2.3 Total Protein To determine the total collagen content as a fraction of total
Quantification protein abundance, the total protein content has to be determined
for C. elegans (Fig. 2) separately.
Collagen Deposition During Aging 181
3.2.4 Total Collagen Here, we determine the total collagen abundance by digesting the
Quantification sample and directly measuring the concentration of the amino acid
for C. elegans (Fig. 2) hydroxyproline in each sample.
1. The sonicated samples are transferred into heat-stable tubes
provided by the kit manufacturer (QuickZyme Total Collagen
Kit, QuickZyme Biosciences, or equivalent kit from other man-
ufacturers). The provided collagen standard does not require to
be sonicated but is otherwise treated identically as the samples.
2. According to the manufacturer’s instructions, the sample and
standard are mixed with 12 M HCl solution and incubated for
20 h at 95 C.
3. In brief, after the digestion the supernatant is isolated, the acid
content lowered, assay buffer added, and the plate incubated at
room temperature for 20 min followed by the addition of the
detection reagent and subsequent incubation at 60 C for 1 h.
The color change is quantified using a plate reader (see Note 36).
4. The total collagen content per sample in a well can be directly
determined through the measured color change. In a first step,
the collagen reference sample (1.2 mg/ml of rat tail collagen) is
used to generate a standard curve relating the magnitude of the
color change to the collagen content of the well. Over the used
concentration range, this relationship is approximately linear.
With the standard curve available, the collagen content of the
samples can be quantified. It must be noted that the collagen
standard supplied in the QuickZyme Biosciences Kit contains
rat tail collagen. To validate the use of rat tail collagen as a
reference for hydroxyproline levels for C. elegans collagens, the
potential hydroxyproline occurrences on the Y position in the
C. elegans [Gly-X-Y] triple repeats in collagens were estimated
in silico. The relative proline abundance on the Y position is
similar between C. elegans and rat tail collagens (personal com-
munication Jan M. Gebauer). Example results for C. elegans
protein and collagen concentration are shown in Fig. 2b.
5. The total collagen-to-total protein ratio can be determined by
combining the measured collagen and protein concentrations
according to formula (1). In the case of C. elegans, the total
collagen abundance per animal can be calculated by subjecting
182 Alina C. Teuscher et al.
3.3 Methods This method was modified from Leushner et al. [28], for the use in
for Cuticle C. elegans. The two advantages of using this protocol compared to
(Extracellular Matrix) the ST protocol is (1) its simplicity and (2) the SDS-cleaned cuticles
Isolation in C. elegans might still contain Schiff base products or other adducts that might
be of importance of the desired analysis [34]. The major problem
3.3.1 Cuticle Isolation with this protocol is that the C. elegans cuticle tends to stick on
(Freeze-Thaw Protocol) plastic; this can be avoided by using glass materials instead. All steps
after the freeze-thawing should be carried out on ice, except steps
involving SDS. We recommend to use about 12,000 C. elegans per
sample to isolate a sufficient number of cuticles. Please see Subhead-
ing 3.2.2 for a protocol to age-synchronize the C. elegans samples.
An example of an aged C. elegans (7-day adult) is shown in Fig. 3a
for comparison with the isolated cuticles Fig. 3b, c.
1. Use 12,000 adult C. elegans (about 8–12 full culturing plates).
Pipette around 3 ml M9 buffer on each plate. Gently tilt plates
to collect the liquid on one side, and carefully pipette the M9
buffer several times across the plate to loosen up the C. elegans
stuck on the bacterial food lawn. Transfer the animals from the
plates into a 15 ml conical centrifuge tube.
2. Wash the C. elegans three times with 15 ml ddH2O by centri-
fugation at around 1000 rpm (ca. 200 g) for 1 min, and
discard the supernatant (see Note 33).
3. After the last washing step, discard the supernatant again, and
use a glass Pasteur pipette to transfer the C. elegans pellet into a
1.5 ml Eppendorf microfuge tube (see Note 37). Centrifuge
1.5 ml tube at around 100 g for 30 s, discard supernatant,
and fill up with ddH2O to 100 μl.
4. Freeze the tubes in liquid nitrogen for 1 min (or until frozen),
followed by a thawing step at room temperature. Repeat this
freeze-thaw cycle at least three times.
5. Sonicate the samples five times in intervals of 20 s each at an
amplitude of 80% with 20 s breaks on ice in between (Sonoplus
mini20 from Bandelin) (see Note 38).
6. Use a glass Pasteur pipette to transfer the ruptured C. elegans
to 15 ml glass tubes (with screw gaps) (see Note 39), and
suspend them in 10 ml 1% SDS solution, followed by overnight
incubation onto a rotor at 37 C. The 1% SDS should wash out
all the internal cells and organs from the disrupted cuticles
(Fig. 3b).
Collagen Deposition During Aging 183
Fig. 3 Isolated C. elegans cuticles. (a) Differential contrast image of a day 7 adult
C. elegans. Anterior to the left and ventral side down. (b) Intact C. elegans cuticle
after 1% SDS before washing step. Black arrows point to the annuli structure of
the cuticle. White arrowheads point to debris. (c) Intact C. elegans cuticle after
washing. Sometimes other extracellular matrices, such as the basement
membranes of the gonad (white arrowheads), are still attached
3.3.2 Cuticle Isolation This method was adapted from Cox et al. [29]. Please see Subhead-
(ST Buffer Protocol) ing 3.3.1 for the comparison with the other cuticle isolation freeze-
thaw method, and see Subheading 3.2.2 for a protocol to age-
synchronize the C. elegans samples. All steps after the sonication
should be carried out on ice, except steps involving SDS.
1. Collect 12,000 (adult) C. elegans with M9 buffer in a 15 ml
tube as described in the first step of Subheading 3.3.1.
184 Alina C. Teuscher et al.
4 Notes
4.2 Total Collagen 22. Ideally, a 1:10 ratio of wet tissue weight to acid volume is
Quantification recommended, with a minimal volume of 200 μl 6 M HCl
for Mammalian Tissue per tube. For example, 50 mg of tissue mixed with 500 μl of
and C. elegans 6 M HCl can be used.
23. To be able to relate the total collagen content of the sample as
robustly as possible to the total protein abundance, we recom-
mend to measure the later directly in the hydrolyzed sample.
To be able to quantify the free amino acid level present in the
acidic hydrolyzed sample, a suitable kit has to be chosen. We
recommend the Total Protein Assay, QuickZyme Biosciences.
24. When centrifuging C. elegans, either turn off the breaks or turn
down the deceleration to a minimum.
25. In general, complete lysis can take about 4–8 min. Hence, it is
important to regularly check if all C. elegans are lysed. If lysis is
performed for too long, the eggs will suffer from increasing
damage.
26. Sometimes three washes are not enough to wash out all bleach
solution. Hence, check by smelling the 15 ml conical centri-
fuge tube if bleach odor is still present.
27. We recommend the C. elegans mutant spe-9(hc88). Due to a
temperature-sensitive defect in spermatogenesis, the animals
are not able to lay fertilized eggs when shifted to 25 C during
development. To ensure all animals are sterile, L1 animals can
be cultured at 25 C until day 1 or 2 of adulthood.
186 Alina C. Teuscher et al.
28. To avoid food scarcity, check the grow-up plates often, and if
needed, transfer to fresh plates.
29. To check the effectiveness of the intervention, a selected num-
ber of plates can be used for validation by, for example, placing
a fluorescent reporter strain on them.
30. Since the collagen content is changing over the lifespan of
C. elegans, it is crucial to work with age-synchronous popula-
tions. Thus, all eggs and larvae must be removed as best as
possible. This can be achieved by transferring animals often to
new plates and rigorous washing or through filtration.
31. Because adult C. elegans will sink to bottom of the conical
centrifuge tube, shake the 15 ml conical centrifuge tube well
between taking the aliquots for counting. Try to pipette from
the middle of the 15 ml conical centrifuge tube for consistency.
32. Use a pipette with a large orifice to sample C. elegans during
the counting procedure. Since adults grow in size, a too small
pipette tip can lead to an underestimation of older populations.
33. At this step of the protocol is a good time point for a break or
to wait until all time points from before are collected, for
instance, if you collect daily samples over the 2–3 weeks life-
span of C. elegans.
34. It is recommended to fill up all sample C. elegans pellets to
200 μl to simplify the downstream steps.
35. Example sonication configuration using a Bandelin Sonoplus,
UW mini20 device: Amplitude 80%, 30s intervals, pulse 1.0 s
on, 1.0 s off. However, using a more powerful sonicator is
recommended to accelerate C. elegans fragmentation.
36. It is advised to perform a few test reactions prior to processing
the entire batch for both quantification methods to be able to
adjust sample dilution should it be too concentrated.
4.3 Cuticle 37. C. elegans tend to stick on the plastic tip or on the plastic tubes.
(Extracellular Matrix) Hence, make sure to use a glass Pasteur pipette and pipette the
Isolation in C. elegans C. elegans on the bottom of the plastic tube to avoid sticking
on the side of the tube.
38. If the cuticles in the freezing protocol are ruptured too much,
you can consider performing only the freezing and thawing
cycles or only sonication.
39. One of the major problems is losing too many cuticles or
C. elegans. Therefore, use always glass pipettes to transfer
worms and cuticles.
40. Triton-X prevents the cuticles from sticking to the tubes during
the washing steps.
Collagen Deposition During Aging 187
Acknowledgments
References
1. Hynes RO (2009) The extracellular matrix: not 10. Sell DR, Monnier VM (2012) Molecular basis
just pretty fibrils. Science 326:1216–1219 of arterial stiffening: role of glycation–a mini-
2. Ricard-Blum S (2011) The collagen family. review. Gerontology 58:227–237
Cold Spring Harb Perspect Biol 3: 11. Snedeker JG, Snedeker JG, Gautieri A et al
a004978–a004978 (2014) The role of collagen crosslinks in ageing
3. Sivan S-S, Wachtel E, Tsitron E et al (2008) and diabetes–the good, the bad, and the ugly.
Collagen turnover in normal and degenerate Muscles Ligaments Tendons J 4:303–308
human intervertebral discs as determined by 12. Myllyharju J (2004) Collagens, modifying
the racemization of aspartic acid. J Biol Chem enzymes and their mutations in humans, flies
283:8796–8801 and worms. Trends Genet 20:33–43
4. Heinemeier KM, Schjerling P, Heinemeier J 13. Fenske NA, Lober CW (1986) Structural and
et al (2016) Radiocarbon dating reveals mini- functional changes of normal aging skin. J Am
mal collagen turnover in both healthy and Acad Dermatol 15:571–585
osteoarthritic human cartilage. Sci Transl Med 14. Shuster S, Black MM, McVitie E (1975) The
8:346ra90–346ra90 influence of age and sex on skin thickness, skin
5. Toyama BH, Hetzer MW (2013) Protein collagen and density. Br J Dermatol
homeostasis: live long, won’t prosper. Nat 93:639–643
Rev Mol Cell Biol 14:55–61 15. López-Otı́n C, Blasco MA, Partridge L et al
6. Kjaer M, Langberg H, Miller BF et al (2005) (2013) The hallmarks of aging. Cell
Metabolic activity and collagen turnover in 153:1194–1217
human tendon in response to physical activity. 16. Franceschi C, Campisi J (2014) Chronic
J Musculoskelet Neuronal Interact 5:41–52 inflammation (inflammaging) and its potential
7. Myllyharju J, Kivirikko KI (2001) Collagens contribution to age-associated diseases. J Ger-
and collagen-related diseases. Ann Med ontol A Biol Sci Med Sci 69(Suppl 1):S4–S9
33:7–21 17. Wynn TA (2007) Common and unique
8. Fisher GJ, Quan T, Purohit T et al (2009) mechanisms regulate fibrosis in various fibro-
Collagen fragmentation promotes oxidative proliferative diseases. J Clin Investig
stress and elevates matrix metalloproteinase-1 117:524–529
in fibroblasts in aged human skin. Am J Pathol 18. Gutiérrez-Fernández A, Soria-Valles C, Osorio
174:101–114 FG et al (2015) Loss of MT1-MMP causes cell
9. Shoulders MD, Raines RT (2009) Collagen senescence and nuclear defects which can be
structure and stability. Annu Rev Biochem reversed by retinoic acid. EMBO J
78:929–958 34:1875–1888
188 Alina C. Teuscher et al.
Abstract
Fibrosis is characterized by excessive deposition of collagen and additional extracellular matrix (ECM)
components in response to chronic injuries. Liver fibrosis often results from chronic hepatitis C virus
infection and alcohol abuse that can deteriorate to cirrhosis and liver failure. Current noninvasive diagnostic
methods of liver fibrosis are limited in their ability to detect and differentiate between early and intermedi-
ate stages of fibrosis. New biomarkers of fibrosis that reflect ECM turnover are therefore badly needed.
Procollagen C-proteinase enhancer 1 (PCPE-1), a connective tissue glycoprotein that functions as a
positive regulator of C-terminal procollagen processing and subsequent collagen fibril assembly, is a
promising candidate. Its tissue distribution and expression profile overlap those of collagen, and its
expression in fibrosis is upregulated in parallel to the increase in collagen expression. The potential of
PCPE-1 as a biomarker of liver fibrosis was recently established using a CCl4 mouse model of liver fibrosis
by showing that the increase in collagen and PCPE-1 content in the fibrotic mouse liver was reflected by
elevated plasma levels of PCPE-1. This was achieved using a newly developed highly sensitive, specific,
accurate, and reproducible ELISA for mouse PCPE-1, which is based on commercially available antibodies
and is offered as a new research tool in the field. A similar ELISA test was developed for human PCPE-1,
and preliminary results with plasma from liver fibrosis patients revealed increased plasma concentrations of
PCPE-1 in some patients. The protocols of both ELISA tests are outlined herein in great detail to permit
their application by any laboratory with similar interests.
Key words Collagen, Procollagen C-proteinase enhancer 1, PCPE-1, ELISA, Liver fibrosis,
Biomarkers
1 Introduction
Irit Sagi and Nikolaos A. Afratis (eds.), Collagen: Methods and Protocols, Methods in Molecular Biology, vol. 1944,
https://doi.org/10.1007/978-1-4939-9095-5_14, © Springer Science+Business Media, LLC, part of Springer Nature 2019
189
190 Efrat Kessler and Eyal Hassoun
Fig. 1 Plasma concentrations of mPCPE-1 reflect the progression of liver fibrosis in CCl4-treated mice. Mice
were treated biweekly with CCl4 for 6 weeks at which time, treatment was terminated, and the mice were
followed for 7 additional weeks. Liver fibrosis was assessed at the indicated times by Sirius red collagen
staining (a), immunofluorescence (b), and immunoblotting (e). Each panel in a and b displays results for a
single randomly selected mouse from the indicated group. Numbers in brackets in b represent time expressed
in weeks. (c) Plasma concentrations of PCPE-1 after 6 weeks of CCl4 treatment are significantly higher than
those of control mice (n ¼ 12). (d) Plasma concentrations of PCPE-1 in CCl4-treated (~) and control (●) mice
as a function of time. Arrow, termination of CCl4 treatment. Results are presented as mean SD. (n at weeks
0 to 6 ¼ 12; n at weeks 7–10 ¼ 9; n at week 13 ¼ 6). Plasma concentrations of mPCPE-1 were determined by
the sandwich ELISA described herein at the indicated time points. (e) Immunoblot evaluating collagen I and
PCPE-1 content in liver extracts from CCl4-treated and control mice at weeks 6, 10, and 13. Samples from a
single control mouse and two CCl4-treated mice (all randomly selected) are shown. The amounts of protein
applied on each lane were 4 and 36 μg for collagen and PCPE-1 detection, respectively (Reproduced from ref.
19, Open Access)
Fig. 2 Plasma concentrations of mPCPE-1 decline during liver recovery in parallel to the decline of type I
collagen and PCPE-1 liver content. (a) The relative amounts of collagen type I and PCPE-1 were determined by
densitometry of the respective protein bands in the immunoblot shown in Fig. 1e. E/C, ratio of absorbance
units obtained for CCl4-treated (E) and control (C) mice, respectively. The values are mean SD (n ¼ 2).
Values of PCPE-1 plasma concentrations represent the mean SD obtained at weeks 6, 10, and 13; n ¼ 12,
9, and 6, respectively. (b) The relative amounts of PCPE-1 and collagen type I in the liver were derived from the
immunofluorescence analysis shown in Fig. 1b. Values represent relative staining, expressed as the percent-
age of stained area out of the total area in each panel. Values correspond to data derived for a single
representative mouse per group at the specified time point. Plasma concentrations of PCPE-1 are those
obtained for the respective mice. Each value is the mean SD; n ¼ 4 (Reproduced from ref. 19, Open Access)
2 Materials
2.1 Proteins 1. Mouse (m) PCPE-1: Purified from the culture medium of 3T6
and Antibodies mouse fibroblasts [13]. The concentration of the stock solu-
tion we used [19] was 250 μg/ml in 50 mM Tris–HCl,
PCPE-1 in Liver Fibrosis 193
150 mM NaCl, 5 mM CaCl2, 0.1% Brij 35, pH 7.5 (see Note 1).
Purified recombinant mPCPE-1 is available commercially
(R&D 2239-PE) and can be used instead.
2. Human (h) recombinant PCPE-1: Purified from the culture
media of 293-EBNA cells expressing the recombinant protein
[25, 33]. The concentration of the stock solution used by us
[32] was 0.5 mg/ml in 50 mM HEPES, 150 mM NaCl, 5 mM
CaCl2, and pH 7.5. Purified recombinant hPCPE-1 is also
available commercially (R&D 2627-PE).
3. Bovine serum albumin (Sigma A7906).
4. Rat monoclonal antibody against mPCPE-1: The antibody
(R&D MAB2239) is supplied as lyophilized powder and is
stored at 20 C. Before the first use, the content (500 μg) is
dissolved in 1.0 ml of sterile PBS (final concentration 500 μg/
ml) and is stored in aliquots at 80 C.
5. Goat polyclonal rabbit antibody to mPCPE-1: The antibody
(R&D AF2239) is supplied as lyophilized powder and is stored
at 20 C. Before the first use, the content (100 μg) is dis-
solved in 0.5 ml sterile PBS (final concentration 200 μg/ml)
and is stored in aliquots at 80 C.
6. Alkaline phosphatase (APA) conjugated rabbit antibody to
goat IgG (Sigma A4187).
7. Mouse monoclonal antibody 7A1/11 against hPCPE-1: This
antibody was developed in our laboratory and purified by
protein G-Sepharose chromatography followed by affinity
chromatography, i.e., adsorption to and elution from hPCPE-
1-Sepharose beads. The hPCPE-1-specific IgG fraction
obtained by affinity chromatography was stored in aliquots at
80 C at concentrations ranging from 0.15 to 0.4 mg/ml
(depending on the preparation) in 0.05 M HEPES, 0.15 M
NaCl, 5 mM CaCl2, and pH 7.5. Monoclonal antibody
7A1/11 is available commercially (Santa Cruz sc-73001;
Sigma C2122) (see Note 2).
8. Rabbit polyclonal antibody (IgG fraction) against hPCPE-1:
IgG fraction, 16 mg/ml in PBS. Stored in aliquots at 20 C.
Produced and purified in our laboratory using standard proto-
cols. Similar antibodies are available commercially (Protein
Tech 14993-1-AP; Sigma P 6243).
9. APA-conjugated goat antibody against rabbit IgG (Sigma
SAB3700950).
3 Methods
3.1 Sandwich ELISA 1. Adsorption of the first Ab (MAB2239): Dilute the stock solu-
for mPCPE-1 tion 1:100 in carbonate buffer (final concentration 5 μg/ml;
prepare just enough for the number of wells to be used; discard
remaining solution). Pipet 50 μl of the Ab solution into each
well in a 96 well plate using a stepper (see Note 4). Incubate
over night at 4 C.
2. Blocking: On the following day, remove and discard the first
antibody solution and wash the wells twice with PBST. Add to
PCPE-1 in Liver Fibrosis 195
Fig. 3 Standard curves for determination of mouse and human PCPE-1. (a) Sandwich ELISA for mPCPE-1.
Increasing amounts of purified mPCPE-1 were adsorbed to wells pre-coated with a rat monoclonal antibody to
mPCPE-1. Bound mPCPE-1 was detected using a goat antibody to mPCPE-1 and quantified using an
APA-conjugated rabbit anti goat IgG antibody. (b) Sandwich ELISA for hPCPE-1. Increasing amounts of purified
hPCPE-1 were adsorbed to wells pre-coated with mouse mAb 7A11/1 to hPCPE-1. Bound hPCPE-1 was
detected using a rabbit polyclonal Ab to hPCPE-1 and quantified using an APA-conjugated goat anti rabbit IgG
antibody. Each value in a and b represents mean SD; n ¼ 2. mOD, optical density expressed in milli units
(Adapted from refs. 19 (a) and 32 (b), both Open Access)
3.2 Sandwich ELISA 1. Adsorption of the first antibody (mAb 7A11/1): Dilute the
for hPCPE-1 antibody solution in carbonate buffer to a final concentration
of 5 μg/ml (see Note 2). Pipet 50 μl of the Ab solution into
each well in a 96 well plate. Incubate over night at 4 C.
2. Blocking: On the following day, remove and discard the first
antibody solution. Wash the wells twice with PBST. Add to
each well 200 μl of blocking buffer, and incubate for 2 h at
30 C.
PCPE-1 in Liver Fibrosis 197
4 Notes
Table 1
Preparation of mPCPE-1 solutions with recommended concentrations
(2–20 ng/ml; 0.1–1 ng/50 μl) for a calibration curvea
Table 2
Preparation of hPCPE-1 solutions with recommended concentrations
(4–24 ng/ml; 0.2–1.2 ng/50 μl) for a calibration curvea
References
1. Zeisberg M, Kalluri R (2013) Cellular mechan- for improving accuracy in daily clinical practice.
isms of tissue fibrosis. 1. Common and organ- Ann Hepatol 11:426–439
specific mechanisms associate with tissue fibro- 9. Patel K, Shackel NA (2014) Current status of
sis. Am J Physiol Cell Physiol 304:C216–C225 fibrosis markers. Curr Opin Gastroenterol
2. Wynn TA, Ramalingam TR (2012) Mechan- 30:253–259
isms of fibrosis: therapeutic translation for 10. Gressner OA, Weiskirchen R, Gressner AM
fibrotic disease. Nat Med 18:1028–1040 (2007) Biomarkers of liver fibrosis: clinical
3. Lotersztajn S, Julien B, Teixeira-Clerc F et al translation of molecular pathogenesis or based
(2005) Hepatic fibrosis: molecular mechanisms on liver-dependent malfunction tests. Clin
and drug targets. Annu Rev Pharmacol Toxicol Chim Acta 381:107–113
45:605–628 11. Nallagangula KS, Nagaraj SK, Venkataswamy L
4. Manning DS, Afdhal NH (2008) Diagnosis et al (2018) Liver fibrosis: a compilation on the
and quantitation of fibrosis. Gastroenterology biomarkers status and their significance during
134:1670–1681 disease progression. Future Sci OA 4(1):
5. Papastergiou V, Tsochatzis E, Burroughs AK FSO250
(2012) Non-invasive assessment of liver fibro- 12. Karsdal MA, Krarup H, Sand JMB et al (2014)
sis. Ann Gastroenterol 25:218–231 Review article: the efficacy of biomarkers in
6. Bedossa P, Carrat F (2009) Liver biopsy: the chronic fibroproliferative diseases-early diagno-
best, not the gold standard. J Hepatol 50:1–3 sis and prognosis, with liver fibrosis as an exem-
7. Rockey DC, Caldwell SH, Goodman ZD et al plar. Aliment Pharmacol Ther 40:233–249
(2009) American association for the study of 13. Kessler E, Adar R (1989) Type I procollagen
liver diseases. Liver biopsy. Hepatology C-proteinase from mouse fibroblasts: purifica-
49:1017–1044 tion and demonstration of a 55 kDa enhancer
8. Duarte-Rojo A, Altamirano JT, Feld JJ (2012) glycoprotein. Eur J Biochem 186:115–121
Noninvasive markers of fibrosis: key concepts 14. Kessler E, Takahara K, Biniaminov L et al
(1996) Bone morphogenetic protein-1: The
PCPE-1 in Liver Fibrosis 201
Abstract
The tumor microenvironment is a heterogeneous tissue that in addition to tumor cells, contain tumor-
associated cell types such as immune cells, fibroblasts, and endothelial cells. Considerably important in the
tumor microenvironment is its noncellular component, namely, the extracellular matrix (ECM). In partic-
ular, the collagenous matrix is subjected to significant alterations in its composition and structure that create
a permissive environment for tumor growth, invasion, and dissemination. Among tumor-infiltrating
immune cells, tumor-associated macrophages (TAMs) are numerous in the tumor stroma and are locally
educated to mediate important biological functions that profoundly affect tumor initiation, growth, and
dissemination. While the influence of TAMs and mechanical properties of the collagenous matrix on tumor
invasion and progression have been comprehensively investigated individually, their interaction within the
complex tumor microenvironment was overlooked. This review summarizes accumulating evidence that
indicate the existence of an intricate tumorigenic crosstalk between TAMs and collagenous matrix. A better
mechanistic comprehension of this reciprocal interplay may open a novel arena for cancer therapeutics.
1 Introduction
Irit Sagi and Nikolaos A. Afratis (eds.), Collagen: Methods and Protocols, Methods in Molecular Biology, vol. 1944,
https://doi.org/10.1007/978-1-4939-9095-5_15, © Springer Science+Business Media, LLC, part of Springer Nature 2019
203
204 Chen Varol
7 Concluding Remarks
References
1. Lu P, Weaver VM, Werb Z (2012) The extra- 4. Biswas SK, Allavena P, Mantovani A (2013)
cellular matrix: a dynamic niche in cancer pro- Tumor-associated macrophages: functional
gression. J Cell Biol 196(4):395–406. diversity, clinical significance, and open ques-
https://doi.org/10.1083/jcb.201102147 tions. Semin Immunopathol 35(5):585–600.
2. Pickup MW, Mouw JK, Weaver VM (2014) https://doi.org/10.1007/s00281-013-
The extracellular matrix modulates the hall- 0367-7
marks of cancer. EMBO Rep 15 5. Noy R, Pollard JW (2014) Tumor-associated
(12):1243–1253. https://doi.org/10. macrophages: from mechanisms to therapy.
15252/embr.201439246 Immunity 41(1):49–61. https://doi.org/10.
3. Fang M, Yuan J, Peng C, Li Y (2014) Colla- 1016/j.immuni.2014.06.010
gen as a double-edged sword in tumor pro- 6. Varol C, Sagi I (2018) Phagocyte-
gression. Tumour Biol 35(4):2871–2882. extracellular matrix crosstalk empowers
https://doi.org/10.1007/s13277-013- tumor development and dissemination.
1511-7 FEBS J 285(4):734–751. https://doi.org/
10.1111/febs.14317
214 Chen Varol
7. Naba A, Clauser KR, Hoersch S, Liu H, Carr 16. Wyckoff JB, Wang Y, Lin EY, Li JF,
SA, Hynes RO (2012) The matrisome: in Goswami S, Stanley ER, Segall JE, Pollard
silico definition and in vivo characterization JW, Condeelis J (2007) Direct visualization
by proteomics of normal and tumor extracel- of macrophage-assisted tumor cell intravasa-
lular matrices. Mol Cell Proteomics 11(4): tion in mammary tumors. Cancer Res 67
M111 014647. https://doi.org/10.1074/ (6):2649–2656. https://doi.org/10.1158/
mcp.M111.014647 0008-5472.CAN-06-1823
8. Mason SD, Joyce JA (2011) Proteolytic net- 17. Xiao Q, Ge G (2012) Lysyl oxidase, extracel-
works in cancer. Trends Cell Biol 21 lular matrix remodeling and cancer metastasis.
(4):228–237. https://doi.org/10.1016/j. Cancer Microenviron 5(3):261–273. https://
tcb.2010.12.002 doi.org/10.1007/s12307-012-0105-z
9. Hoye AM, Erler JT (2016) Structural ECM 18. Velling T, Risteli J, Wennerberg K, Mosher
components in the premetastatic and meta- DF, Johansson S (2002) Polymerization of
static niche. Am J Physiol Cell Physiol 310 type I and III collagens is dependent on fibro-
(11):C955–C967. https://doi.org/10. nectin and enhanced by integrins alpha
1152/ajpcell.00326.2015 11beta 1 and alpha 2beta 1. J Biol Chem
10. Butcher DT, Alliston T, Weaver VM (2009) A 277(40):37377–37381. https://doi.org/10.
tense situation: forcing tumour progression. 1074/jbc.M206286200
Nat Rev Cancer 9(2):108–122. https://doi. 19. Kaplan RN, Riba RD, Zacharoulis S, Bramley
org/10.1038/nrc2544 AH, Vincent L, Costa C, MacDonald DD, Jin
11. Levental KR, Yu H, Kass L, Lakins JN, DK, Shido K, Kerns SA, Zhu Z, Hicklin D,
Egeblad M, Erler JT, Fong SF, Csiszar K, Wu Y, Port JL, Altorki N, Port ER,
Giaccia A, Weninger W, Yamauchi M, Gasser Ruggero D, Shmelkov SV, Jensen KK,
DL, Weaver VM (2009) Matrix crosslinking Rafii S, Lyden D (2005) VEGFR1-positive
forces tumor progression by enhancing integ- haematopoietic bone marrow progenitors ini-
rin signaling. Cell 139(5):891–906. https:// tiate the pre-metastatic niche. Nature 438
doi.org/10.1016/j.cell.2009.10.027 (7069):820–827. https://doi.org/10.1038/
12. Provenzano PP, Inman DR, Eliceiri KW, Knit- nature04186
tel JG, Yan L, Rueden CT, White JG, Keely PJ 20. Chlenski A, Cohn SL (2010) Modulation of
(2008) Collagen density promotes mammary matrix remodeling by SPARC in neoplastic
tumor initiation and progression. BMC Med progression. Semin Cell Dev Biol 21
6:11. https://doi.org/10.1186/1741-7015- (1):55–65. https://doi.org/10.1016/j.
6-11 semcdb.2009.11.018
13. Paszek MJ, Zahir N, Johnson KR, Lakins JN, 21. Arnold SA, Rivera LB, Miller AF, Carbon JG,
Rozenberg GI, Gefen A, Reinhart-King CA, Dineen SP, Xie Y, Castrillon DH, Sage EH,
Margulies SS, Dembo M, Boettiger D, Ham- Puolakkainen P, Bradshaw AD, Brekken RA
mer DA, Weaver VM (2005) Tensional (2010) Lack of host SPARC enhances vascular
homeostasis and the malignant phenotype. function and tumor spread in an orthotopic
Cancer Cell 8(3):241–254. https://doi.org/ murine model of pancreatic carcinoma. Dis
10.1016/j.ccr.2005.08.010 Model Mech 3(1–2):57–72. https://doi.
14. Barcus CE, O’Leary KA, Brockman JL, org/10.1242/dmm.003228
Rugowski DE, Liu Y, Garcia N, Yu M, Keely 22. Carmeliet P, Jain RK (2000) Angiogenesis in
PJ, Eliceiri KW, Schuler LA (2017) Elevated cancer and other diseases. Nature 407
collagen-I augments tumor progressive sig- (6801):249–257. https://doi.org/10.1038/
nals, intravasation and metastasis of 35025220
prolactin-induced estrogen receptor alpha 23. Liu J, Agarwal S (2010) Mechanical signals
positive mammary tumor cells. Breast Cancer activate vascular endothelial growth factor
Res 19(1):9. https://doi.org/10.1186/ receptor-2 to upregulate endothelial cell pro-
s13058-017-0801-1 liferation during inflammation. J Immunol
15. Pickup MW, Laklai H, Acerbi I, Owens P, 185(2):1215–1221. https://doi.org/10.
Gorska AE, Chytil A, Aakre M, Weaver VM, 4049/jimmunol.0903660
Moses HL (2013) Stromally derived lysyl oxi- 24. Montesano R, Orci L, Vassalli P (1983) In
dase promotes metastasis of transforming vitro rapid organization of endothelial cells
growth factor-beta-deficient mouse mam- into capillary-like networks is promoted by
mary carcinomas. Cancer Res 73 collagen matrices. J Cell Biol 97(5 Pt
(17):5336–5346. https://doi.org/10.1158/ 1):1648–1652
0008-5472.CAN-13-0012
Tumorigenic Interplay Between Macrophages and Collagenous Matrix. . . 215
25. Ingber D, Folkman J (1988) Inhibition of 34. Kim YM, Jang JW, Lee OH, Yeon J, Choi EY,
angiogenesis through modulation of collagen Kim KW, Lee ST, Kwon YG (2000) Endosta-
metabolism. Lab Investig 59(1):44–51 tin inhibits endothelial and tumor cellular
26. Myers KA, Applegate KT, Danuser G, Fischer invasion by blocking the activation and cata-
RS, Waterman CM (2011) Distinct ECM lytic activity of matrix metalloproteinase. Can-
mechanosensing pathways regulate microtu- cer Res 60(19):5410–5413
bule dynamics to control endothelial cell 35. Meyaard L (2008) The inhibitory collagen
branching morphogenesis. J Cell Biol 192 receptor LAIR-1 (CD305). J Leukoc Biol 83
(2):321–334. https://doi.org/10.1083/jcb. (4):799–803. https://doi.org/10.1189/jlb.
201006009 0907609
27. Newman AC, Nakatsu MN, Chou W, Ger- 36. Meyaard L, Hurenkamp J, Clevers H, Lanier
shon PD, Hughes CC (2011) The require- LL, Phillips JH (1999) Leukocyte-associated
ment for fibroblasts in angiogenesis: Ig-like receptor-1 functions as an inhibitory
fibroblast-derived matrix proteins are essential receptor on cytotoxic T cells. J Immunol 162
for endothelial cell lumen formation. Mol Biol (10):5800–5804
Cell 22(20):3791–3800. https://doi.org/10. 37. Rosenfeldt H, Grinnell F (2000) Fibroblast
1091/mbc.E11-05-0393 quiescence and the disruption of ERK signal-
28. Bignon M, Pichol-Thievend C, Hardouin J, ing in mechanically unloaded collagen matri-
Malbouyres M, Brechot N, Nasciutti L, ces. J Biol Chem 275(5):3088–3092
Barret A, Teillon J, Guillon E, Etienne E, 38. Provenzano PP, Inman DR, Eliceiri KW,
Caron M, Joubert-Caron R, Monnot C, Keely PJ (2009) Matrix density-induced
Ruggiero F, Muller L, Germain S (2011) mechanoregulation of breast cell phenotype,
Lysyl oxidase-like protein-2 regulates sprout- signaling and gene expression through a
ing angiogenesis and type IV collagen assem- FAK-ERK linkage. Oncogene 28
bly in the endothelial basement membrane. (49):4326–4343. https://doi.org/10.1038/
Blood 118(14):3979–3989. https://doi. onc.2009.299
org/10.1182/blood-2010-10-313296 39. Tilghman RW, Cowan CR, Mih JD,
29. Kalluri R (2003) Basement membranes: struc- Koryakina Y, Gioeli D, Slack-Davis JK, Black-
ture, assembly and role in tumour angiogene- man BR, Tschumperlin DJ, Parsons JT
sis. Nat Rev Cancer 3(6):422–433. https:// (2010) Matrix rigidity regulates cancer cell
doi.org/10.1038/nrc1094 growth and cellular phenotype. PLoS One 5
30. Bonanno E, Iurlaro M, Madri JA, Nicosia RF (9):e12905. https://doi.org/10.1371/jour
(2000) Type IV collagen modulates angio- nal.pone.0012905
genesis and neovessel survival in the rat aorta 40. Mouw JK, Yui Y, Damiano L, Bainer RO,
model. In Vitro Cell Dev Biol Anim 36 Lakins JN, Acerbi I, Ou G, Wijekoon AC,
(5):336–340. https://doi.org/10.1290/ Levental KR, Gilbert PM, Hwang ES, Chen
1071-2690(2000)036<0336:TICMAA>2. YY, Weaver VM (2014) Tissue mechanics
0.CO;2 modulate microRNA-dependent PTEN
31. Herbst TJ, McCarthy JB, Tsilibary EC, expression to regulate malignant progression.
Furcht LT (1988) Differential effects of lami- Nat Med 20(4):360–367. https://doi.org/
nin, intact type IV collagen, and specific 10.1038/nm.3497
domains of type IV collagen on endothelial 41. Leight JL, Wozniak MA, Chen S, Lynch ML,
cell adhesion and migration. J Cell Biol 106 Chen CS (2012) Matrix rigidity regulates a
(4):1365–1373 switch between TGF-beta1-induced apopto-
32. Sudhakar A, Nyberg P, Keshamouni VG, sis and epithelial-mesenchymal transition.
Mannam AP, Li J, Sugimoto H, Cosgrove D, Mol Biol Cell 23(5):781–791. https://doi.
Kalluri R (2005) Human alpha1 type IV col- org/10.1091/mbc.E11-06-0537
lagen NC1 domain exhibits distinct antian- 42. Erler JT, Bennewith KL, Cox TR, Lang G,
giogenic activity mediated by alpha1beta1 Bird D, Koong A, Le QT, Giaccia AJ (2009)
integrin. J Clin Invest 115(10):2801–2810. Hypoxia-induced lysyl oxidase is a critical
https://doi.org/10.1172/JCI24813 mediator of bone marrow cell recruitment to
33. Petitclerc E, Boutaud A, Prestayko A, Xu J, form the premetastatic niche. Cancer Cell 15
Sado Y, Ninomiya Y, Sarras MP Jr, Hudson (1):35–44. https://doi.org/10.1016/j.ccr.
BG, Brooks PC (2000) New functions for 2008.11.012
non-collagenous domains of human collagen 43. Bondareva A, Downey CM, Ayres F, Liu W,
type IV. Novel integrin ligands inhibiting Boyd SK, Hallgrimsson B, Jirik FR (2009)
angiogenesis and tumor growth in vivo. J The lysyl oxidase inhibitor, beta-
Biol Chem 275(11):8051–8061 aminopropionitrile, diminishes the metastatic
216 Chen Varol
colonization potential of circulating breast 55. Withana NP, Blum G, Sameni M, Slaney C,
cancer cells. PLoS One 4(5):e5620. https:// Anbalagan A, Olive MB, Bidwell BN,
doi.org/10.1371/journal.pone.0005620 Edgington L, Wang L, Moin K, Sloane BF,
44. Liotta LA (2016) Adhere, degrade, and move: Anderson RL, Bogyo MS, Parker BS (2012)
the three-step model of invasion. Cancer Res Cathepsin B inhibition limits bone metastasis
76(11):3115–3117. https://doi.org/10. in breast cancer. Cancer Res 72
1158/0008-5472.CAN-16-1297 (5):1199–1209. https://doi.org/10.1158/
45. Gocheva V, Joyce JA (2007) Cysteine cathe- 0008-5472.CAN-11-2759
psins and the cutting edge of cancer invasion. 56. Duong LT, Wesolowski GA, Leung P,
Cell Cycle 6(1):60–64. https://doi.org/10. Oballa R, Pickarski M (2014) Efficacy of a
4161/cc.6.1.3669 cathepsin K inhibitor in a preclinical model
46. Kessenbrock K, Plaks V, Werb Z (2010) for prevention and treatment of breast cancer
Matrix metalloproteinases: regulators of the bone metastasis. Mol Cancer Ther 13
tumor microenvironment. Cell 141 (12):2898–2909. https://doi.org/10.1158/
(1):52–67. https://doi.org/10.1016/j.cell. 1535-7163.MCT-14-0253
2010.03.015 57. Sudhan DR, Siemann DW (2015) Cathepsin
47. Mohamed MM, Sloane BF (2006) Cysteine L targeting in cancer treatment. Pharmacol
cathepsins: multifunctional enzymes in can- Ther 155:105–116. https://doi.org/10.
cer. Nat Rev Cancer 6(10):764–775. 1016/j.pharmthera.2015.08.007
https://doi.org/10.1038/nrc1949 58. Bingle L, Brown NJ, Lewis CE (2002) The
48. Liotta LA, Abe S, Robey PG, Martin GR role of tumour-associated macrophages in
(1979) Preferential digestion of basement tumour progression: implications for new
membrane collagen by an enzyme derived anticancer therapies. J Pathol 196
from a metastatic murine tumor. Proc Natl (3):254–265. https://doi.org/10.1002/
Acad Sci U S A 76(5):2268–2272 path.1027
49. Liotta LA, Tryggvason K, Garbisa S, Hart I, 59. Zhang QW, Liu L, Gong CY, Shi HS, Zeng
Foltz CM, Shafie S (1980) Metastatic poten- YH, Wang XZ, Zhao YW, Wei YQ (2012)
tial correlates with enzymatic degradation of Prognostic significance of tumor-associated
basement membrane collagen. Nature 284 macrophages in solid tumor: a meta-analysis
(5751):67–68 of the literature. PLoS One 7(12):e50946.
https://doi.org/10.1371/journal.pone.
50. Sabeh F, Shimizu-Hirota R, Weiss SJ (2009) 0050946
Protease-dependent versus -independent can-
cer cell invasion programs: three-dimensional 60. Afik R, Zigmond E, Vugman M, Klepfish M,
amoeboid movement revisited. J Cell Biol Shimshoni E, Pasmanik-Chor M, Shenoy A,
185(1):11–19. https://doi.org/10.1083/ Bassat E, Halpern Z, Geiger T, Sagi I, Varol C
jcb.200807195 (2016) Tumor macrophages are pivotal con-
structors of tumor collagenous matrix. J Exp
51. Curino AC, Engelholm LH, Yamada SS, Med 213(11):2315–2331. https://doi.org/
Holmbeck K, Lund LR, Molinolo AA, 10.1084/jem.20151193
Behrendt N, Nielsen BS, Bugge TH (2005)
Intracellular collagen degradation mediated 61. Franklin RA, Liao W, Sarkar A, Kim MV,
by uPARAP/Endo180 is a major pathway of Bivona MR, Liu K, Pamer EG, Li MO
extracellular matrix turnover during malig- (2014) The cellular and molecular origin of
nancy. J Cell Biol 169(6):977–985. https:// tumor-associated macrophages. Science 344
doi.org/10.1083/jcb.200411153 (6186):921–925. https://doi.org/10.1126/
science.1252510
52. Buck MR, Karustis DG, Day NA, Honn KV,
Sloane BF (1992) Degradation of 62. Pollard JW (2004) Tumour-educated macro-
extracellular-matrix proteins by human phages promote tumour progression and
cathepsin B from normal and tumour tissues. metastasis. Nat Rev Cancer 4(1):71–78.
Biochem J 282(Pt 1):273–278 https://doi.org/10.1038/nrc1256
53. Sameni M, Moin K, Sloane BF (2000) Imag- 63. Mantovani A, Ming WJ, Balotta C,
ing proteolysis by living human breast cancer Abdeljalil B, Bottazzi B (1986) Origin and
cells. Neoplasia 2(6):496–504 regulation of tumor-associated macrophages:
the role of tumor-derived chemotactic factor.
54. Sameni M, Dosescu J, Sloane BF (2001) Biochim Biophys Acta 865(1):59–67
Imaging proteolysis by living human glioma
cells. Biol Chem 382(5):785–788. https:// 64. Mantovani A, Sica A (2010) Macrophages,
doi.org/10.1515/BC.2001.094 innate immunity and cancer: balance, toler-
ance, and diversity. Curr Opin Immunol 22
Tumorigenic Interplay Between Macrophages and Collagenous Matrix. . . 217
(2):231–237. https://doi.org/10.1016/j. 74. Lin EY, Nguyen AV, Russell RG, Pollard JW
coi.2010.01.009 (2001) Colony-stimulating factor 1 promotes
65. Hanahan D, Coussens LM (2012) Accessories progression of mammary tumors to malig-
to the crime: functions of cells recruited to the nancy. J Exp Med 193(6):727–740
tumor microenvironment. Cancer Cell 21 75. Zhao L, Lim SY, Gordon-Weeks AN, Tapme-
(3):309–322. https://doi.org/10.1016/j. ier TT, Im JH, Cao Y, Beech J, Allen D,
ccr.2012.02.022 Smart S, Muschel RJ (2013) Recruitment of
66. Mantovani A, Sozzani S, Locati M, Allavena P, a myeloid cell subset (CD11b/Gr1 mid) via
Sica A (2002) Macrophage polarization: CCL2/CCR2 promotes the development of
tumor-associated macrophages as a paradigm colorectal cancer liver metastasis. Hepatology
for polarized M2 mononuclear phagocytes. 57(2):829–839. https://doi.org/10.1002/
Trends Immunol 23(11):549–555 hep.26094
67. Sica A, Saccani A, Bottazzi B, Polentarutti N, 76. Lu X, Kang Y (2009) Chemokine (C-C motif)
Vecchi A, van Damme J, Mantovani A (2000) ligand 2 engages CCR2+ stromal cells of
Autocrine production of IL-10 mediates monocytic origin to promote breast cancer
defective IL-12 production and NF-kappa B metastasis to lung and bone. J Biol Chem
activation in tumor-associated macrophages. J 284(42):29087–29096. https://doi.org/10.
Immunol 164(2):762–767 1074/jbc.M109.035899
68. Mantovani A, Savino B, Locati M, 77. Qian BZ, Li J, Zhang H, Kitamura T,
Zammataro L, Allavena P, Bonecchi R Zhang J, Campion LR, Kaiser EA, Snyder
(2010) The chemokine system in cancer biol- LA, Pollard JW (2011) CCL2 recruits inflam-
ogy and therapy. Cytokine Growth Factor Rev matory monocytes to facilitate breast-tumour
21(1):27–39. https://doi.org/10.1016/j. metastasis. Nature 475(7355):222–225.
cytogfr.2009.11.007 https://doi.org/10.1038/nature10138
69. Lin EY, Pollard JW (2007) Tumor-associated 78. Kitamura T, Qian BZ, Soong D, Cassetta L,
macrophages press the angiogenic switch in Noy R, Sugano G, Kato Y, Li J, Pollard JW
breast cancer. Cancer Res 67 (2015) CCL2-induced chemokine cascade
(11):5064–5066. https://doi.org/10.1158/ promotes breast cancer metastasis by enhanc-
0008-5472.CAN-07-0912 ing retention of metastasis-associated macro-
70. Leek RD, Lewis CE, Whitehouse R, phages. J Exp Med 212(7):1043–1059.
Greenall M, Clarke J, Harris AL (1996) Asso- https://doi.org/10.1084/jem.20141836
ciation of macrophage infiltration with angio- 79. Vaage J, Pepin KG (1985) Morphological
genesis and prognosis in invasive breast observations during developing concomitant
carcinoma. Cancer Res 56(20):4625–4629 immunity against a C3H/he mammary
71. Lewis JS, Landers RJ, Underwood JC, Harris tumor. Cancer Res 45(2):659–666
AL, Lewis CE (2000) Expression of vascular 80. Vaage J, Harlos JP (1991) Collagen produc-
endothelial growth factor by macrophages is tion by macrophages in tumour encapsulation
up-regulated in poorly vascularized areas of and dormancy. Br J Cancer 63(5):758–762
breast carcinomas. J Pathol 192(2):150–158. 81. Ansorge HL, Meng X, Zhang G, Veit G,
https://doi.org/10.1002/1096-9896( Sun M, Klement JF, Beason DP, Soslowsky
2000)9999:9999<::AID-PATH687>3.0. LJ, Koch M, Birk DE (2009) Type XIV colla-
CO;2-G gen regulates fibrillogenesis: premature colla-
72. Colegio OR, Chu NQ, Szabo AL, Chu T, gen fibril growth and tissue dysfunction in
Rhebergen AM, Jairam V, Cyrus N, Bro- null mice. J Biol Chem 284(13):8427–8438.
kowski CE, Eisenbarth SC, Phillips GM, https://doi.org/10.1074/jbc.M805582200
Cline GW, Phillips AJ, Medzhitov R (2014) 82. Gerecke DR, Meng X, Liu B, Birk DE (2003)
Functional polarization of tumour-associated Complete primary structure and genomic
macrophages by tumour-derived lactic acid. organization of the mouse Col14a1 gene.
Nature 513(7519):559–563. https://doi. Matrix Biol 22(3):209–216
org/10.1038/nature13490 83. Liguori M, Solinas G, Germano G,
73. Wyckoff J, Wang W, Lin EY, Wang Y, Pixley F, Mantovani A, Allavena P (2011) Tumor-
Stanley ER, Graf T, Pollard JW, Segall J, Con- associated macrophages as incessant builders
deelis J (2004) A paracrine loop between and destroyers of the cancer stroma. Cancer 3
tumor cells and macrophages is required for (4):3740–3761. https://doi.org/10.3390/
tumor cell migration in mammary tumors. cancers3043740
Cancer Res 64(19):7022–7029. https://doi. 84. Zhu Y, Herndon JM, Sojka DK, Kim KW,
org/10.1158/0008-5472.CAN-04-1449 Knolhoff BL, Zuo C, Cullinan DR, Luo J,
218 Chen Varol
Bearden AR, Lavine KJ, Yokoyama WM, 92. Meng XM, Wang S, Huang XR, Yang C,
Hawkins WG, Fields RC, Randolph GJ, Xiao J, Zhang Y, To KF, Nikolic-Paterson
DeNardo DG (2017) Tissue-resident macro- DJ, Lan HY (2016) Inflammatory macro-
phages in pancreatic ductal adenocarcinoma phages can transdifferentiate into myofibro-
originate from embryonic hematopoiesis and blasts during renal fibrosis. Cell Death Dis 7
promote tumor progression. Immunity 47 (12):e2495. https://doi.org/10.1038/
(2):323–338 e326. https://doi.org/10. cddis.2016.402
1016/j.immuni.2017.07.014 93. Yang J, Lin SC, Chen G, He L, Hu Z, Chan L,
85. Puolakkainen PA, Brekken RA, Muneer S, Trial J, Entman ML, Wang Y (2013) Adipo-
Sage EH (2004) Enhanced growth of pancre- nectin promotes monocyte-to-fibroblast tran-
atic tumors in SPARC-null mice is associated sition in renal fibrosis. J Am Soc Nephrol 24
with decreased deposition of extracellular (10):1644–1659. https://doi.org/10.1681/
matrix and reduced tumor cell apoptosis. ASN.2013030217
Mol Cancer Res 2(4):215–224 94. Weitkamp B, Cullen P, Plenz G, Robenek H,
86. Sangaletti S, Stoppacciaro A, Guiducci C, Rauterberg J (1999) Human macrophages
Torrisi MR, Colombo MP (2003) Leukocyte, synthesize type VIII collagen in vitro and in
rather than tumor-produced SPARC, deter- the atherosclerotic plaque. FASEB J 13
mines stroma and collagen type IV deposition (11):1445–1457
in mammary carcinoma. J Exp Med 198 95. Medbury HJ, James V, Ngo J, Hitos K,
(10):1475–1485. https://doi.org/10.1084/ Wang Y, Harris DC, Fletcher JP (2013) Dif-
jem.20030202 fering association of macrophage subsets with
87. Sangaletti S, Di Carlo E, Gariboldi S, atherosclerotic plaque stability. Int Angiol 32
Miotti S, Cappetti B, Parenza M, Rumio C, (1):74–84
Brekken RA, Chiodoni C, Colombo MP 96. Cieslik KA, Taffet GE, Carlson S,
(2008) Macrophage-derived SPARC bridges Hermosillo J, Trial J, Entman ML (2011)
tumor cell-extracellular matrix interactions Immune-inflammatory dysregulation modu-
toward metastasis. Cancer Res 68 lates the incidence of progressive fibrosis and
(21):9050–9059. https://doi.org/10.1158/ diastolic stiffness in the aging heart. J Mol
0008-5472.CAN-08-1327 Cell Cardiol 50(1):248–256. https://doi.
88. Burke RM, Madden KS, Perry SW, Zettel ML, org/10.1016/j.yjmcc.2010.10.019
Brown EB 3rd (2013) Tumor-associated 97. Bertrand S, Godoy M, Semal P, Van Gansen P
macrophages and stromal TNF-alpha regulate (1992) Transdifferentiation of macrophages
collagen structure in a breast tumor model as into fibroblasts as a result of Schistosoma
visualized by second harmonic generation. J mansoni infection. Int J Dev Biol 36
Biomed Opt 18(8):86003. https://doi.org/ (1):179–184
10.1117/1.JBO.18.8.086003 98. Mass E, Jacome-Galarza CE, Blank T,
89. Allaoui R, Bergenfelz C, Mohlin S, Lazarov T, Durham BH, Ozkaya N,
Hagerling C, Salari K, Werb Z, Anderson Pastore A, Schwabenland M, Chung YR,
RL, Ethier SP, Jirstrom K, Pahlman S, Rosenblum MK, Prinz M, Abdel-Wahab O,
Bexell D, Tahin B, Johansson ME, Geissmann F (2017) A somatic mutation in
Larsson C, Leandersson K (2016) Cancer- erythro-myeloid progenitors causes neurode-
associated fibroblast-secreted CXCL16 generative disease. Nature 549
attracts monocytes to promote stroma activa- (7672):389–393. https://doi.org/10.1038/
tion in triple-negative breast cancers. Nat nature23672
Commun 7:13050. https://doi.org/10. 99. Sinha M, Sen CK, Singh K, Das A, Ghatak S,
1038/ncomms13050 Rhea B, Blackstone B, Powell HM, Khanna S,
90. Schnoor M, Cullen P, Lorkowski J, Stolle K, Roy S (2018) Direct conversion of injury-site
Robenek H, Troyer D, Rauterberg J, Lor- myeloid cells to fibroblast-like cells of granu-
kowski S (2008) Production of type VI colla- lation tissue. Nat Commun 9(1):936.
gen by human macrophages: a new dimension https://doi.org/10.1038/s41467-018-
in macrophage functional heterogeneity. J 03208-w
Immunol 180(8):5707–5719 100. Matsubayashi Y, Louani A, Dragu A,
91. Nikolic-Paterson DJ, Wang S, Lan HY (2014) Sanchez-Sanchez BJ, Serna-Morales E,
Macrophages promote renal fibrosis through Yolland L, Gyoergy A, Vizcay G, Fleck RA,
direct and indirect mechanisms. Kidney Int Heddleston JM, Chew TL, Siekhaus DE,
Suppl 4(1):34–38. https://doi.org/10. Stramer BM (2017) A moving source of
1038/kisup.2014.7 matrix components is essential for de novo
basement membrane formation. Curr Biol
Tumorigenic Interplay Between Macrophages and Collagenous Matrix. . . 219
27(22):3526–3534 e3524. https://doi.org/ 110. Zeng ZS, Cohen AM, Guillem JG (1999)
10.1016/j.cub.2017.10.001 Loss of basement membrane type IV collagen
101. Kalluri R, Zeisberg M (2006) Fibroblasts in is associated with increased expression of
cancer. Nat Rev Cancer 6(5):392–401. metalloproteinases 2 and 9 (MMP-2 and
https://doi.org/10.1038/nrc1877 MMP-9) during human colorectal tumori-
102. Vignaud JM, Marie B, Klein N, Plenat F, genesis. Carcinogenesis 20(5):749–755
Pech M, Borrelly J, Martinet N, Duprez A, 111. Hotary K, Li XY, Allen E, Stevens SL, Weiss
Martinet Y (1994) The role of platelet- SJ (2006) A cancer cell metalloprotease triad
derived growth factor production by tumor- regulates the basement membrane transmi-
associated macrophages in tumor stroma for- gration program. Genes Dev 20
mation in lung cancer. Cancer Res 54 (19):2673–2686. https://doi.org/10.1101/
(20):5455–5463 gad.1451806
103. Pickup M, Novitskiy S, Moses HL (2013) 112. Jiang D, Lim SY (2016) Influence of immune
The roles of TGFbeta in the tumour micro- myeloid cells on the extracellular matrix dur-
environment. Nat Rev Cancer 13 ing cancer metastasis. Cancer Microenviron 9
(11):788–799. https://doi.org/10.1038/ (1):45–61. https://doi.org/10.1007/
nrc3603 s12307-016-0181-6
104. Karlmark KR, Weiskirchen R, Zimmermann 113. Coussens LM, Tinkle CL, Hanahan D, Werb
HW, Gassler N, Ginhoux F, Weber C, Z (2000) MMP-9 supplied by bone marrow-
Merad M, Luedde T, Trautwein C, Tacke F derived cells contributes to skin carcinogene-
(2009) Hepatic recruitment of the inflamma- sis. Cell 103(3):481–490
tory Gr1+ monocyte subset upon liver injury 114. Huang S, Van Arsdall M, Tedjarati S,
promotes hepatic fibrosis. Hepatology 50 McCarty M, Wu W, Langley R, Fidler IJ
(1):261–274. https://doi.org/10.1002/ (2002) Contributions of stromal
hep.22950 metalloproteinase-9 to angiogenesis and
105. Hesketh M, Sahin KB, West ZE, Murray RZ growth of human ovarian carcinoma in mice.
(2017) Macrophage phenotypes regulate scar J Natl Cancer Inst 94(15):1134–1142
formation and chronic wound healing. Int J 115. Hiratsuka S, Nakamura K, Iwai S,
Mol Sci 18(7):E1545. https://doi.org/10. Murakami M, Itoh T, Kijima H, Shipley JM,
3390/ijms18071545 Senior RM, Shibuya M (2002) MMP9 induc-
106. Knipper JA, Willenborg S, Brinckmann J, tion by vascular endothelial growth factor
Bloch W, Maass T, Wagener R, Krieg T, receptor-1 is involved in lung-specific metas-
Sutherland T, Munitz A, Rothenberg ME, tasis. Cancer Cell 2(4):289–300
Niehoff A, Richardson R, 116. Giraudo E, Inoue M, Hanahan D (2004) An
Hammerschmidt M, Allen JE, Eming SA amino-bisphosphonate targets MMP-9-
(2015) Interleukin-4 receptor alpha signaling expressing macrophages and angiogenesis to
in myeloid cells controls collagen fibril assem- impair cervical carcinogenesis. J Clin Invest
bly in skin repair. Immunity 43(4):803–816. 114(5):623–633. https://doi.org/10.1172/
https://doi.org/10.1016/j.immuni.2015. JCI22087
09.005 117. Du R, Lu KV, Petritsch C, Liu P, Ganss R,
107. Zhou X, Franklin RA, Adler M, Jacox JB, Passegue E, Song H, Vandenberg S, Johnson
Bailis W, Shyer JA, Flavell RA, Mayo A, RS, Werb Z, Bergers G (2008) HIF1alpha
Alon U, Medzhitov R (2018) Circuit design induces the recruitment of bone marrow-
features of a stable two-cell system. Cell 172 derived vascular modulatory cells to regulate
(4):744–757 e717. https://doi.org/10. tumor angiogenesis and invasion. Cancer Cell
1016/j.cell.2018.01.015 13(3):206–220. https://doi.org/10.1016/j.
108. Fields GB (2013) Interstitial collagen catabo- ccr.2008.01.034
lism. J Biol Chem 288(13):8785–8793. 118. Deryugina EI, Zajac E, Juncker-Jensen A,
https://doi.org/10.1074/jbc.R113.451211 Kupriyanova TA, Welter L, Quigley JP
109. Collins HM, Morris TM, Watson SA (2001) (2014) Tissue-infiltrating neutrophils consti-
Spectrum of matrix metalloproteinase expres- tute the major in vivo source of angiogenesis-
sion in primary and metastatic colon cancer: inducing MMP-9 in the tumor microenviron-
relationship to the tissue inhibitors of metal- ment. Neoplasia 16(10):771–788. https://
loproteinases and membrane type-1-matrix doi.org/10.1016/j.neo.2014.08.013
metalloproteinase. Br J Cancer 84 119. Madsen DH, Jurgensen HJ, Siersbaek MS,
(12):1664–1670. https://doi.org/10.1054/ Kuczek DE, Grey Cloud L, Liu S,
bjoc.2001.1831 Behrendt N, Grontved L, Weigert R, Bugge
TH (2017) Tumor-associated macrophages
220 Chen Varol
derived from circulating inflammatory mono- 125. Postlethwaite AE, Kang AH (1976)
cytes degrade collagen through cellular Collagen-and collagen peptide-induced che-
uptake. Cell Rep 21(13):3662–3671. motaxis of human blood monocytes. J Exp
https://doi.org/10.1016/j.celrep.2017.12. Med 143(6):1299–1307
011 126. Wiesner C, Le-Cabec V, El Azzouzi K,
120. Gocheva V, Wang HW, Gadea BB, Shree T, Maridonneau-Parini I, Linder S (2014)
Hunter KE, Garfall AL, Berman T, Joyce JA Podosomes in space: macrophage migration
(2010) IL-4 induces cathepsin protease activ- and matrix degradation in 2D and 3D set-
ity in tumor-associated macrophages to pro- tings. Cell Adhes Migr 8(3):179–191
mote cancer growth and invasion. Genes Dev 127. Van Goethem E, Poincloux R, Gauffre F,
24(3):241–255. https://doi.org/10.1101/ Maridonneau-Parini I, Le Cabec V (2010)
gad.1874010 Matrix architecture dictates three-
121. Vasiljeva O, Papazoglou A, Kruger A, dimensional migration modes of human
Brodoefel H, Korovin M, Deussing J, macrophages: differential involvement of pro-
Augustin N, Nielsen BS, Almholt K, teases and podosome-like structures. J Immu-
Bogyo M, Peters C, Reinheckel T (2006) nol 184(2):1049–1061. https://doi.org/10.
Tumor cell-derived and macrophage-derived 4049/jimmunol.0902223
cathepsin B promotes progression and lung 128. Kaplan G (1983) In vitro differentiation of
metastasis of mammary cancer. Cancer Res human monocytes. Monocytes cultured on
66(10):5242–5250. https://doi.org/10. glass are cytotoxic to tumor cells but mono-
1158/0008-5472.CAN-05-4463 cytes cultured on collagen are not. J Exp Med
122. Doyle AD, Yamada KM (2016) Mechanosen- 157(6):2061–2072
sing via cell-matrix adhesions in 3D microen- 129. Wesley RB 2nd, Meng X, Godin D, Galis ZS
vironments. Exp Cell Res 343(1):60–66. (1998) Extracellular matrix modulates mac-
https://doi.org/10.1016/j.yexcr.2015.10. rophage functions characteristic to atheroma:
033 collagen type I enhances acquisition of resi-
123. Jin H, Varner J (2004) Integrins: roles in dent macrophage traits by human peripheral
cancer development and as treatment targets. blood monocytes in vitro. Arterioscler
Br J Cancer 90(3):561–565. https://doi. Thromb Vasc Biol 18(3):432–440
org/10.1038/sj.bjc.6601576 130. Patel NR, Bole M, Chen C, Hardin CC, Kho
124. Franco C, Britto K, Wong E, Hou G, Zhu AT, Mih J, Deng L, Butler J, Tschumperlin D,
SN, Chen M, Cybulsky MI, Bendeck MP Fredberg JJ, Krishnan R, Koziel H (2012)
(2009) Discoidin domain receptor 1 on Cell elasticity determines macrophage func-
bone marrow-derived cells promotes macro- tion. PLoS One 7(9):e41024. https://doi.
phage accumulation during atherogenesis. org/10.1371/journal.pone.0041024
Circ Res 105(11):1141–1148. https://doi.
org/10.1161/CIRCRESAHA.109.207357
Chapter 16
Abstract
Chronic inflammatory diseases are often associated with organ fibrosis, a progressive condition in which
excessive deposition of extracellular matrix (ECM), mainly composed of collagen I (Col I), is deposited by
activated fibroblasts and severely impairs tissue architecture and function, eventually resulting in organ
failure. Moreover, enhanced collagen deposition by activated fibroblasts and increased stiffness of the
extracellular matrix were demonstrated to be associated with tumor progression and metastasis. In order
to quantitatively analyze fibrotic activation of fibroblasts and collagen deposition, it is essential to assess
collagen content. While various histological methods allow assessment of collagen in tissue sections (e.g.,
Masson trichrome and Sirius red), reliable measurement and quantification of collagen levels in vitro remain
a challenge in the field. In this protocol, we utilize intracellular staining of Col1α and flow cytometry
analysis to analyze collagen content in primary fibroblasts isolated from fresh single cell suspensions of
metastases-bearing lungs.
1 Introduction
Irit Sagi and Nikolaos A. Afratis (eds.), Collagen: Methods and Protocols, Methods in Molecular Biology, vol. 1944,
https://doi.org/10.1007/978-1-4939-9095-5_16, © Springer Science+Business Media, LLC, part of Springer Nature 2019
221
222 Noam Cohen and Neta Erez
2 Materials
2.1 Lung Dissection 1. Surgery tools (washed with detergent and then dipped in 70%
ethanol).
2. Styrofoam surface to rest the mice on during dissection.
3. Pins or needles to hold mice during dissection.
4. Plastic jar with lid for digestion.
5. Magnetic stir bar.
Analysis of Col1α Levels in Primary Fibroblasts 223
6. Incubator at 37 C.
7. Multipoint magnetic stirrer.
8. PBS.
9. 10 cm plate.
2.3 Cell Culture 1. Collagen mixture: 0.05 mg rat tail collagen I in 1 ml 0.02 N
acetic acid. Adjust volumes as required in specific steps (see
Note 1).
2. 10% FCS DMEM.
3. DMEM.
4. Six-well plate.
5. 10 cm plate.
6. Trypsin-EDTA.
7. PBS.
3 Methods
The protocol described is suitable for six mice, but can be easily
adjusted for smaller or lager mouse numbers.
3.1 Lung Dissection Since isolated fibroblasts are cultured, it is best to perform the
dissection in a laminar flow hood, under sterile conditions.
1. Euthanize mice using carbon dioxide (CO2) inhalation.
224 Noam Cohen and Neta Erez
Fig. 1 Lung dissection, sequential steps from left to right: pin mouse to diaper coated Styrofoam surface,
dissect out skin, cut through the ribs with scissors and use forceps to disconnect the lungs
3.3 Cell Culture 1. On day 2, wash cells with PBS and change to fresh media to
dispose of any cell debris.
2. On day 3 (or when cells are ~80% confluent, usually takes
1–2 days after isolation), coat six 10 cm plates with collagen
mixture (7 ml per plate), and incubate at 37 C incubator for
30 min (see Note 4) (Fig. 3).
3. Detach cells with trypsin-EDTA and split into 10 cm plates
(one well into one plate).
4. On day 4, wash cells with PBS and change to fresh media.
5. When cells are 80–90% confluent (usually takes 1–2 days),
detach cells, count them, and seed 300,000 cells per well in a
six-well plate for desired experiments (see Note 5).
6. On the next day, wash cell twice with PBS, and incubate with
SFM for 6 h (see Note 6).
7. Wash out the SFM and incubate cells with your treatment
media for 24 h.
226 Noam Cohen and Neta Erez
Fig. 3 Bright field image of cultured lung fibroblasts at ~90% confluency. Magnification 100
Fig. 4 FACS analysis of Col1α in NLF analyzed by Kaluza® 1.5 Flow Analysis Software (Beckman Coulter, Inc.)
4 Notes
References
Abstract
The triple-helical structure of collagen has been accurately reproduced in numerous chemical and recombi-
nant model systems. Triple-helical peptides have found application for dissecting collagen-stabilizing forces,
isolating receptor and protein binding sites in collagen, evaluating collagen-mediated cell signaling activ-
ities, mechanistic examination of collagenolytic proteases, and developing novel biomaterials and drug
delivery vehicles. Due to their inherent stability to general proteolysis, triple-helical peptides present an
opportunity as in vivo inhibitory agents. The present chapter provides methods for the construction of
collagen-based triple-helical peptides designed as matrix metalloproteinase inhibitors.
Key words Collagen, Matrix metalloproteinases, Triple-helical peptides, Triple-helical inhibitor, Pep-
tide-based drugs, Transition state analog
1 Introduction
1.1 Collagen Collagen is the most abundant protein in animals and is the major
structural protein found in the connective tissues such as basement
membranes, tendons, ligaments, cartilage, bone, and skin. The
defining feature of collagen is the supersecondary structure, com-
posed of three parallel extended left-handed polyproline type II
alpha chains of primarily repeating Gly-Xxx-Yyy triplets. Three
left-handed strands intertwine in a right-handed fashioned around
a common axis to form a triple helix. The packing of the triple-
helical coiled-coil structure favors Gly in every third position. In the
collagen Gly-Xxx-Yyy triplet, the residue in the Xxx position is
often L-proline (Pro), and the residue in the Yyy position is often
4(R)-hydroxy-L-proline (Hyp), accounting for 20% of the total
amino acid composition in collagen [1].
Depending on the primary structure of the alpha chains [2],
collagens have been classified into two main categories,
Irit Sagi and Nikolaos A. Afratis (eds.), Collagen: Methods and Protocols, Methods in Molecular Biology, vol. 1944,
https://doi.org/10.1007/978-1-4939-9095-5_17, © Springer Science+Business Media, LLC, part of Springer Nature 2019
229
230 Gregg B. Fields
1.3 THPs Designed THPs represent small, well-folded peptidic structures. There has
as Inhibitors been a recent surge in the advancement of peptide drugs, as they
tend to have lower toxicities and greater efficacies than existing
drugs [21–24]. The size of THPs makes immunogenicity unlikely;
in fact, with the exclusion of specific glycosylated sequences, colla-
gen peptides are non-immunogenic. The triple-helical nature of the
inhibitor renders it reasonably stable to proteolysis, as observed
in vitro in mouse, rat, and human serum and/or plasma and
in vivo in rats [25–29]. The stability of THPs has allowed for
their administration orally [30].
Application of THPs as biomaterials and drug delivery vehicles
has been described recently [27, 31–33]. The use of THPs as
inhibitors has been explored to a considerably lesser extent. When
identifying integrin binding sites, THPs have been shown to inhibit
integrin binding to native collagens [34–36]. THPs that bind to
leukocyte-associated Ig-like receptor-1 (LAIR-1) induce inhibition
of T-cell activation [37], while THPs that bind to human osteoclast-
associated receptor (OSCAR) inhibit osteoclastogenesis [38].
Fig. 1 Tetrahedral intermediate (boxed) and statin and phosphorus-based transition state analog inhibitors.
Used with permission from [40]
Fig. 2 Model structure of a triple-helical peptide inhibitor (THPI) of MMPs. The non-hydrolyzable PO2H–CH2
bond, which functions as a transition state analog, is indicated in purple. Used with permission from [40]
Table 1
Inhibition of MMP-2 and MMP-9 by THPIs
Table 2
Inhibition of MMPs by GlyΨ{PO2H–CH2}Xxx THPIs
Table 3
Inhibition of MMPs by heterotrimeric THPIs
NI ¼ No inhibition
b
2 Materials
Fig. 3 Structures of Fmoc-phosphinate dipeptide building blocks (3), Fmoc-Flp (4), and Fmoc-mep (5).
The phosphinate dipeptides are analogs of Gly-Val (3a), Gly-Leu (3b), and Gly-Ile (3c). Used with permission
from [49]
2.2 HPLC 1. All analytical runs are performed using a Vydac C18 column
Characterization (5 μm, 300 Å, 150 mm 4.6 mm) or similar.
and Purification 2. Preparative runs are carried out using a Vydac C18 column
(15–20 μm, 300 Å, 250 mm 22 mm) or similar.
236 Gregg B. Fields
2.3 Mass The molecular weight of peptides and peptide conjugates are deter-
Spectrometry mined using matrix-assisted laser desorption/ionization time-of-
flight mass spectrometry (MALDI-TOF MS). MALDI-TOF MS
analysis is performed using an Applied Biosystems Voyager
DE-PRO Biospectrometry workstation or Bruker Microflex LF
mass spectrometer using sinapinic acid (SA), α-cyano-4-hydroxy-
cinnamic acid (HCCA), or 2,5-dihydroxybenzoic acid (DHB) as
matrix.
3 Methods
3.1.1 General Amino Acid For Fmoc-amino acid couplings, the following molar excess is
Coupling Cycles recommended: Fmoc-AA ¼ 5 equiv., HCTU ¼ 4.9 equiv.,
HOBt ¼ 4.9 equiv., and N-methylmorpholine (NMM) or
DIPEA ¼ 10 equiv. Recommended coupling time using conven-
tional automatic peptide synthesizer is 1 h, while microwave-
assisted synthesis proceeds for 5 min at 50 C and 25 W.
3.1.3 General Peptide- 1. Place the peptide-resin in a round bottom flask under inert
Resin Cleavage Protocol atmosphere (Ar) in a fume hood.
2. Mix 50 μL of H2O and 50 μL thioanisole with 900 μL of TFA
to create the cleavage cocktail.
3. Cleave the peptide from the resin using 1.5 mL of cleavage
cocktail per 100 mg of dry resin.
4. Cleavage and side-chain deprotection of the peptide is accom-
plished by treating the resin for 3–4 h (see Note 1).
5. Filter the resin and collect the filtrate.
6. Crude peptides are precipitated from the filtrate with cold
MTBE and centrifuged.
7. Dissolve the pellet in H2O and lyophilize.
3.1.4 Solid-Phase General methods Subheadings 3.1.1 and 3.1.2 are followed, with
Synthesis of GlyΨ the following modifications.
{PO2H–CH2}Ile-His-Lys-Gln
1. Synthesis is performed on a Liberty automated microwave
THPI and GlyΨ {PO2H–CH2}
peptide synthesizer using NovaPEG Rink Amide resin (loading
Ile-Tyr-Phe-Gln THPIs
0.5 mmol/g).
2. The coupling of usual Fmoc-amino acids is achieved with
5 equiv. of each amino acid, 4.9 equiv. of HCTU (0.5 M in
DMF), and 8.0 equiv. of NMM (2 M in DMF) using micro-
wave power of 25 W at 50 C with the reaction time of 300 s
(double coupling for all Arg residues first at 0 W, 25 C for
25 min followed by 25 W, 50 C for 300 s).
3. The removal of Fmoc-protecting group is accomplished using
10% piperazine in DMF (microwave power of 35 W at 75 C
and the reaction time of 30 s, followed by a second treatment
under the same conditions for 180 s).
4. The unusual amino acids Fmoc-4R-fluoro-L-proline (Fmoc-
Flp-OH) and Fmoc-4R-methyl-L-proline (Fmoc-mep-OH)
are coupled using 3 equiv. of amino acid, 2.9 equiv. of
HCTU, and 6 equiv. of NMM with microwave power of
25 W at 50 C for 300 s. The coupling of Fmoc-mep-OH is
repeated.
5. The Fmoc-protected phosphinic dipeptides are attached using
3 equiv. dipeptide, 3 equiv. DIPCDI, 6 equiv. HOBt, and
DIPEA to adjust pH to 8 at 40 C for 40 min and room
temperature overnight.
6. Following dipeptide addition the peptide-resin is capped using
0.5 M Ac2O, 0.125 M DIPEA, and 0.015 M HOBt in DMF at
room temperature for 1 h.
7. The deprotection of Fmoc group, immediately after incorpora-
tion of the phosphinic dipeptide into the sequence, is
Collagen Peptide Inhibitors 239
3.1.5 Solid-Phase General methods Subheadings 3.1.1 and 3.1.2 are followed, with
Synthesis of α1(V)GlyΨ the following modifications.
{PO2H–CH2}Val [mep14,32,
1. Synthesis is performed on a CEM liberty blue automated
Flp15,33] THPI
microwave peptide synthesizer using NovaPEG Rink Amide
resin (loading 0.48 mmol/g).
2. The coupling of usual Fmoc-amino acids is achieved with
5 equiv. of each amino acid (0.2 M), 5 equiv. of DIPCDI
(0.5 M in DMF), and 5 equiv. of Oxyma (1 M in DMF).
Fmoc-amino acid coupling cycles are carried out for 15 s at
75 C and 110 s at 90 C.
3. Fmoc deprotection is achieved with 10% piperazine (w/v) in
ethanol:N-methyl-2-pyrrolidone (10:90) for 15 s at 75 C and
50 s at 90 C.
4. Fmoc-Flp-OH is incorporated using 2.5 equiv. of amino acid
for 600 s at 25 C and 300 s at 75 C.
5. Fmoc-mep-OH and Fmoc-GlyΨ{PO2H–CH2}Val are
incorporated using 2.5 equiv. of each compound for 1500 s
at 25 C and 600 s at 75 C.
6. Coupling of Fmoc-Pro-OH following GlyΨ{PO2H–CH2}Val
is performed for 1500 s at 25 C and 600 s at 75 C for using
5 equiv. of amino acid.
7. The final cleavage of peptide from the resin and side-chain
deprotection is carried out using 7 mL of triisopropylsilane-
phenol-H2O-TFA (2:5:5:88) under inert atmosphere
(Ar) for 3 h.
3.1.6 Synthesis General methods Subheadings 3.1.1 and 3.1.2 are followed, with
of Single-Stranded Peptide the following modifications.
1 (SSP1)
1. Synthesis of SSP1 (Fig. 4) is performed in 0.1 mmol scale on
NovaPEG Rink Amide resin (0.48 mmol/g).
2. The coupling of Fmoc-(2S)-propargylglycine (Fmoc-Pra-OH)
is carried out at 45 C for 45 min using 25 W microwave power.
3. The coupling of Pro14 is repeated.
240 Gregg B. Fields
(GPO)nGPQGLAGQRGIR(GPO)5G-HN
(GPO)nGPQGLAGQRGIR(GPO)5G-Lys-Lys-NH2
N3
(GPO)5GPQGLAGQRGIR(GPO)5-Pra-NH2
Cu(I)
(GPO)nGPQGLAGQRGIR(GPO)5G-HN
(GPO)nGPQGLAGQRGIR(GPO)5G-Lys-Lys-NH2
N
N
N
(GPO)5GPQGLAGQRGIR(GPO)5-Pra-NH2
3.1.7 Synthesis General methods Subheadings 3.1.1 and 3.1.2 are followed, with
of Single-Stranded Peptide the following modifications.
2 (SSP2)
1. Synthesis of SSP2 (Figs. 5 and 6) was performed on TentaGel
resin (0.26 mmol/g).
2. The coupling of Fmoc-Pra is as for SSP1. Residues Arg24 and
Arg27 are double coupled, where the first coupling is for
25 min at 25 C using no power followed by 5 min at 25 C
using 25 W power, and the second coupling is for 5 min at
50 C with microwave power of 25 W.
3. Fmoc removal at Ala21 is achieved by initial treatment for 30 s
and final treatment for 3 min at 75 C with microwave power of
35 W.
4. Coupling of the Fmoc-GlyΨ{P(OAd)(OH)–CH2}Ile-OH
dipeptide (3 equiv.) is performed using N,N0 -diisopropyl-
carbodiimide (DIC)/HOBt/DIPEA (3 equiv./6 equiv./3
equiv.) for 80 min at 40 C with microwave power of 25 W
and then left overnight at ambient temperature.
5. The removal of the Fmoc group, immediately after incorpora-
tion of the dipeptide into the sequence and for the rest of the
synthesis, is performed in two stages: Initial and final
Collagen Peptide Inhibitors 241
(GPO)6GPQGLAGQRGIR(GPO)5G-HN
(GPO)6GPQGLAGQRGIR(GPO)5G-Lys-Lys-NH2 DSP4
N3
(GPO)5GPQ{GΨ(PO2H-CH2)I}AGQRGIR(GPO)5G-Pra-NH2 SSP2
Cu(I)
(GPO)6GPQGLAGQRGIR(GPO)5G-HN
(GPO)6GPQGLAGQRGIR(GPO)5G-Lys-Lys-NH2
THCP8
N
N
N
(GPO)5GPQ{GΨ(PO2H-CH2)I}AGQRGIR(GPO)5G-Pra-NH2
Fig. 5 Schematic of the click-based assembly of THCP8. Assembly was achieved with DSP4 and SSP2. Used
with permission from [48]
(GPO)6GPQGLAGQRGIR(GPO)5G-HN
(GPO)6GPQGLAGQRGIR(GPO)5G-Lys-Lys-NH2 DSP5
N3
(GPO)5GPQ{GΨ(PO2H-CH2)I}AGQRGIR(GPO)5G-Pra-NH2 SSP2
Cu(I)
(GPO)6GPQGLAGQRGIR(GPO)5G-HN
(GPO)6GPQGLAGQRGIR(GPO)5G-Lys-Lys-NH2
THCP9
N
N
N
(GPO)5GPQ{GΨ(PO2H-CH2)I}AGQRGIR(GPO)5G-Pra-NH2
Fig. 6 Schematic of the click-based assembly of THCP9. Assembly was achieved with DSP5 and SSP2. Used
with permission from [48]
242 Gregg B. Fields
3.1.8 Synthesis General methods Subheadings 3.1.1 and 3.1.2 are followed, with
of Double-Stranded the following modifications.
Peptide 3 (DSP3)
1. The synthesis of DSP3 (Fig. 4) is performed on NovaPEG Rink
Amide resin (0.6 mmol/g).
2. Fmoc-Lys(ivDde)-OH is initially coupled to the resin followed
by Fmoc-Lys(N3)-OH. Both couplings are carried out using
3 equiv. of amino acid, 2.7 equiv. of HCTU, 2.8 equiv. of
HOBt, and 6 equiv. of NMM at 50 C for 20 min with
microwave power of 25 W.
3. The resin-bound dipeptide is removed from the synthesizer,
treated twice with 4 mL of 2% hydrazine hydrate in DMF for
1 h each time to remove the ivDde group, washed with DMF,
and transferred back to the synthesizer reaction vessel.
4. Coupling of the remaining residues is extended to 15 min, and
Pro14 was double coupled.
3.1.9 Synthesis General methods Subheadings 3.1.1 and 3.1.2 are followed, with
of Double-Stranded the following modifications.
Peptide 4 (DSP4)
1. Synthesis of DSP4 (Figs. 4 and 5) is carried out by adding one
more Gly-Pro-Hyp repeat on resin-bound DSP3 applying the
standard peptide synthesis protocol.
3.1.10 Synthesis General methods Subheadings 3.1.1 and 3.1.2 are followed, with
of Double-Stranded the following modifications.
Peptide 5 (DSP5)
1. Synthesis of DSP5 (Fig. 6) is performed on TentaGel resin
(0.26 mmol/g) using the procedure outlined for DSP3 but
first coupling Fmoc-Lys(N3)-OH followed by Fmoc-Lys
(ivDde)-OH.
3.1.11 Synthesis General methods Subheadings 3.1.1 and 3.1.2 are followed, with
of Single-Stranded Peptide the following modifications.
10 (SSP10)
1. Synthesis of SSP10 (Fig. 7) is performed on TentaGel S Ram
resin (0.26 mmol/g).
2. Two consecutive Fmoc-Lys(N3) residues are coupled to the
resin as described for DSP3.
Collagen Peptide Inhibitors 243
(GPO)5GPQGLAGQRGIR(GPO)5G-Pra-NH2 SSP11
N3
(GPO)6GPQ{GΨ(PO2H-CH2)I}AGQRGIR(GPO)5G-Lys-Lys-NH2 SSP10
N3
(GPO)5GPQGLAGQRGIR(GPO)5G-Pra-NH2 SSP11
Cu(I)
(GPO)5GPQGLAGQRGIR(GPO)5G-Pra-NH2
N
N
N
(GPO)6GPQ{GΨ(PO2H-CH2)I}AGQRGIR(GPO)5G-Lys-Lys-NH2 THCP12
N
N
N
(GPO)5GPQGLAGQRGIR(GPO)5G-Pra-NH2
Fig. 7 Schematic of the click-based assembly of THCP12. Assembly was achieved with SSP10 and SSP11.
Used with permission from [48]
3.1.12 Synthesis General methods Subheadings 3.1.1 and 3.1.2 are followed, with
of Single-Stranded Peptide the following modifications.
11 (SSP11)
1. The synthesis of SSP11 (Fig. 7) is performed in 0.1 mmol scale
on TentaGel S Ram resin (0.26 mmol/g) by following the
procedure described for SSP1, except an extra Gly residue is
included after the Pra.
244 Gregg B. Fields
3.3 Enzyme Activation of all MMPs takes place in the TS buffer. See also Note 2.
Activation
3.5 Inhibitor Assays 1. Peptide substrate and inhibitor solutions are prepared using
TSB*Zn and left overnight at 4 C to allow the proper folding
of the peptide triple helix.
248 Gregg B. Fields
ð1Þ
4 Notes
Acknowledgments
References
residues that facilitate matrix metalloproteinase 53. Lauer-Fields JL, Broder T, Sritharan T,
Collagenolytic activity. J Biol Chem Nagase H, Fields GB (2001) Kinetic analysis
284:24017–24024 of matrix metalloproteinase triple-helicase
44. Bertini I, Fragai F, Luchinat C, Melikian M, activity using fluorogenic substrates. Biochem-
Toccafondi M, Lauer JL, Fields GB (2012) istry 40:5795–5803
Structural basis for matrix metalloproteinase 54. Del Valle JR, Goodman M (2003) Asymmetric
1 catalyzed Collagenolysis. J Am Chem Soc hydrogenations for the synthesis of Boc-Pro-
134:2100–2110 tected-4-Alkylprolinols and Prolines. J Org
45. Lauer-Fields JL, Whitehead JK, Li S, Hammer Chem 68:3923–3931
RP, Brew K, Fields GB (2008) Selective modu- 55. Neumann U, Kubota H, Frei K, Ganu V, Lep-
lation of matrix metalloproteinase 9 (MMP-9) pert D (2004) Characterization of Mca-Lys-
functions via exosite inhibition. J Biol Chem pro-Leu-Gly-Leu-Dpa-Ala-Arg-NH2, a fluoro-
283:20087–20095 genic substrate with increased specificity con-
46. Bhowmick M, Sappidi RR, Fields GB, Lepore stants for collagenases and tumor necrosis
SD (2011) Efficient synthesis of Fmoc- factor converting enzyme. Anal Biochem
protected Phosphinic Pseudodipeptides: build- 328:166–173
ing blocks for the synthesis of matrix metallo- 56. Bhowmick M, Fields GB (2013) Stabilization
proteinase inhibitors (MMPIs). Biopolymers of triple-helical peptides for in vivo applica-
(Pept Sci) 96:1–3 tions. Methods Mol Biol 1081:167–194
47. Bhowmick M, Fields GB (2012) Synthesis of 57. Fields GB, Tian Z, Barany G (1993) Principles
Fmoc-Gly-Ile Phosphinic Pseudodipeptide: and practice of solid-phase peptide synthesis.
residue specific conditions for construction of In: Grant GA (ed) synthetic peptides: a User’s
matrix metalloproteinase inhibitor building guide. W.H. Freeman & Co, New York, pp
blocks. Int J Pept Res Ther 18:335–339 77–183
48. Bhowmick M, Stawikowska R, Tokmina- 58. Fields CG, Lovdahl CM, Miles AJ, Matthias-
Roszyk D, Fields GB (2015) Matrix metallo- Hagen VL, Fields GB (1993) Solid-phase syn-
proteinase inhibition by Heterotrimeric triple- thesis and stability of triple-helical peptides
helical peptide transition state analogs. Chem- incorporating native collagen sequences. Bio-
biochem 16:1084–1092 polymers 33:1695–1707
49. Bhowmick M, Tokmina-Roszyk D, Onwuha- 59. Fields CG, Mickelson DJ, Drake SL, McCarthy
Ekpete L, Harmon K, Robichaud T, Fuerst R, JB, Fields GB (1993) Melanoma cell adhesion
Stawikowska R, Steffensen B, Roush WR, and spreading activities of a synthetic
Wong H, Fields GB (2017) Second generation 124-residue triple-helical “mini-collagen”. J
triple-helical peptide transition state analog Biol Chem 268:14153–14160
matrix metalloproteinase inhibitors. J Med 60. Grab B, Miles AJ, Furcht LT, Fields GB (1996)
Chem 60:3814–3827 Promotion of fibroblast adhesion by triple-
50. Lauer-Fields JL, Sritharan T, Stack MS, helical peptide models of type I collagen-derived
Nagase H, Fields GB (2003) Selective hydroly- sequences. J Biol Chem 271:12234–12240
sis of triple-helical substrates by matrix 61. Lauer-Fields JL, Tuzinski KA, Shimokawa K,
metalloproteinase-2 and -9. J Biol Chem Nagase H, Fields GB (2000) Hydrolysis of
278:18140–18145 triple-helical collagen peptide models by matrix
51. Minond D, Lauer-Fields JL, Cudic M, Overall metalloproteinases. J Biol Chem
CM, Pei D, Brew K, Visse R, Nagase H, Fields 275:13282–13290
GB (2006) The roles of substrate thermal sta- 62. Lauer-Fields JL, Nagase H, Fields GB (2000)
bility and P2 and P0 1 subsite identity on matrix Use of Edman degradation sequence analysis
metalloproteinase triple-helical peptidase activ- and matrix-assisted laser desorption/ionization
ity and collagen specificity. J Biol Chem mass spectrometry in designing substrates for
281:38302–38313 matrix metalloproteinases. J Chromatogr A
52. de Castro Brás LE, Cates CA, DeLeon-Pennell 890:117–125
KY, Ma Y, Iyer RP, Halade GV, 63. Malkar NB, Lauer-Fields JL, Borgia JA, Fields
Yabluchanskiy A, Fields GB, Weintraub ST, GB (2002) Modulation of triple-helical stability
Lindsey ML (2014) Citrate synthase is a novel and subsequent melanoma cellular responses by
in vivo matrix Metalloproteinase-9 substrate single-site substitution of fluoroproline deriva-
that regulates mitochondrial function in the tives. Biochemistry 41:6054–6064
post-myocardial infarction left ventricle. Anti- 64. Yu Y-C, Tirrell M, Fields GB (1998) Minimal
oxid Redox Signal 21:1974–1985 lipidation stabilizes protein-like molecular
architecture. J Am Chem Soc 120:9979–9987
252 Gregg B. Fields
65. Yu Y-C, Berndt P, Tirrell M, Fields GB (1996) inhibitors in drug discovery. John Wiley &
Self-assembling amphiphiles for construction Sons, Inc., Hoboken, NJ, pp 178–213
of protein molecular architecture. J Am Chem 68. Yiallouros I, Vassiliou S, Yiotakis A, Zwilling R,
Soc 118:12515–12520 Stöcker W (1998) Phosphinic peptides, the
66. Palmier MO, Van Doren SR (2007) Rapid first potent inhibitors of astacin, behave as
determination of enzyme kinetics from fluores- extremely slow-binding inhibitors. Biochem J
cence: overcoming the inner filter effect. Anal 331:375–379
Biochem 371:43–51 69. Guy CA, Fields GB (1997) Trifluoroacetic acid
67. Copeland RA (2005) Tight binding inhibition. cleavage and Deprotection of resin-bound pep-
In: Copeland RA (ed) Evaluation of enzyme tides following synthesis by Fmoc chemistry.
Methods Enzymol 289:67–83
INDEX
A E
Age-synchronizing .....................174, 176, 179, 182, 183 ELISA ..................................................191, 192, 194–199
Aging ...................................... v, 136, 146, 169–186, 209 Exon skipping.................................................................... 4
Amino propeptide of type III procollagen Extracellular matrix (ECM)....................................... v, 23,
(PIIINP) ............................................................ 190 57, 61, 63, 65, 71, 72, 115–126, 129–132, 135,
157–164, 169–171, 175, 182–184, 186, 189,
B 190, 204–206, 208, 211, 213, 221, 222, 230
Biomarkers................................................. 87–90, 93, 191
F
Biomedical properties ..................................................... 17
Biophysical characterization .................................. 23, 203 Freeze-cracking ............................................172, 176–177
Biopsy ..................................................145, 147, 153, 190
Bone...................................................................39, 40, 69, G
73, 95, 99, 143, 229 γ-glutamyl transferase (GGT)....................................... 190
Bone resorption............................................................. 135 Gibson assembly..................................................... 7, 8, 10
Glutamyl endoproteinase (GluC) .............. 118, 120, 122
C
Glutaraldehyde (GTA) ............................................ 22, 24,
Caenorhabditis elegans (C. elegans)..................... 169–186 27–29, 31, 33–35, 42, 48, 130
Calcium phosphate.......................................................... 40
Cancer-associated fibroblasts (CAFs)..........208–212, 221 H
Carboxyl propeptide of type I procollagen Herovici staining ......................................... 171, 175, 177
(PICP)................................................................ 190
Histopathology .................................................v, 135–144
Cell adhesion ............................................ v, 160–161, 205 Hybridization ....................................................... 135–144
Chondrocytes ...............................................158–160, 164 Hydroxyprolines............................ 14, 21, 60, 61, 63, 69,
Cloning ...........................................................3, 7, 10, 101 70, 82–84, 87, 88, 136, 170, 171, 177, 178, 181
Co-immunoprecipitations .......................... 102, 110, 111
Collagenase............................ 80, 83, 158, 159, 210, 223 I
Collagen fibers..................................................... 145, 150,
152, 153, 204, 221 Immature crosslinks ........................................... 80, 83–85
Collagen hybridizing peptide (CHP) ................. 135–143 Immunostaining............................................................ 137
Collagenopathies.........................................................3, 96 In situ..................................................... 40, 135–144, 158
Collagen proteostasis network ........................95, 96, 110 Integrins .........................v, 157–164, 205, 208, 211, 231
Collagen remodeling .................207, 208, 210, 212, 213 Interactomes....................................................v, 3, 95–112
Collagen VI ................................................................... 164 Isobaric Tag for Relative and Absolute
Collagen type I .........................18, 23, 24, 173, 192, 209 Quantitation (iTRAQ) .................... 112, 116, 117
Colons................................................................... 129–132
L
Cross-linking ........................................................ v, 23–26,
29, 31, 33–35, 221 Liver fibrosis ............................................. v, 189–200, 209
Cryogenic TEM (cryoTEM) ................ 40, 44–48, 50, 52 Lung fibrosis.................................................................. 204
Cuticles ....................................... 171, 175, 182–184, 186 Lung metastases ............................................................ 204
Cytotoxicity ..................................................................... 24 Lysyl oxidase (LOX) ......................................23, 204, 209
like-1 .......................................................................... 79
D like-2 ........................................................................ 205
Decellularization .................................................. 129–132 like-3 .......................................................................... 79
Dystrophic epidermolysis bullosa (DEB) ........................ 4 like-4 .......................................................................... 79
Irit Sagi and Nikolaos A. Afratis (eds.), Collagen: Methods and Protocols, Methods in Molecular Biology, vol. 1944,
https://doi.org/10.1007/978-1-4939-9095-5, © Springer Science+Business Media, LLC, part of Springer Nature 2019
253
COLLAGEN
254 Index
M Skin .................................................................. v, 4, 18, 20,
21, 34, 70, 80, 83–85, 91, 95, 145–154, 170, 171,
Macrophages .............................. 24, 29, 31, 33, 203–213 173, 209, 224, 229
Mass spectrometry ...............................................v, 57, 61, Solid phase extraction (SPE) ................80, 81, 84, 85, 92
79–93, 95–112, 115, 117, 122, 123, 236, 244 Stochastic optical reconstruction microscopy
Matrix metalloproteinases (MMPs) ........................ v, 136, (STORM) .........................................40, 48–51, 53
190, 206, 210, 231–234, 245
Matrix superstructures ...................................................... 4 T
Mature crosslinks ............................................... 80, 83–85
Multiphoton microscopy ..................................v, 145–154 Tandem mass tag (TMT).............................112, 115–126
Terminal amine isotopic labeling of substrates
P (TAILS)..................................................... 115–126
Tissue ................................................ v, 11, 19–21, 23, 24,
Peptide-based drugs...................................................... 231 29, 31, 33, 39, 57, 60, 61, 63, 65, 66, 71, 73,
Procollagen C-proteinase enhancer-1 79–93, 95, 129, 135–137, 139–142, 145,
(PCPE-1) .................................................. 189–200 151–153, 169–186, 189, 190, 204, 206, 208,
Proteomics.................................v, 95–112, 122, 207, 213 210–212, 222, 225, 229, 230, 234
Transition state analog......................................... 231, 232
R
Transmission electron microscopy (TEM) ................... 40,
Radio frequency (RF) ...........................58, 68, 71, 85, 86 42–47, 51, 52, 161–164
Triple-helical inhibitor ......................................... 229–249
S Tumor-associates macrophages (TAMs)............. 208–213
Tumor microenvironment ....................... v, 203–213, 221
Scaffolds......................................................... v, 23–35, 95,
130, 203–205, 210, 213
U
Scanning electron microscope (SEM) ................ 129–132
Second harmonic generation (SHG) ..........................135, Ultra-high performance liquid chromatography-triple
145, 146, 148–153 quadrupole mass spectrometry
Single reaction monitoring (SRM) ..........................85, 86 (UHPLC-MS/MS) ................... 80, 82–83, 85–90