Nutrition and Chronic Conditions
Nutrition and Chronic Conditions
Nutrition and Chronic Conditions
Nutrition and
Chronic Conditions
Edited by
Omorogieva Ojo
Printed Edition of the Special Issue Published in Nutrients
www.mdpi.com/journal/nutrients
Nutrition and Chronic Conditions
Nutrition and Chronic Conditions
Editorial Office
MDPI
St. Alban-Anlage 66
4052 Basel, Switzerland
This is a reprint of articles from the Special Issue published online in the open access journal Nutrients
(ISSN 2072-6643) in 2018 (available at: https://www.mdpi.com/journal/nutrients/special issues/
nutrition and chronic conditions)
For citation purposes, cite each article independently as indicated on the article page online and as
indicated below:
LastName, A.A.; LastName, B.B.; LastName, C.C. Article Title. Journal Name Year, Article Number,
Page Range.
c 2019 by the authors. Articles in this book are Open Access and distributed under the Creative
Commons Attribution (CC BY) license, which allows users to download, copy and build upon
published articles, as long as the author and publisher are properly credited, which ensures maximum
dissemination and a wider impact of our publications.
The book as a whole is distributed by MDPI under the terms and conditions of the Creative Commons
license CC BY-NC-ND.
Contents
Omorogieva Ojo, Osarhumwese Osaretin Ojo, Fajemisin Adebowale and Xiao-Hua Wang
The Effect of Dietary Glycaemic Index on Glycaemia in Patients with Type 2 Diabetes:
A Systematic Review and Meta-Analysis of Randomized Controlled Trials
Reprinted from: Nutrients 2018, 10, 373, doi:10.3390/nu10030373 . . . . . . . . . . . . . . . . . . . 1
Hanan A. Alfawaz, Kaiser Wani, Abdullah M. Alnaami, Yousef Al-Saleh, Naji J. Aljohani,
Omar S. Al-Attas, Majed S. Alokail, Sudhesh Kumar and Nasser M. Al-Daghri
Effects of Different Dietary and Lifestyle Modification Therapies on Metabolic Syndrome
in Prediabetic Arab Patients: A 12-Month Longitudinal Study
Reprinted from: Nutrients 2018, 10, 383, doi:10.3390/nu10030383 . . . . . . . . . . . . . . . . . . . 16
Li-Li Wang, Qi Wang, Yong Hong, Omorogieva Ojo, Qing Jiang, Yun-Ying Hou,
Yu-Hua Huang and Xiao-Hua Wang
The Effect of Low-Carbohydrate Diet on Glycemic Control in Patients with Type 2
Diabetes Mellitus
Reprinted from: Nutrients 2018, 10, 661, doi:10.3390/nu10060661 . . . . . . . . . . . . . . . . . . . 63
Renée Wilson, Jinny Willis, Richard B. Gearry, Alan Hughes, Blair Lawley, Paula Skidmore,
Chris Frampton, Elizabeth Fleming, Angie Anderson, Lizzie Jones, Gerald W. Tannock and
Anitra C. Carr
SunGold Kiwifruit Supplementation of Individuals with Prediabetes Alters Gut Microbiota and
Improves Vitamin C Status, Anthropometric and Clinical Markers
Reprinted from: Nutrients 2018, 10, 895, doi:10.3390/nu10070895 . . . . . . . . . . . . . . . . . . . 76
v
Magali Leyvraz, Carmelle Mizéhoun-Adissoda, Dismand Houinato, Naby Moussa Baldé,
Albertino Damasceno, Bharathi Viswanathan, Mary Amyunzu-Nyamongo, Jared Owuor,
Arnaud Chiolero and Pascal Bovet
Food Consumption, Knowledge, Attitudes, and Practices Related to Salt in Urban Areas in Five
Sub-Saharan African Countries
Reprinted from: Nutrients 2018, 10, 1028, doi:10.3390/nu10081028 . . . . . . . . . . . . . . . . . . 105
vi
About the Special Issue Editor
Omorogieva Ojo has a Ph.D. in nutrition from the University of Greenwich, London, a post-graduate
diploma in diabetes from the University of Surrey, Roehampton and a graduate certificate in Higher
Education from the University of Greenwich. Prior to these qualifications, Dr. Ojo was awarded
his BSc and MSc in animal science from the University of Ibadan, Nigeria. He is currently a Senior
Lecturer in Diabetes Care and Management in the Faculty of Education and Health, the University
of Greenwich and he teaches across a range of courses and programmes. He was previously a
nutrition specialist at the Home Enteral Nutrition Team, Lewisham Primary Care Trust, London, a
post-doctoral research fellow in the School of Science, University of Greenwich, London, and taught
at the College of Agriculture, Asaba, Nigeria. He is an internationally acclaimed expert in nutrition
and diabetes and these are the primary focus of his teaching and research activities, including Ph.D.
supervision. Dr. Ojo has been a keynote speaker at international conferences and he is on the Editorial
Board of many international journals including Nutrients.
vii
Preface to ”Nutrition and Chronic Conditions”
This book on Nutrition and Chronic Conditions aims to provide insight into the effect of nutrition
in the development, care, and management of chronic conditions. This is in recognition of the impact
that nutrition has on chronic conditions, such as diabetes, cardiovascular disease, dementia, stroke,
and inflammatory bowel disease, which continues to generate interest among researchers. There is
evidence that diet is a modifiable risk factor for these diseases, which manifest either as single entities
or in co-morbid states in individuals and populations around the world. In particular, the prevalence
of diabetes and cardiovascular disease is on the increase, especially in developed countries, but also
in developing economies, partly due to lifestyle changes, including diet. For example, ischaemic
heart disease is the leading cause of death globally. When combined with stroke, these conditions
accounted for 15 million deaths in 2015 and are the world’s greatest killers. In addition, there were an
estimated 422 million adults who were living with diabetes in 2014 compared to 108 million in 1980.
These chronic conditions and their associated complications have significant implications
for morbidity and mortality, and incur huge costs to the health services around the world.
The composition of the diet, the proportion and types of macronutrients and micronutrients present
in the diet are major contributors to these diseases. In addition, the beneficial effects of nutritional
interventions have been well documented although differences remain among researchers with
respect to their overall impact. The evaluation of the role of nutrition in chronic conditions draws
on its effect on body weight and body composition, glycaemic and insulin excursions, vascular
remodeling, and gastro-intestinal dysfunction.
Internationally acclaimed experts in the field of nutrition and chronic conditions have
contributed chapters to this book that should provide the evidence base for practice and research.
Therefore, this book is aimed at patients, students, and healthcare professionals, including nurses,
doctors, public health practitioners, and dietitians/nutritionists. The book has sixteen chapters
covering original research and reviews, which should guide healthcare professionals in their areas
of practice. These include evaluations of the effects of various nutritional interventions, dietary
and lifestyle modifications on cognitive decline, anthropometric parameters, metabolic syndrome,
diabetes, and glycaemic control. In addition, assessment of food consumption, knowledge, attitudes,
and practices related to salt, dairy intake and acne vulgaris, food perceptions, and dietary changes in
chronic conditions are the key topics of interest in this book.
Omorogieva Ojo
Special Issue Editor
ix
nutrients
Review
The Effect of Dietary Glycaemic Index on Glycaemia
in Patients with Type 2 Diabetes: A Systematic
Review and Meta-Analysis of Randomized
Controlled Trials
Omorogieva Ojo 1, *, Osarhumwese Osaretin Ojo 2 , Fajemisin Adebowale 3 and Xiao-Hua Wang 4
1 Department of Adult Nursing and Paramedic Science, University of Greenwich, London SE9 2UG, UK
2 Healthcare, Care UK, HMP Wormwood Scrubs, London W12 0AE, UK; [email protected]
3 Department of Animal Production and Health, Federal University of Technology, PMB, Akure 704,
Ondo State, Nigeria; [email protected]
4 The School of Nursing, Soochow University, Suzhou 215006, China; [email protected]
* Correspondence: [email protected]; Tel.: +44-020-8331-8626; Fax: +44-020-8331-8060
Abstract: Background: The increasing prevalence of diabetes in the United Kingdom and worldwide
calls for new approaches to its management, and diets with low glycaemic index have been proposed
as a useful means for managing glucose response. However, there are conflicting reports and
differences in the results of studies in terms of their effectiveness. Furthermore, the impact of
low-glycaemic index diets and their long-term use in patients with type 2 diabetes remains unclear.
Objectives: The objective of this study was to conduct a systematic review and meta-analysis of
the effect of low-glycaemic index diets in patients with type 2 diabetes. Methods: Search methods:
Randomised controlled studies were selected from a number of databases (EBSCOHost with links
to Health Research databases, PubMed, and grey literature) based on the Population, Intervention,
Comparator, Outcomes and Study designs (PICOS) framework. The search terms included synonyms
and Medical Subject Headings (MeSH) and involved the use of Boolean operators (AND/OR) which
allowed the combination of words and search terms. Selection criteria: As per the selection criteria,
the following types of articles were selected: studies on randomised controlled trials, with year
of publication between 2008 and 2018, including patients with type 2 diabetes. Thus, studies
involving patients with gestational and type 1 diabetes were excluded, as were observational studies.
Nine articles which met the inclusion criteria were selected for the systematic review, whereas
only six articles which met the criteria were included in the meta-analysis. Data collection and
analysis: Studies were evaluated for quality and risk of bias. In addition, heterogeneity, meta-analysis,
and sensitivity tests of the extracted data were carried out using Review Manager 5.3 (Review
Manager, 2014). Results: The findings of the systematic review showed that the low-glycaemic
index (low-GI) diet resulted in a significant improvement (<0.05) in glycated haemoglobin (HbA1c)
in two studies: low-GI diet Δ = −0.5% (95% CI, −0.61% to −0.39%) vs. high-cereal fibre diet
Δ = −0.18% (95% CI, −0.29% to −0.07%); and low-GI legume diet Δ = −0.5% (95%, −0.6%
to −0.4%) vs. high-wheat fibre diet Δ = −0.3% (95% Cl, −0.4 to −0.2%). There was a slight
improvement in one study (low glycaemic response = 6.5% (6.3–7.1) vs. control = 6.6% (6.3–7.0) and no
significant difference (p > 0.05) in four studies compared with the control diet. Four studies showed
improvements in fasting blood glucose in low-GI diets compared to higher-GI diets or control: low-GI
diet = 150.8 ± 8.7 vs. higher-GI diet = 157.8 ± 10.4 mg/dL, mean ± SD p = 0.43; low-GI diet = 127.7
vs. high-cereal fibre diet = 136.8 mg/dL, p = 0.02; low-GI diet = 6.5 (5.6–8.4) vs. standard diabetic
diet = 6.7 (6.1–7.5) mmol/L, median and interquartile range p > 0.05; and low-GI diet = 7.3 ± 0.3 vs.
conventional carbohydrate exchange diet = 7.7 ± 0.4 mmol/L, mean ± SEM (Standard Error of Mean)
p < 0.05. The results of the meta-analysis and sensitivity tests demonstrated significant differences
(p < 0.001 and p < 0.001, respectively) between the low-GI diet and the higher-GI diet or control
diet in relation to glycated haemoglobin. Differences between the low-GI diet and higher-GI diet or
control were significant (p < 0.05) with respect to the fasting blood glucose following meta-analysis.
Conclusion: The low-GI diet is more effective in controlling glycated haemoglobin and fasting blood
glucose compared with a higher-GI diet or control in patients with type 2 diabetes.
Keywords: glycaemic index; glycated haemoglobin; fasting blood glucose; type 2 diabetes;
randomised controlled trials; meta-analysis; systematic review
1. Introduction
The increasing prevalence of diabetes and its impact on morbidity and mortality have become
global problems [1,2]. About 422 million adults worldwide were reported to live with diabetes in
2016, and the global prevalence rose from 4.7% in 1980 to 8.5% in 2014 [1]. In the United Kingdom,
the prevalence of type 2 diabetes more than doubled from 2.39% in the year 2000 to 5.32% in 2013 [2].
The management of type 2 diabetes and its related complications, including retinopathy, kidney
dysfunction, neuropathy, and foot problems accounts for about 10% of the entire National Health
Service (NHS) budget in the UK [2]. Presently, 11% of U.S. adult population has diabetes and the
total estimated costs associated with the condition in 2012 were US $245 billion due to direct medical
costs and reduced worker productivity [3]. Several factors, including genetic predisposition and
environmental factors, have been implicated in the aetiology of diabetes [3,4]. This is particularly true
in the case of type 2 diabetes, which accounts for over 90% of all forms of diabetes and where lifestyle
has a profound effect on its manifestation [5]. Usually, lifestyle factors such as diet and physical
activities can be modified in terms of the choices that individuals make. The composition of diet with
respect to the quality of the nutrients including carbohydrates, protein, fats, minerals and vitamins is
important in determining nutritive value and usefulness in human health [6].
2
Nutrients 2018, 10, 373
(HbA1C) provides a measure of the average glycaemia over the preceding 3 months, the fasting blood
glucose is a measure of blood glucose level following at least 8 h of fasting, and is usually taken before
breakfast [17].
1.4. Objectives
This is a systematic review and meta-analysis which evaluates the effect of the low-glycaemic
index diet in patients with type 2 diabetes
Research question: Is a low-GI diet effective in improving glycaemia in patients with type 2
diabetes compared with a higher-GI diet?
2. Methods
3
Nutrients 2018, 10, 373
Combining
Patient/Population Intervention Comparator Study Designs
Search Terms
Low-glycaemic Higher-glycaemic Randomised
Patients with diabetes
index diet index diet or control controlled trial
#1 Randomised
controlled trial OR
Patients with diabetes Glycaemic index OR
controlled clinical trial
OR type 2 diabetes OR glycemic index OR
OR randomized OR
diabetes OR diabetes glycaemic load OR Column 1 and
placebo OR drug
complications OR glycaemic indices or Column 2 and
therapy OR randomly
diabetes mellitus, glycaemic index Column 3
OR trial OR groups
type 2 OR number or glycaemic
#2 “Animals”
diabetes mellitus index numbers
NOT “Humans”
#3 #1 NOT #2
4
Nutrients 2018, 10, 373
In addition, only studies involving adults with type 2 diabetes and the use of the dietary glycaemic
index were included. Studies written in English from across the world have been included as diabetes
is a worldwide problem.
Identification
(n =2347) (n = 3543)
N = 7 (Studies on glycaemic
load)
15
N = 6 (Studies involving
carbohydrate diets)
Studies included in systematic
review
(N = 9)
Included
presentation)
Studies included in Meta-analysis
N = 2 (Studies not providing
(N = 6)
data before intervention)
Therefore, other studies involving patients with type 1 diabetes or gestational diabetes and animal
studies were excluded from this review (Table 2). Similarly, studies involving children with diabetes or
healthy adults without diabetes were also excluded. Studies which were not randomised and those
involving dietary supplements have been excluded from this review.
5
Nutrients 2018, 10, 373
Table 2. Criteria for considering studies for the review based on the Population, Intervention,
Comparator, Outcomes and Study designs (PICOS) structure.
6
Nutrients 2018, 10, 373
outcome data (attrition bias), selective reporting (reporting bias), and other bias [27]. The risk of
bias was assessed by Review Manager 5.3 software [24].
3. Results
With respect to the systematic review, only nine articles [28–36] met the criteria for inclusion
(Table 3). However, six of these studies [28–31,35,36] were used for the meta-analysis to test the effect
of low-GI diet on glycated haemoglobin and fasting blood glucose in patients with type 2 diabetes.
The others were excluded as they did not meet the criteria for meta-analysis such as reporting their
results in the form of median and interquartile or not providing data before intervention.
The length of study ranged from 2 weeks to 22 months. In terms of the interventions,
these involved comparing the low-GI diet with the higher-GI diet or control (conventional carbohydrate
exchange, high-cereal fibre diet, high-wheat fibre diet, standard diabetic diet, American diabetes
association diet) (Table 3).
In most of the studies [28,30,34–36] that reported dietary glycaemic index values, the low-GI
diet had significantly (p < 0.05) lower values than the higher-GI diet or the control. The study by
Jenkins et al. [29] also showed that the low-GI diet resulted in lower GI values than the high-cereal
fibre diet, although the level of statistical difference was not stated (Table 3). However, differences in
dietary GI values were not significant (p > 0.05) in one study [31], while data were not available in two
of the studies [32,33] although the authors stated that the intervention diets involved low glycaemic
index or low glycaemic response.
In studies reporting on the effect of dietary GI on the HbA1c levels, diets with low GI were shown
to result in a significant improvement (p < 0.05) in HbA1c levels in two studies [29,30] compared with
the higher-GI diet or the control. One study [33] reported slight improvement in HbA1c in the low-GI
diet group compared with control, while differences between low-GI diet and higher-GI diet or control
were not significantly different in four studies [31,34–36].
The effect of low-GI diets on fasting blood glucose compared to higher-GI diets or control diets
was evident in seven of the studies selected [28–30,32–34,36]. While four studies [28,29,34,36] showed a
greater improvement in fasting blood glucose in the low-GI diet compared with higher-GI diet, some of
the differences were not statistically significant. Furthermore, there was a lower fasting blood glucose
level in the higher-GI diet or control compared with low-GI diet in two studies [30,33]. The fasting
blood glucose levels were not significantly different in the low-GI diet compared with control in one
other study [32].
7
Table 3. Summary of studies included in the systematic review.
8
(127–141) (95% CI) (77–80) (95% CI)
Parallel 58 (1.3) diet = 9.2 (8.0) Low-GI legume diet vs. −0.4%) vs. high-wheat
Jenkins et al. [30] 3 months 121 End of study End of study
Design High-wheat fibre High-wheat fibre high-wheat fibre diet fibre diet Δ = −0.3% (95%
Low-GI legume diet = Low-GI legume diet = 66
diet = 61 (1.0) diet = 8.6 (0.8) Cl, −0.4 to −0.2%)
132(126–138) (95% CI) vs. (64–67) (95% CI) vs.
p < 0.001
high-wheat fibre diet = 127 high-wheat fibre diet = 82
(121–133) (95% CI) (81–83) (95% CI)
p = 0.001 p < 0.001
## Baseline ## Baseline
Low-GI diet = Low-GI diet =
8.74 ± 0.29% vs. baseline 79.35 ± 1.36 vs. ADA diet
Low-GI diet vs. American ADA diet = 8.1 ± 0.28% = 82.03 ± 1.31
12 Parallel
Ma et al. [31] 40 # 53.53 ± 8.40 # 9.32 ± 9.66 Diabetes Association diet 12 months No data 12 months
months Design
(ADA) Low-GI diet = Low-GI diet =
8.39 ± 0.30% vs. 12-month 76.64 ± 1.46 vs. ADA diet
ADA diet = 7.67 ± 0.28% = 80.36 ± 1.40
p = 0.08 p = 0.07
Table 3. Cont.
9
Wolever et al. 12 Parallel Low-GI diet vs. higher-GI = 61.5 ± 0.4 Study Low-GI
162 Higher-GI diet = No data Low-GI diet = No data
[35] months Design diet diet = 55.1 ± 0.4
60.4 ± 1.1 6.34 ± 0.05%
Higher-GI diet = 63.2 ±
Higher-GI diet =
0.4 p < 0.001
6.34 ± 0.05%
p > 0.05
## Baseline
**## Baseline
Low-GI diet =
Low-GI diet = 7.33 ± 2.23
7.68 ± 1.13% # Week 12 Low-GI diet =
Low-GI diet vs. CCE = 7.01 ± 1.79
Parallel CCE = 7.51 ± 1.24% 57 ± 6
Yusof et al. [36] 12 weeks 100 53.5 No data conventional carbohydrate Week 12
Design Week 12 Week 12 CCE = 64 ± 5
exchange (CCE) Low-GI diet = 7.3 ± 0.3
Low-GI diet = 7.2 ± 0.1% p < 0.001
CCE = 7.7 ± 0.4
CCE = 7.2 ± 0.2%
p < 0.05
p > 0.05
Abbreviations: ADA (American Diabetes Association); CCE (conventional carbohydrate exchange); FGB (fasting blood glucose); glycated haemoglobin (HbA1c); GI (glycaemic index);
GR (glycaemic response); low-GR (low glycaemic response); Δ (change); * (FBG, mg/dL); ** FBG (mmol/L); # (Mean ± SD); ## (mean ± SEM); ### (Median) (25th–75th percentile);
#### (Mean and 95% CI, confidence interval).
Nutrients 2018, 10, 373
10
Nutrients 2018, 10, 373
Figure 4. A forest plot showing the effect of low-GI diet on glycated haemoglobin (%).
Figure 5. A forest plot showing the effect of low-GI diet on fasting blood glucose (mg/dL).
11
Nutrients 2018, 10, 373
Regarding the heterogeneity test, the high p value (p = 0.36) and the chi-squared statistic (2.04)
relative to the degree of freedom (2) would suggest that there was low heterogeneity between the
studies. This was confirmed by the results of the I2 test. The heterogeneity test for fasting blood glucose
showed I2 = 2% for meta-analysis and I2 = 45% for the sensitivity test, indicating low heterogeneity of
the studies included.
4. Discussion
Based on the findings of the systematic review, the low-GI diet resulted in greater improvement
in glycated haemoglobin [29,30] and fasting blood glucose [28,29,34,36] compared to higher-GI diet
or control in patients with type 2 diabetes. In the study by Yusof et al. [36], although the effect on
HbA1c was not significantly different between the low-GI diet and the higher-GI diet or control,
the improvement within the low-GI group was more pronounced and of clinical benefit. However,
compared with the American Diabetes Association (ADA) diet, the low-GI diet achieved equivalent
control of HbA1c using less medication [31] and for patients on diet alone with optimal glycaemic
control, long-term HbA1c was not affected by altering the GI [35]. The findings of this systematic
review confirm the results of earlier systematic review by Thomas and Elliot [8] which suggests that
lowering the glycaemic index of food may improve glycated haemoglobin in patients with diabetes.
However, the difference between this review and the Thomas and Elliot [8] study is with respect to
the second outcome measure. While fasting glucose was the outcome of interest in this review, it was
fructosamine in the review by Thomas and Elliot [8]. Regarding the risk of bias, 100% of the studies
showed low risk of selection bias while all the studies demonstrated 100% low risk of bias or unclear
risk of bias in relation to blinding, incomplete outcome data, selective reporting, and other potential
sources of bias except for one study that showed a high risk of bias with respect to the blinding of
participants and personnel.
There was no statistically significant difference between the low-GI and higher-GI diets in relation
to HbA1c and fasting blood glucose in the study by Visek et al. [34]. Acute glycaemic control was
variable over the 3-day period in the study by Gonçalves Reis and Dullius [32] following the adoption
of low-GI diet. While the first day of the study demonstrated a significant difference in acute glycaemic
control, differences were not significant on the second or third day [32]. Stenvers et al. [33] found
no beneficial effect of the low-glycaemic response liquid meal with respect to fasting blood glucose.
The differences observed between these studies in terms of their results may be due to the limitations of
using HbA1c in identifying the daily changes in glycaemia [17]. The use of HbA1c may not detect the
harmful effects of excessive postprandial hyperglycaemic excursions and the risk of hypoglycaemia [7].
According to Chiu and Taylor [7], the contribution of postprandial glucose may be up to 70% in
daily hyperglycaemia, and the contribution of fasting glucose increases as glycaemic control worsens.
This may explain why the association between dietary GI and HbA1c is not consistent across studies [7].
Therefore, the use of a meta-analysis of both fasting blood glucose and HbA1c levels to evaluate the
effect of dietary GI in patients with diabetes is a useful strategy of eliminating the limitation of using
HbA1c to assess GI exposure. Other factors to consider include the nature of the starch, particle size,
pH, the amount of fibre, fat, and protein, and the cooking method and time, which may affect the GI
of food and its effect on blood glucose response, and lead to differences in outcomes of studies [15].
The differences in the classification of low- and high-GI diets may also have effect on outcomes of
studies [37].
The Food and Agricultural Organisation [9] has recommended the use of the glycaemic index
of foods in clinical applications in patients with diabetes, and that the glycaemic index be used as
a useful indicator of the impact of food on the blood glucose response. Recently, the American
Diabetes Association (ADA) [38] recommended that patients with diabetes consume carbohydrates
from vegetables, fruits, legumes, whole grains, and dairy products. The ADA [38] also recommended
that emphasis should be placed on foods which are higher in fibre and lower in glycaemic load as
opposed to other sources, especially those with added sugar. The glycaemic load is the product of
12
Nutrients 2018, 10, 373
the glycaemic index of food or diet and the grams of available carbohydrate in that food or diet
divided by 100 [9,15]. These recommendations are in line with the findings of the current systematic
review and meta-analysis, which showed that low-GI diets were more effective in controlling glycated
haemoglobin than higher-GI diets. The position of the ADA [38] with respect to the mixed results of
glycaemic index or glycaemic load in patients with diabetes is also true in relation to the findings of
the studies included in this review. Foods with low GI have demonstrated beneficial effects on glucose
control in short-term trials in patients with type 2 diabetes [13]. However, the higher intake of sucrose
or fructose and the long-term use of high-GI diets can place higher metabolic demands on the body in
relation to higher insulin requirements [6,39,40].
6. Conclusions
The findings of this systematic review and meta-analysis have shown that low-GI diets are more
effective in controlling HbA1c and fasting blood glucose compared with higher-GI diets or control
diets in patients with type 2 diabetes. Although the outcomes of the individual studies were sometimes
different with respect to the variables of interest, the results of the meta-analysis and sensitivity tests
have demonstrated significant differences (p < 0.001 and p < 0.001, respectively) between low-GI diets
and higher-GI diets or control diets in relation to glycated haemoglobin. In addition, differences
between the low-GI diet and the higher-GI diet or control were significant (p < 0.05) with respect to the
fasting blood glucose following meta-analysis.
Author Contributions: The concept for this article was agreed upon by O.O., O.O.O., F.A. and X.-H.W. O.O.
O.O.O., F.A., and X.-H.W. participated in the searches and in extracting data from the articles reviewed. O.O.
and X.-H.W. carried out the data analysis. O.O. wrote the initial draft, which was reviewed and revised by O.O.,
O.O.O., F.A., and X.-H.W.
Conflicts of Interest: The authors declare no conflict of interest.
References
1. Zghebi, S.S.; Steinke, D.T.; Carr, M.J.; Rutter, M.K.; Emsley, R.A.; Ashcroft, D.M. Examining trends in type 2
diabetes incidence, prevalence and mortality in the UK between 2004 and 2014. Diabetes Obes. Metab. 2017,
19, 1537–1545. [CrossRef] [PubMed]
2. Sharma, M.; Nazareth, I.; Petersen, I. Trends in incidence, prevalence and prescribing in type 2 diabetes
mellitus between 2000 and 2013 in primary care: A retrospective cohort study. BMJ Open 2016, 6, e010210.
[CrossRef] [PubMed]
3. Haynes-Maslow, L.; Leone, L.A. Examining the relationship between the food environment and adult
diabetes prevalence by county economic and racial composition: An ecological study. BMC Public Health
2017, 17, 648. [CrossRef] [PubMed]
4. Hill, J. Management of diabetes in South Asian communities in the UK. Prim. Health Care 2007, 17, 49–55.
[CrossRef]
5. National Collaborating Centre for Chronic Conditions (NCCCC). Type 2 Diabetes: National Clinical Guideline
for Management in Primary and Secondary Care (Update); Royal College of Physicians: London, UK, 2008.
13
Nutrients 2018, 10, 373
6. Russell, W.R.; Baka, A.; Björck, I.; Delzenne, N.; Gao, D.; Griffiths, H.R.; Weickert, M.O. Impact of Diet
Composition on Blood Glucose Regulation. Crit. Rev. Food Sci. Nutr. 2016, 56, 541–590. [CrossRef] [PubMed]
7. Chiu, C.; Taylor, A. Dietary hyperglycemia, glycemic index and metabolic retinal diseases. Prog. Retin.
Eye Res. 2011, 30, 18–53. [CrossRef] [PubMed]
8. Thomas, D.E.; Elliott, E.J. The use of low-glycaemic index diets in diabetes control. Br. J. Nutr. 2010, 104,
797–802. [CrossRef] [PubMed]
9. Food and Agricultural Organisation (FAO). Carbohydrates in Human Nutrition. Report of a Joint FAO/WHO
Expert Consultation; FAO (Food and Nutrition paper–66); FAO: Rome, Italy, 1998. Available online: http:
//www.fao.org/docrep/w8079e/w8079e00.htm (accessed on 16 January 2018).
10. Chang, K.T.; Lampe, J.W.; Schwarz, Y.; Breymeyer, K.L.; Noar, K.A.; Song, X.; Neuhouser, M.L. Low Glycemic
Load Experimental Diet More Satiating Than High Glycemic Load Diet. Nutr. Cancer 2012, 64, 666–673.
[CrossRef] [PubMed]
11. Mohan, V.; Anjana, R.M.; Gayathri, R.; Ramya Bai, M.; Lakshmipriya, N.; Ruchi, V.; Sudha, V. Glycemic
Index of a Novel High-Fiber White Rice Variety Developed in India—A Randomized Control Trial Study.
Diabetes Technol. Ther. 2016, 18, 164–170. [CrossRef] [PubMed]
12. Similä, M.E.; Valsta, L.M.; Kontto, J.P.; Albanes, D.; Virtamo, J. Low-, medium- and high-glycaemic index
carbohydrates and risk of type 2 diabetes in men. Br. J. Nutr. 2011, 105, 1258–1264. [CrossRef] [PubMed]
13. Jung, C.; Choi, K.M. Impact of High-Carbohydrate Diet on Metabolic Parameters in Patients with Type 2
Diabetes. Nutrients 2017, 9. [CrossRef] [PubMed]
14. Ikpotokin, O.S.; Adeleye, O.S.; Aliu, E.D.; Osayande, A.B. Ehiabhi, S. Dietary factors in fasting blood glucose
levels and weight gain in female Sprague Dawley in rats. J. Clin. Nutr. Diet. 2017, 3, 2. [CrossRef]
15. Esfahani, A.; Wong, J.W.; Mirrahimi, A.; Villa, C.R.; Kendall, C.C. The application of the glycemic index
and glycemic load in weight loss: A review of the clinical evidence. IUBMB Life 2011, 63, 7–13. [CrossRef]
[PubMed]
16. Fabricatore, A.N.; Ebbeling, C.B.; Wadden, T.A.; Ludwig, D.S. Continuous glucose monitoring to assess
the ecologic validity of dietary glycemic index and glycemic load. Am. J. Clin. Nutr. 2011, 94, 1519–1524.
[CrossRef] [PubMed]
17. Dunning, T. Care of People with Diabetes a Manual of Nursing Practice, 4th ed.; Wiley Blackwell: Hoboken, NJ,
USA, 2014.
18. Villegas, R.; Liu, S.; Gao, Y.; Yang, G.; Li, H.; Zheng, W.; Shu, X.O. Prospective study of dietary carbohydrates,
glycemic index, glycemic load, and incidence of type 2 diabetes mellitus in middle-aged Chinese women.
Arch. Intern. Med. 2007, 167, 2310–2316. [CrossRef] [PubMed]
19. Krishnan, S.; Rosenberg, L.; Singer, M.; Hu, F.B.; Djoussé, L.; Cupples, L.A.; Palmer, J.R. Glycemic index,
glycemic load, and cereal fiber intake and risk of type 2 diabetes in US black women. Arch. Intern. Med. 2007,
167, 2304–2309. [CrossRef] [PubMed]
20. Ludwig, D.S. The glycemic index: Physiological mechanisms relating to obesity, diabetes, and cardiovascular
disease. JAMA 2002, 287, 2414–2423. [CrossRef] [PubMed]
21. Sacks, F.M.; Carey, V.J.; Anderson, C.M.; Miller, E.; Copeland, T.; Charleston, J.; Appel, L.J. Effects of high vs
low glycemic index of dietary carbohydrate on cardiovascular disease risk factors and insulin sensitivity:
The OmniCarb randomized clinical trial. JAMA 2014, 312, 2531–2541. [CrossRef] [PubMed]
22. Greenwood, D.C.; Threapleton, D.E.; Evans, C.L.; Cleghorn, C.L.; Nykjaer, C.; Woodhead, C.; Burley, V.J.
Glycemic Index, Glycemic Load, Carbohydrates, and Type 2 Diabetes. Diabetes Care 2013, 36, 4166–4171.
[CrossRef] [PubMed]
23. Fabricatore, A.N.; Wadden, T.A.; Ebbeling, C.B.; Thomas, J.G.; Stallings, V.A.; Schwartz, S.; Ludwig, D.S.
Targeting dietary fat or glycemic load in the treatment of obesity and type 2 diabetes: A randomized
controlled trial. Diabetes Res. Clin. Pract. 2011, 92, 37–45. [CrossRef] [PubMed]
24. The Nordic Cochrane Centre. Review Manager (RevMan) [Computer Program]; Version 5.3.; The Nordic
Cochrane Centre, The Cochrane Collaboration: Copenhagen, Denmark, 2014.
25. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G.; The PRISMA Group. Preferred Reporting Items for
Systematic Reviews and Meta-Analyses: The PRISMA Statement. Ann. Intern. Med. 2009, 151, 264–269.
[CrossRef] [PubMed]
26. Critical Appraisal Skills Programme (CASP). Randomised Controlled Trial Checklist. 2017. Available online:
http://docs.wixstatic.com/ugd/dded87_4239299b39f647ca9961f30510f52920.pdf (accessed on 18 January 2018).
14
Nutrients 2018, 10, 373
27. Higgins, J.P.T.; Green, S. Cochrane Handbook for Systematic Reviews of Interventions; Wiley-Blackwell: Hoboken,
NJ, USA, 2009.
28. Gomes, J.G.; Fabrini, S.P.; Alfenas, R.G. Low glycemic index diet reduces body fat and attenuates
inflammatory and metabolic responses in patients with type 2 diabetes. Arch. Endocrinol. Metab. 2017, 61,
137–144. [CrossRef] [PubMed]
29. Jenkins, D.A.; Kendall, C.C.; McKeown-Eyssen, G.; Josse, R.G.; Silverberg, J.; Booth, G.L.; Leiter, L.A. Effect
of a low-glycemic index or a high-cereal fiber diet on type 2 diabetes: A randomized trial. JAMA 2008, 300,
2742–2753. [CrossRef] [PubMed]
30. Jenkins, D.A.; Kendall, C.C.; Augustin, L.A.; Mitchell, S.; Sahye-Pudaruth, S.; Blanco Mejia, S.; Josse, R.G.
Effect of legumes as part of a low glycemic index diet on glycemic control and cardiovascular risk factors in
type 2 diabetes mellitus: A randomized controlled trial. Arch. Intern. Med. 2012, 172, 1653–1660. [CrossRef]
[PubMed]
31. Ma, Y.; Olendzki, B.C.; Merriam, P.A.; Chiriboga, D.E.; Culver, A.L.; Li, W.; Pagoto, S.L. A randomized
clinical trial comparing low-glycemic index versus ADA dietary education among individuals with type 2
diabetes. Nutrition 2008, 24, 45–56. [CrossRef] [PubMed]
32. Gonçalves Reis, C.E.; Dullius, J. Glycemic acute changes in type 2 diabetics caused by low and high glycemic
index diets. Nutr. Hosp. 2011, 26, 546–552. [PubMed]
33. Stenvers, D.J.; Schouten, L.J.; Jurgens, J.; Endert, E.; Kalsbeek, A.; Fliers, E.; Bisschop, P.H. Breakfast
replacement with a low-glycaemic response liquid formula in patients with type 2 diabetes: A randomised
clinical trial. Br. J. Nutr. 2014, 112, 504–512. [CrossRef] [PubMed]
34. Visek, J.; Lacigova, S.; Cechurova, D.; Rusavy, Z. Comparison of a Low-Glycemic Index vs. Standard Diabetic
Diet; Biomedical Papers of the Medical Faculty of The University Palacky; The University Palacky: Olomouc,
Czechoslovakia, 2014; Volume 158, pp. 112–116.
35. Wolever, T.; Gibbs, A.; Mehling, C.; Chiasson, J.; Connelly, P.; Josse, R.; Ryan, E. The Canadian Trial of
Carbohydrates in Diabetes (CCD), a 1-y controlled trial of low-glycemic-index dietary carbohydrate in type 2
diabetes: No effect on glycated hemoglobin but reduction in C-reactive protein. Am. J. Clin. Nutr. 2008, 87,
114–125. [CrossRef] [PubMed]
36. Yusof, B.M.; Talib, R.A.; Kamaruddin, N.A.; Karim, N.A.; Chinna, K.; Gilbertson, H. A low-GI diet is
associated with a short-term improvement of glycaemic control in Asian patients with type 2 diabetes.
Diabetes Obes. Metab. 2009, 11, 387–396. [CrossRef] [PubMed]
37. Miller, C.K.; Headings, A.; Peyrot, M.; Nagaraja, H. A behavioural intervention incorporating specific
glycaemic index goals improves dietary quality, weight control and glycaemic control in adults with type 2
diabetes. Public Health Nutr. 2011, 14, 1303–1311. [CrossRef] [PubMed]
38. American Diabetes Association. Life style management: Standards of Medical Care in Diabetes–2018.
Diabetes Care 2018, 41 (Suppl. 1), S38–S50.
39. Ojo, O.; Brooke, J. Evaluation of the role of enteral nutrition in managing patients with diabetes: A systematic
review. Nutrients 2014, 6, 5142–5152. [CrossRef] [PubMed]
40. Widanagamage, R.D.; Ekanayake, S.; Welihinda, J. Carbohydrate-rich foods: Glycaemic indices and the effect
of constituent macronutrients. Int. J. Food Sci. Nutr. 2009, 60, 215–223. [CrossRef] [PubMed]
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
15
nutrients
Article
Effects of Different Dietary and Lifestyle
Modification Therapies on Metabolic Syndrome
in Prediabetic Arab Patients: A 12-Month
Longitudinal Study
Hanan A. Alfawaz 1,2 , Kaiser Wani 1,3 , Abdullah M. Alnaami 1,3 , Yousef Al-Saleh 4 ,
Naji J. Aljohani 5 , Omar S. Al-Attas 1,3 , Majed S. Alokail 1,3 , Sudhesh Kumar 6
and Nasser M. Al-Daghri 1,3, *
1 Prince Mutaib Chair for Biomarkers of Osteoporosis, Biochemistry Department, College of Science,
King Saud University, Riyadh 11451, Saudi Arabia; [email protected] (H.A.A.);
[email protected] (K.W.); [email protected] (A.M.A.); [email protected] (O.S.A.-A.);
[email protected] (M.S.A.)
2 Department of Food Science and Nutrition, College of Food Science & Agriculture, King Saud University,
Riyadh 11451, Saudi Arabia
3 Biomarkers Research Program, Biochemistry Department, College of Science, King Saud University,
Riyadh 11451, Saudi Arabia
4 College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11461, Saudi Arabia;
[email protected]
5 Specialized Diabetes and Endocrine Center, King Fahad Medical City, Faculty of Medicine, King Saud bin
Abdulaziz University for Health Sciences, Riyadh 11525, Saudi Arabia; [email protected]
6 Division of Metabolic and Vascular Health, Clinical Sciences Research Institute, University Hospitals
Coventry and Warwickshire Trust, Walsgrave, Coventry CV2 2DX, UK; [email protected]
* Correspondence: [email protected]; Tel.: +966-11-467-5939
Abstract: This three-arm, randomized, controlled study aimed to determine the differences in the
effects of general advice (GA) on lifestyle change, intensive lifestyle modification programme (ILMP)
and GA + metformin (GA + Met) in reducing the prevalence of full metabolic syndrome (MetS)
in subjects with prediabetes; 294 Saudis with prediabetes (fasting glucose 5.6–6.9 mmol/L) were
initially randomized, 263 completed 6 months and 237 completed 12 months. They were allocated
into three groups: GA group which received a standard lifestyle change education; ILMP which
followed a rigorous lifestyle modification support on diet and physical activity; and a GA + Met
group. Anthropometric and biochemical estimations were measured. Full MetS (primary endpoint)
and its components (secondary endpoint) were screened at baseline, 6 and 12 months. Full MetS
in the ILMP group decreased by 26% (p < 0.001); in GA + Met group by 22.4% (p = 0.01) and in GA
group by 8.2% (p = 0.28). The number of MetS components decreased significantly in the ILMP and
GA + Met groups (mean change 0.81, p < 0.001 and 0.35, p = 0.05, respectively). Between-group
comparison revealed a clinically significant decrease in MetS components in favor of the ILMP group
(−0.58 (−0.88–0.28), p < 0.001). This study highlights the clinical potency of ILMP versus other
diabetes prevention options in reducing MetS in Saudi adults with elevated fasting glucose.
1. Introduction
Type 2 diabetes mellitus (T2DM) has a huge impact on the health status of patients and the
overall health care cost of the country. One big opportunity to reduce such an impact is to reduce
the incidence of the disease by focusing on high-risk people such as those with impaired glucose
regulation. This condition is characterized either by impaired fasting glucose (IFG) (fasting glucose
levels 5.6–6.9 mmol/L) or impaired glucose tolerance (IGT) (2-h oral glucose tolerance test (OGTT)
7.8–11 mmol/L) [1]. Every year, about 5–10% of people with IGT progress to T2DM [2]. Fortunately,
landmark clinical trials like the Diabetes Prevention Programme (DPP) [3] and others [4,5] have
revealed the effectiveness of lifestyle modifications in reducing the incidence of T2DM. The major focus
of these interventions were weight loss and physical activity. However, a recent meta-analysis [6,7]
concluded that when translated into routine clinical settings, these expensive programmes have more
effect on weight reduction and less effect on diabetes risk reduction. Clearly, such intervention studies
need to be done, not only to add to the literature to better predict the outcomes on a global health-care
perspective but also to devise large-scale effective and low cost intervention programmes on a regional
level such as in Saudi Arabia, where data are limited.
Identification of individual risk factors associated with the increased effectiveness of lifestyle
intervention programmes may help in devising them in a more economical and practical way. Metabolic
Syndrome (MetS), an amalgam of cardiovascular risk factors, is a major health problem globally. It is
defined by the National Cholesterol Education Program Adult Treatment Panel III (NCEP ATP III) as the
presence of at least three out of five risk factors (central obesity, hyperglycemia, low HDL-cholesterol,
hypertriglyceridemia and elevated blood pressure) [8]. The prevalence of MetS is increasing rapidly
worldwide, including Saudi Arabia, as a result of rapid economic growth and Westernization of
diet [9,10].
The risk of developing incident T2DM in patients with MetS is manifold compared to those
without this condition [11]. Also, all components of MetS are independent risk factors for developing
T2DM [12]. Prevention of T2DM in subjects with impaired glucose regulation through lifestyle
modifications in diet, physical activity or by giving drugs like metformin have been studied in the past,
however, the applicability of these treatments in prevention or reversal of MetS is largely unknown,
particularly in a population like Saudi Arabia where the prevalence of both T2DM and MetS is
high [13,14]. We hypothesize that lifestyle modifications in diet and physical activity have a role
in preventing or reversal of Mets in subjects with impaired glucose regulation. Hence, this study
aimed to investigate the differences in the effectiveness of lifestyle modifications (diet and physical
activity) and drug therapy (metformin), in an adult Saudi population with impaired glucose regulation,
in preventing or reversing full MetS and its individual components.
17
Nutrients 2018, 10, 383
identified as one of the five components of MetS (NCEP ATP III criteria) [15]. Interested participants
were referred to the concerned physician who used the guidelines stated under section “prevention
or delay of type 2 diabetes” in “standards for medical care in diabetes” [16] to screen the candidates.
Subjects who were already on anti-hyperglycemic treatment; pregnant or lactating women; with known
renal, hepatic, pulmonary, cardiac, etc., complications were excluded.
A computer-generated serial number, randomly assigned to one of the three intervention groups
(GA, ILMP, and GA + Met), was given blindly to each participant. All participants were allocated (1:1:1)
to receive one of the three interventions. True allocation concealment was done since the research
personnel involved cannot adjust randomization. The total duration of this lifestyle intervention
programme was 12 months. Fasting blood samples and anthropometric data was collected at
recruitment, at 6-month and at the end of the programme. Out of the 294 initially recruited, the
data for 217 was used in this study. A flow chart summarizing the programme is provided in Figure 1.
Figure 1. Flow chart detailing the participation of subjects and their allocation to treatment groups.
18
Nutrients 2018, 10, 383
2.2. Intervention
All participants had an orientation session with concerned physician and a dietician where they
were educated about risk of developing T2DM and the current scenario of diabetes worldwide and
Saudi Arabia. They were advised to adopt lifestyle modifications in their dietary habits; weight
reduction; exercise; increased physical activity etc. This knowledge sharing included distributing
pamphlets and booklets with information related to lifestyle modifications in earlier programmes,
done elsewhere [17,18]. In addition to this, every four months, seminars were conducted at the
auditorium of the respective hospitals in which the investigators of the current study educated the
participants about lifestyle modifications to prevent T2DM. This intervention process was unified
across the two study centers.
The participants in the general advice (GA) group received the usual instructions to lifestyle
change as described above. In addition to this the participants in intensive lifestyle modification
programme (ILMP) group were followed with a rigorous lifestyle modification support published
earlier [19,20], which included:
(a) Individual consultation with the dietician was done to assess the participant’s food intake.
Special dietary charts were supplied to participants explaining how to reduce the total fat intake
to less than 30% of energy consume and increase the fiber intake to 15 g/1000 Kcal.
(b) Guidelines for physical activity were supplied as pamphlets to each participant. Also, each
participant was given a pedometer (081564483, Patterson Medical) and recommended at least
5000 steps per day to gradually increase as tolerance develops.
(c) Individual consultation with an expert on vitamin D to educate about the benefits of optimal
levels of vitamin D for good health. They were recommended to expose to sunlight for at least
30 min either before 10 a.m. and/or after 3 p.m. twice a week.
The intervention in ILMP group was monitored regularly which were scheduled every three
months during the course of the programme.
The third intervention group (GA + Met) was provided with the same general advice and were
given 500 mg of metformin hydrochloride, twice a day. The participants in ILMP and GA + Met groups
were regularly contacted by research assistants to reinforce instructions through phone calls.
19
Nutrients 2018, 10, 383
Cholesterol Education Programme Adult Treatment Panel III” (NCEP ATP III) as having atleast three
of the five components [15]:
Two other variables were tested in this study for the intervention effects between groups. One was
the total number of MetS components (taken as a continuous variable) and the other was the MetS
risk-score. The MetS risk-score was constructed for the evaluation of continuous MetS status, calculated
using the formula with cut-off values employed to define each component of MetS, with consideration
to age and gender as follows:
MetS risk-score = ((waist/101.6 for males or 88.9 for males) + (fasting glucose/5.6) −
(HDL-Cholesterol/1.03 for males or 1.30 for females) + (triglyceride/1.7) + (systolic BP/130) + (diastolic
BP/85)) × (Age/45 for males or 50 for females).
The Receiver Operating Characteristic (ROC) analysis was employed to test this score for
predicting MetS in our data, with full MetS positive (≥3 MetS components) versus full MetS negative
(<3 MetS components). The ROC revealed an area under the curve (AOC) of 0.890 with 95% confidence
interval of 0.86 and 0.92 and a p-value of <0.001. The cut-off of MetS risk-score for predicting MetS,
obtained in ROC analysis, was 3.85 (Supplementary Figure S1, Table S1–S2).
3. Results
A total of 294 (98 in each group) Saudi adults with impaired fasting glucose were initially randomized
and 237 (94 in GA, 75 in ILMP and 68 in GA + Met) completed the entire 12 months of this
intervention. The most common reasons for drop out included loss to follow-up and poor compliance.
After excluding persons with missing data >5% in any parameter, the data for 217 subjects (85 in GA,
73 in ILMP and 59 in GA + Met) were used for analysis (Figure 1).
20
Nutrients 2018, 10, 383
21
Nutrients 2018, 10, 383
Table 1. Cont.
22
Nutrients 2018, 10, 383
in GA and GA + Met groups, respectively, while it decreased by 1.4% (n = 1) in the ILMP group.
The highest change was seen in component 2 (hyperglycemia) where from baseline to end of the study,
hyperglycemia was reduced by 22.4% (n = 19), 38.4% (n = 28) and 39% (n = 23), respectively, in GA,
ILMP, and GA+ILMP groups respectively. Full MetS was also significantly reduced by 8.2% (n = 7),
26% (n = 19, p < 0.001), and 22.4% (n = 13, p = 0.013), respectively, in GA, ILMP, and GA + Met groups.
3.3. Odds of Having MetS and Its Components at Follow-Up Compared to Baseline
Table 3 shows the odds of having MetS and its individual components at follow-up compared to
baseline in each group. Supplementary Table S3 shows the odds of having MetS for these covariates,
independent of each other.
Table 3. Odds ratio representing the risk of having MetS and its individual components at follow-up
compared to baseline in respective groups.
23
Nutrients 2018, 10, 383
Table 3. Cont.
The odds of central obesity and hypertriglyceridemia showed marginal changes in all groups,
with odds ratio (95% confidence interval (C.I.)) of 0.84 (0.4, 1.8) and 0.86 (0.5, 1.3), respectively, in GA
group; 0.80 (0.3, 2.2) and 0.80 (0.5, 1.4) in ILMP group and 0.74 (0.2, 3.5) and 1.0 (0.6, 1.8) in GA + Met
group. The odds of low HDL-cholesterol increased (1.66 (0.8, 3.5)) in GA group, but was reduced both
in ILMP (0.79 (0.4, 1.4)) and GA + Met (0.86 (0.4, 1.7)) groups. The odds of hypertension increased
in GA + Met group (2.36 (1.2, 4.6), p-value = 0.012) and GA group (1.28 (0.5, 3.1)) but was modestly
decreased in ILMP group (0.88 (0.4, 2.1)). The odds of full MetS was significantly reduced both in
ILMP (0.25 (0.1, 0.4), p-value < 0.001) and GA + Met (0.32 (0.1, 0.7), p-value = 0.005) groups only.
Figure 2 shows the odds ratio of MetS and its components at the end of the study compared to
baseline for different treatment groups. The odds for MetS were significantly reduced in ILMP group
followed by GA + Met group.
3.4. Intervention Effects in Total Number of MetS Components and MetS Risk Factor
Table 4 shows the intervention effects in the total number of MetS components and the MetS
risk-score. The number of MetS components significantly decreased from baseline to the end of the
study in the ILMP group (mean change (standard error)) of 0.81 (0.13), p-value < 0.001. Similarly,
24
Nutrients 2018, 10, 383
it decreased in GA + Met group by 0.35 (0.18), p-value = 0.05. Between-group comparison revealed
that the decrease in the number of MetS components in ILMP was statistically more significant than
GA (p < 0.001). Between (GA + Met) and GA groups, this decrease was not significant. Intervention
effects between groups for MetS risk score showed the same trend between ILMP vs. GA group (0.31
(−0.53, −0.09); p = 0.003).
Figure 2. Odds ratio (OR) of MetS and its components for three treatment groups. The model is adjusted
for age, sex, BMI and baseline covariates: waist, systolic and diastolic BP, fasting glucose, HbA1c, and
vitamin D.
Table 4. Intervention effects in number of MetS components and the MetS risk-score.
25
Nutrients 2018, 10, 383
Figure 3 shows the intervention effects of the three treatment strategies on MetS, represented by
bar graphs showing changes in total number of MetS components and MetS risk-score overtime.
Figure 3. Intervention effects in total number of MetS components (A) and MetS risk-score (B) for
three treatment strategies calculated at baseline and follow-up. Data is adjusted for age, sex and BMI,
systolic and diastolic BP, waist, fasting glucose, HbA1c, and vitamin D (all baseline values). The overall
change in ILMP and GA + Met groups versus GA group reported as Mean Change (95% confidence
interval). Within group intervention effect is shown as * for p < 0.05 or ** for p < 0.01.
4. Discussion
In this study, Saudi adults with impaired glucose regulation were given treatment based on
lifestyle modifications in diet and exercise or low doses of metformin to assess the status of MetS and
its individual components over time. Changing lifestyle and reducing the burden of chronic diseases in
this population is of great public health importance, however, the impact of such programmes on full
MetS and components of MetS in the pre-diabetic Saudi population has not been studied. It is notable
that, for the past two decades since these lifestyle intervention studies started, the study outcome
variables in most of them has been the incidence of diabetes. In this study, we chose a broader set of
26
Nutrients 2018, 10, 383
outcome measures and focused on the status of MetS and its components together, as well as those
independently considered as risk factors for the incidence of T2DM. To the best of our knowledge,
this is the first such study in Saudi Arabia which emphasizes the changes in the status of MetS and its
components through lifestyle modifications. A similar study was done by the Diabetes Prevention
Program (DPP) research group in 2005 [21] and focused on lifestyle intervention and metformin-based
therapies for MetS.
Mean fasting glucose was significantly reduced in the ILMP and GA + Met groups while in
the GA group this reduction was modest. Similarly, the prevalence of hyperglycemia was reduced
significantly by 38.4% and 39% in the ILMP and GA + Met groups, respectively. This improvement in
mean fasting glucose is similar to previous studies done in USA [22], Italy [23], and Iran [24] where,
on an average, fasting glucose was reduced by 6 mg/dl from baseline to the end of the study in the
dietary intervention group. Lifestyle modifications, including replacing high-glycemic index diet with
low-glycemic index diet like vegetables, dairies, and whole grains, etc. might have played a role in the
reduction in mean fasting glucose [25]. However, as expected, the mean reduction in fasting glucose in
the ILMP group is lesser than that found in the GA + Met group. Though the molecular mechanism
of the action of metformin remains debated, a consensus is on the direct action of metformin on
hepatic glucose production and improving insulin sensitivity in muscle and fat cells [26]. Furthermore,
metformin’s action is not limited to glucose reduction and insulin sensitivity, it is also an effective
weight loss drug due to its effect on appetite and its regulation of fat oxidation, storage in liver and
adipose tissue [27]. In our study, the mean weight loss from baseline to the end of the study was
4.15 kg in GA + Met group and this was higher than the ILMP group (1.86 kg).
The ILMP group showed modest improvements in all of the other components of MetS, namely,
central obesity, low HDL-cholesterol, hypertriglyceridemia and hypertension. Waist circumference
also decreased from baseline to the end of study in the ILMP group by an average of 1.86 cm.
This change in waist circumference is close to a mean decrease of 2.7 cm found in a meta-analysis
of six interventions [28]. The odds for low HDL-cholesterol was 1.66 (95% confidence interval 0.8 to
3.5) in the GA group, indicating a higher risk in the absence of an intensive lifestyle change program.
Similarly, the odds for hypertriglyceridemia at the end of the study versus baseline in the metformin
group was 1.0 (0.6, 1.8), which together with the odds obtained for low HDL-cholesterol indicated
that GA + Met induce less effects on lipid levels than the ILMP. This was in agreement with studies
like [29] which reported significantly larger improvement in lipid indexes in the ILMP group than in
the GA + Met group.
The prevalence of hypertension was significantly higher at the end of the study compared to
baseline in the GA + Met group (O.R. 2.36 (1.2, 4.6), p-value 0.012). The literature on the effect of
metformin on hypertension has yielded conflicting results. While some [30] suggest metformin has no
intrinsic effect on blood pressure, others, like a recent meta-analysis from 26 studies [31], suggested
that metformin could effectively lower systolic blood pressure especially in those with IGT. There
are also reports on the role of metformin in reversing pulmonary hypertension through inhibition of
aromatase synthesis [32]. In any case, the findings in this study related to hypertension in the GA + Met
group should be looked at keeping in mind that most of the study participants (72.3% of all subjects
and 71.2% of GA + Met group) are females. Women normally tend to have lower blood pressure than
men, related to sex differences in concentrations of angiotensin II and sodium reabsorption in distal
nephron [33].
The decrease in the prevalence of full MetS at the end of the study compared to baseline was
greater in ILMP (−26%, p< 0.001) and GA + Met groups (−22.4%, p = 0.013) than the GA group
(−8.2%, p = 0.281). Furthermore, the analysis of the number of MetS components as well as MetS
risk-score suggests that although ILMP was superior to GA + Met, both are more effective compared
to GA. The biological mechanism whereby ILMP or metformin exerts its protective effect from MetS is
complex and unclear; however, greater intakes of dietary fibers, low-glycemic index foods, vitamins,
27
Nutrients 2018, 10, 383
more physical activity, etc. in ILMP all contribute to the reduction in low-grade inflammation, normally
associated with MetS [34,35].
The MetS risk-score, employed in testing the hypothesis of the study regarding MetS reversal/
reduction with three different treatments, had high discriminative power for presence/absence of
MetS (AOC of 0.890, 95% C.I. of 0.86 to 0.92 and a p-value of <0.001, Supplementary Figure S1,
Tables S1–S2). A continuous risk-score for predicting MetS has been developed by approaches such as
taking standardized residuals (z-scores) or scores from principal component analysis of risk factors [36,37].
These scores, although precise, are complex and require specialized statistical software. While the
authors attempted an easy-to-calculate continuous MetS risk-score, it should be used with caution as
the values apply to the population and local laboratory reference values.
The authors acknowledge certain limitations of the study. Firstly, the study was done on subjects
with IGT and, hence, the results may not hold true for those with normal glucose regulation but having
full MetS manifestations. Such intensive lifestyle change programmes should be conducted in these
categories to have a better understanding of the benefits. Secondly, the study does not give details
about the actual changes in lifestyle that each participant in this programme carried out in terms of
variables related to diet or physical activity, measured before intervention and follow up. Despite
the study being based on self-monitoring; it showed a significant reversal in MetS in the ILMP group
which suggests that, if guided properly, people are willing to change from a sedentary to a healthy
lifestyle. The study also offered a unique statistical approach in assessing MetS and its components
and by evaluating the change in the total number of MetS components as an outcome variable. Also,
the MetS risk-score devised in this study, if further researched, may provide a simplistic estimate for
assessing MetS in an individual.
5. Conclusions
Intensive lifestyle modifications or low dose metformin for a period of 12 months significantly
reduces MetS manifestation in individuals with prediabetes, with lifestyle modifications being superior
to metformin, as the latter’s potency is limited to weight loss and reduction of hyperglycemia, while the
former improves all the components of MetS together as well as independently.
References
1. American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care 2014, 37,
S81–S90.
2. Prediabetes and the Potential to Prevent Diabetes. Lancet 2012, 379. Available online: http://www.thelancet.
com/journals/lancet/article/PIIS0140-6736%2812%2960960-X/fulltext (accessed on 15 March 2018).
3. Bray, G.A.; Ehrmann, D.A.; Goldberg, R.B.; Diabetes Prevention Program Research Group. Reduction in the
incidence of type 2 diabetes with lifestyle intervention or metformin. N. Engl. J. Med. 2002, 346, 393–403.
28
Nutrients 2018, 10, 383
4. Pan, X.R.; Li, G.W.; Hu, Y.H.; Wang, J.X.; Yang, W.Y.; An, Z.X.; Hu, Z.X.; Xiao, J.Z.; Cao, H.B.; Liu, P.A. Effects
of diet and exercise in preventing NIDDM in people with impaired glucose tolerance: The Da Qing IGT and
Diabetes Study. Diabetes Care 1997, 20, 537–544. [CrossRef] [PubMed]
5. Ramachandran, A.; Snehalatha, C.; Mary, S.; Mukesh, B.; Bhaskar, A.; Vijay, V. The Indian Diabetes Prevention
Programme shows that lifestyle modification and metformin prevent type 2 diabetes in Asian Indian subjects
with impaired glucose tolerance (IDPP-1). Diabetologia 2006, 49, 289–297. [CrossRef] [PubMed]
6. Cardona-Morrell, M.; Rychetnik, L.; Morrell, S.L.; Espinel, P.T.; Bauman, A. Reduction of diabetes risk in
routine clinical practice: Are physical activity and nutrition interventions feasible and are the outcomes
from reference trials replicable? A systematic review and meta-analysis. BMC Public Health 2010, 10, 653.
[CrossRef] [PubMed]
7. Johnson, M.; Jones, R.; Freeman, C.; Woods, H.B.; Gillett, M.; Goyder, E.; Payne, N. Can diabetes prevention
programmes be translated effectively into real-world settings and still deliver improved outcomes?
A synthesis of evidence. Diabet. Med. 2013, 30, 3–15. [CrossRef] [PubMed]
8. Expert Panel on Detection, Evaluation. Executive summary of the Third Report of the National Cholesterol
Education Program (NCEP) expert panel on detection, evaluation, and treatment of high blood cholesterol
in adults (Adult Treatment Panel III). JAMA 2001, 285, 2486–2497.
9. Al-Qahtani, D.A.; Imtiaz, M.L. Prevalence of metabolic syndrome in Saudi adult soldiers. Saudi Med. J. 2005,
26, 1360–1366. [PubMed]
10. Al-Nozha, M.; Al-Khadra, A.; Arafah, M.R.; Al-Maatouq, M.A.; Khalil, M.Z.; Khan, N.B.; Al-Mazrou, Y.Y.;
Al-Marzouki, K.; Al-Harthi, S.S.; Abdullah, M.; et al. Metabolic syndrome in Saudi Arabia. Saudi Med. J.
2005, 26, 1918–1925. [PubMed]
11. Wilson, P.W.; D’Agostino, R.B.; Parise, H.; Sullivan, L.; Meigs, J.B. Metabolic syndrome as a precursor of
cardiovascular disease and type 2 diabetes mellitus. Circulation 2005, 112, 3066–3072. [CrossRef] [PubMed]
12. Hanson, R.L.; Imperatore, G.; Bennett, P.H.; Knowler, W.C. Components of the “metabolic syndrome” and
incidence of type 2 diabetes. Diabetes 2002, 51, 3120–3127. [CrossRef] [PubMed]
13. Al-Daghri, N.M.; Al-Attas, O.S.; Alokail, M.S.; Alkharfy, K.M.; Yousef, M.; Sabico, S.L.; Chrousos, G.P.
Diabetes mellitus type 2 and other chronic non-communicable diseases in the central region, Saudi Arabia
(Riyadh cohort 2): A decade of an epidemic. BMC Med. 2011, 9, 76. [CrossRef] [PubMed]
14. Aljohani, N.J. Metabolic syndrome: Risk factors among adults in Kingdom of Saudi Arabia. J. Fam.
Community Med. 2014, 21, 170–175. [CrossRef] [PubMed]
15. Huang, P.L. A comprehensive definition for metabolic syndrome. Dis. Models Mech. 2009, 2, 231–237.
[CrossRef] [PubMed]
16. American Diabetes Association. Standards of Medical Care in Diabetes. Diabetes Care 2017, 40.
Available online: http://care.diabetesjournals.org/content/diacare/suppl/2016/12/15/40.Supplement_
1.DC1/DC_40_S1_final.pdf (accessed on 2 February 2018).
17. Tuomilehto, J.; Lindström, J.; Eriksson, J.G.; Valle, T.T.; Hämäläinen, H.; Ilanne-Parikka, P.;
Keinänen-Kiukaanniemi, S.; Laakso, M.; Louheranta, A.; Rastas, M. Prevention of type 2 diabetes mellitus by
changes in lifestyle among subjects with impaired glucose tolerance. N. Engl. J. Med. 2001, 344, 1343–1350.
[CrossRef] [PubMed]
18. Kosaka, K.; Noda, M.; Kuzuya, T. Prevention of type 2 diabetes by lifestyle intervention: A Japanese trial in
IGT males. Diabetes Res. Clin. Pract. 2005, 67, 152–162. [CrossRef] [PubMed]
19. Al-Daghri, N.M.; Alfawaz, H.; Aljohani, N.J.; Al-Saleh, Y.; Wani, K.; Alnaami, A.M.; Alharbi, M.; Kumar, S.
A 6-month “self-monitoring” lifestyle modification with increased sunlight exposure modestly improves
vitamin D status, lipid profile and glycemic status in overweight and obese Saudi adults with varying
glycemic levels. Lipids Health Dis. 2014, 13, 87. [CrossRef] [PubMed]
20. Al-Daghri, N.M.; Alfawaz, H.; Aljohani, N.J.; Wani, K.; Alharbi, M.; Al-Saleh, Y.; Al-Attas, O.S.; Alokail, M.S.
Age-specific improvements in impaired fasting glucose and vitamin D status using a lifestyle intervention
programme in overweight and obese Saudi subjects. Int. J. Clin. Exp. Med. 2016, 9, 19850–19857.
21. Orchard, T.J.; Temprosa, M.; Goldberg, R.; Haffner, S.; Ratner, R.; Marcovina, S.; Fowler, S. The effect of
metformin and intensive lifestyle intervention on the metabolic syndrome: The Diabetes Prevention Program
randomized trial. Ann. Intern. Med. 2005, 142, 611–619. [CrossRef] [PubMed]
29
Nutrients 2018, 10, 383
22. Watkins, L.L.; Sherwood, A.; Feinglos, M.; Hinderliter, A.; Babyak, M.; Gullette, E.; Waugh, R.;
Blumenthal, J.A. Effects of exercise and weight loss on cardiac risk factors associated with syndrome X.
Arch. Intern. Med. 2003, 163, 1889–1895. [CrossRef] [PubMed]
23. Esposito, K.; Marfella, R.; Ciotola, M.; Di Palo, C.; Giugliano, F.; Giugliano, G.; D’armiento, M.; D’andrea, F.;
Giugliano, D. Effect of a Mediterranean-style diet on endothelial dysfunction and markers of vascular
inflammation in the metabolic syndrome: A randomized trial. JAMA 2004, 292, 1440–1446. [CrossRef]
[PubMed]
24. Azadbakht, L.; Mirmiran, P.; Esmaillzadeh, A.; Azizi, T.; Azizi, F. Beneficial effects of a Dietary Approaches
to Stop Hypertension eating plan on features of the metabolic syndrome. Diabetes Care 2005, 28, 2823–2831.
[CrossRef] [PubMed]
25. Brand-Miller, J.; Hayne, S.; Petocz, P.; Colagiuri, S. Low–glycemic index diets in the management of diabetes.
Diabetes Care 2003, 26, 2261–2267. [CrossRef] [PubMed]
26. Rena, G.; Hardie, D.G.; Pearson, E.R. The mechanisms of action of metformin. Diabetologia 2017, 60, 1577–1585.
[CrossRef] [PubMed]
27. Malin, S.K.; Kashyap, S.R. Effects of metformin on weight loss: Potential mechanisms. Curr. Opin. Endocrinol.
Diabetes Obes. 2014, 21, 323–329. [CrossRef] [PubMed]
28. Yamaoka, K.; Tango, T. Effects of lifestyle modification on metabolic syndrome: A systematic review and
meta-analysis. BMC Med. 2012, 10, 138. [CrossRef] [PubMed]
29. American Diabetes Association. Impact of intensive lifestyle and metformin therapy on cardiovascular
disease risk factors in the diabetes prevention program. Diabetes Care 2005, 28, 888–894.
30. Wulffelé, E.M.; Kooy, A.; Zeeuw, D.D.; Stehouwer, C.; Gansevoort, R. The effect of metformin on blood
pressure, plasma cholesterol and triglycerides in type 2 diabetes mellitus: A systematic review. J. Intern. Med.
2004, 256, 1–14. [CrossRef] [PubMed]
31. Zhou, L.; Liu, H.; Wen, X.; Peng, Y.; Tian, Y.; Zhao, L. Effects of metformin on blood pressure in nondiabetic
patients: A meta-analysis of randomized controlled trials. J. Hypertens. 2017, 35, 18–26. [CrossRef] [PubMed]
32. Dean, A.; Nilsen, M.; Loughlin, L.; Salt, I.P.; MacLean, M.R. Metformin reverses development of pulmonary
hypertension via aromatase inhibition. Hypertension 2016, 68, 446–454. [CrossRef] [PubMed]
33. Kagan, A.; Faibel, H.; Ben-Arie, G.; Granevitze, Z.; Rapoport, J. Gender differences in ambulatory blood
pressure monitoring profile in obese, overweight and normal subjects. J. Hum. Hypertens. 2007, 21, 128–134.
[CrossRef] [PubMed]
34. Gleeson, M.; Bishop, N.C.; Stensel, D.J.; Lindley, M.R.; Mastana, S.S.; Nimmo, M.A. The anti-inflammatory
effects of exercise: Mechanisms and implications for the prevention and treatment of disease. Nat. Rev.
Immunol. 2011, 11, 607–615. [CrossRef] [PubMed]
35. Messier, S.P.; Mihalko, S.L.; Legault, C.; Miller, G.D.; Nicklas, B.J.; DeVita, P.; Beavers, D.P.; Hunter, D.J.;
Lyles, M.F.; Eckstein, F. Effects of intensive diet and exercise on knee joint loads, inflammation, and clinical
outcomes among overweight and obese adults with knee osteoarthritis: The IDEA randomized clinical trial.
JAMA 2013, 310, 1263–1273. [CrossRef] [PubMed]
36. Andersen, L.B.; Harro, M.; Sardinha, L.B.; Froberg, K.; Ekelund, U.; Brage, S.; Anderssen, S.A. Physical
activity and clustered cardiovascular risk in children: A cross-sectional study (The European Youth Heart
Study). Lancet 2006, 368, 299–304. [CrossRef]
37. Katzmarzyk, P.T.; Perusse, L.; Malina, R.M.; Bergeron, J.; Despres, J.; Bouchard, C. Stability of indicators of
the metabolic syndrome from childhood and adolescence to young adulthood: The Quebec Family Study.
J. Clin. Epidemiol. 2001, 54, 190–195. [CrossRef]
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
30
nutrients
Article
Dietary Inflammatory Index and Type 2 Diabetes
Mellitus in Adults: The Diabetes Mellitus Survey
of Mexico City
Edgar Denova-Gutiérrez 1 , Paloma Muñoz-Aguirre 2 , Nitin Shivappa 3,4,5 , James R. Hébert 3,4,5 ,
Lizbeth Tolentino-Mayo 1 , Carolina Batis 6 and Simón Barquera 1, *
1 Nutrition and Health Research Center, National Institute of Public Health, Cuernavaca 62100, Mexico;
[email protected] (E.D.-G.); [email protected] (L.T.-M.)
2 Center for Research on Population Health , National Institute of Public Health, Cuernavaca 62100, Mexico;
[email protected]
3 Cancer Prevention and Control Program, University of South Carolina, Columbia, SC 29208, USA;
[email protected] (N.S.); [email protected] (J.R.H.)
4 Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South
Carolina, Columbia, SC 29208, USA
5 Connecting Health Innovations LLC, Columbia, SC 29250, USA
6 CONACYT-Nutrition and Health Research Center, National Institute of Public Health, Cuernavaca 62100,
Mexico; [email protected]
* Correspondence: [email protected]; Tel.: +52-(777)-329-3017 or +52-(777)-311-2219
Abstract: Diet and inflammation are both associated with type 2 diabetes mellitus (T2DM). In the
present study, we aimed to assess the relation between the dietary inflammatory index (DII) and
the presence of T2DM in Mexican adults participating in the Diabetes Mellitus Survey administered
in Mexico City (DMS-MC). The study involved 1174 subjects (48.5% men) between 20–69 years of
age. A validated semi-quantitative food frequency questionnaire was employed to evaluate dietary
intake and to compute DII. The DII is based on scientific evidence about the association between
dietary compounds and six established inflammatory biomarkers. Multivariate logistic regression
models were used to estimate the odds ratios (ORs) and 95% confidence intervals (95% CIs) of DII
in relation to T2DM. Our results suggest that subjects in the highest quintile of the DII had higher
odds of T2DM (OR = 3.02; 95% CI: 1.39, 6.58; p = 0.005) compared to subjects in the lowest quintile
of DII scores. Assessing possible effect modification, an association with T2DM was evident when
comparing DII quintile 5 to quintile 1 for participants aged ≥ 55 years (OR = 9.77; 95% CI: 3.78, 25.50;
p = 0.001). These results suggest that a pro-inflammatory diet is associated with significantly higher
odds of T2DM among adult Mexicans.
Keywords: type 2 diabetes mellitus; dietary inflammatory index; obesity; Mexican population; survey
1. Introduction
Low-grade, systemic inflammation, which provides the substrate for many chronic diseases,
is characterized by elevated pro-inflammatory markers, including interleukin-6 (IL-6), and tumor
necrosis factor-alpha (TNF-α) [1]. Chronic systemic inflammation has been linked to non-communicable
diseases such as cancer, cardiovascular disease, and diabetes [2]. Globally, type 2 diabetes mellitus
(T2DM) has become a serious public health problem, affecting 382 million people in 2013 [3] and had
led to a loss of approximately 64 million disability adjusted life-years (DALYs) in 2015 [4]. Currently,
80% of people with T2DM live in low-income and middle-income countries [5]. In Mexico, T2DM
prevalence has reached 14.7% of the population and two million DALYs lost in 2013 [5,6].
Recent evidence indicates that risk factors can induce chronic inflammation, which are also related
with T2DM such as adiposity [7], a sedentary lifestyle [8], and diet [9]. In the past several decades,
certain individual dietary factors have been widely investigated regarding their association with
diabetes risk and inflammation, including high intake of saturated fatty acids, sugar-sweetened
beverages, and starchy food, combined with low consumption of fruits, vegetables, and whole
grains [10]. More recently, alternative approaches have been used in order to capture the complicated
nutrient interactions and cumulative effects in the food matrix [11]. Based on evidence linking diet with
inflammation and chronic conditions such as T2DM, we used a literature-derived, population-based
dietary inflammatory index (DII) [12] to evaluate the potential inflammatory properties of diet. The DII
has been validated with different inflammatory biomarkers [13,14]. Furthermore, it has been associated
with components of metabolic syndrome [15], as well as a variety of chronic disease outcomes [16,17].
To the best of our knowledge, no study has yet explored the relation between the DII and T2DM
in a Mexican population. Thus, we aimed to explore the association between DII and the prevalence of
T2DM in adults participating in the Diabetes Mellitus Survey in Mexico City 2015 (DMS-MC 2015).
32
Nutrients 2018, 10, 385
33
Nutrients 2018, 10, 385
and/or HbA1c values consistent with T2DM diagnostic criteria and were also defined as participants
with T2DM.
34
Nutrients 2018, 10, 385
3. Results
Participants’ baseline characteristics are shown in Table 1. A total of 1174 subjects (48.5% men)
between 20–69 years were included in the present analysis. The overall prevalence of T2DM was 13.6%;
of these, 10.1% were previously diagnosed individuals, whereas 3.5% were individuals with glucose
and/or HbA1c values consistent with T2DM definition at the time of the survey. The mean age among
participants with T2DM was 52.3 years. Subjects with T2DM had a significantly higher prevalence of
obesity, abdominal obesity, had lower levels of physical activity and intake of cereal fiber, and also,
as expected, levels of total cholesterol, triglycerides, and glycated hemoglobin were all significantly
higher in participants with T2DM.
Table 1. Characteristics of the study population: The Diabetes Mellitus Survey of Mexico City, 2015.
35
Nutrients 2018, 10, 385
Table 1. Cont.
According to DII score quintiles, subjects with the most pro-inflammatory diet were significantly
older, less educated, had lower SES, smoked less, had a higher prevalence of obesity, abdominal obesity,
and T2DM (Table 2). The most pro-inflammatory diet was characterized by a higher consumption
of carbohydrates (56.1%; (95% CI: 54.3, 57.8) vs. 53.8% (52.6, 55.1)), red meat and processed meat
(78.3 g/day (66.3, 90.2) vs. 40.1 g/day (33.7, 46.5)), refined cereals (173.9 g/day (147.1, 200.6) vs.
109.2 g/day (96.0, 122.5)), and soft drinks (401.7 mL/day (343.3, 460.1) vs. 69.5 mL/day (51.6, 87.4))
compared to the most anti-inflammatory diet. All of these differences were statistically significant. Also,
significantly decreasing trends were observed for the anti-inflammatory nutrients: fiber, vitamin C,
vitamin D, and magnesium (Table 3).
After adjusting for age and sex, the odds of having T2DM, across all DII score quintiles were
1.00, 1.63, 1.80, 1.84, and 2.29 (95% CI: 1.11, 4.75; p = 0.01). Finally, after additional adjustment for
co-variables, we observed that subjects with the most pro-inflammatory diet had approximately three
times greater odds of having T2DM (OR: 3.02, 95% CI: 1.39, 6.58; p = 0.005), compared to individuals in
the lowest DII quintile (Table 4).
Additionally, a sensitivity analysis included individuals with glucose and/or HbA1c values
consistent with T2DM definition at the time of the survey. In this case, we observed that subjects with
the most pro-inflammatory diet had greater odds of having T2DM (OR = 3.56; 95% CI: 1.13, 9.11) (data
not shown).
In addition, we evaluated the effect-modifying role of age; we observed a greater association with
T2DM contrasting DII extreme quintiles (Q5 vs. Q1 ) for participants aged ≥ 55 years (OR = 9.77; 95%
CI: 3.78, 25.50; p = 0.001) (Figure 1).
36
Table 2. Characteristics of participants according to quintiles of the dietary inflammatory index: The Diabetes Mellitus Survey of Mexico City, 2015.
37
High school 24.5 21.3 27.2 29.8 31.7 <0.001
Bachelor’s degree or higher 59.9 52.6 45.5 42.6 33.9
Smoking status, %
Current 52.9 51.8 47.8 39.5 31.7
Past 10.5 6.1 10.0 17.9 15.2 <0.001
Never 36.6 42.1 42.2 42.6 53.1
Physical activity, %
Inactive 25.7 27.8 29.5 26.5 23.8
0.14
Active/highly active 74.3 72.2 70.5 73.5 76.2
Body mass index (kg/m2 ) b 28.1 (0.38) 28.3 (0.40) 28.7 (0.47) 28.9 (0.45) 29.8 (0.48) 0.007
Body mass index, %
Normal (<25.0 kg/m2 ) 29.2 25.3 23.5 25.4 20.8
Overweight (≥25.0 to <30.0 kg/m2 ) 38.5 38.8 41.4 38.6 36.2 0.005
Obesity (≥30.0 kg/m2 ) 32.3 35.8 35.1 36.0 43.0
Table 2. Cont.
38
error (SE) in parentheses, unless otherwise specified.
Table 3. Nutrient and food consumption according to quintiles of the dietary inflammatory index: in the Diabetes Mellitus Survey of Mexico City, 2015.
Carbohydrates intake (% energy) b 53.8 (52.6, 55.1) 55.0 (53.7, 56.4) 56.0 (55.2, 58.4) 56.1 (54.7, 57.5) 56.1 (54.3, 57.8) <0.001
Protein intake (% energy) 14.4 (13.9, 14.8) 13.9 (13.4, 14.6) 13.7 (13.1, 14.0) 13.0 (12.6, 13.5) 12.6 (11.7, 13.7) <0.001
Total fat intake (% energy) 31.8 (30.9, 32.9) 30.8 (29.8, 31.6) 30.3 (29.2, 31.3) 31.2 (30.1, 32.3) 31.3 (29.9, 32.7) 0.08
Saturated fats (% energy) 11.4 (10.9, 11.9) 11.7 (11.2, 12.2) 11.3 (10.9, 11.8) 11.3 (10.8, 11.8) 12.3 (11.5, 13.2) 0.51
Monounsaturated fats (% energy) 11.2 (10.8, 11.6) 10.8 (10.4, 11.2) 10.6 (10.1, 11.1) 10.9 (10.4, 11.4) 10.9 (10.3, 11.4) 0.20
Polyunsaturated fats (% energy) 7.6 (7.3, 7.9) 7.2 (6.9, 7.5) 6.9 (6.6, 7.2) 6.8 (6.5, 7.1) 6.7 (6.5, 7.0) <0.001
Fiber (g/day) 42.7 (40.0, 45.4) 31.5 (27.8, 35.2) 24.7 (23.5, 25.9) 24.2 (22.6, 25.9) 13.3 (12.7, 14.0) <0.001
Alcohol consumption (g/day) 15.2 (9.4, 21.0) 11.8 (7.2, 16.4) 6.6 (2.2, 11.0) 6.6 (1.2, 13.2) 2.5 (1.2, 3.9) <0.001
Magnesium (mg/day) 600.3 (568.8, 631.8) 453.5 (402.1, 504.8) 365.5 (350.3, 380.7) 355.9 (333.7, 388.2) 219.5 (208.6, 230.5) <0.001
Vitamin C (mg/day) 374.4 (344.5, 404.2) 288.0 (258.2, 317.8) 194.5 (181.0, 208.2) 160.6 (141.4, 179.7) 82.0 (72.9, 91.2) <0.001
Vitamin A (μg/day) 1900.2 (1666.1, 2134.2) 1219.9 (1071.6, 1368.2) 830.1 (772.7, 887.6) 701.5 (635.1, 767.9) 396.3 (360.0, 432.6) <0.001
Vitamin E (mg/day) 13.0 (12.3, 13.6) 8.9 (7.6, 10.1) 6.5 (6.2, 6.8) 6.5 (6.2, 7.3) 3.4 (3.4, 3.9) <0.001
Vitamin D (μg/day) 7.7 (7.1, 8.3) 5.1 (4.4, 5.7) 4.4 (3.9, 4.9) 3.8 (3.4, 4.2) 2.5 (2.5, 3.1) <0.001
Vegetables (g/day) 370.0 (321.2, 418.8) 245.5 (212.8, 278.4) 139.4 (121.3, 157.5) 112.2 (95.9, 128.5) 69.3 (60.0, 78.6) <0.001
Fruits (g/day) 327.6 (293.3, 362.0) 259.9 (225.9, 294.0) 178.8 (162.2, 195.5) 134.5 (114.3, 154.6) 74.2 (64.4, 84.0) <0.001
Legumes (g/day) 67.4 (49.5, 85.3) 51.0 (29.2, 72.6) 31.6 (26.0, 37.2) 33.7 (26.6, 40.8) 21.2 (16.2, 26.1) <0.001
39
Fish and seafood (g/day) 24.1 (15.0, 33.1) 13.9 (4.7, 23.0) 10.8 (5.9, 15.7) 10.4 (5.1, 15.7) 3.2 (1.5, 4.9) <0.001
Dairy products (g/day) 242.5 (209.9, 275.1) 225.8 (188.9, 262.6) 177.9 (142.3, 213.5) 172.5 (134.3, 210.8) 139.2 (109.3, 169.1) <0.001
Red meat and processed meat (g/day) 40.1 (33.7, 46.5) 46.9 (41.3, 52.6) 55.0 (44.4, 65.5) 55.3 (41.8, 68.7) 78.3 (66.3, 90.2) <0.001
Eggs (g/day) 28.9 (23.8, 33.9) 30.8 (25.1, 36.5) 32.2 (25.2, 39.2) 35.5 (28.4, 42.6) 34.3 (26.6, 41.9) <0.001
Refined cereals (g/day) 109.2 (96.0, 122.5) 145.0 (129.8, 160.3) 145.4 (125.5, 165.3) 158.2 (122.7, 193.6) 173.9 (147.1, 200.6) <0.001
Potatoes (g/day) 9.7 (5.2, 14.2) 11.8 (7.7, 15.8) 11.5 (7.8, 15.3) 8.3 (4.9, 11.7) 7.1 (5.1, 9.2) 0.87
Soft drinks (mL/day) 69.5 (51.6, 87.4) 146.9 (109.6, 184.1) 219.5 (175.7, 263.3) 262.4 (202.2, 322.6) 401.7 (343.3, 460.1) <0.001
a p-values were determined using ANOVA test. b Data are given as means, with 95% CI in parentheses.
Nutrients 2018, 10, 385
Table 4. Odds ratio (OR) and 95% confidence intervals (CI) for the relation between the dietary
inflammatory index and T2DM in the Diabetes Mellitus Survey of Mexico City, 2015.
%%
%$ Female 3.06 0.91, 10.80
-
, Age2
+ < 55 years 2.88 1.05, 8.08
* 0.001
) ≥ 55 years 9.77 3.78, 25.50
(
' BMI3
&
% < 25.0 kg/m2 2.66 0.53-6.42
0.13
$
≥ 25.0 kg/m2 5.10 1.66, 15.60
.))
/))
.&) $!&
/&) $!&
!
Physical activity4
Low 3.16 1.16, 7.02
0.02
2.85 0.90, 11.01
Figure 1. Subgroup analysis. Odds ratios (95% CI) for the association between extreme quintiles of
the dietary inflammatory index (DII) and type 2 diabetes mellitus (T2DM). 1 Adjusted for age (years),
physical activity (inactive vs. active/highly active), hours of television watching; tobacco use (current,
past, and never), socioeconomic status (low, medium, and high), education (elementary and secondary
education, high school, and Bachelor’s degree or higher), family history of diabetes mellitus (yes vs. no),
personal history of hypertension (yes vs. no), medication use (yes vs. no), multivitamin use (yes vs. no),
alcohol intake (gr/day), body mass index (<25.0 vs. ≥25.0 kg/m2 ). 2 Adjusted for sex, physical activity
(inactive vs. active/highly active), hours of television watching; tobacco use (current, past, and never),
socioeconomic status (low, medium, and high); education (elementary and secondary education, high
school, and Bachelor’s degree or higher), family history of diabetes mellitus (yes vs. no), personal
history of hypertension (yes vs. no), medication use (yes vs. no), multivitamin use (yes vs. no), alcohol
intake (gr/day), body mass index (<25.0 vs. ≥25.0 kg/m2 ). 3 Adjusted for age (years), sex, physical
activity (inactive vs. active/highly active), hours of television watching, tobacco use (current, past,
and never), socioeconomic status (low, medium, and high); education (elementary and secondary
education, high school, and Bachelor’s degree or higher), family history of diabetes mellitus (yes vs. no),
personal history of hypertension (yes vs. no), medication use (yes vs. no), multivitamin use (yes vs. no),
alcohol intake (gr/day). 4 Adjusted for age (years), sex, hours of television watching, tobacco use
(current, past, and never); socioeconomic status (low, medium, and high); education (elementary and
secondary education, high school, and Bachelor’s degree or higher), family history of diabetes mellitus
(yes vs. no), personal history of hypertension (yes vs. no), medication use (yes vs. no), multivitamin
use (yes vs. no), alcohol intake (gr/day), body mass index (<25.0 vs. ≥25.0 kg/m2 ).
40
Nutrients 2018, 10, 385
4. Discussion
In the Mexico City Diabetes Mellitus Survey, we observed that participants in the DII
score highest quintile, representing the most pro-inflammatory diet, had higher odds of T2DM
(independent of other diabetes risk factors) compared with participants in the DII score lowest
quintile (maximum anti-inflammatory potential). When we stratified by age (<55 years vs. ≥55 years),
we observed positive associations between DII and T2DM, with larger magnitude of association among
participants ≥55 years of age.
In the present study, we used a previously derived and validated DII score [13] to appraise the
capacity of a pro-inflammatory diet on T2DM. The DII scores computed from a Mexican population
included 27 food parameters, ranged from −5.49 to +4.12 with a mean of −0.68. This result is
consistent with prior publications conducted in Italy, France, and Spain [16,17,29,30]. For example,
Ramallal and colleagues at the University of Navarra cohort study reported a DII score ranging from
−5.14 to +3.97 [30].
We also evaluated associations between food groups and nutrient intake and the DII, and found
that subjects in the highest DII quintile (most pro-inflammatory diet) consumed more red and processed
meat, eggs, refined cereals, and soft drinks, and had a lower intakes of vegetables, fruits, fish,
and seafood. Similar to these results, the “PREvención con DIeta MEDiterránea” (PREDIMED) study
found that consumption of vegetables and fruit was less frequent among men and women in the
highest DII quintile [29].
Regarding nutrient intake, we observed that subjects with the most pro-inflammatory diet
had lower intakes of polyunsaturated fatty acids, fiber, magnesium, and some vitamins. Similar
to our results, the PREDIMED study found that subjects in the highest DII quintile consumed fewer
polyunsaturated fatty acids, vitamins, and fiber. Moreover, other studies using the dietary pattern
or dietary score approach, have observed an inverse association between healthy diets or patterns
(mainly characterized by fruits, vegetables, whole grains and fiber) and inflammation and T2DM,
as well as a positive association with Western patterns or unhealthy diet scores [9,10,30,31].
In the present analysis, we studied the association between DII and T2DM. We observed that
subjects in the highest DII quintile had approximately three times greater odds of T2DM compared
with subjects in the lowest DII quintile. The findings in our analysis agree with prior studies that
evaluate the relation between diet and T2DM. For example, in the Insulin Resistance Atherosclerosis
Cohort study, including subjects with a diet high in red meat, low-fiber cereals, fried potatoes, eggs,
cheese, and low in wine had approximately 4.5 times greater risk of T2DM comparing extreme
quartiles of the dietary pattern [31]. Similar results were observed in the Nurses’ Health Study [32],
where a pattern higher in sugar-sweetened soft drinks, refined grains, and processed meat but low in
vegetables was associated with an increased risk of T2DM (OR = 3.09; 95% CI: 1.99, 4.79; compared to
diet characterized by lower consumption of these dietary components). On the other hand, multiple
prospective studies have reported an inverse association between the adherence to Mediterranean
diet (with anti-inflammatory effects) [33] and the risk of T2DM [34]. Furthermore, a recent study [35]
that evaluated the relation between DII and the risk of prediabetes found that subjects in the highest
DII tertile had higher odds of prediabetes (OR = 18.88; 95% CI: 7.02, 50.82) compared to those who
consumed a more anti-inflammatory diet. Additionally, most of the highly consumed food groups in
the most pro-inflammatory DII quintile (red and processed meat, refined cereals, and soft drinks) have
been associated with T2DM and with inflammatory markers [10]. For example, soft drinks have been
linked to T2DM and inflammatory biomarkers due to their significant contribution to the glycemic
load [36]. Whereas, low intakes of whole grains, fruits, and vegetables in the highest DII quintile
have been related with reduced diabetes risk, probably mediated by a reduction of CRP, and certain
interleukins, and by an improvement in the endothelial function [37].
Other results in the present study indicate that a pro-inflammatory effect of diet on T2DM
could be particularly unfavorable among older (≥55 years) or inactive individuals (<150 min/week).
Although not statistically significant, there was a suggestion of an interaction between DII and
41
Nutrients 2018, 10, 385
overweight/obesity in relation to T2DM. Despite the lack of significance, this result could be
biologically important. Similar results were obtained in the Multiethnic Cohort (MEC) Study. In this
study, the stratified analysis suggests that, among men, the risk for T2DM increased with higher
consumption of fat and meat patterns among overweight (Hazard ratio = 1.49; 95% CI: 1.23, 1.81)
and obese (HR = 1.57; 95% CI: 1.16, 2.12) individuals [38]. In women, similar results were observed,
although these results were not statistically significant. In our case, this finding should be interpreted
with caution; it is difficult to determine, with our study design, the role of BMI in the causal sequence
(i.e., whether it is a confounder or an effect modifier). The relation between DII and T2DM remained
strong after adjustment for BMI, indicating that the DII may be associated with diabetes in all
individuals, but especially among obese subjects.
The connection between DII and T2DM could be explained through the effect of a pro-inflammatory
diet on insulin resistance that has been linked to the inflammatory process [39]. In this sense, prior
studies suggest a positive relationship between markers of chronic inflammation, including CRP, IL-1β,
IL-6, and TNF-α, and insulin resistance [39]. Furthermore, it has been established that insulin resistance
increases levels of these adhesion molecules in diabetic and nondiabetics individuals [40]. On the other
hand, the functional effects of food or food groups and nutrients like meat, processed meat, refined
cereals, and soft drinks, which corresponded with high DII scores in our study, have been also shown
to influence systemic inflammation [32].
Strengths of our study include: (1) the random stratified cluster design of the DMS-MC; (2) the use
of previously validated questionnaires; (3) the inclusion of many potential demographic, behavioral,
and other factors as potential effect modifiers or confounders in the multivariate analysis; and (4)
the use of a validated DII score specially constructed to assess the inflammatory potential of any
diet [12,13]. As reported in other studies [17,29], the DII can be adapted for use in different populations
including the Mexican population providing results that can be compared to those from studies based
in diverse populations in many parts of the world.
Some potential weaknesses of our study need to be highlighted. Because of its cross-sectional
design, this study cannot infer causality. Therefore, these results need to be further investigated in
future longitudinal studies. Although the SFFQ used in this study had been previously validated [20],
other possible limitations are related to information bias. In this regard, it is important to note that
dietary factors used to calculate the DII were evaluated from a single measurement of SFFQ, which is
subject to random error that would tend to underestimate the true association between DII and T2DM
in our study. As observed in previous studies [29], some dietary components such as saffron, thyme,
turmeric, and others were not available to compute the DII in the present analysis; however, as they
are not habitually consumed in large quantities or common in Mexican diet, they may not have
an important impact on the DII score. Another probable weakness could be related to the T2DM
definition; nevertheless, we provided appropriate allowance for the prevalent ascertainment in the
present analysis. In other words, prevalent cases of T2DM (previously diagnosed) were included only
if they were recently diagnosed (less than 11 months), had fasting glucose concentrations ≥126 mg/dL,
and had poor glycemic control (HbA1c ≥ 6.5) at the time of the survey. Additionally, we conducted
a sensitivity analysis including only subjects diagnosed at the time of the survey and we observed
similar results (OR = 3.56; 95% CI: 1.13, 9.11). We adjusted for many potentially confounding factors,
however, residual confounding due to measurement error, mainly in assessing some self-reported
lifestyle variables or due to insufficient control in statistical models, might have produced some degree
of bias in our results. However, it is improbable that such a bias would explain the consistently robust
association observed between DII and T2DM.
5. Conclusions
Our data suggest that a higher DII score (revealing a more pro-inflammatory diet) was associated
with increased odds of T2DM compared with DII scores in the lowest quintile of the DII (indicative of
an anti-inflammatory diet) among participants from the DMS-MC. In addition, we observed that
42
Nutrients 2018, 10, 385
Acknowledgments: This study was possible thanks to an unrestricted research grant obtained by SB from
Novo Nordisk (number Project: 1295. Cities Changing Diabetes: Mexico City Representative Diabetes survey)
and a NIH-Fogarty grant (NIH-Fogarty RO3 TW009061) to the National Institute of Public Health. Shivappa and
Hébert were supported by grant number R44DK103377 from the United States National Institute of Diabetes and
Digestive and Kidney Diseases.
Author Contributions: S.B. and E.D.-G. designed the study; E.D.-G., S.B., L.T.-M. conducted the research; E.D.-G.,
P.M.-A. and N.S. performed the statistical analyses; E.D.-G. wrote the first draft of the manuscript; E.D.-G., P.M.-A.,
N.S., J.R.H., L.T.-M., C.B., and S.B. critically reviewed and commented on the manuscript. All authors read and
approved the final version of the manuscript.
Conflicts of Interest: E. Denova-Gutiérrez, P. Muñoz-Aguirre, L. Tolentino-Mayo, C. Batis, S. Barquera, declare
no conflict of interest. James R. Hébert owns controlling interest in Connecting Health Innovations LLC (CHI),
a company planning to license the right to his invention of the dietary inflammatory index (DIITM ) from the
University of South Carolina in order to develop computer and smart phone applications for patient counseling
and dietary intervention in clinical settings. Dr. Nitin Shivappa is an employee of CHI.
References
1. Calder, P.C.; Albers, R.; Antoine, J.M.; Blum, S.; Bourdet-Sicard, R.; Ferns, G.A.; Folkerts, G.; Friedmann, P.S.;
Frost, G.S.; Guarner, F.; et al. Inflammatory disease processes and interactions with nutrition. Br. J. Nutr.
2009, 101, S1–S45. [CrossRef] [PubMed]
2. Fung, T.T.; McCullough, M.L.; Newby, P.K.; Manson, J.E.; Meigs, J.B.; Rifai, N.; Willett, W.C.; Hu, F.B.
Diet-quality scores and plasma concentrations of markers of inflammation and endotelial dysfunction. Am. J.
Clin. Nutr. 2005, 82, 163–173. [CrossRef] [PubMed]
3. Zimmet, P.Z.; Magliano, D.J.; Herman, W.H.; Shaw, J.E. Diabetes: A 21st century challenge. Lancet Diabetes
Endocrinol. 2014, 2, 56–64. [CrossRef]
4. Kassebaum, N.J.; DALYs and GBD collaborators, 2015. Global, regional, and national disability-adjusted
life-years (DALYs) for 315 diseases and injuries and healthy life expectancy (HALE), 1990–2015: A systematic
analysis for the Global Burden of Disease Study 2015. Lancet 2016, 388, 1603–1658. [CrossRef]
5. Guariguata, L.; Whiting, D.R.; Hambleton, I.; Beagley, J.; Linnenkamp, U.; Shaw, J.E. Global estimates of
diabetes prevalence for 2013 and projections for 2035. Diabetes Res. Clin. Pract. 2014, 103, 137–149. [CrossRef]
[PubMed]
6. Gómez-Dantés, H.; Fullman, N.; Lamadrid-Figueroa, H.; Cahuana-Hurtado, L.; Darney, B.; Avila-Burgos, L.;
Correa-Rotter, R.; Rivera, J.A.; Barquera, S.; González-Pier, E.; et al. Dissonant health transition in the states
of Mexico, 1990–2013: A systematic analysis for the Global Burden of Disease Study 2013. Lancet 2016, 388,
2386–2402. [CrossRef]
7. Visser, M.; Bouter, L.M.; McQuillan, G.M.; Wener, M.H.; Harris, T.B. Elevated C-reactive protein levels in
overweight and obese adults. JAMA 1999, 282, 2131–2135. [CrossRef] [PubMed]
8. Petersen, A.M.; Pedersen, B.K. The anti-inflammatory effect of exercise. J. Appl. Physiol. 2005, 98, 1154–1162.
[CrossRef] [PubMed]
9. Calder, P.C.; Ahluwalia, N.; Brouns, F.; Buetler, T.; Clement, K.; Cunningham, K.; Esposito, K.; Jönsson, L.S.;
Kolb, H.; Lansink, M.; et al. Dietary factors and low-grade inflammation in relation to overweight and
obesity. Br. J. Nutr. 2011, 106, S5–S78. [CrossRef] [PubMed]
10. Schulze, M.B.; Hu, F.B. Primary prevention of diabetes: What can we done and how much can be prevented?
Annu. Rev. Public Health 2005, 26, 445–467. [CrossRef] [PubMed]
11. Fung, T.T.; Schulze, M.; Manson, J.E.; Willett, W.C.; Hu, F.B. Dietary patterns, meat intake, and the risk of
type 2 diabetes in women. Arch. Intern. Med. 2004, 164, 2235–2240. [CrossRef] [PubMed]
12. Shivappa, N.; Steck, S.E.; Hurley, T.G.; Hussey, J.R.; Hébert, J.R. Designing and developing a literature-
derived, population-based dietary inflammatory index. Public Health Nutr. 2014, 17, 1689–1696. [CrossRef]
[PubMed]
43
Nutrients 2018, 10, 385
13. Shivappa, N.; Steck, S.E.; Hurley, T.G.; Hussey, J.R.; Ma, Y.; Ockene, I.S.; Tabung, F.; Hébert, J.R. A population
based dietary inflammatory index predicts levels of C-reactive protein in the seasonal variation of blood
cholesterol study (SEASONS). Public Health Nutr. 2014, 17, 1825–1833. [CrossRef] [PubMed]
14. Shivappa, N.; Hébert, J.R.; Rietzschel, E.R.; De Buyzere, M.L.; Langlois, M.; Debruyne, E.; Marcos, A.;
Huybrechts, I. Associations between dietary inflammatory index and inflammatory markers in the Asklepios
Study. Br. J. Nutr. 2015, 113, 665–671. [CrossRef] [PubMed]
15. Alkerwi, A.; Shivappa, N.; Crichton, G.; Hébert, J.R. No significant independent relationships with
cardiometabolic biomarkers were detected in the Observation of Cardiovascular Risk Factors in Luxemburgo
Study population. Nutr. Res. 2014, 34, 1058–1065. [CrossRef] [PubMed]
16. Shivappa, N.; Zucchetto, A.; Montella, M.; Serraino, D.; Steck, S.E.; La Vecchia, C.; Hébert, J.R. Inflammatory
potential of diet and risk of colorectal cancer: A case-control study from Italy. Br. J. Nutr. 2015, 114, 152–158.
[CrossRef] [PubMed]
17. Neufcourt, L.; Assmann, K.E.; Fezeu, L.K.; Touvier, M.; Graffouillère, L.; Shivappa, N.; Hébert, J.R.;
Wirth, M.D.; Hercberg, S.; Galan, P.; et al. Prospective association between the dietary inflammatory index
and cardiovascular diseases in the SUpplémentation en VItamines et Minéraux AntioXydants (SU.VI.MAX)
cohort. J. Am. Heart Assoc. 2016, 5, e002735. [CrossRef] [PubMed]
18. Romero-Martínez, M.; Shamah-Levy, T.; Franco-Nuñez, A.; Villalpando, S.; Cuevas-Nasu, L.; Gutiérrez, J.P.;
Rivera-Dommarco, J.Á. National Health and Nutrition Survey 2012: Design and coverage. Salud Publica Mex.
2013, 55, S332–S340. [CrossRef] [PubMed]
19. Rosner, B.; Willett, W.C. Interval estimates for correlation coefficients corrected for within-person variation:
Implications for study design and hypothesis testing. Am. J. Epidemiol. 1988, 127, 377–386. [CrossRef] [PubMed]
20. Denova-Gutiérrez, E.; Ramírez-Silva, I.; Rodríguez-Ramírez, S.; Jiménez-Aguilar, A.; Shamah-Levy, T.;
Rivera-Dommarco, J.A. Validity of a food frequency questionnaire to assess food intake in Mexican adolescent
and adult population. Salud Publica Mex. 2016, 58, 617–628. [CrossRef] [PubMed]
21. Hernández-Ávila, J.E.; González-Avilés, L.; Rosales-Mendoza, E. Manual de Usuario. SNUT Sistema de
Evaluación de Hábitos Nutricionales y Consumo de Nutrimentos; Instituto Nacional de Salud Pública: Cuernavaca,
México, 2003. (In Spanish)
22. Hebert, J.R.; Ockene, I.S.; Hurley, T.G.; Luippold, R.; Well, A.D.; Harmatz, M.G. Development and testing of
a seven-day dietary recall. J. Clin. Epidemiol. 1997, 50, 925–937. [CrossRef]
23. Twetz, N. Textbook of Clinical Chemistry; WB Saunders: Philadelphia, PA, USA, 1999.
24. American Diabetes Association. Standards of medical care in diabetes-2014. Diabetes Care 2014, 37, S14–S80.
[CrossRef]
25. Alberti, K.G.; Zimmet, P.; Shaw, J. The metabolic syndrome: A new worldwide definition. Lancet 2005, 366,
1059–1062. [CrossRef]
26. Martinez, S.M.; Ainsworth, B.E.; Elder, J.P. A review of physical activity measures used among US Latinos:
Guidelines for developing culturally appropriate measures. Ann. Behav. Med. 2008, 36, 195–207. [CrossRef]
[PubMed]
27. International Physical Activity Questionnaire. Guidelines for Data Processing and Analysis of the International
Physical Activity Questionnaire (IPAQ)-Short and Long Forms. Available online: http://www.ipaq.ki.se/
scoring.pdf (accessed on 30 May 2016).
28. World Health Organization. Global Recommendations on Physical Activity for Health. Available online:
http://whqlibdoc.who.int/publications/2010/9789243599977_spa.pdf (accessed on 29 May 2016).
29. Ruiz-Canela, M.; Zazpe, I.; Shivappa, N.; Hébert, J.R.; Sánchez-Tainta, A.; Corella, D.; Salas-Salvadó, J.;
Fitó, M.; Lamuela-Raventós, R.M.; Rekondo, J.; et al. Dietary inflammatory index and anthropometric
measures of obesity in a population sample at high cardiovascular risk from the PREDIMED (PREvención
con DIeta MEDiterránea) trial. Br. J. Nutr. 2015, 113, 984–995. [CrossRef] [PubMed]
30. Ramallal, R.; Toledo, E.; Martínez-González, M.A.; Hernández-Hernández, A.; García-Arellano, A.;
Shivappa, N.; Hébert, J.R.; Ruiz-Canela, M. Dietary inflammatory index and incidence of cardiovascular
disease in the SUN Cohort. PLoS ONE 2015, 10, e0135221. [CrossRef] [PubMed]
31. Liese, A.D.; Weis, K.E.; Schulz, M.; Tooze, J.A. Food intake patterns associated with incident type 2 diabetes.
Diabetes Care 2009, 32, 263–268. [CrossRef] [PubMed]
44
Nutrients 2018, 10, 385
32. Schulze, M.B.; Hoffmann, K.; Manson, J.E.; Willett, W.C.; Meigs, J.B.; Weikert, C.; Heidemann, C.;
Colditz, G.A.; Hu, F.B. Dietary pattern, inflammation, and incidence of type 2 diabetes in women. Am. J.
Clin. Nutr. 2005, 82, 675–684. [CrossRef] [PubMed]
33. Casas, R.; Sacanella, E.; Urpí-Sardà, M.; Chiva-Blanch, G.; Ros, E.; Martínez-González, M.A.; Covas, M.I.;
Lamuela-Raventos, R.M.; Salas-Salvadó, J.; Fiol, M.; et al. The effects of the Mediterranean diet on biomarkers
of vascular wall inflammation and plaque vulnerability in subjects with high risk for cardiovascular disease.
A randomize trial. PLoS ONE 2014, 9, e100084. [CrossRef] [PubMed]
34. Mozaffarian, D.; Marfisi, R.; Levantesi, G.; Silletta, M.G.; Tavazzi, L.; Tognoni, G.; Valagussa, F.; Marchioli, R.
Incidence of new-onset diabetes and impaired fasting glucose in patients with recent myocardial infarction
and the effect of clinical and lifestyle risk factors. Lancet 2007, 370, 667–675. [CrossRef]
35. Vahid, F.; Shivappa, N.; Karamati, M.; Naeini, A.J.; Hebert, J.R.; Davoodi, S.H. Association between Dietary
Inflammatory Index (DII) and risk of prediabetes: A case-control study. Appl. Physiol. Nutr. Metab. 2017, 42,
399–404. [CrossRef] [PubMed]
36. Schulze, M.B.; Manson, J.E.; Ludwig, D.S.; Colditz, G.A.; Stampfer, M.J.; Willett, W.C.; Hu, F.B. Sugar-sweetend
beverages, weight gain, and incidence of type 2 diabetes in young and middle-aged women. JAMA 2004, 292,
927–934. [CrossRef] [PubMed]
37. Whincup, P.H.; Donin, A.S. Cereal fibre and type 2 diabetes: Time now for randomised control trials?
Diabetologia 2015, 58, 1383–1385. [CrossRef] [PubMed]
38. Erber, E.; Hopping, B.N.; Grandinetti, A.; Park, S.Y.; Kolonel, L.N.; Maskarinec, G. Dietary pattern and risk
for diabetes. The multiethnic cohort. Diabetes Care 2010, 33, 532–538. [CrossRef] [PubMed]
39. Festa, A.; D’Agostino, R., Jr.; Howard, G.; Mykkänen, L.; Tracy, R.P.; Haffner, S.M. Chronic subclinical
inflammation as part of the insulin resistance syndrome: The Insulin ResistanceAtherosclerosis Study (IRAS).
Circulation 2000, 102, 42–47. [CrossRef] [PubMed]
40. Gearing, A.J.; Newman, W. Circulating adhesion molecules in disease. Immunol. Today 1993, 14, 506–512.
[CrossRef]
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
45
nutrients
Article
Effects of Virgin Olive Oils Differing in Their
Bioactive Compound Contents on Metabolic
Syndrome and Endothelial Functional Risk
Biomarkers in Healthy Adults: A Randomized
Double-Blind Controlled Trial
Estefania Sanchez-Rodriguez 1 , Elena Lima-Cabello 2 , Sara Biel-Glesson 3 ,
Jose R. Fernandez-Navarro 3 , Miguel A. Calleja 3 , Maria Roca 4 , Juan A. Espejo-Calvo 5 ,
Blas Gil-Extremera 6 , Maria Soria-Florido 7 , Rafael de la Torre 8 , Montserrat Fito 7,9 ,
Maria-Isabel Covas 9,10 , Juan de Dios Alche 2 , Emilio Martinez de Victoria 11 , Angel Gil 1,9, * and
Maria D. Mesa 1
1 Department of Biochemistry and Molecular Biology II, Institute of Nutrition and Food Technology
“José Mataix”, Biomedical Research Center, University of Granada, Parque Tecnológico de la Salud,
Avenida del Conocimiento s/n, 18100 Armilla, Granada, Spain; [email protected] (E.S.-R.);
[email protected] (M.D.M.)
2 Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín (CSIC),
Profesor Albareda 1, 18008 Granada, Spain; [email protected] (E.L.-C.);
[email protected] (J.d.D.A.)
3 Fundación Pública Andaluza para la Investigación Biosanitaria de Andalucía Oriental “Alejandro Otero”
(FIBAO), Avenida de Madrid 15, 18012 Granada, Spain; sbiel@fibao.es (S.B.-G.);
[email protected] (J.R.F.-N.); [email protected] (M.A.C.)
4 Food Phytochemistry Department, Instituto de la Grasa, Consejo Superior de Investigaciones Científicas
(CSIC), University Campus Pablo de Olavide, 41013 Sevilla, Spain; [email protected]
5 Instituto para la Calidad y Seguridad Alimentaria (ICSA), Avenida de la Hispanidad 17, 18320 Santa Fe,
Granada, Spain; [email protected]
6 Department of Medicine, University of Granada, Avenida de la Investigación 11, 18071 Granada, Spain;
[email protected]
7 Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), Dr.
Aiguader 88, 08003 Barcelona, Spain; [email protected] (M.S.-F.); mfi[email protected] (M.F.)
8 Integrative Pharmacology and Systems Neuroscience Research Group, IMIM (Hospital del Mar Research
Institute), Universitat Pompeu Fabra (CEXS-UPF), Dr. Aiguader 88, 08003 Barcelona, Spain; [email protected]
9 Spanish Biomedical Research Networking Centre, Physiopathology of Obesity and Nutrition (CIBEROBN),
Instituto de Salud Carlos III, Monforte de Lemos 3-5, 28029 Madrid, Spain; [email protected]
10 NUPROAS Handelsbolag, Nackã, Sweden, NUPROAS HB, Apartado de Correos 93, 17242 Girona, Spain
11 Department of Physiology, Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research
Center, University of Granada, Parque Tecnológico de la Salud, Avenida del Conocimiento s/n,
18100 Armilla, Granada, Spain; [email protected]
* Correspondence: [email protected]; Tel.: +34-958-246-187
Abstract: The aim of this study was to evaluate the effect of virgin olive oils (VOOs) enriched with
phenolic compounds and triterpenes on metabolic syndrome and endothelial function biomarkers
in healthy adults. The trial was a three-week randomized, crossover, controlled, double-blind,
intervention study involving 58 subjects supplemented with a daily dose (30 mL) of three oils:
(1) a VOO (124 ppm of phenolic compounds and 86 ppm of triterpenes); (2) an optimized VOO
(OVOO) (490 ppm of phenolic compounds and 86 ppm of triterpenes); and (3) a functional olive oil
(FOO) high in phenolic compounds (487 ppm) and enriched with triterpenes (389 ppm). Metabolic
syndrome and endothelial function biomarkers were determined in vivo and ex vivo. Plasma high
density lipoprotein cholesterol (HDLc) increased after the OVOO intake. Plasma endothelin-1 levels
decreased after the intake of the three olive oils, and in blood cell cultures challenged. Daily intake
of VOO enriched in phenolic compounds improved plasma HDLc, although no differences were
found at the end of the three interventions, while VOO with at least 124 ppm of phenolic compounds,
regardless of the triterpenes content improved the systemic endothelin-1 levels in vivo and ex vivo.
No effect of triterpenes was observed after three weeks of interventions. Results need to be confirmed
in subjects with metabolic syndrome and impaired endothelial function (Clinical Trials number
NCT02520739).
Keywords: olive oil; virgin olive oil; olive oil polyphenols; maslinic acid; oleanolic acid;
cardiovascular diseases; endothelial function; phenolic compounds; triterpenes; metabolic syndrome
1. Introduction
Metabolic syndrome (MS) is a cluster of associated metabolic and clinical disturbances that
tend to occur together [1]. This syndrome is commonly represented by the combination of obesity
(particularly abdominal), hyperglycemia, dyslipidemia, and hypertension [2]. MS is associated with
an increased risk of cardiovascular disease (CVD), which is the main cause of disability and mortality
in industrialized countries, and is associated with a chronic inflammatory response characterized by
abnormal cytokine production leading to endothelial dysfunction [3].
There is consolidated clinical evidence that the Mediterranean diet (MD) is associated with
a lower risk of CVDs, including myocardial infarction, stroke and cardiovascular death [4].
Mayneris-Perxachs et al. [5] proposed the MD as a successful tool for the prevention and treatment of
MS and related comorbidities. A large study in a high cardiovascular risk population has found that the
MD supplemented with virgin olive oil (VOO) protects people from vascular disease, suggesting a key
role of olive oil [6]. A recent systematic review and meta-analysis provided evidence that olive oil might
exert beneficial effects on endothelial function and biomarkers of inflammation, thus representing a key
ingredient contributing to the MD cardiovascular-protective effects [7]. Olive oil is not only a source of
monounsaturated fatty acids (MUFAs) but also an important source of bioactive compounds, such as
phenols and triterpenes [8,9]. Previous studies suggested a protective effect of olive oil phenolic
compounds on endothelial dysfunction [10], whereas olive oil triterpenes could be useful for the
prevention of multiple diseases related to cell oxidative damage [11]. Maslinic and oleanolic acids
are the principal triterpenes found in VOO. The potential of these olive oil triterpenic acids for use as
a therapeutic strategy to improve vascular function and treating CVD has been recently reviewed [12].
However, to our best knowledge, no clinical trial has been performed to provide evidence of their
benefits in healthy adults, according to EFSA requirement [13]. The present study which acronyms is
NUTRAOLEUM, aimed to evaluate the effect of VOO enriched with bioactive compounds, such as
phenolic compounds and triterpenes, on MS and endothelial function biomarkers in healthy adults.
We reported that daily intake of VOO enriched in phenolic compounds during three weeks improved
plasma high density lipoprotein cholesterol levels (HDLc), one of the features of metabolic syndrome,
although no differences were found at the end of the three interventions. In addition, VOO with at
least 124 ppm of phenolic compounds, regardless of the triterpenes content, improved the systemic
endothelin-1 levels in vivo and ex vivo, while no additional effect of triterpenes was observed.
2.1. Subjects
Information about subjects, sample size and their eligibility and dietary control have been
referenced in detail elsewhere [14]. In brief, fifty-eight intention-to-treat subjects were eligible.
47
Nutrients 2018, 10, 626
The inclusion criteria were as follows: people in good health on the basis of a physical examination and
basic biochemical and hematological analyses, and willingness to provide written informed consent.
The exclusion criteria were as follows: smoking, intake of antioxidant supplements, aspirin or any
other drug with established antioxidant properties, hyperlipidemia, obesity (body mass index (BMI)
>30 kg/m2 ), diabetes, hypertension, celiac or other intestinal disease, any condition limiting mobility,
life-threatening diseases, or any other disease or condition that would impair compliance. Five subjects
declined to participate for personal reasons before olive oil type allocation. Fifty-three subjects (27 men
and 26 women) aged from 20 to 50 years from the general population of Granada were enrolled in the
study from February 2014 to July 2014 and were assigned into groups. Two subjects did not complete
the study. After the first intervention, one subject refused to continue for personal reasons, and the
other did not follow the protocol correctly. At the end of the experimental period, 51 subjects remained
in the study (Figure 1) [14]. All subjects provided written informed consent according to the principles
of the Declaration of Helsinki, and the local institutional review board, the Ethics Committee Research
Centre of Granada, approved the protocol (13/11 C38).
Figure 1. CONSORT Based Flow Diagram of the recruitment, enrollment and randomization process.
48
Nutrients 2018, 10, 626
San Francisco de Asís Cooperative (Montefrío (Granada), Spain) provided the olive oils used in the
present study. Daily doses of 30 mL of the three types of raw olive oils, as recommended by the
US Food and Drug Administration [15], were distributed over three meals. Olive oils were blindly
prepared in special containers; the three types of olive oil were labeled “A”,”B”, and “C”. Containers
with the corresponding 30 mL olive oil daily dose and enough amount of the same oil for cooking
during each intervention period were delivered to the subjects at the beginning of each intervention
period. The subjects were randomly assigned to three orders of administration of olive oil, paired
by gender and age, using the block-randomization method of a software program for sequence
generation [14]. The randomization lists were concealed in a lightproof sealed envelope. The sealed
envelopes were kept by the independent statistician during the study, avoiding the breaking of the
seal. Thus, the subjects, investigators, and outcome assessors were blinded and could not foresee
the treatment allocation throughout the study. Hence, blinding of outcome assessment was also
ensured. According to previous published studies, olive oils were sequentially administered over three
periods of 3 weeks [16] preceded by two weeks of washout periods [17] during which the subjects
were requested to avoid olives and olive oil consumption. A nutritionist personally advised subjects
on replacing all types of habitually consumed raw fats using only the assigned oil. The advantage
of using a crossover design is that each subject served as his or her own control. Olive oils were
specially prepared for the trial and differed only in phenolic compound and triterpenes contents
(Table 1). The oils were prepared in dark, sealed containers and were similar in appearance and
color, thus ensuring blinding of the subjects and study personnel. The trial has been registered at
ClinicalTrials.gov ID: NCT02520739.
49
Nutrients 2018, 10, 626
50
Nutrients 2018, 10, 626
(CV): 10.3%), resistin (CV: 7.7%), soluble intercellular adhesion molecule (sICAM-1) (CV: 6.1%) and
soluble vascular adhesion molecule (sVCAM-1) (CV: 5.4%), (Cat. #HADK1MAG-61K).
Endothelin-1 was determined as a secondary outcome by ELISA (CV): 7.2%) (R&D Systems,
Minneapolis, MN, USA; Cat. DET100) in both plasma and whole blood culture supernatants.
3. Results
51
Nutrients 2018, 10, 626
Table 2. Clinical and biochemical characteristics of subjects at baseline according to olive oil
administration sequence.
Table 3. Average daily energy and selected nutrient intake of subjects at baseline according to olive oil
administration sequence.
52
Nutrients 2018, 10, 626
Table 4. Average daily energy and selected nutrient intake of subjects after the three olive
oil interventions.
53
Table 5. Metabolic syndrome and endothelial function biomarkers before and after each olive oil intervention in healthy adults.
Waist circumference, cm 77.4 ± 0.7 77.2 ± 0.7 77.2 ± 0.7 76.8 ± 0.7 77.4 ± 0.7 76.9 ± 0.7
HDLc, mg/dL 58 ± 2 59 ± 2 57 ± 2 60 ± 2 * 58 ± 2 60 ± 2
Males 52 ± 2 53 ± 2 52 ± 2 52 ± 2 52 ± 2 54 ± 2
Females 64 ± 2 65 ± 2 63 ± 2 67 ± 2 * 65 ± 2 66 ± 2
LDLc, mg/dL 105 ± 4 108 ± 4 a,b 104 ± 4 106 ± 4 a 104 ± 4 111 ± 4 b
Total cholesterol, mg/dL 179 ± 5 182 ± 5 177 ± 5 183 ± 5 178 ± 5 186 ± 5 *
Triacylglycerols, mg/dL 72 ± 7 75 ± 7 * 74 ± 7 81 ± 7 * 75 ± 7 75 ± 7
Glucose, mg/dL 91 ± 2 91 ± 2 91 ± 2 91 ± 2 90 ± 2 91 ± 2
Adiponectin, mg/L 13.74 ± 1.7 12.8 ± 1.7 12.48 ± 1.7 13.6 ± 1.71 12.62 ± 1.71 13.74 ± 1.71
Resistin, μg/L 14.1 ± 1.1 14.1 ± 1.1 14.5 ± 1.1 13.8 ± 1.1 13.8 ± 1.1 14.1 ± 1.1
SBP, mmHg 117 ± 3 115 ± 3 *,a 119 ± 3 116 ± 3 a,b 114 ± 3 118 ± 3 *,b
DBP, mmHg 72 ± 2 72 ± 2 75 ± 2 73 ± 2 72 ± 2 74 ± 2
Pulse pressure, mmHg 45 ± 1 43 ± 1 44 ± 1 42 ± 1 42 ± 1 45 ± 1
Endothelin-1, pg/mL 1.53 ± 0.15 1.38 ± 0.15 * 1.58 ± 0.15 1.41 ± 0.15 * 1.49 ± 0.15 1.35 ± 0.15 *
54
sICAM-1, ng/mL 68.09 ± 3.19 67.11 ± 3.19 65.17 ± 3.20 67.73 ± 3.19 65.1 ± 3.20 66.7 ± 3.21
sVCAM-1, ng/mL 451 ± 22 443 ± 23 435 ± 22 451 ± 22 431 ± 22 442 ± 23
Values are expressed as the adjusted means ± SEMs. LMM was used to compare pre-intervention vs. post-interventions with each oil data, and data after the three interventions
(post-interventions). * Significant differences between pre-intervention vs. post-intervention data within each intervention with the three olive oils. Different superscript letters indicate
significant differences between post-intervention results (a,b ). p < 0.05 was considered significant. DBP, diastolic blood pressure; FOO, functional olive oil; HDLc, high density lipoprotein
cholesterol; LDLc, low density lipoprotein cholesterol; OVOO, optimized virgin olive oil; SBP, systolic blood pressure; SEM, standard error of the mean; sICAM-1, soluble intercellular
adhesion molecule; sVCAM-1, soluble vascular cell adhesion molecule; VOO, virgin olive oil.
Nutrients 2018, 10, 626
Figure 2. Plasma endothelin-1 ex vivo changes (post-intervention minus pre-intervention data) when
stimulated with PHA, LPS or PMA + IO in whole blood cultures from healthy adults. Values are
expressed as the means ± SEMs. ANOVA was used to compare differences between interventions
and induction treatments. The Tukey post-hoc test was used for multiple comparisons among groups.
p < 0.05 was considered significant. FOO, functional olive oil; IO, ionomycin; OVOO, optimized virgin
olive oil; PHA, phytohemagglutinin; PMA, phorbol 12-myristate 13-acetate; VOO, virgin olive oil.
Supplemental Figure S1 shows the supernatant endothelin-1 concentrations that were induced
ex vivo with PHA, LPS, and PMA + IO in whole blood cultures from the subjects before and after the
three interventions. After the VOO, OVOO, and FOO interventions, challenging with LPS or PMA
+ IO induced a significantly lower increase of endothelin-1 secretion in whole blood cultures, while
challenging with PHA induced a significantly lower increase of endothelin-1 secretion only after the
OVOO and FOO interventions.
55
Nutrients 2018, 10, 626
about four times higher than those recovered after the VOO and OVOO interventions (p = 0.004 and
p < 0.001, respectively, for maslinic acid, and p = 0.026 and p < 0.001, respectively, for oleanolic acid).
Urine concentrations of total hydroxytyrosol (the sum of hydroxytyrosol and its glucuronide and
sulfate conjugates) were higher after the OVOO (2444 mM, p = 0.003) and FOO (2876 mM, p = 0.002)
interventions than after the VOO (115 mM, p = 0.011) intervention.
Figure 3. In vivo urinary triterpenes changes (post-intervention minus pre-intervention data) for each
olive oil intervention in healthy adults. Values are expressed as the means ± SEMs. ANOVA was
used to compare differences between the three interventions. Different superscript letters indicate
significant differences between the interventions for oleanolic acid (a,b ) and for maslinic acid (c,d ).
p < 0.05 was considered significant. FOO, functional olive oil; OVOO, optimized virgin olive oil; VOO,
virgin olive oil.
4. Discussion
The NUTRAOLEUM study is the first human nutritional clinical trial concerning the effects of
VOO that is high in phenolic compounds and enriched with triterpenes, maslinic and oleanolic acids,
from olive exocarp, on MS features and endothelial function risk biomarkers in comparison with
standard VOO in healthy subjects.
The PREDIMED study reported that a MD supplemented with at least 50 g of dietary VOO
caused a reversion of MS after a median follow-up of 4.8 years [25], reducing the rate of CVD events
by 30% compared with a low-fat diet control group [26]. In addition, this MD enriched with VOO
and without energy restrictions reduced the diabetes risk among individuals at high cardiovascular
risk [27]. Consumption of VOO close to 2.7, 164 or 366 ppm/day of phenolic compounds in humans,
a 0.03% of hydroxytyrosol in a rodent model, and 50 ppm/day of hydroxytyrosol in a murine model
have been reported to improve the blood lipid profile, although results in mice were not conclusive [28],
possibly due to differences in the phenolic content of the olive oil and the physio-pathology of the
studied animals. Polyphenols from olive oils have been shown to provide additional benefits on
HDLc, other than those provided by the MUFA content. However, contradictory data exists on these
benefits. In 2015, a meta-analysis reported no effect on HDLc concentration after the intake of VOO
with at least 150 ppm of phenolic compounds [29], while, in accordance with our results, a recent
systematic review [30] concluded that plasma HDLc was increased in different studies consuming
from 2.28 to 75 g/day of olive oil. In the Eurolive Study, a European multicenter study, three olive oils
(refined, medium and high) differing in their phenolic compound content (2.7, 164 and 366 ppm/day of
phenolic content, respectively) increased HDLc and decreased triacylglycerol concentrations [16]. The
increase in HDLc was linear with the olive oil polyphenol content. This additional benefit has also been
56
Nutrients 2018, 10, 626
described in healthy and hypercholesterolemic subjects treated with polyphenol-enriched VOO [31]
when excluding patients treated with hypolipidemic medication. Additionally, the increase of olive oil
polyphenols in the lipoprotein fraction may increase HDL size, stability, and antioxidant status [32].
It has been reported that the daily intake of olive oils enriched with its own polyphenols (250 ppm
or 500 ppm), as well as the intake of olive oil enriched in polyphenols from thyme (250 ppm/day)
decrease LDLc and improve the lipoprotein subclass distribution and associated ratios [33]. Other
authors have described that a one-year intervention with a MD enriched with VOO improved several
LDL characteristics related to its atherogenicity (resistance against oxidation, size, composition, and
cytotoxicity) but did not modify the plasma LDLc concentrations in a subsample of subjects at high
cardiovascular risk in the PREDIMED study [34]. Our results are consistent with this null effect of
VOO on LDLc. On the other hand, besides the weak increase in plasma triacylglycerols observed
after interventions with both VOO and OVOO, this was without clinical significance given that levels
were low at baseline and remain low at the end of the three interventions, and thus it did not increase
cardiovascular risk in these healthy subjects (triacylglycerols < 150 mg/dL) [35]. Our results are
in contrast with the triacylglycerol improvement reported in the EUROLIVE Study [16]. A recent
meta-analysis and systematic review focused on high polyphenol VOO concluded no effect on plasma
triacylglycerol concentrations [29]; in addition, Saibandith et al. [36] stated that the effect of olive oil
polyphenols on plasma triacylglycerols remained unclear. However, longer studies are needed to
clarify this issue.
To the best of our knowledge, this is the first clinical trial evaluating the effect of olive
oil triterpenes on plasma lipids and endothelial function biomarkers. Previous animal studies
have reported different effects of oleanolic acid on plasma lipids, depending on the experimental
animal model [37]. Based on our results, FOO enriched with triterpenes did not modify plasma
HDLc, LDLc, or triacylglycerols, but increased plasma total cholesterol concentrations, although
no inter-group significance was found. However, although these modifications do not represent
an increase in cardiovascular risk since the values are low enough to be considered safe (total
cholesterol < 190 mg/dL) [35], further human clinical trials are needed to demonstrate the potential
benefit of olive phenolic compounds and triterpenes on plasma lipid concentrations over longer
intervention periods.
Endothelial dysfunction is a critical early event in the development of atherosclerosis [38].
An imbalance between vasodilating and vasoconstricting molecules, such as nitric oxide and
endothelin-1, respectively, contributes to the pathogenesis of hypertension and its complications [39].
Our results indicate that plasma endothelin-1 levels decreased after the VOO, OVOO and FOO
interventions, and these results were also confirmed throughout ex vivo blood culture experiments.
Although endothelin-1 is mainly produced from vascular endothelial cell, several studies have
suggested that blood cells, such as polymorphonuclear neutrophils [40] and T-Cells [41], and also
macrophages [42] are responsible from circulating levels of endothelin-1. In addition, ex vivo
experiment has demonstrated the release of the mature peptide after LPS and LPS + PMA
stimulation [40]; Mencarelli et al. [43] have confirmed these results. As demonstrated here, endothelin-1
production was decreased after the three interventions. This effect may be beneficial for cardiovascular
risk affected people, since the effect of endothelin-1 has been documented on endothelial and
inflammatory cells, which contributes to pathophysiological processes such as vascular hypertrophy,
cell proliferation, fibrosis and inflammation [44–46]. In humans, the consumption of VOO has shown
benefits on blood pressure and endothelial function [47]. In agreement with our results, a meta-analysis
stated that olive oils with at least 150 ppm of phenolic compounds exert a moderate effect on lowering
SBP and no effects on DBP [29]. Regarding triterpenes, besides the weak SBP increase observed after
this intervention, no clinical significance was observed as blood pressure remained under 130 mmHg
and did not increase cardiovascular risk [35]. It is reported that SBP varies to a greater degree than
DBP [48,49]. For this reason, we calculated pulse pressure and found no significant effect of FOO.
A beneficial effect of triterpenes on endothelial function [50] and blood pressure [51] has been described
57
Nutrients 2018, 10, 626
in animal models of hypertension, but further studies are required to explore the mechanisms involved
in the effect of specific components of VOO on endothelin-1 regulation.
In vitro studies showed that minor olive oil components, specifically hydroxytyrosol and its
metabolites, down-regulate the secretion of E-selectin, p-selectin, sICAM-1, and sVCAM-1, affecting
endothelial function [52]. However, and in agreement with our results, no differences in sICAM-1 or
sVCAM-1 plasma concentrations were reported after 50 mL of VOO or refined olive oil consumption
containing (161 or 14.67 ppm/day of phenolic compounds, respectively) in coronary heart disease
patients [53]. Another study found a significant reduction in sICAM-1 but not in sVCAM-1
concentrations after the intake of 50 mL/day of olive oil [54]. Recently, it has been proposed that the
intake of 25 mL/day of VOO containing 366 ppm of phenolic compounds modulates the expression
of several genes related to the renin-angiotensin-aldosterone system [55]. Therefore, further longer
studies are needed to reach final conclusions about the effect of VOO minor compounds on molecules
modulating endothelial function.
One of the limitations of the present study is that young and healthy subjects are recruited as
target population. The present intervention does not appear sufficient to draw definitive conclusions.
Therefore, new studies in older subject affected by metabolic syndrome and endothelial dysfunction
would be interesting in order to evaluate the beneficial effect of VOO components. Although no
differences in dietary intakes were observed during the three interventions, measurements of dietary
intake relied on self-reporting and were therefore subjective. Another limitation of this study is that
dietary records for the washout periods were not recorded, thus we cannot analyzed dietary intakes
during these periods. In addition, our subjects live in the south of Spain, where MD and VOO are
highly consumed. Therefore, a 3-week intervention is not long enough to cause significant changes.
Further studies are required to find conclusions related to the bioactive compounds presents in olive
oils, and to explore the mechanisms involved in these effects of specific components of VOO on MS
and endothelial function.
5. Conclusions
In conclusion, olive oil rich in polyphenols increased HDLc levels in females, although no
differences were found at the end of the three interventions, and improved an endothelial function
biomarker both in vivo and ex vivo. No additional benefits were obtained from triterpenes VOO
enrichment after 3-wk supplementation. However, further longer studies are warranted on olive
oil triterpenes and their health benefits in older subjects particularly in those affected by metabolic
syndrome and endothelial dysfunction.
58
Nutrients 2018, 10, 626
Conflicts of Interest: The authors declare no conflict of interest. The founding sponsors had no role in the design
of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, and in the
decision to publish the results.
Appendix A
References
1. Sarafidis, P.A.; Nilsson, P.M. The metabolic syndrome: A glance at its history. J. Hypertens. 2006, 24, 621–626.
[CrossRef] [PubMed]
2. Alberti, K.G.; Zimmet, P.; Shaw, J. The metabolic syndrome—A new worldwide definition. Lancet 2005, 366,
1059–1062. [CrossRef]
3. Freitas Lima, L.C.; Braga, V.A.; do Socorro de França Silva, M.; Cruz, J.C.; Sousa Santos, S.H.;
de Oliveira Monteiro, M.M.; Balarini, C.M. Adipokines, diabetes and atherosclerosis: An inflammatory
association. Front. Physiol. 2015, 6, 1–15. [CrossRef] [PubMed]
4. Gardener, H.; Wright, C.B.; Gu, Y.; Demmer, R.T.; Boden-Albala, B.; Elkind, M.S.; Sacco, R.L.; Scarmeas, N.
Mediterranean-style diet and risk of ischemic stroke, myocardial infarction, and vascular death: The Northern
Manhattan Study. Am. J. Clin. Nutr. 2011, 94, 1458–1464. [CrossRef] [PubMed]
5. Mayneris-Perxachs, J.; Sala-Vila, A.; Chisaguano, M.; Castellote, A.I.; Estruch, R.; Covas, M.I.; Fitó, M.;
Salas-Salvadó, J.; Martínez-González, M.A.; Lamuela-Raventós, R.; et al. Effects of 1-Year Intervention with
a Mediterranean Diet on Plasma Fatty Acid Composition and Metabolic Syndrome in a Population at High
Cardiovascular Risk. PLoS ONE 2014, 9, e85202. [CrossRef] [PubMed]
59
Nutrients 2018, 10, 626
6. Estruch, R.; Ros, E.; Salas-Salvadó, J.; Covas, M.; Corella, D.; Arós, F.; Gómez-Gracia, E.; Ruiz-Gutiérrez, V.;
Fiol, M.; Lapetra, J.; et al. Primary prevention of cardiovascular disease with a Mediterranean diet. N. Engl.
J. Med. 2013, 368, 1279–1290. [CrossRef] [PubMed]
7. Schwingshackl, L.; Christoph, M.; Hoffmann, G. Effects of Olive Oil on Markers of Inflammation and
Endothelial Function-A Systematic Review and Meta-Analysis. Nutrients 2015, 7, 7651–7675. [CrossRef]
[PubMed]
8. Covas, M.I.; de la Torre, K.; Farré-Albaladejo, M.; Kaikkonen, J.; Fitó, M.; López-Sabater, C.; Pujadas-Bastardes, M.A.;
Joglar, J.; Weinbrenner, T.; Lamuela-Raventós, R.M.; et al. Postprandial LDL phenolic content and LDL
oxidation are modulated by olive oil phenolic compounds in humans. Free Radic. Biol. Med. 2006, 40, 608–616.
[CrossRef] [PubMed]
9. Tresserra-Rimbau, A.; Medina-Remón, A.; Pérez-Jiménez, J.; Martínez-González, M.A.; Covas, M.I.;
Corella, D.; Salas-Salvadó, J.; Gómez-Gracia, E.; Lapetra, J.; Arós, F.; et al. Dietary intake and major
food sources of polyphenols in a Spanish population at high cardiovascular risk: The PREDIMED study.
Nutr. Metab. Cardiovasc. Dis. 2013, 23, 953–959. [CrossRef] [PubMed]
10. Storniolo, C.E.; Roselló-Catafau, J.; Pintó, X.; Mitjavila, M.T.; Moreno, J.J. Polyphenol fraction of extra virgin
olive oil protects against endothelial dysfunction induced by high glucose and free fatty acids through
modulation of nitric oxide and endothelin-1. Redox Biol. 2014, 2, 971–977. [CrossRef] [PubMed]
11. Sánchez-Quesada, C.; López-Biedma, A.; Warleta, F.; Campos, M.; Beltrán, G.; Gaforio, J.J. Bioactive
Properties of the Main Triterpenes Found in Olives, Virgin Olive Oil, and Leaves of Olea europaea. J. Agric.
Food Chem. 2013, 61, 12173–12182. [CrossRef] [PubMed]
12. Rodríguez-Rodríguez, R. Oleanolic acid and related triterpenoids from olives on vascular function: Molecular
mechanisms and therapeutic perspectives. Curr. Med. Chem. 2015, 22, 1414–1425. [CrossRef] [PubMed]
13. EFSA Panel on Dietetic Products, Nutrition, and Allergies (NDA). General guidance for stakeholders on the
evaluation of Article 13.1, 13.5 and 14 health claims. EFSA J. 2011, 9, 1–24.
14. Biel, S.; Mesa, M.-D.; de la Torre, R.; Espejo, J.-A.; Fernández-Navarro, J.-R.; Fitó, M.; Sánchez-Rodriguez, E.;
Rosa, C.; Marchal, R.; Alche, J.D.; et al. The NUTRAOLEOUM Study, a randomized controlled trial,
for achieving nutritional added value for olive oils. BMC Complement. Altern. Med. 2016, 16, 404. [CrossRef]
[PubMed]
15. FDA. U.S. Food and Drug Administration. Available online: https://www.fda.gov/food/labelingnutrition/
ucm073992.htm#cardio (accessed on 28 March 2018).
16. Covas, M.-I.; Nyyssönen, K.; Poulsen, H.E.; Kaikkonen, J.; Zunft, H.-J.F.; Kiesewetter, H.; Gaddi, A.;
de la Torre, R.; Mursu, J.; Bäumler, H.; et al. The Effect of Polyphenols in Olive Oil on Heart Disease
Risk Factors. Ann. Intern. Med. 2006, 145, 333–341. [CrossRef] [PubMed]
17. Fielding, C.J.; Havel, R.J.; Todd, K.M.; Yeo, K.E.; Schloetter, M.C.; Weinberg, V.; Frost, P.H. Effects of dietary
cholesterol and fat saturation on plasma lipoproteins in an ethnically diverse population of healthy young
men. J. Clin. Investig. 1995, 95, 611–618. [CrossRef] [PubMed]
18. Yang, Y.J.; Kim, M.K.; Hwang, S.H.; Ahn, Y.; Shim, J.E.; Kim, D.H. Relative validities of 3-day food records
and the food frequency questionnaire. Nutr. Res. Pract. 2010, 4, 142–148. [CrossRef] [PubMed]
19. Base de Datos de Composición de Alimentos (BEDCA). Available online: http://www.bedca.net/ (accessed
on 28 March 2018).
20. Huisman, H.W.; Schutte, A.E.; van Rooyen, J.M.; Schutte, R.; Malan, L.; Fourie, C.M.T.; Malan, N.T.
The Association of Red Blood Cell Counts with Endothelin-1 in African and Caucasian Women.
Clin. Exp. Hypertens. 2009, 31, 1–10. [CrossRef] [PubMed]
21. Walter, R.; Mark, M.; Gaudenz, R.; Harris, L.G.; Reinhart, W.H. Influence of nitrovasodilators and
endothelin-1 on rheology of human blood in vitro. Br. J. Pharmacol. 1999, 128, 744–750. [CrossRef] [PubMed]
22. Pozo, O.J.; Pujadas, M.; Gleeson, S.B.; Mesa-García, M.D.; Pastor, A.; Kotronoulas, A.; Fitó, M.; Covas, M.I.;
Navarro, J.R.F.; Espejo, J.A.; et al. Liquid chromatography tandem mass spectrometric determination of
triterpenes in human fluids: Evaluation of markers of dietary intake of olive oil and metabolic disposition of
oleanolic acid and maslinic acid in humans. Anal. Chim. Acta 2017, 990, 84–95. [CrossRef] [PubMed]
23. Senn, S. Cross-over Trials in Clinical Research, 2nd ed.; John Wiley & Sons, Ltd.: Chichester, UK, 2002; pp. 35–53.
24. Liu, G.F.; Lu, K.; Mogg, R.; Mallich, M.; Mehrotra, D.V. Should baseline be a covariate or dependent variable
in analyses of change from baseline in clinical trials? Stat. Med. 2009, 28, 2509–2530. [CrossRef] [PubMed]
60
Nutrients 2018, 10, 626
25. Estruch, R.; Salas-Salvadó, J. Towards an even healthier mediterranean diet. Nutr. Metab. Cardiovasc. Dis.
2013, 23, 1163–1166. [CrossRef] [PubMed]
26. Babio, N.; Toledo, E.; Estruch, R.; Ros, E.; Martínez-González, M.A.; Castañer, O.; Bulló, M.; Corella, D.;
Arós, F.; Gómez-Gracia, E.; et al. Mediterranean diets and metabolic syndrome status in the PREDIMED
randomized trial. CMAJ 2014, 186, E649–E657. [CrossRef] [PubMed]
27. Salas-Salvadó, J.; Bulló, M.; Estruch, R.; Ros, E.; Covas, M.-I.; Ibarrola-Jurado, N.; Corella, D.;
Arós, F.; Gómez-Gracia, E.; Ruiz-Gutiérrez, V.; et al. Prevention of Diabetes with Mediterranean Diets.
Ann. Intern. Med. 2014, 160, 1–10. [CrossRef] [PubMed]
28. Peyrol, J.; Riva, C.; Amiot, M. Hydroxytyrosol in the Prevention of the Metabolic Syndrome and Related
Disorders. Nutrients 2017, 9, 306. [CrossRef] [PubMed]
29. Hohmann, C.D.; Cramer, H.; Michalsen, A.; Kessler, C.; Steckhan, N.; Choi, K.; Dobos, G. Effects of high
phenolic olive oil on cardiovascular risk factors: A systematic review and meta-analysis. Phytomedicine 2015,
22, 631–640. [CrossRef] [PubMed]
30. Rondanelli, M.; Giacosa, A.; Morazzoni, P.; Guido, D.; Grassi, M.; Morandi, G.; Bologna, C.; Riva, A.;
Allegrini, P.; Perna, S. MediterrAsian Diet Products that Could Raise HDL-Cholesterol: A Systematic Review.
BioMed. Res. Int. 2016, 2016, 1–15. [CrossRef] [PubMed]
31. Farràs, M.; Castañer, O.; Martín-Peláez, S.; Hernáez, Á.; Schröder, H.; Subirana, I.; Muñoz-Aguayo, D.;
Gaixas, S.; de la Torre, R.; Farré, M.; et al. Complementary phenol-enriched olive oil improves HDL
characteristics in hypercholesterolemic subjects. A randomized, double-blind, crossover, controlled trial.
The VOHF study. Mol. Nutr. Food Res. 2015, 59, 1758–1770. [CrossRef] [PubMed]
32. Hernaez, A.; Fernandez-Castillejo, S.; Farras, M.; Catalan, U.; Subirana, I.; Montes, R.; Sola, R.;
Munoz-Aguayo, D.; Gelabert-Gorgues, A.; Diaz-Gil, O.; et al. Olive Oil Polyphenols Enhance High-Density
Lipoprotein Function in Humans: A Randomized Controlled Trial. Arterioscler. Thromb. Vasc. Biol. 2014, 34,
2115–2119. [CrossRef] [PubMed]
33. Fernández-Castillejo, S.; Valls, R.-M.; Castañer, O.; Rubió, L.; Catalán, Ú.; Pedret, A.; Macià, A.;
Sampson, M.L.; Covas, M.-I.; Fitó, M.; et al. Polyphenol rich olive oils improve lipoprotein particle
atherogenic ratios and subclasses profile: A randomized, crossover, controlled trial. Mol. Nutr. Food Res.
2016, 60, 1544–1554. [CrossRef] [PubMed]
34. Hernáez, Á.; Castañer, O.; Goday, A.; Ros, E.; Pintó, X.; Estruch, R.; Salas-Salvadó, J.; Corella, D.; Arós, F.;
Serra-Majem, L.; et al. The Mediterranean Diet decreases LDL atherogenicity in high cardiovascular risk
individuals: A randomized controlled trial. Mol. Nutr. Food Res. 2017, 61, 1601015. [CrossRef] [PubMed]
35. Nordestgaard, B.G.; Langsted, A.; Mora, S.; Kolovou, G.; Baum, H.; Bruckert, E.; Watts, G.F.; Sypniewska, G.;
Wiklund, O.; Borén, J.; et al. Fasting is not routinely required for determination of a lipid profile: Clinical
and laboratory implications including flagging at desirable concentration cut-points—A joint consensus
statement from the European Atherosclerosis Society and European Federation of Clinical Chemistry and
Laboratory Medicine. Eur. Heart J. 2016, 37, 1944–1958. [PubMed]
36. Saibandith, B.; Spencer, J.; Rowland, I.; Commane, D. Olive Polyphenols and the Metabolic Syndrome.
Molecules 2017, 22, 1082. [CrossRef] [PubMed]
37. Luo, H.; Liu, J.; Ouyang, Q.; Xuan, C.; Wang, L.; Li, T.; Liu, J. The effects of oleanolic acid on atherosclerosis
in different animal models. Acta Biochim. Biophys. Sin. 2017, 49, 349–354. [CrossRef] [PubMed]
38. Ross, R. The pathogenesis of atherosclerosis: A perspective for the 1990s. Nature 1993, 362, 801–809.
[CrossRef] [PubMed]
39. Gkaliagkousi, E.; Gavriilaki, E.; Triantafyllou, A.; Douma, S. Clinical Significance of Endothelial Dysfunction
in Essential Hypertension. Curr. Hypertens. Rep. 2015, 17, 85. [CrossRef] [PubMed]
40. Cambiaggi, C.; Mencarelli, M.; Muscettola, M.; Grasso, G. Gene expression of endothelin-1 (ET-1) and release
of mature peptide by activated human neutrophils. Cytokine 2001, 14, 230–233. [CrossRef] [PubMed]
41. Shinagawa, S.; Okazaki, T.; Ikeda, M.; Yudoh, K.; Kisanuki, Y.Y.; Yanagisawa, M.; Kawahata, K.; Ozaki, S. T
cells upon activation promote endothelin 1 production in monocytes via IFN-γ and TNF-α. Sci. Rep. 2017, 7,
14500. [CrossRef] [PubMed]
42. Massai, L.; Carbotti, P.; Cambiaggi, C.; Mencarelli, M.; Migliaccio, P.; Muscettola, M.; Grasso, G.
Prepro-endothelin-1 mRNA and its mature peptide in human appendix. Gastrointest. Liver Physiol. 2003, 284,
G340–G348. [CrossRef] [PubMed]
61
Nutrients 2018, 10, 626
43. Mencarelli, M.; Pecorelli, A.; Carbotti, P.; Valacchi, G.; Grasso, G.; Muscettola, M. Endothelin receptor A
expression in human inflammatory cells. Regul. Pept. 2009, 158, 1–5. [CrossRef] [PubMed]
44. Achmad, T.H.; Rao, G.S. Chemotaxis of human boold monocytes toward endothelin-1 and the influence of
calcium channel blockers. Biochem. Biophys. Res. Commun. 1992, 189, 994–1000. [CrossRef]
45. Ruetten, H.; Thiemermann, C. Endothelin-1 stimulates the biosynthesis of tumour necrosis factor in
macrophages: ET-receptors, signal transduction and inhibitionby dexamethasone. J. Physiol. Pharmacol. 1997,
48, 675–688. [PubMed]
46. Zouki, C.; Baron, C.; Fournie, A.; Filep, J.G. Endothelin-1 enhances neutrophil adhesion to human coronary
artery endothelial cells: Role of ETA receptors and platelet activating factor. Br. J. Pharmacol. 1999, 127,
969–979. [CrossRef] [PubMed]
47. Zern, T.L.; Fernandez, M.L. Cardioprotective effects of dietary polyphenols. J. Nutr. 2005, 135, 2291–2294.
[CrossRef] [PubMed]
48. Musini, V.M.; Wright, J.M. Factors affecting blood pressure variability: Lessons learned from two systematic
reviews of randomized controlled trials. PLoS ONE 2009, 4, e5673. [CrossRef] [PubMed]
49. Handler, J. The Importance of Accurate Blood Pressure Measurement. Perm. J. 2009, 13, 51–54. [CrossRef]
[PubMed]
50. Rodriguez-Rodriguez, R.; Herrera, M.D.; de Sotomayor, M.A.; Ruiz-Gutierrez, V. Pomace Olive Oil Improves
Endothelial Function in Spontaneously Hypertensive Rats by Increasing Endothelial Nitric Oxide Synthase
Expression. Am. J. Hypertens. 2007, 20, 728–734. [CrossRef] [PubMed]
51. Valero-Muñoz, M.; Martín-Fernández, B.; Ballesteros, S.; de la Fuente, E.; Quintela, J.C.; Lahera, V.;
de las Heras, N. Protective effect of a pomace olive oil concentrated in triterpenic acids in alterations related
to hypertension in rats: Mechanisms involved. Mol. Nutr. Food Res. 2014, 58, 376–383. [CrossRef] [PubMed]
52. Dell’Agli, M.; Fagnani, R.; Mitro, N.; Scurati, S.; Masciadri, M.; Mussoni, L.; Galli, G.V.; Bosisio, E.;
Crestani, M.; De Fabiani, E.; et al. Minor Components of Olive Oil Modulate Proatherogenic Adhesion
Molecules Involved in Endothelial Activation. J. Agric. Food Chem. 2006, 54, 3259–3264. [CrossRef] [PubMed]
53. Fitó, M.; Cladellas, M.; de la Torre, R.; Martí, J.; Muñoz, D.; Schröder, H.; Alcántara, M.; Pujadas-Bastardes, M.;
Marrugat, J.; López-Sabater, M.C.; et al. Anti-inflammatory effect of virgin olive oil in stable coronary disease
patients: A randomized, crossover, controlled trial. Eur. J. Clin. Nutr. 2007, 62, 570–574. [CrossRef] [PubMed]
54. Papageorgiou, N.; Tousoulis, D.; Psaltopoulou, T.; Giolis, A.; Antoniades, C.; Tsiamis, E.; Miliou, A.;
Toutouzas, K.; Siasos, G.; Stefanadis, C. Divergent anti-inflammatory effects of different oil acute
consumption on healthy individuals. Eur. J. Clin. Nutr. 2011, 65, 514–519. [CrossRef] [PubMed]
55. Martín-Peláez, S.; Castañer, O.; Konstantinidou, V.; Subirana, I.; Muñoz-Aguayo, D.; Blanchart, G.; Gaixas, S.;
de la Torre, R.; Farré, M.; Sáez, G.T.; et al. Effect of olive oil phenolic compounds on the expression of blood
pressure-related genes in healthy individuals. Eur. J. Nutr. 2017, 56, 663–670. [CrossRef] [PubMed]
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
62
nutrients
Article
The Effect of Low-Carbohydrate Diet on Glycemic
Control in Patients with Type 2 Diabetes Mellitus
Li-Li Wang 1 , Qi Wang 1 , Yong Hong 1 , Omorogieva Ojo 2 , Qing Jiang 1 , Yun-Ying Hou 1 ,
Yu-Hua Huang 3 and Xiao-Hua Wang 1, *
1 School of Nursing, Medical College, Soochow University, Suzhou 215006, China;
[email protected] (L.-L.W.); [email protected] (Q.W.); [email protected] (Y.H.);
[email protected] (Q.J.); [email protected] (Y.-Y.H.)
2 Department of Adult Nursing and Paramedic Science, University of Greenwich, London SE9 2UG, UK;
[email protected]
3 Medical College, Soochow University, Suzhou 215006, China; [email protected]
* Correspondence: [email protected]; Tel.: +86-138-1488-0208
Abstract: Objective: In China, a low-fat diet (LFD) is mainly recommended to help improve blood
glucose levels in patients with type 2 diabetes mellitus (T2DM). However, a low-carbohydrate diet
(LCD) has been shown to be effective in improving blood glucose levels in America and England.
A few studies, primarily randomized controlled trials, have been reported in China as well. Method:
Firstly, we designed two ‘six-point formula’ methods, which met the requirements of LCD and LFD,
respectively. Fifty-six T2DM patients were recruited and randomly allocated to the LCD group
(n = 28) and the LFD group (n = 28). The LCD group received education about LCD’s six-point
formula, while the LFD group received education about LFD’s six-point formula. The follow-up
time was three months. The indicators for glycemic control and other metabolic parameters were
collected and compared between the two groups. Results: Forty-nine patients completed the study.
The proportions of calories from three macronutrients the patients consumed met the requirements
of LCD and LFD. Compared to the LFD group, there was a greater decrease in HbA1c level in the
LCD group (−0.63% vs. −0.31%, p < 0.05). The dosages of insulin and fasting blood glucoses (FBG)
in the third month were lower than those at baseline in both groups. Compared with baseline values,
body mass index (BMI) and total cholesterol (TC) in the LCD group were significantly reduced in the
third month (p < 0.05); however, there were no statistically significant differences in the LFD group.
Conclusions: LCD can improve blood glucose more than LFD in Chinese patients with T2DM. It can
also regulate blood lipid, reduce BMI, and decrease insulin dose in patients with T2DM. In addition,
the six-point formula is feasible, easily operable, and a practical educational diet for Chinese patients
with T2DM.
Keywords: diabetes mellitus; diet; carbohydrate; blood glucose; HbA1c; fasting blood glucose;
postprandial blood glucose
1. Introduction
Dietary intervention is a strategy to manage diabetes mellitus (DM) [1], as it can reduce the
burden on islet cells and thus improve blood glucose levels, lipid profiles, and cognitive status [2–4].
However, good adherence to diabetic diets is the premise of diet therapy. In China, a low-fat diet (LFD)
is mainly recommended to help improve blood glucose levels in patients with type 2 diabetes mellitus
(T2DM) [5]. Studies have shown that LFD could reduce glycated hemoglobin (HbA1c) by as much as
0.8–2.8% [6–8].
On the other hand, a low-carbohydrate diet (LCD) is a dietary strategy that refers to carbohydrate
intake of between 30–200 g/day or calories from carbohydrates/total calories of <45%, supplementing
instead with fat or protein [9]. This has been found to be effective in the treatment of obesity,
and apart from significantly reducing weight, it can also effectively improve blood lipid and insulin
resistance [10]. In recent years, the American Diabetes Association and Diabetes UK have both
confirmed the effectiveness of LCD in reducing weight, improving blood glucose, and regulating blood
lipid in patients with DM [11,12]. In Japan, Yamada [13] reported that HbA1c and triglyceride (TG)
levels in patients with T2DM decreased significantly in the LCD group without calorie-restriction,
compared to the LFD group with calorie-restriction. This indicates that LCD made patients with DM
have less desire to eat due to a feeling of satiety. However, only limited studies relating to the use of
LCD in patients with DM, especially randomized controlled trials, have been reported in China.
Based on research evidence, only 29.8% of Chinese patients with T2DM comply with a diabetic
diet advised by their doctors and dietitians [14]. In addition, we found that certain types of foods were
strictly limited and patients with DM were finding it hard to understand the caloric values of foods
consumed, thus making it difficult to adhere to the diet. Thus, it is necessary to develop an easy and
more effective method to support these patients. Firstly, we designed the ‘six-point formula’ to help
patients master LCD and LFD. We then let them record details of their diets and hand over to us the
task of calculating the caloric values of foods. Based on this, we explored the effect of two DM diets
(LCD and LFD) on hyperglycemia.
2.1. Subjects
Participants with T2DM were recruited from the community and the First Affiliated Hospital of
Soochow University. The inclusion criteria were the following: Patients older than 18 years, had been
diagnosed with T2DM, had no change in oral antidiabetic drugs or insulin in half a month before the
intervention, were able to communicate, had volunteered to participate in this study, and are able to
provide informed consent. Those excluded were patients who ate nuts regularly (≥4 day/week ) [15];
were allergic to food, especially nuts; had difficulty in chewing nuts (such as those with few teeth);
received other dietary interventions or had severe conditions including indigestion, heart failure,
renal failure, malignant tumours, severe cerebrovascular disease, ketosis, digestive dysfunction, liver
dysfunction or severe gallbladder and pancreatic diseases; and those whose fasting blood glucose
(FBG) were more than 16.7 mmol/L [16] during the interventions.
64
Nutrients 2018, 10, 661
2.7. Intervention
Firstly, our team developed a preliminary dietary education handbook for patients with T2DM
based on evidence from literature and guidelines regarding T2DM dietary management [5,20].
Secondly, two endocrinologists, four diabetic nurse specialists, and one dietician reviewed and
modified the handbook. Finally, five T2DM inpatients of different ages and educational levels reviewed
the handbook to ensure that patients with T2DM understood it and that it could help improve their
dietary adherence. The major content of the handbook was a concise formula that included six points.
Detailed contents of the six-point formula are shown in Figure 1. Other educational contents about
foods included how to distinguish vegetables and staple food (such as potato and broad bean); ways
to cook food; and symptoms, prevention and treatment of hypoglycemia.
In the one-on-one education session, the researcher and the patients reviewed the handbook.
Using the LCD handbook, the researcher focused on instructing patients to restrict intake of staple
food/meal (1 Liang) per day in the LCD group. The reduced staple food/meal was replaced by
consuming 60 g/day nuts for males and 50 g/day for females, respectively. Nuts were uniformly
purchased, weighed, vacuum-packed, and distributed every two weeks.
65
Nutrients 2018, 10, 661
For patients in the LFD group, we provided participants with a handbook about LFD, and instructed
them on a pithy formula of six points.
Follow up was conducted once a week in the first month of the intervention and once every two
weeks in the second and third months. The duration of follow up was about 10 min. The main focus
of the follow-up was to review the patients’ compliance to the diet program and to support them to
adhere to it (in patients with poor compliance). It also involved collecting data of the modification
of hypoglycemic agents and the occurrence of hypoglycemia. If a patient’s diet did not meet the
requirements of the dietary program in the intervention period, they were excluded from the study.
Figure 1. The detailed contents of the six-point formula of two groups. Notes: 1 jin = 10 liang = 500 g,
Chinese conventional units of weight. Staple food/meal refers to foods rich in carbohydrates, mainly
three kinds of steamed bread, noodles and rice in China. LFD: Low-fat diet; LCD: Low-carbohydrate diet.
66
Nutrients 2018, 10, 661
3. Results
67
Nutrients 2018, 10, 661
Table 1. Cont.
68
Nutrients 2018, 10, 661
carbohydrates (39%) met the standard of LCD (<45%). The 26% of calories from fat met the standard of
LFD, while the calories from protein were almost similar in the two groups (p > 0.05, Table 3) (Figure 3).
Table 3. Comparison of the calories from three macronutrients consumed by the patients.
Figure 3. The percentage of the calories from carbohydrates (39%) met the standard of LCD (<45%) in
the LCD group, while the 26% calories from fat met the standard of LFD. LCD: Low-carbohydrate diet;
LFD: Low-fat diet
Glycated Hemoglobin
Compared to the baseline, HbA1c levels in both the LCD group and LFD group decreased
significantly (0.63 ± 1.18% and 0.31 ± 0.70%), respectively. At the baseline, HbA1c levels were
not significantly different between the two groups. However, after the intervention, HbA1c levels
in the LCD group decreased significantly (p < 0.05, Table 4), when compared to the LFD group.
The Intention-To-Treat (ITT) in relation to HbA1c levels was performed to ensure the stability of the
above results. The ITT results were found to be in agreement with the earlier findings (Table 5).
69
Nutrients 2018, 10, 661
Table 5. Comparison of glycated hemoglobin (%) between the two groups in ITT.
Figure 4. The changing trends of the FBG in the LCD and LFD Groups. FBG: fasting blood glucoses
Table 6. Comparison of fasting blood glucose (mmol/L) between the two groups.
70
Nutrients 2018, 10, 661
Figure 5. The changing trends of the postprandial 2-h blood glucose in the LCD and LFD Groups.
71
Nutrients 2018, 10, 661
Table 10. Comparison of insulin dose (insulin unit, IU) between the two groups.
Table 11. Comparison of other antidiabetic drugs between the two groups.
4. Discussion
The use of LCD in human nutrition and health is a dietary strategy that ensures that carbohydrate
intake is restricted. However, in a Chinese dietary plan, most staple foods have high glycemic
index [20,21]. Therefore, it would seem that LCD may not be accepted easily among Chinese patients
with diabetes mellitus (DM). In consideration, we initially designed the ‘six-point formula’ to help
patients improve dietary adherence. We found that the participants showed good adherence to the
intervention, and no significant difference with respect to dietary adherence between two groups (LCD
versus LFD) was observed. The proportions of energy provided by the three macronutrients met the
requirements of LCD and LFD. It was indicated that the ‘six-point formula’ of the DM diet was feasible
for Chinese T2DM patients.
72
Nutrients 2018, 10, 661
5. Limitation
There are some limitations to the study. Firstly, the method used to evaluate the energy intake
of food may not have been robust enough. At the baseline, we obtained data of caloric intake from
73
Nutrients 2018, 10, 661
patients’ memories, which meant that it was probably underestimated. Secondly, measurement
differences might exist in FBG and postprandial 2-h blood glucose levels, which were measured at
home by the patients themselves using different blood glucose meters. Thirdly, the prolonged effect of
LCD on the prognosis of DM was not observed due to short follow-up time. Finally, a control group
without a treatment was not considered in the study design.
6. Conclusions
LCD can improve blood glucose more than LFD in Chinese patients with T2DM. It can also
regulate blood lipids, reduce BMI, and decrease insulin doses in patients with T2DM. In addition,
the six-point formula is feasible, easily operable, and is a practical educational diet for Chinese patients
with T2DM.
Author Contributions: L.-L.W. and Q.W. collected and analyzed data and wrote the initial draft, which was
revised by X.-H.W., Y.-H.H., Y.H., O.O., Q.J., Y.-Y.H. All authors participated in research design and quality control.
L.-L.W. and Q.W. contributed equally to this study. X.-H.W. was the corresponding author.
Acknowledgments: We thank the patients with T2DM who participated in the study. Thanks also to Huijuan Zhou,
Xiaoyan Zhang and Li Wang, who provided us with study sites to ensure that the study was conducted. This study
was supported by Suzhou Science and Technology Project, China (Grant number SYS201513).
Conflicts of Interest: The authors declare no conflict of interest.
References
1. Breen, C.; Ryan, M.; Gibney, M.J.; O’Shea, D. Diabetes-related nutrition knowledge and dietary intake among
adults with type 2 diabetes. Br. J. Nutr. 2015, 114, 439–447. [CrossRef] [PubMed]
2. Ley, S.H.; Hamdy, O.; Mohan, V.; Hu, F.B. Prevention and management of type 2 diabetes: Dietary
components and nutritional strategies. Lancet 2014, 383, 1999–2007. [CrossRef]
3. Seetharaman, S.; Andel, R.; McEvoy, C.; Aslan, A.K.D.; Finkel, D.; Pedersen, N.L. Blood Glucose, Diet-Based
Glycemic Load and Cognitive Aging Among Dementia-Free Older Adults. J. Gerontol. A Biol. Sci. Med. Sci.
2015, 70, 471. [CrossRef] [PubMed]
4. Seetharaman, S. The Influences of Dietary Sugar and Related Metabolic Disorders on Cognitive Aging and
Dementia. Mol. Basis Nutr. Aging 2016, 331–344. [CrossRef]
5. Chinese Diabetes Society. Guidelines for the prevention and treatment of type 2 diabetes in China.
Chin. Med. J. 2017, 10, 4–67.
6. Cao, A.H.; Sun, L.Z.; Cui, J.W. Effects of A Low-Carbohydrate Diet and A Low-Fat Diet on Weight and
Glycemic Control in Type 2 Diabetics Mellitus. Chin. Gener. Pract. 2011, 14, 52–56.
7. Cao, A.L.; Xin, B. Effects of low-fat diet on body mass, blood lipids and sugar control in obese patients with
type 2 diabetes mellitus. Clin. Focus 2012, 27, 1025–1031.
8. Wang, Y.L.; Yao, Y.N.; Yang, X.L. Clinical study of the changing of bodyweight (BW) and fasting blood
glucose (FBG) in obese patients with type 2 diabetes on a low-carbohydrate diet (LCD). J. Xinjiang Med. Univ.
2009, 32, 914–916.
9. Hite, A.H.; Berkowitz, V.G.; Berkowitz, K. Low-carbohydrate diet review: Shifting the paradigm.
Nutr. Clin. Pract. 2011, 26, 300. [CrossRef] [PubMed]
10. Forsythe, C.E.; Phinney, S.D.; Fernandez, M.L.; Quann, E.E.; Wood, R.J.; Bibus, D.M.; Kraemer, W.J.;
Feinman, R.D.; Volek, J.S. Comparison of Low Fat and Low Carbohydrate Diets on Circulating Fatty
Acid Composition and Markers of Inflammation. Lipids 2008, 43, 65–77. [CrossRef] [PubMed]
11. Dyson, P.A.; Kelly, T.; Deakin, T.; Duncan, A.; Frost, G.; Harrison, Z.; Khatri, D.; Kunka, D.; McArdle, P.;
Mellor, D. Diabetes UK evidence-based nutrition guidelines for the prevention and management of diabetes.
Diabet. Med. 2011, 28, 1282–1288. [CrossRef] [PubMed]
12. Wheeler, M.L.; Dunbar, S.A.; Jaacks, L.M.; Wahida, K.; Mayer-Davis, E.J.; Judith, W.R., Jr.; William, S.
Macronutrients, Food Groups, and Eating Patterns in the Management of Diabetes. Diabet. Care 2010, 35,
434–445. [CrossRef] [PubMed]
74
Nutrients 2018, 10, 661
13. Yamada, Y.; Uchida, J.; Izumi, H.; Tsukamoto, Y.; Inoue, G.; Watanabe, Y.; Irie, J.; Yamada, S.
A non-calorie-restricted low-carbohydrate diet is effective as an alternative therapy for patients with type 2
diabetes. Int. Med. 2012, 53, 13. [CrossRef]
14. Zhu, H.M.; Ji, C. The status and compliance of medical nutrition education in patients with type 2 diabetes.
J. Nurs. 2015, 50–52.
15. Asghari, G.; Ghorbani, Z.; Mirmiran, P.; Azizi, F. Nut consumption is associated with lower incidence of
type 2 diabetes: The Tehran Lipid and Glucose Study. Diabet. Metab. 2017, 43, 18. [CrossRef] [PubMed]
16. Chen, L. A brief discussion on the increase of fasting blood glucose. Health Guide 2014, 20, 34–35.
17. Daving, Y.; Andrén, E.; Nordholm, L.; Grimby, G. Reliability of an interview approach to the Functional
Independence Measure. Clin. Rehabil. 2001, 15, 301. [CrossRef] [PubMed]
18. Turner, R.; Holman, R.; Cull, C.; Stratton, I.; Matthews, D.; Frighi, V.; Manley, S.; Neil, A.; McElroy, K.;
Wright, D. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional
treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes
Study (UKPDS) Group. Lancet 1998, 352, 837.
19. Association WOHAD. Defining and reporting hypoglycemia in diabetes: A report from the American
Diabetes Association Workgroup on Hypoglycemia. Diabet. Care 2005, 28, 1245.
20. Cai, W. Modern Nutrition; Fudan University Press: Shanghai, China, 2011.
21. Akhoundan, M.; Shadman, Z.; Jandaghi, P.; Aboeerad, M.; Larijani, B.; Jamshidi, Z.; Ardalani, H.; Nikoo, M.K.
The Association of Bread and Rice with Metabolic Factors in Type 2 Diabetic Patients. PLoS ONE 2016,
11, e0167921. [CrossRef] [PubMed]
22. Cohen, A.E.; Johnston, C.S. Almond ingestion at mealtime reduces postprandial glycemia and
chronic ingestion reduces hemoglobin A in individuals with well-controlled type 2 diabetes mellitus.
Metab. Clin. Exp. 2011, 60, 1312. [CrossRef] [PubMed]
23. Li, S.C.; Liu, Y.H.; Liu, J.F.; Chang, W.H.; Chen, C.M.; Chen, C.Y. Almond consumption improved glycemic
control and lipid profiles in patients with type 2 diabetes mellitus. Metab. Clin. Exp. 2011, 60, 474–479.
[CrossRef] [PubMed]
24. Sacks, F.M.; Carey, V.J.; Anderson, C.A.; Miller, E.R.; Copeland, T.; Charleston, J.; Harshfield, B.J.; Laranjo, N.;
McCarron, P.; Swain, J. Effects of high vs low glycemic index of dietary carbohydrate on cardiovascular
disease risk factors and insulin sensitivity: The OmniCarb randomized clinical trial. JAMA 2014, 312,
2531–2541. [CrossRef] [PubMed]
25. Mayer, S.B.; Jeffreys, A.S.; Olsen, M.K.; McDuffie, J.R.; Feinglos, M.N., Jr.; Yancy, W.S. Two Diets with
Different Hemoglobin A1c and Antiglycemic Medication Effects Despite Similar Weight Loss in Type 2
Diabetes. Diabet. Obes. Metab. 2014, 16, 90–93. [CrossRef] [PubMed]
26. Liu, J.F.; Liu, Y.H.; Chen, C.M.; Chang, W.H.; Chen, C.O. The effect of almonds on inflammation and oxidative
stress in Chinese patients with type 2 diabetes mellitus: A randomized crossover controlled feeding trial.
Eur. J. Nutr. 2013, 52, 927–935. [CrossRef] [PubMed]
27. Vadivel, V.; Kunyanga, C.N.; Biesalski, H.K. Health benefits of nut consumption with special reference to
body weight control. Nutrition 2015, 28, 1089–1097. [CrossRef] [PubMed]
28. Tan, S.Y.; Dhillon, J.; Mattes, R.D. A review of the effects of nuts on appetite, food intake, metabolism,
and body weight. Am. J. Clin. Nutr. 2014, 100 (Suppl. 1), 412S–422S. [CrossRef] [PubMed]
29. Barbour, J.A.; Howe, P.R.; Buckley, J.D.; Bryan, J.; Coates, A.M. Effect of 12 Weeks High Oleic Peanut
Consumption on Cardio-Metabolic Risk Factors and Body Composition. Nutrients 2015, 7, 7381–7398.
[CrossRef] [PubMed]
30. Lovejoy, J.C.; Most, M.M.; Lefevre, M.; Greenway, F.L.; Rood, J.C. Effect of diets enriched in almonds on
insulin action and serum lipids in adults with normal glucose tolerance or type 2 diabetes. Am. J. Clin. Nutr.
2002, 76, 1000–1006. [CrossRef] [PubMed]
31. Westman, E.C.; Feinman, R.D.; Mavropoulos, J.C.; Vernon, M.C.; Volek, J.S.; Wortman, J.A.; Yancy, W.S.;
Phinney, S.D. Low-carbohydrate nutrition and metabolism. Am. J. Clin. Nutr. 2007, 86, 276–284. [CrossRef]
[PubMed]
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
75
nutrients
Article
SunGold Kiwifruit Supplementation of Individuals
with Prediabetes Alters Gut Microbiota and Improves
Vitamin C Status, Anthropometric and
Clinical Markers
Renée Wilson 1 , Jinny Willis 2 , Richard B. Gearry 1,3 , Alan Hughes 4 , Blair Lawley 4 ,
Paula Skidmore 5 , Chris Frampton 1 , Elizabeth Fleming 5 , Angie Anderson 5 , Lizzie Jones 5 ,
Gerald W. Tannock 3,4,6 and Anitra C. Carr 7, *
1 Department of Medicine, University of Otago, Christchurch 8140, New Zealand;
[email protected] (R.W.); [email protected] (R.B.G.);
[email protected] (C.F.)
2 New Zealand Nursing Organisation, Christchurch 8140, New Zealand; [email protected]
3 Microbiome Otago, University of Otago, Dunedin 9054, New Zealand; [email protected]
4 Department of Microbiology and Immunology, University of Otago, Dunedin 9054, New Zealand;
[email protected] (A.H.); [email protected] (B.L.)
5 Department of Human Nutrition, University of Otago, Dunedin 9054, New Zealand;
[email protected] (P.S.); liz.fl[email protected] (E.F.); [email protected] (A.A.);
[email protected] (L.J.)
6 Riddet Centre of Research Excellence, Massey University, Palmerston North 4442, New Zealand
7 Department of Pathology & Biomedical Science, University of Otago, Christchurch 8140, New Zealand
* Correspondence: [email protected]; Tel.: +64-3-364-0649
Abstract: Kiwifruit are a nutrient dense food and an excellent source of vitamin C. Supplementation
of the diet with kiwifruit enhances plasma vitamin C status and epidemiological studies have shown
an association between vitamin C status and reduced insulin resistance and improved blood glucose
control. In vitro experiments suggest that eating kiwifruit might induce changes to microbiota
composition and function; however, human studies to confirm these findings are lacking. The aim of
this study was to investigate the effect of consuming two SunGold kiwifruit per day over 12 weeks
on vitamin C status, clinical and anthropometric measures and faecal microbiota composition in
people with prediabetes. This pilot intervention trial compared baseline measurements with those
following the intervention. Participants completed a physical activity questionnaire and a three-day
estimated food diary at baseline and on completion of the trial. Venous blood samples were collected
at each study visit (baseline, 6, 12 weeks) for determination of glycaemic indices, plasma vitamin
C concentrations, hormones, lipid profiles and high-sensitivity C-reactive protein. Participants
provided a faecal sample at each study visit. DNA was extracted from the faecal samples and
a region of the 16S ribosomal RNA gene was amplified and sequenced to determine faecal microbiota
composition. When week 12 measures were compared to baseline, results showed a significant
increase in plasma vitamin C (14 μmol/L, p < 0.001). There was a significant reduction in both
diastolic (4 mmHg, p = 0.029) and systolic (6 mmHg, p = 0.003) blood pressure and a significant
reduction in waist circumference (3.1 cm, p = 0.001) and waist-to-hip ratio (0.01, p = 0.032). Results also
showed a decrease in HbA1c (1 mmol/mol, p = 0.005) and an increase in fasting glucose (0.1 mmol/L,
p = 0.046), however, these changes were small and were not clinically significant. Analysis of faecal
microbiota composition showed an increase in the relative abundance of as yet uncultivated and
therefore uncharacterised members of the bacterial family Coriobacteriaceae. Novel bacteriological
investigations of Coriobacteriaceae are required to explain their functional relationship to kiwifruit
polysaccharides and polyphenols.
Keywords: vitamin C; blood pressure; waist circumference; glucose; glycaemic control; HbA1c;
kiwifruit; gut microbiota; Coriobacteriaceae
1. Introduction
Diabetes is one of the largest global health crises, affecting 415 million adults worldwide, with type
2 diabetes mellitus (T2DM) accounting for at least 90% of all cases of diabetes [1]. People with
diabetes are at a higher risk of developing a number of disabling and life-threatening health
conditions [1]. Prediabetes reflects a stage between normal glucose tolerance (NGT) and T2DM.
Those with prediabetes are at high risk of progressing to T2DM, although this is not inevitable [2].
In routine practice, prediabetes is characterized by an increase in HbA1c. Results from the 2008/2009
New Zealand Adult Nutrition Survey (NZANS) provided data on the prevalence of diabetes and
prediabetes using the American Diabetes Association (ADA) criteria and found that the prevalence of
diabetes and prediabetes in New Zealand was 7.0% and 25.5% respectively [3]. The alarmingly high
prevalence of people with prediabetes necessitates further research to explore modifiable causative
factors for prediabetes.
Hyperglycaemia (elevated blood glucose concentrations) and insulin resistance are implicated in
the pathogenesis of micro- and macrovascular complications [4]. Maintaining normal blood glucose
levels through diet is one of the main goals in the management of a glucose metabolism disorder
(diabetes or prediabetes) [5]. Higher plasma vitamin C is associated with reduced insulin resistance and
improved blood glucose control [6–10]. Since hyperglycaemia is associated with increased oxidative
stress, a role for antioxidants such as vitamin C in the prevention of T2DM and/or the reduction
of complications is a reasonable proposition. However, there have been mixed findings reported in
randomised controlled trials (RCTs) of supplementation with vitamin C on glycaemic control and
insulin sensitivity [11–13]. A recent meta-analysis of 15 RCTs investigating vitamin C supplementation
and insulin resistance and biomarkers of glycaemic control (fasting glucose, HbA1c) found that doses
of ≥200 mg/day vitamin C significantly reduced glucose concentrations in patients with T2DM,
particularly if the intervention was for more than 30 days and in older individuals [14]. Furthermore,
a recent 12 month RCT found that treating those with T2DM with both metformin and vitamin C was
more effective at reducing HbA1c and risk factors for diabetes-related long-term complications than
treating with metformin alone [15].
SunGold kiwifruit are one of the best dietary sources of vitamin C (160 mg/100 g) among fruit and
vegetables [16]. Regular consumption of two SunGold kiwifruit has been shown to significantly
increase the plasma vitamin C levels in men with inadequate levels of plasma vitamin C [17].
Virtually all fruits represent a source of sugars and therefore moderate consumption is recommended
in people with T2DM. However, there are differing amounts of sugar and other nutrients in various
fruit and portion sizes vary markedly. Kiwifruit, eaten as a whole fruit, have a low glycaemic impact
and are, therefore, thought to be a suitable choice for those with prediabetes and T2DM [18].
In addition to having a positive effect on glycaemic control, gold kiwifruit contain dietary fibre
(1.4 g/100 g in raw Zespri SunGold kiwifruit) [16] and polyphenols which resist digestion by human
enzymes and are degraded by bacteria in the digestive tract, stimulating the growth or activity of
certain bacteria in the colon. The fibre in kiwifruit comprises both soluble (e.g., pectin) and insoluble
(e.g., hemicelluloses and celluloses) components which make up cell walls [19].
T2DM has been reported to be associated with an alteration in the usual balance of bowel
bacterial species, described as a bacterial dysbiosis [20]. Differences in the gut microbiota have been
demonstrated between individuals with T2DM and healthy individuals in previous research [21,22].
Further, the finding of differences in gut microbiota between healthy individuals, individuals with
prediabetes and those with T2DM suggests a potential association between gut ecology and the
progression from normal glucose tolerance to T2DM [23]. A logical extension is the hypothesis that
77
Nutrients 2018, 10, 895
manipulation, by dietary change, of the relative abundances of particular bacterial taxa within the gut
microbiota could play a role in managing T2DM [24].
On the basis of in vitro experiments, SunGold Kiwifruit consumption has been speculated to
affect the composition and functioning of the bowel microbiota [25]. However, the outcomes of this
study need to be confirmed in human intervention studies. Therefore, the aim of the present study
was to determine whether gold kiwifruit consumption by a prediabetic cohort altered gut microbiota
composition, increased plasma vitamin C concentrations and improved glycaemic control or prevented
further deterioration of glucose intolerance.
78
Nutrients 2018, 10, 895
everything they ate or drank over three non-consecutive days (two weekdays and one weekend day)
to analyse their usual dietary intake.
After the lead-in phase, Zespri SunGold kiwifruit (Gold3, Actinidia chinensis) were delivered to
participants weekly. Participants were provided with twice as many fruit as were required and were
advised that extra fruit could be shared with family and friends but to ensure that they had enough
to last them until the next delivery. The participants were asked to store the fruit in a refrigerator
or cool place. The kiwifruit were selected to be of the same size and quality. The average weight of
one SunGold kiwifruit was 132 g (whole fruit) and 95 g for the flesh (28% accounts for the skin, [16]).
Participants were asked to consume the flesh of two SunGold kiwifruit every day for twelve weeks.
Strategies to avoid forgetting the fruit were discussed such as eating them at the same time every day
and having two kiwifruit out on the bench every day so the participants knew whether or not they had
eaten them. Participants were asked not to eat any kiwifruit other that the two study fruit during the
12 weeks. As the study was a free-living situation, participants were asked to consume the fruit as part
of their usual diet. Participants were asked not to eat the skin and to eat the flesh raw and not crushed
or blended in a smoothie. Participants were asked to maintain their normal dietary and lifestyle habits
for the duration of the trial.
Following screening and the lead-in week there were three study visits; baseline, week six and
week twelve (Figure 1). Table 1 shows the information that was collected at each study visit.
79
Nutrients 2018, 10, 895
2.4.1. Glucose
Fasting glucose was measured in blood collected in fluoride oxalate venoject tubes by standard
methods (Glucose Hexokinase Enzymatic Assay, Abbott c series analyser, Abbott Park, IL, USA) at
an IANZ laboratory. The coefficient of variation associated with the measurement of glucose in plasma
is 1.5% at 6.78 mmol/L [28].
2.4.2. Vitamin C
Frozen plasma samples were rapidly defrosted, acidified and stabilised with perchloric acid and
a metal chelator. The supernatants were then treated with a reducing agent prior to analysis to recover
any vitamin C that had become oxidised during handling or storage [29]. The samples were analysed by
80
Nutrients 2018, 10, 895
the gold standard method of high performance liquid chromatography with electrochemical detection
as described previously [17,29].
2.4.3. HbA1c
Determined in EDTA blood by standard methods (Bio-rad Variant HPLC, Bio-Rad, Hercules, CA, USA)
at an IANZ laboratory. The coefficient of variation associated with the measurement of HbA1c at the IANZ
is 0.66% at HbA1c of 38 mmol/mol [30].
2.4.5. hs-CRP
The inflammatory marker hs-CRP was measured using end-point nephelometry at an IANZ laboratory.
2.4.6. Hormones
EDTA plasma was collected and centrifuged for 10 minutes at 1000× g (2500 rpm) at 4 ◦ C. The plasma
was frozen at −80 ◦ C and stored for batched analyses. Ghrelin, leptin and adiponectin were determined by
the Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch.
Ghrelin was measured by an in-house RIA following extraction from plasma using Sep Pak C18
cartridges, as described previously [31]. The assay recognises the total circulating ghrelin (i.e., both
octanoyl and non-octanoyl forms). The cross reactivities of other peptides in the assay, including
vasointestinal peptide, prolactin, galanin, growth hormone releasing hormone, neuropeptide Y, brain
natriuretic peptide, atrial natriuretic peptide, endothelin-1 and angiotensin II were all less than 0.03%.
The RIA had a mean detection limit of 10.8 ± 0.8 pmol/L and mean ED50 of 136.2 ± 10.0 pmol/L over
23 consecutive assays.
Leptin and adiponectin were measured using commercial ELISA from BioVendor (Brno, Czech Republic),
Research and Diagnostic products (RD191001100 Human Leptin ELISA and RD191023100 Human
Adiponectin ELISA) according to the manufacturer’s instructions.
Insulin was measured using the Roche Cobas e411 method in an IANZ laboratory. After storage
at −80 ◦ C, thawed plasma was pre-treated using 25% polyethylene glycol to precipitate antibodies.
81
Nutrients 2018, 10, 895
gene and sequencing on a MiSeq (Illumina, San Diego, CA, USA) instrument. DNA sequences were
processed using the QIIME v. 1.9.1 and vsearch v.2.0.2 suite of programs [32,33]. Genus level taxonomy
was obtained by filtering Operational Taxonomic Unit (OTU) tables containing taxonomic data
generated using the RDP classifier, extracting representative sequences and using BLAST to identify
genus level matches, where possible, within the NCBI database. Alpha- and beta-diversity analysis
of phylogenetic data compared the coverage and richness (number of phylotypes) and similarity or
difference in the composition of the stool microbiota between subjects and treatment groups.
2.6. Questionnaires
82
Nutrients 2018, 10, 895
the two SunGold kiwifruit daily (week 12). At week 12 both fresh and total fruit, with and without
the SunGold kiwifruit, were created for comparison with baseline fruit intake. Kiwifruit consumption
recorded in the week 12 food diary was used as an indication of compliance.
Percent energy from macronutrients per day was calculated from the average total daily dietary
intake as follows [35]:
3. Results
The average age of the participants was 66 years and ranged from 44 to 85 years old. There was
an equal number of male and female participants and the majority of participants were European
(81%) (Table 2). There was a mix of educational qualifications, which is expected given the age range
of the participants. There were fewer participants currently smoking (15%) compared to the number of
participants who reported being ex- (39%) or non-smokers (46%). The majority of participants reported
consuming alcohol (73%) (Table 2).
Characteristics n = 26
Age (years) (mean ± SD) 66 ± 9
Gender
Female % (n) 50 (13)
Male % (n) 50 (13)
Ethnicity
European % (n) 81 (21)
Māori % (n) 8 (2)
Samoan % (n) 4 (1)
Asian/Chinese % (n) 4 (1)
Other % (n) 4 (1)
Qualification
No qualification % (n) 27 (7)
83
Nutrients 2018, 10, 895
Characteristics n = 26
Secondary school % (n) 19 (5)
Post-secondary certificate, diploma or trade diploma % (n) 42 (11)
University % (n) 12 (3)
Smoking Status
Current smoker % (n) 15 (4)
Ex-smoker % (n) 39 (10)
Non-smoker % (n) 46 (12)
Alcohol Status
Current drinker % (n) 73 (19)
Ex-drinker % (n) 12 (3)
Non-drinker % (n) 15 (4)
84
Nutrients 2018, 10, 895
Table 3. Cont.
100%
80%
% of Individuals
60% ш SDT
40% RDI-<SDT
EAR-<RDI
20%
<EAR
0%
Week 0 Week 12
Time
Figure 2. Dietary vitamin C intake of individuals at weeks 0 and week 12 meeting the estimated average
requirement (EAR) (30 mg/day), recommended dietary intake (RDI) (45 mg/day) and suggested dietary
target (SDT) to reduce chronic disease risk (220 mg/day for men and 190 mg/day for women) [36].
85
Nutrients 2018, 10, 895
dose reduced by half from week 6 to 12 and another participant had it increased between baseline and
week 6 but then reduced back to the baseline dose between week 6 and 12.
86
Nutrients 2018, 10, 895
(r = −0.450, p = 0.024), insulin (r = −0.520, p = 0.008) and waist circumference (r = −0.399, p = 0.048)
were inversely associated with plasma vitamin C concentrations.
100%
90%
80%
70%
% of Individuals
60%
Saturating
50%
Adequate
40%
Inadequate
30%
20% Marginal
10%
0%
Week 0 Week 12
Time
Figure 3. Plasma vitamin C status of individuals at weeks 0 and 12 classified as having saturating
(≥70 μmol/L), adequate (50–69 μmol/L), inadequate (24–49 μmol/L) and marginal (11–23 μmol/L)
plasma vitamin C. There were no participants classified as having deficient (<11 μmol/L) plasma
vitamin C concentrations [38].
87
Nutrients 2018, 10, 895
Beta-diversity (unique fraction metric (Unifrac) weighted and unweighted) was used to compare
similarities in microbiota composition over time. A comparison of distances within and between time
points did not reveal significant changes in community structure with time (Figure 4B,C). Unweighted
unifrac distances (kinds of bacteria and phylogenetic relationships) indicated that the microbiotas of the
participants tended to be more similar to each other after 12 weeks intervention than at baseline or 6 weeks
(Figure 4B). Applying a weighting (based on the relative abundance of OTUs) suggested that the microbiota
of participants are more similar to each other at both 6 and 12 weeks than at baseline (Figure 4C).
Figure 4. Faecal microbiota diversity metrics. (A) Rarefaction curves based on Observed Species metric of
alpha diversity, showing coverage of DNA sequences obtained from faecal DNA. Means and 95% CI are
shown. (B,C) Beta diversity metrics comparing microbiota similarity distances within and between groups.
Means and SEM are shown. (B) Unweighted Unifrac (kinds of bacteria and phylogenetic relatedness).
(C) Weighted Unifrac (kinds of bacteria, phylogenetic relatedness and relative abundances).
88
Nutrients 2018, 10, 895
The complete results of taxonomic analysis of the faecal microbiota at the Family level are supplied
in Supplementary Material Table S1. Sequences from members of the bacterial phylum Actinobacteria
and a bacterial family within this taxon, Coriobacteriaceae, were more abundant in faecal samples
collected during the kiwifruit intervention compared to baseline (Figure 5A,B). The original OTU table
contained 10 clusters identified as Coriobacteriacae, including one OTU each in the genera; Atopobium,
Collinsella, Eggerthella, Gordonibacter and Senegalimassilia. The other five OTUs were ‘unidentified’
(i.e., never cultured and characterised). Combining the abundances from these OTUs replicated the
significant increase in relative abundance of the Family Coriobacteriaceae observed with time (Figure 5C).
Thus, it appears that as yet uncultivated member(s) of the Coriobacteriaceae contributed to a significant
increase in relative abundance during the intervention.
Figure 5. Relative abundances of bacterial groups in faeces of participants at baseline and during
supplementation of the diet with kiwifruit. Box and whiskers plots showing individual values and
means as horizontal lines. (A) Phylum Actinobacteria. (B) Family Coriobacteriaceae. (C) OTUs of
uncharacterized bacteria of the Family Coriobacteriaceae. Statistical evaluation by Wilcoxon matched
pairs test using Prism 7.
89
Nutrients 2018, 10, 895
Dietary intervention of two SunGold kiwifruit per day resulted in significantly greater faecal
water content at weeks 6 (76%, p < 0.001) and 12 (74%, p = 0.01) compared to baseline (68%).
4. Discussion
In this study, consuming two SunGold kiwifruit per day was associated with a significant increase in
plasma vitamin C and fasting glucose, and a decrease in HbA1c, however, these latter two changes were
small and, although statistically significant, were not clinically significant. There was a significant reduction
in both diastolic and systolic blood pressure. A significant reduction in waist circumference and waist-to-hip
ratio was also seen, despite small and non-significant reductions in average BMI and fat mass.
Consumption of two SunGold kiwifruit per day (total of 190 g flesh) for three months provided
approximately 300 mg of vitamin C daily from the kiwifruit alone. This increase in dietary vitamin
C from the SunGold kiwifruit resulted in most participants meeting the New Zealand Ministry of
Health’s suggested dietary target to reduce chronic disease risk [35]. This increase in dietary vitamin C
resulted in a significant increase in plasma vitamin C concentrations (mean increase of 14 μmol/L).
The increase in plasma vitamin C concentrations was greatest in those who had started the trial with
inadequate plasma vitamin C concentration (i.e., <50 μmol/L), compared to those who already had
adequate plasma vitamin C concentrations at baseline. This result is consistent with findings from an
earlier kiwifruit trial where those with lower baseline plasma vitamin C concentrations showed an
earlier and greater response to supplementation, compared to those with adequate plasma vitamin C
concentrations at baseline, who showed little effect from supplementation [17]. Three participants (13%)
had only adequate plasma vitamin C concentrations at week 12 which may indicate non-compliance
or be related to some lifestyle factor, such as smoking, or chronic disease [39].
While all participants met the ADA criteria for prediabetes using HbA1c (39–46 mmol/mol)
at baseline, there was still a range of anthropometric and laboratory measures for participants.
For example, fasting glucose ranged from 3.9 to 7.5 mmol/L, insulin ranged from 15 to 220 pmol/L
and waist circumference ranged from 70 to 125 cm. Fasting glucose, insulin and waist circumference
were all inversely associated with plasma vitamin C concentrations at baseline. This result is consistent
with previous research [6–10,37,38].
It has been suggested that vitamin C concentrations at baseline may represent a critical factor in
predicting the metabolic responses to nutritional interventions tackling oxidative stress and may in
part explain why there have been inconsistent findings in RCTs investigating supplementation with
vitamin C and glycaemic control [17,39]. Individuals in this study were supplemented with vitamin
C-rich kiwifruit; however, there were only three participants with marginal vitamin C levels and no
participants with deficient levels at baseline. This may explain why no clinically significant differences
in metabolic markers were observed between baseline and week 12. Furthermore, the length of the
intervention may need to be longer than 12 weeks to see biological changes [14].
There was a small statistically significant reduction in both diastolic and systolic blood pressure over the
duration of the study. This result is unlikely to be due to changes in blood pressure medication as only two
participants reported changes to the dose of their blood pressure medication during the study. One of these
participants actually had their blood pressure medication dose reduced by half between week 6 and 12 and
the other participant was taking the same dose at week 12 as they were at baseline despite having it increased
earlier in the study. Those with inadequate vitamin C levels at baseline (i.e., <50 μmol/L) had a greater
reduction in blood pressure (decrease of 7 ± 9 mmHg diastolic and 10 ± 11 mmHg systolic) compared to
those who had adequate levels (decrease of 2 ± 8 mmHg diastolic and 5 ± 10 mmHg systolic) when week
12 was compared to baseline. This may be related to their level of oxidative stress as hypertension has been
related to increased oxidative stress and reduced antioxidant status [40]. Kiwifruit contain a wide range of
natural antioxidants; they are rich in vitamin C and also contain vitamin E, polyphenols and flavonoids
which are all potent antioxidants [41]. This result is consistent with a RCT of smokers, who are also known to
have lower vitamin C concentrations, which showed three kiwifruit per day for eight weeks was associated
with reductions of 10 mmHg in systolic blood pressure and 9 mmHg in diastolic blood pressure [42].
90
Nutrients 2018, 10, 895
Despite the small increase in fruit intake at week 12, there were no significant differences in energy,
fibre, protein, total fat and total carbohydrate intake when dietary data at week 12 was compared to baseline.
This result suggests participants did not make major changes to their diet during the trial. The addition
of the SunGold kiwifruit also had no significant impact on their macronutrient intake as it is relatively
low in calories (63 kcal/100 g), protein (1.0 g/100 g), fat (0.3 g/100 g) and carbohydrate (16 g/100 g) [16].
However, SunGold kiwifruit does contain 12 g/100 g of sugar [16], which may have contributed to the
significantly higher intake of fructose (increase of 6 g) and glucose (increase of 6 g) at week 12 compared to
baseline. SunGold kiwifruit also contains vitamin E (1.4 mg/100 g) and folate (31 μg/100 g), so would have
contributed to the higher intake of these vitamins at week 12 compared to baseline.
Although faecal microbiota diversity was stable across the 12-week intervention when viewing
the cohort as a whole, Coriobacteriaceae family members showed a significant increase in relative
abundance during the course of the study. The assignment of these bacteria to uncharacterised
bacterial species was particularly interesting because it suggests that as yet uncultivated bacteria that
use substrates associated with kiwifruit (such as pectins) exist in the human faecal microbiota. Future
work should include efforts to cultivate these bacteria and to determine their functional characteristics.
The Coriobacteriaceae are in general a poorly studied bacterial family, however they can chemically
transform plant polyphenols [43] and thus their activities may be of interest in promoting human
health [44]. Certainly, future work should investigate the potential link between Coriobacteriaceae and
polyphenolic compounds in the faeces of kiwifruit-fed subjects and chemical derivatives of these
substances in the blood circulation. While the increased relative abundances of Coriobacteriaceae were
small, this does not preclude an important role for the bacteria in bowel ecology because low abundance
bacteria in microbial communities can have a disproportionally large effect relative to abundance [45].
Our human intervention trial did not confirm the reported in vitro effect of kiwifruit fermentation [25].
In the vitro study, Bacteroides spp., Parabacteroides spp. and Bifidobacterium spp. were reported to increase
in cultures containing kiwifruit and inoculated with human faeces. Increased Coriobacteriaceae relative
abundances were the only impact of dietary modification seen in the participants in our study.
Dietary intervention in the form of two gold kiwifruit per day led to an increase in stool water
content at both 6 and 12 weeks. This indicates a shortened gut transit time (laxative effect) and has been
linked to decreases in community diversity in other studies. Stool consistency is strongly associated
with gut microbiota richness and composition, enterotypes and bacteria growth rates [46]. In our study,
the observation may be confounded by the fact that kiwifruit contains 1.4 g/100 g of fibre [16], which is
comprised of both soluble and insoluble components at a ratio of approximately 1:3 [19]. The soluble
fibre is made up almost exclusively of pectic polysaccharides that have the ability to retain water
and form gels, which increases the size and softness of the faeces and aids stimulation of peristaltic
movements [19]. The soluble fibre may be the cause of the increase in faecal water seen in this study.
Results from several clinical trials also show that kiwifruit promotes laxation [47–50].
This was the first study to measure changes in the gut microbiota and vitamin C status associated
with a whole fruit (SunGold kiwifruit) in people with prediabetes. The participants were free-living
representing a realistic intervention for the general population. The use of a whole food rather
than an extract/supplement in this study also makes it more affordable and available for people.
Although there was no control group in this study, participants acted as their own control with baseline
measurements for comparison and dietary intake was consistent over time. In this study plasma
vitamin C status corroborated the food diaries as an objective measure of compliance. The study
duration of three months enabled the use of HbA1c as a measure of glycaemic control and allowed us
to investigate temporary and longer-term shifts in the microbiota.
As this was a pilot exploratory study with a limited number of participants it may not have
been sufficient to identify some clinically relevant associations. Additionally, we did not pre-screen
participants for vitamin C intake or status and, as a result, half the study cohort already had adequate
plasma vitamin C status at baseline. Furthermore, because we did not include an arm with vitamin C
supplementation alone, it is difficult to ascertain the contribution of the vitamin C content of the fruit
91
Nutrients 2018, 10, 895
to the observed effects. When assessing dietary intakes there are also limitations around misreporting
and a potential bias in recording “good/bad” foods.
5. Conclusions
Supplementation with two SunGold kiwifruit per day significantly increased plasma vitamin C
concentrations and provided small improvements in several markers of metabolic and cardiovascular
health. The results of faecal microbiota analysis point to the need for novel bacteriological
investigations of Coriobacteriaceae in order to explain their faecal abundances and to investigate
mechanistic relationships with regard to kiwifruit polysaccharides and polyphenols. Due to the
small sample size of this pilot study a larger study is indicated, particularly focusing on recruiting
participants with low vitamin C status at baseline and determinations of polyphenol derivatives in
faeces and blood.
References
1. International Diabetes Federation. IDF Diabetes Atlas 2015. Available online: www.diabetesatlas.org
(accessed on 12 June 2017).
2. World Health Organization, Diabetes Fact Sheet N◦ 312 [Internet], World Health Organization Media Centre.
Available online: http://www.who.int/mediacentre/factsheets/fs312/en/index.html (accessed on 12 June 2017).
3. Coppell, K.J.; Mann, J.I.; Williams, S.M.; Jo, E.; Drury, P.L.; Miller, J.C.; Parnell, W.R. Prevalence of diagnosed
and undiagnosed diabetes and prediabetes in New Zealand: Findings from the 2008/09 Adult Nutrition
Survey. N. Z. Med. J. 2013, 126, 23–42. [PubMed]
4. Brownlee, M. Biochemistry and molecular cell biology of diabetic complications. Nature 2001, 414, 813.
[CrossRef] [PubMed]
5. Evert, A.B.; Boucher, J.L.; Cypress, M.; Dunbar, S.A.; Franz, M.J.; Mayer-Davis, E.J.; Neumiller, J.J.;
Nwankwo, R.; Verdi, C.L.; Urbanski, P.; et al. Nutrition therapy recommendations for the management of
adults with diabetes. Diabetes Care 2014, 37, S120–S143. [CrossRef] [PubMed]
6. Sargeant, L.; Wareham, N.; Bingham, S.; Day, N. Vitamin C and hyperglycemia in the European prospective
investigation into cancer-Norfolk (EPIC-Norfolk) study: A population-based study. Diabetes Care 2000, 23,
726–732. [CrossRef] [PubMed]
7. Feskens, E.J.M.; Virtanen, S.M.; Räsänen, L.; Tuomilehto, J.; Stengård, J.; Pekkanen, J.; Nissinen, A.; Kromhout, D.
Dietary factors determining diabetes and impaired glucose tolerance: A 20-year follow-up of the Finnish and
Dutch cohorts of the Seven Countries Study. Diabetes Care 1995, 18, 1104–1112. [CrossRef] [PubMed]
8. Will, J.; Ford, E.; Bowman, B. Serum vitamin C concentrations and diabetes: Findings from the third National
Health and Nutrition Examination Survey, 1988–1994. Am. J. Clin. Nutr. 1999, 70, 49–52. [CrossRef] [PubMed]
9. Harding, A.-H.; Wareham, N.J.; Bingham, S.A.; Khaw, K.; Luben, R.; Welch, A.; Forouhiet, N.G. Plasma
vitamin C level, fruit and vegetable consumption and the risk of new-onset type 2 diabetes mellitus:
The European prospective investigation of cancer-Norfolk prospective study. Arch. Intern. Med. 2008, 168,
1493–1499. [CrossRef] [PubMed]
92
Nutrients 2018, 10, 895
10. Zhou, C.; Na, L.; Shan, R.; Cheng, Y.; Li, Y.; Wu, X.; Sun, C. Dietary Vitamin C Intake Reduces the Risk of Type 2
Diabetes in Chinese Adults: HOMA-IR and T-AOC as Potential Mediators. PLoS ONE 2016, 11, e0163571. Available
online: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5042374/ (accessed on 12 June 2017). [CrossRef]
[PubMed]
11. Ellulu, M.S.; Rahmat, A.; Patimah, I.; Khaza’ai, H.; Abed, Y. Effect of vitamin C on inflammation and metabolic
markers in hypertensive and/or diabetic obese adults: A randomized controlled trial. Drug Des. Dev. Ther. 2015, 9,
3405–3412. [CrossRef] [PubMed]
12. Gutierrez, A.; Duran-Valdez, E.; Robinson, I.; De Serna, D.; Schade, D. Does Short-term Vitamin C Reduce
Cardiovascular Risk in Type 2 Diabetes? Endocr. Pract. 2013, 19, 785–791. [CrossRef] [PubMed]
13. Dakhale, G.N.; Chaudhari, H.V.; Shrivastava, M. Supplementation of vitamin C reduces blood glucose
and improves glycosylated hemoglobin in type 2 diabetes mellitus: A randomized, double-blind study.
Adv. Pharmacol. Sci. 2011, 2011, 195271. [CrossRef] [PubMed]
14. Ashor, A.W.; Werner, A.D.; Lara, J.; Willis, N.D.; Mathers, J.C.; Siervo, M. Effects of vitamin C supplementation on
glycaemic control: A systematic review and meta-analysis of randomised controlled trials. Eur. J. Clin. Nutr. 2017,
71, 1371–1380. [CrossRef] [PubMed]
15. Gillani, S.W.; Sulaiman, S.A.S.; Abdul, M.I.M.; Baig, M.R. Combined effect of metformin with ascorbic
acid versus acetyl salicylic acid on diabetes-related cardiovascular complication: A 12-month single blind
multicenter randomized control trial. Cardiovasc. Diabetol. 2017, 16, 103. [CrossRef] [PubMed]
16. Sivakumaran, S.; Huffman, L.; Sivakumaran, S.; Drummond, L. The nutritional composition of Zespri
SunGold Kiwifruit and Zespri Sweet Green Kiwifruit. Food Chem. 2018, 238, 195–202. [CrossRef] [PubMed]
17. Carr, A.C.; Pullar, J.M.; Moran, S.; Vissers, M.C. Bioavailability of vitamin C from kiwifruit in non-smoking
males: Determination of ‘healthy’ and ‘optimal’ intakes. J. Nutr. Sci. 2012, 1, e14. [CrossRef] [PubMed]
18. Monro, J.A. Kiwifruit, Carbohydrate Availability and the Glycemic Response. Adv. Food Nutr. Res. 2013, 68,
257–271. [PubMed]
19. Sims, I.M.; Monro, J.A. Fiber: Composition, structures and functional properties. Adv. Food Nutr. Res. 2013,
68, 81–99. [PubMed]
20. Qin, J.; Li, Y.; Cai, Z.; Li, S.; Zhu, J.; Zhang, F.; Liang, S.; Zhang, W.; Guan, Y.; Shen, D.; et al. A Metagenome-
wide association study of gut microbiota in type 2 diabetes. Nature 2012, 490, 55–60. [CrossRef] [PubMed]
21. Larsen, N.; Vogensen, F.K.; Van den Berg, F.W.J.; Nielsen, D.S.; Andreasen, A.S.; Pedersen, B.K.; Al-Soud, W.A.;
Sørensen, S.J.; Hansen, L.H.; Jakobsenet, M. Gut Microbiota in Human Adults with Type 2 Diabetes Differs
from Non-Diabetic Adults. PLoS ONE 2010, 5, e9085. [CrossRef] [PubMed]
22. Karlsson, F.H.; Tremaroli, V.; Nookaew, I.; Bergström, G.; Behre, C.J.; Fagerberg, B.; Nielsen, J.; Bäckhed, F.
Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 2013, 498,
99–103. [CrossRef] [PubMed]
23. Zhang, X.; Shen, D.; Fang, Z.; Jie, Z.; Qiu, X.; Zhang, C.; Chen, Y.; Ji, L. Human gut microbiota changes reveal
the progression of glucose intolerance. PLoS ONE 2013, 8, e71108. [CrossRef] [PubMed]
24. Cani, P.D. The gut microbiota manages host metabolism. Nat. Rev. Endocrinol. 2014, 10, 74–76. [CrossRef]
[PubMed]
25. Blatchford, P.; Bentley-Hewitt, K.L.; Stoklosinski, H.; McGhie, T.; Gearry, R.; Gibson, G.; Ansell, J. In vitro
characterisation of the fermentation profile and prebiotic capacity of gold-fleshed kiwifruit. Benef. Microbes
2015, 6, 829–839. [CrossRef] [PubMed]
26. Braatvedt, G.D.; Cundy, T.; Crooke, M.; Florkowski, C.; Mann, J.I.; Lunt, H.; Rodney, J.; Orr-Walker, B.; Timothy, K.;
Drury, P.L. Understanding the new HbA1c units for the diagnosis of Type 2 diabetes. N. Z. Med. J. 2012, 125, 70.
[PubMed]
27. World Health Organization. Section 5: Collecting Step 2 Data: Physical Measurements. Available online:
http://www.who.int/chp/steps/Part3_Section5.pdf?ua=1 (accessed on 19 June 2017).
28. Ricós, C.; Alvarez, V.; Cava, F.; García-Lario, J.V.; Hernández, A.; Jiménez, C.V.; Minchinela, J.; Perich, C.;
Simón, M. Current databases on biological variation: Pros, cons and progress. Scand. J. Clin. Lab. Investig.
1999, 59, 491–500.
29. PullaR, J.M.; Bozonet, S.M.; Carr, A.C. Appropriate Handling, Processing and Analysis of Blood Samples Is
Essential to Avoid Oxidation of Vitamin C to Dehydroascorbic Acid. Antioxid 2018, 7, 29. [CrossRef] [PubMed]
30. Bozkaya, G.; Uzuncan, N.; Bilgili, S.; Demirezen, O. Evaluation of analytical performance of Variant II turbo
HbA1c analyzer according to sigma metrics. J. Med. Biochem. 2018, 37, 1–13. [CrossRef]
93
Nutrients 2018, 10, 895
31. Bang, A.S.; Soule, S.G.; Yandle, T.G.; Richards, A.M.; Pemberton, C.J. Characterisation of proghrelin peptides
in mammalian tissue and plasma. J. Endocrinol. 2007, 192, 313–323. [CrossRef] [PubMed]
32. Caporaso, J.G.; Kuczynski, J.; Stombaugh, J.; Bittinger, K.; Bushman, F.D.; Costello, E.K.; Fierer, N.; Peña, A.G.;
Goodrich, J.K.; Gordonet, J.I.; et al. QIIME allows analysis of high-throughput community sequencing data.
Nat. Methods 2010, 7, 335. [CrossRef] [PubMed]
33. Rognes, T.; Flouri, T.; Nichols, B.; Quince, C.; Mahé, F. VSEARCH: A versatile open source tool for
metagenomics. PeerJ 2016, 4, e2584. [CrossRef] [PubMed]
34. Wilson, R.; Willis, J.; Gearry, R.; Skidmore, P.; Fleming, E.; Frampton, C.; Carr, A. Inadequate Vitamin C
status in prediabetes and type 2 diabetes mellitus: Associations with glycaemic control, obesity, and smoking.
Nutrients 2017, 9, 997. [CrossRef] [PubMed]
35. NHMRC. Nutrient Reference Values for Australia and New Zealand Including Recommended Dietary Intakes
Canberra: ACT: National Health and Medical Research Council. 2006. Available online: https://www.
nhmrc.gov.au/_files_nhmrc/file/publications/17122_nhmrc_nrv_update--dietary_intakes--web.pdf (accessed
on 12 June 2017).
36. IPAQ. Guidelines for Data Processing and Analysis of the International Physical Activity Questionnaire
(IPAQ): Short and Long Forms. 2005. Available online: https://sites.google.com/site/theipaq/ (accessed on
12 June 2017).
37. Will, J.C.; Byers, T. Does diabetes mellitus increase the requirement for vitamin C? Nutr. Rev. 1996, 54,
193–202. [CrossRef] [PubMed]
38. Kositsawat, J.; Freeman, V.L. Vitamin C and A1c relationship in the National Health and Nutrition
Examination Survey (NHANES) 2003–2006. J. Am. Coll. Nutr. 2011, 30, 477–483. [CrossRef] [PubMed]
39. Lykkesfeldt, J.; Poulsen, H.E. Is vitamin C supplementation beneficial? Lessons learned from randomised
controlled trials. Br. J. Nutr. 2010, 103, 1251–1259. [CrossRef] [PubMed]
40. Tandon, R.; Sinha, M.K.; Garg, H.; Khanna, R.; Khanna, H.D. Oxidative stress in patients with essential
hypertension. Natl. Med. J. India 2005, 18, 297–299. [PubMed]
41. Boland, M. Advances in Food and Nutrition Research. In Nutritional Benefits of Kiwifruit; Moughan, P.J., Ed.;
Elsevier Science: Burlington, MA, USA, 2013.
42. Karlsen, A.; Svendsen, M.; Seljeflot, I.; Laake, P.; Duttaroy, A.K.; Drevon, C.A.; Arnesen, H.; Tonstad, S.;
Blomhoffet, R. Kiwifruit decreases blood pressure and whole-blood platelet aggregation in male smokers.
J. Hum. Hypertens. 2012, 27, 126. [CrossRef] [PubMed]
43. Clavel, T.; Lepage, P.; Charrier, C. The family coriobacteriaceae. In The Prokaryotes, 4th ed.; Rosenberg, E.,
DeLong, E.F., Lory, S., Stackebrandt, E., Thompson, F., Eds.; Springer International Publishing: Berlin, Germany,
2014; pp. 201–238.
44. Del Rio, D.; Costa, L.G.; Lean, M.E.J.; Crozier, A. Polyphenols and health: What compounds are involved?
Nutr. Metab. Cardiovasc. Dis. 2010, 20, 1–6. [CrossRef] [PubMed]
45. Power, M.E.; Tilman, D.; Estes, J.A.; Menge, B.A.; Bond, W.J.; Mills, L.S.; Daily, G.; Castilla, G.C.; Lubchenco, J.;
Paine, R.T. Challenges in the Quest for Keystones. BioScience 1996, 46, 609–620. [CrossRef]
46. Tigchelaar, E.F.; Bonder, M.J.; Jankipersadsing, S.A.; Fu, J.; Wijmenga, C.; Zhernakova, A. Gut microbiota
composition associated with stool consistency. Gut 2015, 65, 540–542. [CrossRef] [PubMed]
47. Rush, E.C.; Patel, M.; Plank, L.D.; Ferguson, L.R. Kiwifruit promotes laxation in the elderly. Asia Pac. J. Clin. Nutr.
2002, 11, 164–168. [CrossRef] [PubMed]
48. Chan, A.O.; Leung, G.; Tong, T.; Wong, N.Y. Increasing dietary fiber intake in terms of kiwifruit improves
constipation in Chinese patients—Increasing dietary fiber intake in terms of kiwifruit improves constipation
in Chinese patients. World J. Gastroenterol. 2007, 13, 4771–4775. [CrossRef] [PubMed]
49. Chang, C.-C.; Lin, Y.-T.; Lu, Y.-T.; Liu, Y.-S.; Liu, J.-F. Kiwifruit improves bowel function in patients with
irritable bowel syndrome with constipation. Asia Pac. J. Clin. Nutr. 2010, 19, 451–457. [PubMed]
50. Cunillera, O.; Almeda, J.; Mascort, J.J.; Basora, J.; Marzo-Castillejo, M. Improvement of functional constipation
with kiwifruit intake in a Mediterranean patient population: An open, non-randomized pilot study.
Revista Española de Nutrición Humana y Dietética 2015, 19, 58–67. [CrossRef]
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
94
nutrients
Review
Nutritional Interventions as Beneficial Strategies to
Delay Cognitive Decline in Healthy
Older Individuals
Blanka Klímová 1, * and Martin Vališ 2
1 Department of Applied Linguistics, University of Hradec Kralove, Rokitanskeho 62,
Hradec Kralove 500 03, Czech Republic
2 Department of Neurology, University Hospital Hradec Kralove, Sokolska 581,
Hradec Kralove 500 05, Czech Republic; [email protected]
* Correspondence: [email protected]; Tel.: +420-604-737-645
Abstract: Current demographic trends indicate that the population is aging. The aging process
is inevitably connected with cognitive decline, which manifests itself in worsening working
memory, processing speed, and attention. Therefore, apart from pharmacological therapies,
non-pharmacological approaches which can influence cognitive performance (such as physical
activities or healthy diet), are being investigated. The purpose of this study is to explore the types
of nutritional interventions and their benefits in the prevention and delay of cognitive delay in
healthy older individuals. The methods used in this study include a literature review of the available
studies on the research topic found in Web of Science, Scopus, and MEDLINE. The findings show that
nutritional intervention has a positive impact on cognitive function in healthy older people. However,
it seems that the interactions between more than one nutrient are most effective. The results reveal
that specifically the Mediterranean diet appears to be effective in this respect. Moreover, the findings
also indicate that multi-domain interventions including diet, exercise, cognitive training, and vascular
risk monitoring have a far more significant effect on the enhancement of cognitive functions among
healthy older individuals.
1. Introduction
Currently, the number of older people is rapidly increasing. For instance, in 2017 there
were 962 million people aged 60+ years worldwide, with their number increasing by 3% every
year. The largest proportion of the aging population can be found in Europe (25%). By 2050,
all continents except Africa will have one quarter or more of their population aged over 60 years [1].
This demographic trend of aging population will cause significant social and economic problems [2].
Therefore, there are efforts to develop relevant strategies and guidelines both at national and
international levels in order to delay this process of aging and prolong the active age of older
individuals, as well as to maintain quality of life [3].
The aging process is inevitably connected with cognitive decline, which manifests itself in
worsening working memory, processing speed, and attention [4], i.e., in so-called fluid intelligence,
which deteriorates at different rate in each individual [5]. The rate and severity of cognitive decline
varies from normal age-related change to chronic neurodegenerative diseases such as Alzheimer’s
disease for which at present there is no cure [6]. Therefore, non-pharmacological approaches which
can influence fluid intelligence, such as physical activities or healthy diet, are being investigated [6–8].
Evidence-based studies [9,10] indicate that specifically the Mediterranean diet (MedDiet) rich in
olive oil and nuts, seems to contribute to the prevention of cognitive impairment among healthy
older people. However, other food products, for example, avocados [11], or nutritional supplements
such as consumption of flavanone-rich 100% orange juice [12] or omega-3 fatty acid [13] also play
an important role in the prevention of cognitive decline in healthy older individuals. In addition,
nutritional interventions in comparison with the pharmacological treatment, are usually well-tolerated,
easy-to-implement, cost-effective, and safe for long-term use [13].
The purpose of this study is to explore the types of nutrition intervention and its benefits in the
prevention and delay of cognitive delay in healthy older individuals.
2. Methods
The authors conducted a literature review of the research studies on the basis of the key words in
three acknowledged databases: Web of Science, Scopus, and MEDLINE. This review was performed
over the period from 2014 to June 2018 since several review studies had been already written on
this topic [14,15]. The key words were as follows: diet AND cognitive decline AND healthy older
people, diet AND cognitive decline AND healthy elderly, nutrition intervention AND cognitive
decline AND healthy older people, nutrition intervention AND cognitive decline AND healthy elderly,
nutrition AND cognitive decline AND healthy older people, nutrition AND cognitive decline AND
healthy elderly, diet AND dementia AND healthy older people, diet AND dementia AND healthy
elderly, nutrition intervention AND dementia AND healthy older people, nutrition intervention AND
dementia AND healthy elderly, nutrition AND dementia AND healthy older people, nutrition AND
dementia AND healthy elderly.
The majority of the studies were detected in the Web of Science database (97 studies), followed
by Scopus (66 studies) and MEDLINE (45 studies). However, in MEDLINE it is possible to look only
for clinical trials, and therefore the selection was easier. Altogether, 208 studies were found via the
database search and 11 from other available sources (i.e., web pages, conference proceedings and
books outside the scope of the databases described above). The titles of all studies were then checked
in order to discover whether they focused on the research topic or not and irrelevant studies were
excluded. In addition, the duplicate studies were also excluded. Afterwards, the authors checked the
content of the abstracts whether the study examined the research topic. Thirty-two studies/articles
were selected for the full-text analysis. Only 12 studies were then able to be used for detailed analysis
of the research topic. The selection of these studies is described below (Figure 1).
96
Nutrients 2018, 10, 905
The study was included if it was a randomized controlled trial, and matched the corresponding
period, i.e., from 2014 up to June 2018. Furthermore, the study was included if it involved healthy
older people, i.e., those without any cognitive impairment or dementia aged 50+ years, and focused
on the research topic, i.e., nutritional intervention in the delay and prevention of cognitive decline
in healthy older individuals. All studies had to be written in English. Thus, studies such as [5] were
excluded, as well as cross-sectional descriptive and cohort studies, for example [16–18].
3. Results
Altogether 12 randomized controlled trials (RCT) were found. The majority of were from Europe
(i.e., Finland, Germany, Italy, Spain, and the UK) [10,12,13,19–24] and the rest from the USA and
Australia [11,25,26]. The nutrition intervention involved dietary supplements (i.e., cocoa flavanols,
Vitis vinifera, docosahexaenoic acid (DHA)-rich fish oil, or omega-3 fatty acid), vegetable (e.g., avocado),
berry and orange beverages, and the MedDiet. Two of the studies were multidomain lifestyle
intervention studies including, apart from healthy diet, physical exercises, social activities, cognitive
training, and vascular risk management [20,24]. The intervention period in the studies ranged from
five weeks to five years. The subject samples also varied; the smallest sample of subjects consisted
of 37 healthy older individuals and the largest included 775 healthy older people. The efficacy of the
nutrition intervention focused on the prevention and delay of cognitive decline in the studies was
measured with available validated cognitive assessment tools such as a battery of cognitive tests aimed
at assessing working memory, attention, or processing speed. Table 1 below provides an overview of
the main findings of these studies. They are summarized in alphabetical order of their first author.
97
Table 1. Overview of the twelve selected studies focused on cognitive decline and its prevention by nutrition intervention.
98
health was discussed;
goal-setting (GS)—an
interview in which they The Lifetime of Experiences
To evaluate a goal-setting
set behaviour change Questionnaire (LEQ), The results show that at 12-month
intervention aimed at 75 healthy elderly
goals relating to Physical Activities Scale for follow-up, the two goal-setting
Clare et al. [20] promoting increased (IC—27 subjects;
physical, cognitive and the Elderly (PASE), a groups increased their level of
RCT cognitive and physical 12 months. GS—24 subjects;
social activity, health battery of cognitive tests, physical (effect size 0.37) and
(UK) activity and improving GM—24 subjects);
and nutrition; and audio-recordings of the cognitive (effect size 0.15) activity
mental and physical age: 50+ years.
goal-setting with interviews, statistical relative to controls.
fitness, diet and health.
mentoring (GM)—the analysis.
goal-setting interview
followed by bi-monthly
telephone mentoring.
The one-to-one
interviews lasted for 90
min.
Table 1. Cont.
99
long-chain 22 healthy older
To investigate the impact Visuospatial
polyunsaturated subjects in the The findings reveal that the daily
Kulzow et al. [13] of omega-3 fatty acid object-location-memory
omega-3 fatty acids intervention intake of LC-n3-FA has a positive
RCT supplementation on 26 weeks. task (LOCATO), standard
(LC-n3-FA) in the group and 22 in impact on memory functions in
(Germany) memory functions in neuropsychological tests,
intervention group and the control group, healthy older people (p = 0.049).
healthy older adults. statistical analysis.
placebo in the control age: 50–75 years.
group.
The findings show that the intake
631 healthy older
of several vitamins and minerals
Lehtisalo et al. To discuss the success of Dietary intervention subjects in the
remained unchanged or increased
[21] dietary counselling counselling: 3 intervention Food records, statistical
Two years. in the intervention group and that
RCT intervention among individual and 8 group group and 629 in analysis.
the dietary counselling may have a
(Finland) healthy older individuals. sessions. the control group,
positive impact on age-related diet
age: 60–77 years.
quality and cognitive performance.
A drink containing 993 90 cognitively
To evaluate the effect of The results reveal that regular CF
Mastroiacovo et al. mg high flavanol (HF), intact elderly A battery of
flavanol consumption on consumption can reduce some
[22] 520 mg intermediate subjects divided neuropsychological tests,
cognitive performance in Eight weeks. measures of age-related cognitive
RCT flavanol (IF), or 48 mg into three groups blood pressure measures,
cognitively intact elderly dysfunction, possibly through an
(Italy) low flavanol (LF) cocoa (HF, IF, LF); age: statistical analysis.
people. improvement in insulin sensitivity.
flavanols (CFs). 61–85 years.
Table 1. Cont.
100
subjects (392 A battery of
Sindi et al. [24] telomere length (LTL) intervention (diet, baseline LTL, particularly for
control, 383 neuropsychological tests,
RCT modified the cognitive exercise, cognitive Two years. executive functioning, indicating
intervention), at blood samples, statistical
(Finland) benefits of a 2-year training, and vascular that the multi-domain lifestyle
the age of 30–77 analysis.
multidomain lifestyle risk management) and intervention was especially
years.
intervention. control (general health beneficial among higher-risk
advice) groups. individuals.
Participants were
randomly assigned to a 447 cognitively
To investigate whether a Mediterranean diet healthy volunteers
Mediterranean diet supplemented with (233 women
Valls-Pedret et al. In an older population, a
supplemented with extra virgin olive oil (1 (52.1%); mean age,
[10] A neuropsychological test Mediterranean diet supplemented
antioxidant-rich foods L/week), a Five years. 66.9 years); three
RCT battery, statistical analysis. with olive oil or nuts is associated
influences cognitive Mediterranean diet groups: two
(Spain) with improved cognitive function.
function compared with a supplemented with intervention
control diet. mixed nuts (30 g/day), groups and one
or a control diet (advice control group.
to reduce dietary fat).
RCT: randomized controlled trial.
Nutrients 2018, 10, 905
Apart from one study [26], results of all RCTs indicate that the nutritional interventions had a
positive impact on the cognitive performance of healthy older individuals, specifically on their working
memory and attention [10,11,23]. Moreover, the findings of the multidomain lifestyle intervention
studies reveal that such interventions exhibit significant impacts on cognitive functioning in later
life [20,24]. The results also show that the change in the cognitive performance can be already detected
after five weeks of intervention [23]. However, this is questionable because according to Kurz and van
Baelen [27], the relevant period for medication assessment by regulatory authorities is 24 weeks at
a minimum.
4. Discussion
The findings described in Table 1 show that nutrition intervention has a positive impact on
cognitive functioning of healthy older people. The trials with the exception of four RCTs [10,19,23,26],
concentrated only on one type of nutritional supplement (i.e., Vitis vinifera, docosahexaenoic acid
(DHA)-rich fish oil, omega-3 fatty acid, avocado, and berry). Three RCTs [12,22,25] focused on
high flavanol-rich drinks, which appeared to have quite a positive effect on cognitive performance
among healthy older individuals. In fact, all nutrients described in Table 1 are part of the traditional
multi-nutrient MedDiet pattern, and the research [28] indicates that the interaction of specific foods
and nutrients, especially in the MedDiet, is more powerful on the aging brain than individual nutrients
or a low-fat diet. In fact, the MedDiet seems to be a nutritional model for healthy dietary habits
since it contains all the needed nutrients: monounsaturated fatty acids, polyunsaturated fatty acids,
antioxidants (e.g., allium sulphur compounds, anthocyanins, beta-carotene-flavonoids, catechins,
carotenoids, indoles, or lutein), vitamins (A, B1, 6, 9, 12, D, E), and minerals (magnesium, potassium,
calcium, iodine, zinc, selenium) [28]. In addition, the combination of these nutrients positively
affects pathological neurodegenerative processes such as oxidative stress, neuroinflammation, insulin
resistance, and reduced cerebral blood flow [29]. Nevertheless, the MedDiet appears to improve
cognitive performance in case there is a high adherence to this diet [30–32]. Furthermore, the MedDiet
has a positive impact on neuropsychological [19] and physical state of older people [16] and on vascular
diseases and diabetes [22–24,33]. Although the results mainly concerned western developed countries,
similar research (however, non-RCT) was conducted among healthy older adults in Asian countries
such as Taiwan [34] and Japan [35]. The findings of these studies also show a positive correlation
between the healthy diet rich in vegetables, soy products, fruit, and fish and the prevention and delay
of cognitive decline in older age. Wright et al. [36] in their study carried out among 2090 African
Americans and whites emphasized that higher diet quality was associated with higher performance
on tests of attention and cognitive flexibility, visuospatial ability, and perceptual speed. This was also
evidenced by Smyth et al. [18] who in their cohort study claimed that higher diet quality is associated
with a reduced risk of cognitive decline. Hosking et al. [37] report that adequate nutrition is essential
for cognitive development in childhood and cognitive ability in childhood is strongly associated with
cognitive performance across the lifetime.
Moreover, it seems that multi-nutrient dietary intervention should be implemented in the delay
of cognitive decline among healthy older individuals. The multi-nutrient intervention is also one of
the priorities discussed at the First WHO Ministerial Conference on Global Action against Dementia in
March 2015 [38]. Furthermore, Shilsky et al. [39] suggest in their study that healthcare systems should
play a more significant role in integrating nutrition care for healthy older individuals and that the
nutrition assessment should be incorporated in the medical records. This was also confirmed by the
discussed study [21], for which findings revealed that the dietary counselling had a positive impact on
age-related diet quality and cognitive performance.
The findings of the reviewed RCTs [20,24] indicate that the multi-domain interventions including
healthy diet, physical exercises, cognitive training, and vascular risk monitoring have a far more
significant effect on the enhancement of cognitive functions among healthy older individuals [40].
In fact, these non-pharmacological strategies (healthy diet, physical exercises and cognitive training)
101
Nutrients 2018, 10, 905
have been confirmed by other research studies [7,8] and they should be all integrated at an optimal
level into daily regime of healthy older individuals in order to prolong their active and quality life.
This was also proposed at the International Conference on Nutrition and the Brain in Washington in
2013 [41].
On the contrary, research implies that dietary patterns rich in fat and sugar, with high intake of
meat/poultry or eggs, have negative, harmful effects on cognitive functioning in older age [34,35,37].
The limitations of the reviewed studies consist in the small sample sizes, different types of
nutrition interventions and outcome measures, and a lack of follow-up assessments, as well as
the fact that not all studies were specifically designed to examine cognitive performance. All these
insufficiencies might generate overestimated conclusions in this review study [42,43]. Therefore, more
RCTs should be conducted to prove the efficacy of nutritional interventions on the prevention and
delay of cognitive decline.
Author Contributions: Conceptualization, B.K. and M.V.; Methodology, M.V.; Software, B.K.; Validation, B.K.
and M.V.; Formal Analysis, B.K.; Investigation, B.K.; Resources, B.K.; Data Curation, B.K.; Writing—Original
Draft Preparation, B.K.; Writing—Review and Editing, M.V.; Visualization, M.V.; Supervision, M.V.; Project
Administration, M.V.; Funding Acquisition, M.V.
Funding: This research received no external funding.
Acknowledgments: This paper was supported by the research project Excellence 2018, Faculty of Informatics and
Management, University of Hradec Kralove, Czech Republic, as well as by MH CZ – DRO (UHHK 00179906) and
PROGRES Q40 run at the Medical Faculty Charles University, Czech Republic.
Conflicts of Interest: The authors declare no conflicts of interest.
References
1. United Nations. Ageing. Available online: http://www.un.org/en/sections/issues-depth/ageing/
(accessed on 17 June 2018).
2. Klimova, B.; Maresova, P.; Valis, M.; Hort, J.; Kuca, K. Alzheimer’s disease and language impairments: Social
intervention and medical treatment. Clin. Interv. Aging 2015, 10, 1401–1408. [PubMed]
3. Marešová, P.; Klímová, B.; Kuča, K. Alzheimer’s disease: Cost cuts call for novel drugs development and
national strategy. Čes. Slov. Farm. 2015, 64, 25–30.
4. Klimova, B. Computer-Based Cognitive Training in Aging. Front. Aging Neurosci. 2016, 8, 313. [CrossRef]
[PubMed]
5. Hardman, R.J.; Kennedy, G.; Macpherson, H.; Scholey, A.B.; Pipingas, A. A randomised controlled trial
investigating the effects of Mediterranean diet and aerobic exercise on cognition in cognitively healthy older
people living independently within aged care facilities: The Lifestyle Intervention in Independent Living
Aged Care (LIILAC) study protocol [ACTRN12614001133628]. Nutr. J. 2015, 14, 53. [PubMed]
6. Knight, A.; Bryan, J.; Wilson, C.; Hodgson, J.; Murphy, K. A randomised controlled intervention trial
evaluating the efficacy of a Mediterranean dietary pattern on cognitive function and psychological wellbeing
in healthy older adults: The MedLey study. BMC Geriatr. 2015, 15, 55. [CrossRef] [PubMed]
7. Klimova, B.; Maresova, P.; Kuca, K. Non-pharmacological approaches to the prevention and treatment of
Alzheimer’s disease with respect to the rising treatment costs. Curr. Alzheimer Res. 2016, 13, 1249–1258.
[CrossRef] [PubMed]
8. Klimova, B.; Valis, M.; Kuca, K. Cognitive decline in normal aging and its prevention: A review on
non-pharmacological lifestyle strategies. Clin. Interv. Aging 2017, 12, 903–910. [CrossRef] [PubMed]
9. Martinez-Lapiscina, E.H.; Clavero, P.; Toledo, E.; San Julian, B.; Sanchez-Tainta, A.; Corella, D.;
Lamuela-Raventos, R.M.; Martinez, J.A.; Martinez-Gonzalez, M.A. Virgin olive oil supplementation and
long-term cognition: The PREDIMED-NAVARRA randomized, trial. J. Nutr. Health Aging 2013, 17, 544–552.
[CrossRef] [PubMed]
10. Valls-Pedret, C.; Sala-Vila, A.; Serra-Mir, M.; Corella, D.; de la Torre, R.; Martínez-González, M.A.;
Martínez-Lapiscina, E.H.; Fitó, M.; Pérez-Heras, A.; Salas-Salvadó, J.; et al. Mediterranean Diet and
Age-Related Cognitive Decline: A Randomized Clinical Trial. JAMA Intern. Med. 2015, 175, 1094–1103.
[CrossRef] [PubMed]
102
Nutrients 2018, 10, 905
11. Scott, T.M.; Rasmussen, H.M.; Chen, O.; Johnson, E.J. Avocado consumption increases macular pigment
density in older adults: A randomized, controlled trial. Nutrients 2016, 9, 919. [CrossRef] [PubMed]
12. Kean, R.J.; Lamport, D.J.; Dodd, G.F.; Freeman, J.E.; Williams, C.M.; Ellis, J.A.; Butler, L.T.; Spencer, J.P.
Chronic consumption of flavanone-rich orange juice is associated with cognitive benefits: An 8-wk,
randomized, double-blind, placebo-controlled trial in healthy older adults. Am. J. Clin. Nutr. 2015,
101, 506–514. [CrossRef] [PubMed]
13. Külzow, N.; Witte, A.V.; Kerti, L.; Grittner, U.; Schuchardt, J.P.; Hahn, A.; Flöel, A. Impact of omega-3 fatty
acid supplementation on memory functions in healthy older adults. J. Alzheimers Dis. 2016, 51, 713–725.
[CrossRef] [PubMed]
14. Swaminathan, A.; Jicha, G.A. Nutrition and prevention of Alzheimer’s dementia. Front. Aging Neurosci.
2014, 6, 282. [CrossRef] [PubMed]
15. Hardman, R.J.; Kennedy, G.; Macpherson, H.; Scholey, A.B.; Pipingas, A. Adherence to a Mediterranean-style
diet and effects on cognition in adults: A qualitative evaluation and systematic review of longitudinal and
prospective trials. Front. Nutr. 2016, 3, 22. [CrossRef] [PubMed]
16. Zaragoza-Marti, A.; Ferrer-Cascales, R.; Hurtado-Sanchez, J.A.; Laguna-Perez, A.; Cabanero-Martinez, M.J.
Relationship between adherence to the Mediterranean diet and health-related quality of life and life
satisfaction among older adults. J. Nutr. Health Aging 2018, 22, 89–96. [CrossRef] [PubMed]
17. Clare, L.; Wu, Y.T.; Teale, J.C.; MacLeod, C.; Matthews, F.; Brayne, C.; Woods, B.; CFAS-Wales Study Team.
Potentially modifiable lifestyle factors, cognitive reserve, and cognitive function in later life: A cross-sectional
study. PLoS Med. 2017, 14, e1002259. [CrossRef] [PubMed]
18. Smyth, A.; Dehghan, M.; O’Donnell, M.; Anderson, C.; Teo, K.; Gao, P.; Sleight, P.; Dagenais, G.;
Probstfield, J.L.; Mente, A.; et al. Healthy eating and reduced risk of cognitive decline: A cohort from
40 countries. Neurology 2015, 84, 2258–2265. [CrossRef] [PubMed]
19. Calapai, G.; Bonina, F.; Bonina, A.; Rizza, L.; Mannucci, C.; Arcoraci, V.; Laganà, G.; Alibrandi, A.;
Pollicino, C.; Inferrera, S.; et al. A Randomized, double-blinded, clinical trial on effects of a Vitis vinifera
extract on cognitive function in healthy older adults. Front. Pharmacol. 2017, 8, 776. [CrossRef] [PubMed]
20. Clare, L.; Nelis, S.M.; Jones, I.R.; Hindle, J.V.; Thom, J.M.; Nixon, J.A.; Cooney, J.; Jones, C.L.; Edwards, R.T.;
Whitaker, C.J. The Agewell trial: A pilot randomised controlled trial of a behaviour change intervention to
promote healthy ageing and reduce risk of dementia in later life. BMC Psychiatry 2015, 15, 25. [CrossRef]
[PubMed]
21. Lehtisalo, J.; Ngandu, T.; Valve, P.; Antikainen, R.; Laatikainen, T.; Strandberg, T.; Soininen, H.; Tuomilehto, J.;
Kivipelto, M.; Lindström, J. Nutrient intake and dietary changes during a 2-year multi-domain lifestyle
intervention among older adults: Secondary analysis of the Finnish Geriatric Intervention Study to Prevent
Cognitive Impairment and Disability (FINGER) randomised controlled trial. Br. J. Nutr. 2017, 118, 291–302.
[CrossRef] [PubMed]
22. Mastroiacovo, D.; Kwik-Uribe, C.; Grassi, D.; Necozione, S.; Raffaele, A.; Pistacchio, L.; Righetti, R.; Bocale, R.;
Lechiara, M.C.; Marini, C.; et al. Cocoa flavanol consumption improves cognitive function, blood pressure
control, and metabolic profile in elderly subjects: The Cocoa, Cognition, and Aging (CoCoA) Study—A
randomized controlled trial. Am. J. Clin. Nutr. 2015, 101, 538–548. [CrossRef] [PubMed]
23. Nilsson, A.; Salo, I.; Plaza, M.; Björck, I. Effects of a mixed berry beverage on cognitive functions and
cardiometabolic risk markers; A randomized cross-over study in healthy older adults. PLoS ONE 2017,
12, e0188173. [CrossRef] [PubMed]
24. Sindi, S.; Ngandu, T.; Hovatta, I.; Kåreholt, I.; Antikainen, R.; Hänninen, T.; Levälahti, E.; Laatikainen, T.;
Lindström, J.; Paajanen, T.; et al. Baseline telomere length and effects of a multidomain lifestyle intervention
on cognition: The FINGER randomized controlled trial. J. Alzheimers Dis. 2017, 59, 1459–1470. [CrossRef]
[PubMed]
25. Brickman, A.M.; Khan, U.A.; Provenzano, F.A.; Yeung, L.K.; Suzuki, W.; Schroeter, H.; Wall, M.; Sloan, R.P.;
Small, S.A. Enhancing dentate gyrus function with dietary flavanols improves cognition in older adults.
Nat. Neurosci. 2014, 17, 1798–1803. [CrossRef] [PubMed]
26. Danthiir, V.; Hosking, D.E.; Nettelbeck, T.; Vincent, A.D.; Wilson, C.; O’Callaghan, N.; Calvaresi, E.; Clifton, P.;
Wittert, G.A. An 18-mo randomized, double-blind, placebo-controlled trial of DHA-rich fish oil to prevent
age-related cognitive decline in cognitively normal older adults. Am. J. Clin. Nutr. 2018, 107, 754–762.
[CrossRef] [PubMed]
103
Nutrients 2018, 10, 905
27. Kurz, A.; van Baelen, B. Ginkgo biloba compared with cholinesterase inhibitors in the treatment of dementia:
A review based on meta-analyses by the Cochrane collaboration. Dement. Geriatr. Cogn. Disord. 2004,
18, 217–226. [CrossRef] [PubMed]
28. Knight, A.; Bryan, J.; Murphy, K. Is the Mediterranean diet a feasible approach to preserving cognitive
function and reducing risk of dementia for older adults in Western countries: New insights and future
directions. Ageing Res. Rev. 2016, 25, 85–101. [CrossRef] [PubMed]
29. Scarmeas, N.; Stern, N.; Tang, M.X.; Mayeux, R.; Luchsinger, J.A. Mediterranean diet and risk for Alzheimer’s
disease. Ann. Neurol. 2006, 59, 912–921. [CrossRef] [PubMed]
30. Tagney, C.C. DASH and Mediterranean-type dietary patterns to maintain cognitive health. Curr. Nutr. Rep.
2014, 3, 51–61. [CrossRef] [PubMed]
31. Mosconi, L.; Murray, J.; Tsui, W.H.; Li, Y.; Davis, M.; Williams, S.; Pirraglia, E.; Spector, N.; Osorio, R.S.;
Glodzik, L.; et al. Mediterranean diet and magnetic resonance imaging-assessed brain atrophy in cognitively
normal individuals at risk for Alzheimer’s disease. J. Prev. Alzheimers Dis. 2014, 1, 23–32. [PubMed]
32. Frisardi, V.; Panza, F.; Seripa, D.; Imbimbo, B.P.; Vendemiale, G.; Pilotto, A.; Solfrizzi, V. Nutraceutical
properties of Mediterranean diet and cognitive decline: Possible underlying mechanisms. J. Alzheimers Dis.
2010, 22, 715–740. [CrossRef] [PubMed]
33. Pope, S.K.; Shue, V.M.; Beck, C. Will a healthy lifestyle help prevent Alzheimer’s disease? Annu. Rev.
Public Health 2003, 24, 111–132. [CrossRef] [PubMed]
34. Tsai, H.J. Dietary patterns and cognitive decline in Taiwanese aged 65 years and older. Int. J. Geriatr. Psychiatry
2015, 30, 523–530. [CrossRef] [PubMed]
35. Okubo, H.; Inagaki, H.; Gondo, Y.; Kamide, K.; Ikebe, K.; Masui, Y.; Arai, Y.; Ishizaki, T.; Sasaki, S.;
Nakagawa, T.; et al. Association between dietary patterns and cognitive function among 70-year-old
Japanese elderly: A cross-sectional analysis of the SONIC study. Nutr. J. 2017, 16, 56. [CrossRef] [PubMed]
36. Wright, R.S.; Waldstein, S.R.; Kuczmarski, M.F.; Pohlig, R.T.; Gerassimakis, C.S.; Gaynor, B.; Evans, M.K.;
Zonderman, A.B. Diet quality and cognitive function in an urban sample: Findings from the Healthy Aging
in Neighborhoods of Diversity across the Life Span (HANDLS) study. Public Health Nutr. 2017, 20, 92–101.
[CrossRef] [PubMed]
37. Hosking, D.E.; Nettelbeck, T.; Wilson, C.; Danthiir, V. Retrospective lifetime dietary patterns predict cognitive
performance in community-dwelling older Australians. Br. J. Nutr. 2014, 112, 228–237. [CrossRef] [PubMed]
38. Shah, H.; Albanese, E.; Duggan, C.; Rudan, I.; Langa, K.M.; Carrillo, M.C.; Chan, K.Y.; Joanette, Y.; Prince, M.;
Rossor, M.; et al. Research priorities to reduce the global burden of dementia by 2025. Lancet Neurol. 2016,
15, 1285–1294. [CrossRef]
39. Shlisky, J.; Bloom, D.E.; Beaudreault, A.R.; Tucker, K.L.; Keller, H.H.; Freund-Levi, Y.; Fielding, R.A.;
Cheng, F.W.; Jensen, G.L.; Wu, D.; et al. Nutritional considerations for healthy aging and reduction in
age-related chronic disease. Adv. Nutr. 2017, 8, 17–26. [CrossRef] [PubMed]
40. Ngandu, T.; Lehtisalo, J.; Solomon, A.; Levalahti, E.; Ahtiluoto, S.; Antikainen, R.; Bäckman, L.; Hänninen, T.;
Jula, A.; Laatikainen, T.; et al. A 2 year multidomain intervention of diet, exercise, cognitive training,
and vascular risk monitoring versus control to prevent cognitive decline in at-risk people (FINGER): A
randomised controlled trial. Lancet 2015, 385, 2255–2263. [CrossRef]
41. Barnard, N.D.; Bush, A.I.; Ceccarelli, A.; Cooper, J.; de Jager, C.A.; Fraser, G.; Fraser, G.; Kesler, S.; Levin, S.M.;
Lucey, B.; et al. Dietary and lifestyle guidelines for the prevention of Alzheimer’s disease. Neurobiol. Aging
2014, 35, S74–S78. [CrossRef] [PubMed]
42. Melby-Lervag, M.; Hulme, C. There is no convincing evidence that working memory training is effective:
A reply to Au et al. (2014) and Karbach and Verhaeghen (2014). Psychon. Bull. Rev. 2016, 23, 324–330.
[CrossRef] [PubMed]
43. Melby-Lervag, M.; Hulme, C. Is working memory training effective? A meta-analytic review. Dev. Psychol.
2013, 49, 270–291. [CrossRef] [PubMed]
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
104
nutrients
Article
Food Consumption, Knowledge, Attitudes, and
Practices Related to Salt in Urban Areas in Five
Sub-Saharan African Countries
Magali Leyvraz 1 , Carmelle Mizéhoun-Adissoda 2 , Dismand Houinato 3 , Naby Moussa Baldé 4 ,
Albertino Damasceno 5 , Bharathi Viswanathan 6 , Mary Amyunzu-Nyamongo 7 , Jared Owuor 7 ,
Arnaud Chiolero 1,8 and Pascal Bovet 1,6, *
1 Institute of Social and Preventive Medicine (IUMSP), Canton University Hospital (CHUV), 1010 Lausanne,
Switzerland; [email protected] (M.L.); [email protected] (A.C.)
2 School of Nutrition and Dietetics, Faculty of Health Science, University of Abomey-Calavi,
Cotonou 01 BP 526, Benin; [email protected]
3 Laboratory of Noncommunicable and Neurologic Diseases Epidemiology, Faculty of Health Science,
University of Abomey-Calavi, Cotonou 01 BP 526, Benin; [email protected]
4 Department of Endocrinology and Diabetes, Donka University Hospital, Conakry, Guinea;
[email protected] or [email protected]
5 Department of Medicine, Eduardo Mondlane University, Maputo, Mozambique; [email protected]
6 Ministry of Health, Victoria, Republic of Seychelles; [email protected]
7 African Institute for Health and Development (AIHD), Nairobi 00100, Kenya;
[email protected] (M.A.-N.); [email protected] (J.O.)
8 Institute of Primary Health Care (BIHAM), University of Bern, 3012 Bern, Switzerland
* Correspondence: [email protected]; Tel.: +41-21-314-72-72
Abstract: High salt intake is a major risk factor of hypertension and cardiovascular disease. Improving
knowledge, attitudes, and practices (KAP) related to salt intake in the general population is a key
component of salt reduction strategies. The objective of this study was to describe and compare
the KAP of adults related to salt in urban areas of five countries in sub-Saharan Africa. The survey
included 588 participants aged 25 to 65 years who were selected using convenience samples in the
urban areas of Benin, Guinea, Kenya, Mozambique, and Seychelles. Socio-demographic and food
consumption were assessed using a structured closed-ended questionnaire administered by survey
officers. Height, weight, and blood pressure were measured. Food consumption varied largely
between countries. Processed foods high in salt, such as processed meat, cheese, pizzas, and savory
snacks were consumed rather infrequently in all the countries, but salt-rich foods, such as soups
or bread and salty condiments, were consumed frequently in all countries. The majority of the
participants knew that high salt intake can cause health problems (85%) and thought that it is
important to limit salt intake (91%). However, slightly over half (56%) of the respondents regularly
tried to limit their salt intake while only 8% of the respondents thought that they consumed too
much salt. Salt and salty condiments were added most of the time during cooking (92% and 64%,
respectively) but rarely at the table (11%). These findings support the need for education campaigns
to reduce salt added during cooking and for strategies to reduce salt content in selected manufactured
foods in the region.
Keywords: salt; sodium; hypertension; knowledge; attitudes; practices; diet; Africa; Benin; Guinea;
Mozambique; Kenya; Seychelles
1. Introduction
Cardiovascular disease (CVD) is the leading cause of deaths worldwide including in low-income
and middle-income countries (LMICs) [1]. It is estimated that high salt intake accounts for 9.5%
of all CVD deaths globally due to its effect on blood pressure [2]. In sub-Saharan Africa (SSA),
approximately 800,000 deaths per year are due to CVD and 6% of these deaths are attributable to high
salt intake [2]. The disease burden related to elevated salt intake is expected to further increase over the
next decade in LMICs [3] due to the growing and aging populations and trends towards urbanization
and westernization of the diet in these countries.
Reducing dietary salt intake in the population is a key strategy to reduce the CVD burden [4].
Strategies to reduce salt intake at the population level include awareness campaigns advising
individuals to reduce their salt consumption, food labeling, and reformulation of selected industrially
produced foods [5]. Improving knowledge, attitudes, and practices (KAP) related to salt intake in the
population is an important part of any salt reduction strategy [6]. In particular, dietary advice can
reduce salt intake, blood pressure, and other CVD risk factors [7,8]. In 2015, 40 countries worldwide
were implementing some organized policy or program to reduce salt consumption in the population
but only one program was implemented in SSA (i.e., in South Africa) [9].
Only a few studies have examined KAP related to salt intake in SSA countries [10–14].
These studies have generally found low levels of KAP related to salt intake. However, most of
these studies were conducted in specific population groups (e.g., hypertensive patients) and in selected
settings. Given the scarcity of data on KAP related to salt intake and the high burden of CVD in
LMICs [15], it was recently recommended that more studies should be conducted to assess dietary
patterns and key sources of sodium in these countries [16].
The objective of this study was to describe and compare the food consumption, KAP related to
salt intake, and associated factors in adults from the general population in urban areas of five SSA
countries. This information is expected to be useful for guiding the development of salt reduction
programs and policies in SSA.
106
Nutrients 2018, 10, 1028
obtained from each participant. In each country, the study was approved by the locally relevant
institutional ethical review boards.
Weight was measured with an electronic weighing scale to the nearest 0.1 kg. Height was
measured with a fixed height rod to the nearest 0.1 cm in all countries. Overweight and obesity were
defined for body mass index (BMI) between 25 and 29 or ≥30 kg/m2 , respectively. Blood pressure
was measured with an electronic blood pressure device and high blood pressure defined as
systolic/diastolic blood pressure ≥140/90 mmHg or taking treatment for hypertension.
We assessed associations between KAP variables and selected predictors using Spearman
correlation coefficients and using stratified analysis. We used the chi-square test to test for differences
between categories. Means for all countries were weighted so that each country had the same weight.
The level of significance was set at 0.05. Analyses were conducted using Stata 14.1 (StataCorp,
College Station, TX, USA).
Publication of data on KAP from this study was delayed because the assessment of salt excretion,
which was originally another goal of this study in addition to KAP, had to be cancelled due to funding
and other issues. Moreover, sample size was limited when compared to the initially four-time larger
anticipated sample size due to discontinued funding by the donor.
3. Results
107
Nutrients 2018, 10, 1028
A number of processed food items with known high salt content including processed meat, cheese,
pizzas, breakfast cereals, and savory snacks were consumed rather infrequently in all the countries.
However, soup and bread, which often contain high amounts of salt, were consumed frequently in
all countries. As mentioned in the next paragraph, salt-rich condiments (e.g., Maggi cubes and food
spreads such as Marmite/Vegemite), which are often added in soups and other dishes, were also used
frequently in all countries.
108
Nutrients 2018, 10, 1028
problem that can arise from high salt intake (66%). Most of the participants thought it was important
to limit salt intake (91% ‘very important’ and ‘somehow important’). However, only 56% of the
respondents often tried to limit their salt intake. Moreover, only a small proportion of respondents
thought that they consumed too much salt (8%), while a substantial proportion of the respondents
thought they consumed too little salt (26%).
Table 3. Knowledge, attitudes, and practices related to salt intake according to country (in %).
Knowledge, Attitudes, and Practices All 1 Benin Guinea Kenya Mozambique Seychelles
Knowledge
High salt intake can cause serious health problems
Yes 85 (82–88) 93 (88–97) 66 (57–74) 87 (79–92) 88 (78–93) 92 (86–95)
No 5 (3–7) 2 (1–7) 5 (2–11) 6 (3–13) 4 (1–12) 6 (3–11)
Don’t know 10 (8–13) 4 (2–9) 28 (21–37) 7 (3–14) 8 (4–17) 2 (1–6)
Health problems are associated with high salt intake
≥1 problem known 66 (61–70) 87 (78–92) 64 (52–74) 48 (38–59) 93 (81–98) 50 (41–59)
None known 34 (30–39) 13 (8–22) 36 (26–48) 52 (41–62) 7 (2–19) 50 (41–59)
It is important to limit salt intake
Very important 9 (7–12) 7 (4–13) 20 (13–28) 7 (3–14) 7 (3–15) 5 (2–10)
Somehow important 12 (9–14) 4 (1–8) 23 (16–32) 14 (9–23) 0 (0–0) 17 (12–24)
Not really important 79 (76–82) 89 (83–93) 57 (48–66) 79 (70–86) 93 (85–97) 78 (70–84)
Attitudes
Try to limit salt
Often 55 (51–59) 76 (68–82) 47 (38–57) 54 (44–63) 66 (54–76) 32 (25–40)
Sometimes 16 (13–19) 11 (7–18) 18 (12–26) 26 (18–36) 3 (1–11) 22 (16–29)
Not really 29 (25–33) 13 (8–20) 35 (26–44) 20 (13–29) 31 (21–43) 46 (38–54)
Perceived amount of salt consumed
Too little 26 (22–30) 2 (1–6) 55 (46–64) 36 (27–45) 30 (21–41) 7 (4–12)
About right 59 (55–63) 96 (92–99) 20 (14–28) 52 (43–62) 60 (48–70) 67 (59–74)
Too much 8 (6–10) 1 (0–5) 5 (2–11) 12 (7–20) 10 (5–20) 11 (7–17)
Don’t know 7 (5–9) 1 (0–5) 19 (13–27) 0 (0–0) 0 (0–0) 16 (11–23)
Practices
Salt is added during cooking
Never 2 (1–3) 2 (1–6) 3 (1–8) 2 (0–8) 0 (0–0) 1 (0–5)
Sometimes (1–2 times/week) 7 (5–9) 1 (0–5) 27 (20–36) 3 (1–9) 3 (1–10) 0 (0–0)
Often (most meals) 19 (16–22) 5 (2–10) 18 (12–26) 30 (22–39) 27 (18–38) 15 (10–21)
Always (all meals) 73 (69–76) 92 (86–96) 52 (43–61) 65 (56–74) 70 (59–79) 84 (77–89)
Salty condiments are used during cooking 2
Never 9 (6–11) 6 (3–12) 8 (4–14) 21 (14–31) 4 (1–11) 4 (2–9)
Sometimes (1–2 times/week) 26 (23–30) 7 (4–13) 29 (22–38) 32 (23–42) 25 (16–36) 38 (30–46)
Often (most meals) 22 (18–25) 9 (5–15) 11 (6–18) 24 (17–34) 25 (16–36) 40 (32–48)
Always (all meals) 42 (38–47) 78 (70–84) 50 (41–59) 17 (11–26) 47 (36–58) 18 (13–25)
Salt is added to food at the table
Never 66 (62–70) 71 (63–78) 46 (37–55) 46 (36–56) 82 (72–89) 87 (80–91)
Sometimes (1–2 times/week) 23 (20–27) 24 (17–31) 33 (25–42) 37 (28–47) 12 (6–21) 9 (6–15)
Often (most meals) 6 (4–8) 2 (1–6) 14 (8–21) 8 (4–15) 4 (1–11) 2 (1–6)
Always (all meals) 5 (3–7) 3 (1–7) 8 (4–14) 9 (5–16) 3 (1–10) 2 (1–6)
Consumption of foods high in salt 3
Never 34 (30–38) 1 (0–6) 41 (32–50) 16 (10–24) 83 (73–90) 28 (21–36)
1–2 times/week 15 (13–18) 7 (4–13) 38 (30–48) 18 (11–27) 1 (0–9) 12 (8–18)
3–4 times/week 29 (25–32) 5 (2–10) 9 (5–16) 54 (45–64) 16 (9–26) 58 (50–66)
Every day/almost every day 21 (18–25) 86 (79–91) 6 (3–12) 12 (7–20) 0 (0–0) 2 (1–6)
1 Estimates for all were weighted so that data from each country had the same weight. 2 These condiments included
bouillon cubes, Aromat powder, soy sauce, food spreads (e.g., Vegemite, Marmite), and similar items. 3 These foods
included salted fish, salted meat, salami, salted peanuts, food spreads, pizza, and other typical local meals rich
in salt.
Most participants reported that salt was added to the foods most of the time during cooking
(92% ‘often’ and ‘always’). Salty condiments such as bouillon cubes, aroma enhancing powders, and
109
Nutrients 2018, 10, 1028
sauces were used frequently in all countries especially in Benin, Guinea, and Mozambique (87%, 61%,
and 72% added ‘often’ and ‘always’). In contrast, few participants reported adding salt to meals at the
table (11% ‘often’ and ‘always’).
4. Discussion
Overall, the study shows that the distribution of intake of food items varied widely between
countries. Several processed food items with known or presumably high salt content (such as pizzas
and savory snacks) were consumed rather infrequently (<3 times/week), but soups and bread were
consumed frequently in all countries (>3 times/week), which may suggest substantial salt intake.
The study also shows a fairly good level of KAP in relation to salt intake in urban settings among five
countries in Africa. However, there were some gaps. A fairly modest proportion of persons added
salt at the table. The large majority of participants were aware of health risks related to salt intake
and recognized the importance of limiting dietary salt intake. Yet, less than one in ten participants
believed they consumed too much salt. We did not find substantial associations within KAP variables
or between KAP variables and socio-demographic characteristics, except for an inverse association
between the knowledge of the need to restrict salt intake and the discretionary use of salt at the table.
Food consumption differed largely between countries, which underlies the difficulty of developing
dietary questionnaires and nutritional guidelines that could apply to all countries. However, certain
processed foods such as soups and bread were consumed frequently in all countries. These findings
suggest the need for voluntary or mandatory reformulation strategies to reduce the salt content of
selected manufactured foods that are both commonly consumed and have high salt content. A study
conducted in the early 2000s in South Africa reported that bread was a main source of dietary salt in
this country [18]. As a result, the South African government regulated the maximum levels of salt
permitted in a wide range of industrially processed food categories, including breads, in order to reduce
salt intake in the population [19,20]. Reformulation policies aimed at reducing salt in manufactured
foods can be highly cost-effective [21] and are recommended by the WHO’s Global Action Plan for
the Prevention and Control of Non-Communicable Diseases [4]. Several countries have implemented
policies to reformulate selected manufactured foods with subsequent reductions of the salt intake
at the population level [22–24]. However, none of the five countries included in this survey have
implemented such reformulation strategies [9]. South Africa is the only country in the African region
that has taken regulatory steps to mandatorily reduce the salt content of selected foods [9]. Findings of
our study also emphasize the need for continued education campaigns in order to encourage people
to limit dietary salt intake. Such campaigns are also useful when advocating for studies assessing
sources of salt intake in a particular population and when advocating for corresponding reformulation
110
Nutrients 2018, 10, 1028
strategies. Policy aimed at reformulating foods frequently consumed and high in salt is a cornerstone
strategy for effective reduction of salt intake in the population.
Knowledge on the detrimental effect of high salt intake was fairly high and higher than could
have been anticipated from previous research [10–14]. However, fairly good knowledge about health
effects of salt did not seem to have translated into strong attitudes and practices with regards to salt
intake reduction. For example, knowledge that salt could be detrimental for health was associated
with only one salt-related practice, i.e., low discretionary use of salt. A study conducted in 2014–2015
in Mozambique also found participants high in knowledge but low in attitudes and practices, in
which is similar to our study [25]. This suggests that campaigns aimed at raising awareness about
the detrimental impact of salt intake on health might better translate in actual salt reduction if
structural measures are also implemented, e.g., programs to reduce salt in the food served in work
or school canteens and measures to limit salt intake in selected manufactured foods (e.g., bread).
This is consistent with a review showing that the implementation of education and awareness-raising
interventions alone is unlikely to be adequate in reducing population salt intake to the recommended
levels, which suggests that behavior change might better occur when combining health education
and public awareness campaigns [26]. The quasi-ubiquitous presence of television and radio in the
surveyed households suggests that these media could be the main instruments to relay such public
awareness campaigns.
The main strengths of this study were the inclusion of population-based samples in five countries
and the use of the same methodology, which allows direct comparison between countries. The study
also has limitations. First, salt intake was not measured in all countries and we cannot report on
the relation between salt-related behaviors and an objective measurement of salt intake. Moreover,
our study did not allow for the quantification of salt added at the table or the contribution of salt in
the form of processed versus non-processed foods. Second, a number of selection biases may have
occurred in the sampling of participants. For example, persons present in a household at the time of
the survey may have been different than persons absent. This may, however, have limited impact,
since food consumption tends to be fairly homogenous at a household level. Individuals from a
low socioeconomic status (e.g., the illiterate persons unable to understand questions) may also have
been under-represented in this study. However, this proportion is likely small and has little overall
impact on the results. Our survey was limited to urban areas and the generalization of the findings
is, therefore, limited to such urban areas. Admittedly, food consumption may differ largely in rural
areas and further studies need to be conducted in these different areas. Yet, urbanization is rapidly
increasing in SSA and findings in this study may reflect dietary habits among large segments of the
population on the continent. Third, the study relied on reported information, which is prone to recall
and other biases that can lead to under-reporting or over-reporting of certain foods and practices.
Fourth, the fairly low numbers of participants precluded meaningful statistical analyses of associations
between KAP variables and socio-demographic characteristics.
Very little data is available on actual salt intake, the sources of salt intake, and KAP related to salt
in SSA. One systematic review aimed to identify all published studies reporting salt intake in countries
of SSA until 2015 [27]. This review found that 81% of the adult populations consumed salt intake above
the recommended maximum 5 g per day [27], which suggests overall high intake in the region. Intake
was higher in urban than in rural populations [27]. With regard to countries included in this study,
the review identified one study in children in Benin in 1996 [28] and one study in Kenya in 1986 in
rural areas [29]. When looking for more recent studies, urinary excretion of salt was assessed in Benin
based on the same study on KAP and in Mozambique using another study. Both studies found high
salt intake (10.2 g and 10.5 g of salt per day in Benin and Mozambique, respectively) [30,31]. A global
modelling study [32] estimated that mean salt intake in all of the five countries in our study, apart
from Kenya, was above the maximum 5 g of salt recommend by the WHO [33].
Future research on KAP related to salt is recommended in these countries among others in the
region. An objective measurement of the actual amount of salt consumed in these populations is
111
Nutrients 2018, 10, 1028
needed to assess whether salt intake is above recommendations and in which population groups.
In addition, further qualitative studies should examine specific practices that may favor salt intake.
Lastly, analyses of salt intake of foods especially in bread, instant soups and selected local foods as
well as market studies are needed to identify the main sources of dietary salt in these populations in
order to guide reformulation guidelines.
5. Conclusions
In conclusion, our study among adults in urban settings found largely different food consumption
patterns between countries but consumption of salt-rich bread, soup, and salty condiments was
frequent in all countries, which suggests that the salt intake could be substantial in all countries.
We found fairly good knowledge related to the detrimental effects of high salt intake, but mixed
findings related to the attitudes and practices related to the reduction of salt intake. These findings
support the need for both education campaigns to promote knowledge, attitudes, and behaviors for the
control of dietary salt intake and reformulation strategies to reduce salt content of selected frequently
eaten foods high in salt.
References
1. World Health Organization. Global Status Report on Noncommunicable Diseases; World Health Organization:
Geneva, Switzerland, 2014.
2. Mozaffarian, D.; Fahimi, S.; Singh, G.M.; Micha, R.; Khatibzadeh, S.; Engell, R.E.; Lim, S.; Danaei, G.;
Ezzati, M.; Powles, J. Global Sodium Consumption and Death from Cardiovascular Causes. N. Engl. J. Med.
2014, 371, 624–634. [CrossRef] [PubMed]
3. Twagirumukiza, M.; Dirk, D.B.; Kips, J.G.; Guy, D.B.; Stichele, R.V.; Van Bortel, L.M. Current and Projected
Prevalence of Arterial Hypertension in Sub-Saharan Africa by Sex, Age and Habitat: An Estimate from
Population Studies. J. Hypertens. 2011, 29, 1243–1252. [CrossRef] [PubMed]
4. World Health Organization. Global Action Plan for the Prevention and Control of Noncommunicable Diseases
2013–2020; World Health Organization: Geneva, Switzerland, 2013.
5. The Shake Technical Package for Salt Reduction. Available online: http://apps.who.int/iris/bitstream/
handle/10665/250135/9789241511346-eng.pdf;jsessionid=5BACD9B4917B297D370C440E99791E7A?
sequence=1 (accessed on 6 August 2018).
6. He, F.J.; Macgregor, G.A. Salt and Sugar: Their Effects on Blood Pressure. Pflugers Arch. Eur. J. Physiol. 2015,
467, 577–586. [CrossRef] [PubMed]
7. Rees, K.; Dyakova, M.; Wilson, N.; Ward, K.; Thorogood, M.; Brunner, E. Dietary Advice for Reducing
Cardiovascular Risk. Cochrane Database Syst. Rev. 2013, 3. [CrossRef]
8. Hooper, L.; Bartlett, C.; Davey, S.G.; Ebrahim, S. Advice to Reduce Dietary Salt for Prevention of
Cardiovascular Disease. Cochrane Database Syst. Rev. 2004, 3. [CrossRef] [PubMed]
112
Nutrients 2018, 10, 1028
9. World Health Organization. Report of the 2015 Global NCD Survey; World Health Organization: Geneva,
Switzerland, 2016.
10. Salaudeen, A.G.; Musa, O.I.; Babatunde, O.A.; Atoyebi, O.A.; Durowade, K.A.; Omokanye, L.O. Knowledge
and Prevalence of Risk Factors for Arterial Hypertension and Blood Pressure Pattern among Bankers and
Traffic Wardens in Ilorin, Nigeria. Afr. Health Sci. 2014, 14, 593–599. [CrossRef] [PubMed]
11. Katibi, I.A.; Olarinoye, J.K.; Kuranga, S.A. Knowledge and Practice of Hypertensive Patients as Seen in a
Tertiary Hospital in the Middle Belt of Nigeria. Niger. J. Clin. Pract. 2010, 13, 159–162. [PubMed]
12. Becker, H.; Bester, M.; Reyneke, N.; Labadarios, D.; Monyeki, K.D.; Steyn, N.P. Nutrition Related Knowledge
and Practices of Hypertensive Adults Attending Hypertensive Clinics at Day Hospitals in the Cape
Metropole. Curationis 2004, 27, 63–69. [CrossRef] [PubMed]
13. Aubert, L.; Bovet, P.; Gervasoni, J.P.; Rwebogora, A.; Waeber, B.; Paccaud, F. Knowledge, Attitudes, and
Practices on Hypertension in a Country in Epidemiological Transition. Hypertension 1998, 31, 1136–1145.
[CrossRef] [PubMed]
14. Kaddumukasa, M.N.; Katabira, E.; Sajatovic, M.; Pundik, S.; Kaddumukasa, M.; Goldstein, L.B. Influence of
Sodium Consumption and Associated Knowledge on Poststroke Hypertension in Uganda. Neurology 2016,
87, 1198–1205. [CrossRef] [PubMed]
15. Olsen, M.H.; Angell, S.Y.; Asma, S.; Boutouyrie, P.; Burger, D.; Chirinos, J.A.; Damasceno, A.; Delles, C.;
Gimenez-Roqueplo, A.P.; Hering, D.; et al. A Call to Action and a Lifecourse Strategy to Address the
Global Burden of Raised Blood Pressure on Current and Future Generations: The Lancet Commission on
Hypertension. Lancet 2016, 388, 2665–2712. [CrossRef]
16. Mancia, G.; Oparil, S.; Whelton, P.K.; McKee, M.; Dominiczak, A.; Luft, F.C.; AlHabib, K.; Lanas, F.;
Damasceno, A.; Prabhakaran, D.; et al. The Technical Report on Sodium Intake and Cardiovascular Disease
in Low- and Middle-Income Countries by the Joint Working Group of the World Heart Federation, the
European Society of Hypertension and the European Public Health Association. Eur. Heart J. 2017, 38,
712–719. [CrossRef] [PubMed]
17. World Health Organization. Who Steps Surveillance Manual; World Health Organization: Geneva, Switzerland,
2008.
18. Charlton, K.E.; Steyn, K.; Levitt, N.S.; Zulu, J.V.; Jonathan, D.; Veldman, F.J.; Nel, J.H. Diet and Blood Pressure
in South Africa: Intake of Foods Containing Sodium, Potassium, Calcium, and Magnesium in Three Ethnic
Groups. Nutrition 2005, 21, 39–50. [CrossRef] [PubMed]
19. Department of Health. Foodstuffs, Cosmetics and Disinfectants Act, 1972 (Act 54 of 1972): Regulations Relating to
the Reduction in Certain Foodstuffs and Related Matters; Government Gazette: Pretoria, South Africa, 2013.
20. Charlton, K.; Webster, J.; Kowal, P. To Legislate or Not to Legislate? A Comparison of the UK and South
African Approaches to the Development and Implementation of Salt Reduction Programs. Nutrients 2014, 6,
3672–3695. [CrossRef] [PubMed]
21. Wang, G.; Labarthe, D. The Cost-Effectiveness of Interventions Designed to Reduce Sodium Intake.
J. Hypertens. 2011, 29, 1693–1699. [CrossRef] [PubMed]
22. Christoforou, A.; Trieu, K.; Land, M.A.; Bolam, B.; Webster, J. State-Level and Community-Level Salt
Reduction Initiatives: A Systematic Review of Global Programmes and Their Impact. J. Epidemiol.
Public Health 2016, 70, 1140–1150. [CrossRef] [PubMed]
23. Trieu, K.; Neal, B.; Hawkes, C.; Dunford, E.; Campbell, N.; Rodriguez-Fernandez, R.; Legetic, B.; McLaren, L.;
Barberio, A.; Webster, J. Salt Reduction Initiatives around the World—A Systematic Review of Progress
Towards the Global Target. PLoS ONE 2015, 10, e0130247. [CrossRef] [PubMed]
24. Webster, J.L.; Dunford, E.K.; Hawkes, C.; Neal, B.C. Salt Reduction Initiatives around the World. J. Hypertens.
2011, 29, 1043–1050. [CrossRef] [PubMed]
25. Jessen, N.; Santos, A.; Damasceno, A.; Silva-Matos, C.; Severo, M.; Padrao, P.; Lunet, N. Knowledge and
Behaviors Regarding Salt Intake in Mozambique. Eur. J. Clin. Nutr. 2018, 1. [CrossRef] [PubMed]
26. Trieu, K.; McMahon, E.; Santos, J.A.; Bauman, A.; Jolly, K.A.; Bolam, B.; Webster, J. Review of Behaviour
Change Interventions to Reduce Population Salt Intake. Int. J. Behav. Nutr. Phys. Act. 2017, 14. [CrossRef]
[PubMed]
27. Oyebode, O.; Oti, S.; Chen, Y.F.; Lilford, R.J. Salt Intakes in Sub-Saharan Africa: A Systematic Review and
Meta-Regression. Popul. Health Metr. 2016, 14. [CrossRef] [PubMed]
113
Nutrients 2018, 10, 1028
28. Melse-Boonstra, A.; Rozendaal, M.; Rexwinkel, H.; Gerichhausen, M.J.; van den Briel, T.; Bulux, J.;
Solomons, N.W.; West, C.E. Determination of Discretionary Salt Intake in Rural Guatemala and Benin
to Determine the Iodine Fortification of Salt Required to Control Iodine Deficiency Disorders: Studies Using
Lithium-Labeled Salt. Am. J. Clin. Nutr. 1998, 68, 636–641. [CrossRef] [PubMed]
29. Poulter, N.R.; Khaw, K.T.; Mugambi, M.; Peart, W.S.; Sever, P.S. Migration-Induced Changes in Blood
Pressure: A Controlled Longitudinal Study. Clin. Exp. Pharmacol. Physiol. 1985, 12, 211–216. [CrossRef]
30. Mizehoun-Adissoda, C.; Houinato, D.; Houehanou, C.; Chianea, T.; Dalmay, F.; Bigot, A.; Aboyans, V.;
Preux, P.M.; Bovet, P.; Desport, J.C. Dietary Sodium and Potassium Intakes: Data from Urban and Rural
Areas. Nutrition 2017, 33, 35–41. [CrossRef] [PubMed]
31. Queiroz, A.; Damasceno, A.; Jessen, N.; Novela, C.; Moreira, P.; Lunet, N.; Padrao, P. Urinary Sodium
and Potassium Excretion and Dietary Sources of Sodium in Maputo, Mozambique. Nutrients 2017, 9, 830.
[CrossRef] [PubMed]
32. Powles, J.; Fahimi, S.; Micha, R.; Khatibzadeh, S.; Shi, P.; Ezzati, M.; Engell, R.E.; Lim, S.S.; Danaei, G.;
Mozaffarian, D. Global, Regional and National Sodium Intakes in 1990 and 2010: A Systematic Analysis of
24 H Urinary Sodium Excretion and Dietary Surveys Worldwide. BMJ Open 2013, 3, e003733. [CrossRef]
[PubMed]
33. World Health Organization. Guideline: Sodium Intake for Adults and Children; World Health Organization:
Geneva, Switzerland, 2012.
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
114
nutrients
Article
Lactase Persistence, Milk Intake, and Adult Acne:
A Mendelian Randomization Study of 20,416
Danish Adults
Christian R. Juhl 1 , Helle K. M. Bergholdt 2 , Iben M. Miller 3 , Gregor B. E. Jemec 3 ,
Jørgen K. Kanters 1, *and Christina Ellervik 2,4,5,6, *
1 Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen,
2100 Copenhagen, Denmark; [email protected]
2 Department of Production, Research, and Innovation, Region Zealand, 4180 Sorø, Denmark;
[email protected]
3 Department of Dermatology, Zealand University Hospital, 4000 Roskilde, Denmark;
[email protected] (I.M.M.); [email protected] (G.B.E.J.)
4 Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen,
2100 Copenhagen, Denmark
5 Department of Laboratory Medicine, Boston Children’s Hospital, 300 Longwood Avenue,
Boston, MA 02115, USA
6 Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
* Correspondence: [email protected] (J.K.K.); [email protected] or
[email protected] (C.E.)
Abstract: Whether there is a causal relationship between milk intake and acne is unknown. We tested
the hypothesis that genetically determined milk intake is associated with acne in adults using
a Mendelian randomization design. LCT-13910 C/T (rs4988235) is associated with lactase persistence
(TT/TC) in Northern Europeans. We investigated the association between milk intake, LCT-13910
C/T (rs4988235), and acne in 20,416 adults (age-range: 20–96) from The Danish General Suburban
Population Study (GESUS). The adjusted observational odds ratio for acne in any milk intake vs.
no milk intake was 0.93(95% confidence interval: 0.48–1.78) in females and 0.49(0.22–1.08) in males
aged 20–39 years, and 1.15(95% confidence interval: 0.66–1.99) in females and 1.02(0.61–1.72) in males
above 40 years. The unadjusted odds ratio for acne in TT+TC vs. CC was 0.84(0.43–1.62) in the age
group 20–39 years, and 0.99(0.52–1.88) above 40 years. We did not find any observational or genetic
association between milk intake and acne in our population of adults.
Keywords: acne; acne vulgaris; milk; dairy; diet; Mendelian randomization; adults
1. Introduction
Acne is a common chronic inflammatory skin disease, which is almost universal in adolescence,
with rates up to 85% [1–4]. After adolescence the prevalence decreases but a significant number
of patients are affected by persistent acne or develop new-onset adult acne [5]. Acne is overall
characterized by open comedones, papules, pustules, and nodules [6], but the clinical appearance
varies by age and lifestyle [5,7,8].
The genetic architecture of acne vulgaris is complex and multiple susceptible loci have been
identified reflecting the multifactorial pathogenesis of acne involving the innate immune system,
inflammation, modified lipogenesis, and androgens [9,10]. But there is likely also an environmental
component in the development of acne vulgaris. Several observational studies have investigated the
association of milk intake with acne in children, adolescents, and young adults [11–16]. However, no
previous observational study has been performed in all adult ages.
In individuals of Northern European descent, the genetic variant LCT-13910 C/T (rs4988235)
located 13,910 base pairs upstream of the lactase (LCT) gene on chromosome 2q21-22, within intron 13
of the adjacent MCM6 gene, shows complete correlation with lactase persistence/non-persistence [17].
The T-allele is the lactase-persistent allele, whereas the C allele is the lactase non-persistent allele.
The inheritance is autosomal recessive manner such that individuals homozygous for CC are unable to
digest lactose, whereas individuals with TC or TT are able to digest lactose.
To investigate whether there is a causal relationship between milk intake and acne, large long-term
randomized trials would be needed, but these are costly and it is difficult to uphold the randomization
over time. Instead the epidemiological Mendelian randomization (MR) design offers a feasible
alternative [18]. The underlying principle in the MR design is that genetic variants are randomly
assorted during gamete formation, which is similar to the random assignment of patients to placebo or
active treatment in a clinical intervention trial. In the MR design, confounders are, therefore, balanced
across the genotypes, and the genotypes will serve as a proxy for lifelong exposure.
In this study, we used the Mendelian randomization design to investigate the long-term effect
of milk intake on acne using the lactase persistent (TT + TC)/non-persistent(CC) LCT-13910 C/T
genotype in 20,416 adult individuals from The Danish General Population study (GESUS).
2.1. Participants
The cross-sectional population study The Danish General Population Study (GESUS) was
conducted from January 2010 to October 2013 in Naestved Municipality, Denmark [19]. Criteria
for invitation were age 20+, Danish citizenship and Danish Civil Registration number (CPR number).
All persons aged 30+ were invited and in the age group 20–30 years only a random 25% selection
were invited. 21,205 adults were enrolled, with an overall participation rate of 43%.
In this present study, 20,850 persons was included of whom 98.9% were of Danish descent and
the rest other Scandinavian or European descent. Individuals with missing values for acne diagnosis
(n = 53) or LCT-13910 C/T genotyping (n = 381) were excluded, resulting in inclusion of 20,416 people.
A prerequisite for attending the health examination was a completed self-reported paper-
questionnaire about demographic information, medical history, smoking, skin-condition, and food
intake, among others. The health examination was performed by trained health professionals and took
place at the department of clinical biochemistry at Naestved University Hospital, Denmark. Body mass
index (BMI) was calculated as kg/m2 . Details about the study design of GESUS have been described
elsewhere [19].
Written informed consent was obtained from all participants. The study conforms to the principles
of the Declaration of Helsinki and is approved by the institutional review board, the ethical committee
of Region Zealand (SJ-113, SJ-114, SJ-191) and the Danish Data Protection Agency.
116
Nutrients 2018, 10, 1041
2.4. Genotyping
Every participant in the GESUS study was genotyped for LCT-13910 C/T variant (rs4988235).
The genotyping was done by KASPar allelic discrimination (LGC Genomics) with a call rate of
99% [21]. The genotype LCT-13910 TC and TT are lactase persistent and CC lactase non-persistent.
The LCT-13910 C/T genotype distribution, in the GESUS population, was in Hardy-Weinberg
equilibrium (Supplementary Table S1).
117
Nutrients 2018, 10, 1041
3. Results
118
Table 1. Baseline characteristics for The Danish General Suburban Population study (GESUS) by acne, age groups, and LCT-13910 genotype.
LCT-13910 Genotype
All Acne Control Age Age > 40
n = 20,416 n = 303 n = 20,113 20–39 n = 17,674 CC TC TT
p-Value * p-Value * p-Value
100% 1.5% 98.5% n = 2742 86.6% n = 1246 n = 7377 n = 11,793
**
13.4% 6.1% 36.1% 57.8%
Nutrients 2018, 10, 1041
119
Current Smoker, 3632 75 3557 490 3142 233 1318 2081
0.0014 0.9060 0.6371
n (%) (17.8) (24.8) (17.7) (17.9) (17.8) (18.7) (17.9) (17.7)
Milk Intake, 8.1 10.2 8.1 11.0 7.7 6.1 8.2 8.3
<0.0001 <0.0001 <0.0001
mean (SD), glasses/week (9.1) (14.3) (9.0) (9.9) (8.9) (7.1) (9.1) (9.4)
* The chi-square test or the analysis of variance (ANOVA) test is used to calculate the p-value. ** Rao-Scott modified chi-square test is used for categorized variables and ANOVA for
continuous variables. LCT-13910 genotype (rs4988235): CC = the lactase non-persistent genotype, TC/TT = the lactase persistent genotype.
Table 2. Odds ratio for acne by milk intake in the age group 20–39 years.
Female Male
Milk Intake
Unadjusted Adjusted * Unadjusted Adjusted *
(glasses/week) n Total n Acne n Total n Acne
OR [95% CI] p OR [95% CI] p OR [95% CI] p OR [95% CI] p
0 172 11 1.00 - 1.00 - 122 8 1.00 - 1.00 -
Nutrients 2018, 10, 1041
1–3 274 19 1.09 [0.51:2.35] 0.82 0.98 [0.45:2.13] 0.96 147 4 0.40 [0.12:1.36] 0.14 0.43 [0.13:1.47] 0.18
4–7 375 25 1.05 [0.50:2.18] 0.91 0.98 [0.47:2.06] 0.96 186 11 0.90 [0.35:2.29] 0.82 0.93 [0.36:2.41] 0.89
8–14 397 25 0.98 [0.47:2.05] 0.96 0.90 [0.43:1.90] 0.79 322 7 0.32 [0.11:0.89] 0.03 0.36 [0.13:1.02] 0.06
>14 331 20 0.94 [0.44:2.01] 0.88 0.85 [0.39:1.83] 0.67 416 11 0.39 [0.15:0.98] 0.05 0.40 [0.16:1.02] 0.05
Any 1377 89 1.01 [0.53:1.93] 0.97 0.93 [0.48:1.78] 0.82 1071 33 0.45 [0.20:1.00] 0.05 0.49 [0.22:1.08] 0.08
Low-fat ** 1348 85 0.83 [0.47:1.48] 0.53 0.84 [0.47:1.52] 0.57 1031 31 0.47 [0.23:0.98] 0.04 0.49 [0.23:1.03] 0.06
High-fat *** 58 7 2.06 [0.91:4.67] 0.08 1.42 [0.55:3.69] 0.47 82 5 1.94 [0.74:5.08] 0.18 2.01 [0.75:5.38] 0.17
* Adjusted for age, smoking and body mass index, ** Low-fat (0.1–0.3%), *** High-fat (0.5–3.5%). p-values are calculated as analysis of maximum likelihood estimates.
Table 3. Odds ratio for acne by milk intake for in the age group ≥40 years.
Female Male
120
Milk Intake
Unadjusted Adjusted * Unadjusted Adjusted *
(glasses/week) n Total n Acne n Total n Acne
OR [95% CI] p OR [95% CI] p OR [95% CI] p OR [95% CI] p
0 2802 17 1.00 - 1.00 - 2099 20 1.00 - 1.00 -
1–3 1564 16 1.69 [0.85:3.36] 0.13 1.30 [0.65:2.59] 0.46 1099 10 0.95 [0.45:2.05] 0.90 0.91 [0.42:1.96] 0.80
4–7 2129 22 1.71 [0.91:3.23] 0.10 1.40 [0.74:2.65] 0.31 1592 13 0.86 [0.42:1.73] 0.66 0.82 [0.40:1.68] 0.59
8–14 1816 11 1.00 [0.47:2.14] 1.00 0.76 [0.35:1.63] 0.48 1682 23 1.44 [0.79:2.63] 0.23 1.31 [0.71:2.43] 0.39
>14 1262 12 1.57 [0.75:3.30] 0.23 1.13 [0.53:2.40] 0.75 1629 18 1.16 [0.61:2.20] 0.65 0.99 [0.51:1.91] 0.97
Any 6771 61 1.49 [0.87:2.55] 0.15 1.15 [0.66:1.99] 0.62 6002 64 1.12 [0.68:1.86] 0.66 1.02 [0.61:1.72] 0.94
Low-fat ** 6324 58 1.49 [0.90:2.49] 0.12 1.14 [0.68:1.91] 0.63 5268 57 1.14 [0.72:1.80] 0.59 0.98 [0.61:1.58] 0.94
High-fat *** 590 4 0.82 [0.30:2.26] 0.70 0.97 [0.35:2.68] 0.95 901 10 1.08 [0.56:2.10] 0.82 1.25 [0.64:2.46] 0.51
* Adjusted for age, smoking and body mass index, ** Low-fat (0.1–0.3%), *** High-fat (0.5–3.5%). p-values are calculated as analysis of maximum likelihood estimates.
Table 4. Differences in milk intake, glasses per week, by the LCT-13910 C/T genotype.
121
Nutrients 2018, 10, 1041
Low Upper
Dairy Author Year OR p-Value
95% CI 95% CI
Any intake Di Landro [25] 2016 0.88 0.61 1.28 0.52
Any intake Juhl Current 1.27 0.84 1.92 0.26
Fixed effects pooled odds ratio 1.04 0.79 1.37 0.78
Random effects pooled odds ratio 1.05 0.74 1.49 0.80
Whole milk Di Landro [25] 2016 0.84 0.49 1.44 0.53
Whole milk Juhl Current 1.43 0.76 2.70 0.27
Fixed effects pooled odds ratio 1.05 0.70 1.58 0.82
Random effects pooled odds ratio 1.07 0.64 1.79 0.81
Low-fat milk Di Landro [25] 2016 0.90 0.61 1.32 0.59
Low-fat milk Juhl Current 1.15 0.79 1.68 0.47
Fixed effects pooled odds ratio 1.02 0.78 1.34 0.89
Random effects pooled odds ratio 1.02 0.78 1.34 0.89
4. Discussion
Among 20,419 adults from the Danish general population we found no association between milk
intake and acne, observationally or genetically using the lactase persistent/non-persistent LCT-13910
C/T genotype in a Mendelian randomization design.
In the Mendelian randomization design the genetic variant is used as a proxy for the long-term
differences in milk intake, thereby largely avoiding confounding and reverse causation, which can blur
or distort the underlying true association in observational studies [18]. The LCT-13910 fulfilled the
requirements for using the Mendelian randomization design, as the variant is linked to the intermediate
phenotype (milk intake) in a biologically explainable way [17], there is no known pleiotropic effects
of the variant, and the variant was not associated with confounders. The Mendelian randomization
design mimics a randomized clinical trial and takes advantage of the random assortment of alleles at
conception which ensures random distribution of confounding factors thereby circumventing reverse
causation and most confounding. Thus, the Mendelian randomization design provides an estimate of
the long-term effect of milk intake on acne.
We showed that both the milk consumption and the acne diagnosis declined with age, and that
fewer people drank milk as age increased. Nevertheless, we found a crude prevalence of self-reported
acne of 6%. We also investigated milk intake and acne among adults by combining our current
122
Nutrients 2018, 10, 1041
study with findings from Landro , but the associations were still null for any milk intake, whole
milk, and low-fat milk. In contrast, observational studies of mostly childhood and adolescent acne
have debated, but largely favored, the association between milk intake and acne [11–13,16,26–29].
These studies were heterogeneous with respect to geographical location, cultural dairy influence,
gender, sample size, reporting of dairy frequency and type, and the ascertainment of participants
(dermatology clinics, general population study, online questionnaire). Some of the studies did not run
adjusted analyses [16,26,27], thus confounding may account for the associations. However, reasons for
the discrepant findings between our study and the study by Landro [25] in adults versus the studies in
adolescent acne [11–13,16,26–29] could also be related to different pathogenesis and the appearance of
acne in different age groups. Milk intake increases levels of insulin-like growth factor-1 (IGF1), which is
hypothesized to be a central link between milk intake and stimulation of the sebaceous gland [30].
Thus, as people age and drink less milk, they may be less exposed to IGF1 and, therefore, also less prone
to the development of acne. The appearance of acne varies by age, such that acne precox and acne
tarda are mostly variants with predominant inflammatory papules, pustules and nodules at the lower
face half [5,31], while adolescent acne is mostly characterized by comedoes and papules/pustules at
the entire face (chest and back) [7]. In contrast, acne in adult smokers is characterized by comedoes
and scars [8] and acne fulminans exhibits a severe clinical picture [32]. Even though the acne diagnosis
was questionnaire based as in many previous observational studies [16,28], it has been validated in
a comparable setting with a high specificity but with a sensitivity of only 50% [20]. However, we did
not have information on anatomical acne location or longer period of acne appearance. Additionally,
our study spanned four years including all seasons, thereby compensating for seasonal variation in
acne appearance [33].
The strength of our study is the homogenous population of largely Danish descent or other
Scandinavian descent making confounding from population substructure less likely. Similarly, the
rs4988235 variant is the most common lactase persistent variant in populations of Northern European
descent, despite the many other lactase persistence haplotypes [34]. A limitation of our study included
the low prevalence of lactase non-persistence (6%) and the fact that some of these individuals drank
milk, which could offset and conceal a true association. Milk consumption was based on self-reported
questionnaire data, with possible recall bias.
5. Conclusions
In conclusion, in the Danish General Suburban Population Study (GESUS) of adults we did
not find any observational or genetic association between milk intake and acne using the lactase
persistent/non-persistent LCT-13910 C/T genotype in a Mendelian randomization design.
References
1. White, G.M. Recent findings in the epidemiologic evidence, classification, and subtypes of acne vulgaris.
J. Am. Acad. Dermatol. 1998, 39, S34–S37. [CrossRef]
123
Nutrients 2018, 10, 1041
2. Bhate, K.; Williams, H.C. Epidemiology of acne vulgaris. Br. J. Dermatol. 2013, 168, 474–485. [CrossRef]
[PubMed]
3. Gollnick, H.; Cunliffe, W.; Berson, D.; Dreno, B.; Finlay, A.; Leyden, J.J.; Shalita, A.R.; Thiboutot, D.
Management of acne: A report from a Global Alliance to Improve Outcomes in Acne. J. Am. Acad. Dermatol.
2003, 49, S1–S37. [CrossRef] [PubMed]
4. Ghodsi, S.Z.; Orawa, H.; Zouboulis, C.C. Prevalence, severity, and severity risk factors of acne in high school
pupils: A community-based study. J. Invest. Dermatol. 2009, 129, 2136–2141. [CrossRef] [PubMed]
5. Perkins, A.C.; Maglione, J.; Hillebrand, G.G.; Miyamoto, K.; Kimball, A.B. Acne vulgaris in women:
Prevalence across the life span. J. Womens Health 2012, 21, 223–230. [CrossRef] [PubMed]
6. Williams, H.C.; Dellavalle, R.P.; Garner, S. Acne vulgaris. Lancet 2012, 379, 361–372. [CrossRef]
7. Tuchayi, S.M.; Makrantonaki, E.; Ganceviciene, R.; Dessinioti, C.; Feldman, S.R.; Zouboulis, C.C. Acne
vulgaris. Nat. Rev. Dis. Primers 2015, 1, 15029. [CrossRef] [PubMed]
8. Capitanio, B.; Sinagra, J.L.; Bordignon, V.; Fei, P.C.; Picardo, M.; Zouboulis, C.C. Underestimated clinical
features of postadolescent acne. J. Am. Acad. Dermatol. 2010, 63, 782–788. [CrossRef] [PubMed]
9. Lichtenberger, R.; Simpson, M.A.; Smith, C.; Barker, J.; Navarini, A.A. Genetic architecture of acne vulgaris.
J. Eur. Acad. Dermatol. Venereol. 2017, 31, 1978–1990. [CrossRef] [PubMed]
10. Zouboulis, C.C.; Jourdan, E.; Picardo, M. Acne is an inflammatory disease and alterations of sebum
composition initiate acne lesions. J. Eur. Acad. Dermatol. Venereol. 2014, 28, 527–532. [CrossRef] [PubMed]
11. Adebamowo, C.A.; Spiegelman, D.; Danby, F.W.; Frazier, A.L.; Willett, W.C.; Holmes, M.D. High school
dietary dairy intake and teenage acne. J. Am. Acad. Dermatol. 2005, 52, 207–214. [CrossRef] [PubMed]
12. Adebamowo, C.A.; Spiegelman, D.; Berkey, C.S.; Danby, F.W.; Rockett, H.H.; Colditz, G.A.; Walter, C.W.;
Michelle, D.H. Milk consumption and acne in adolescent girls. Dermatol. Online J. 2006, 12, 1. [PubMed]
13. Adebamowo, C.A.; Spiegelman, D.; Berkey, C.S.; Danby, F.W.; Rockett, H.H.; Colditz, G.A.; Willett, W.C.;
Holmes, M.D. Milk consumption and acne in teenaged boys. J. Am. Acad. Dermatol. 2008, 58, 787–793.
[CrossRef] [PubMed]
14. Grossi, E.; Cazzaniga, S.; Crotti, S.; Naldi, L.; Di Landro, A.; Ingordo, V.; Cusano, F.; Atzori, L.; Tripodi Cutrì, F.;
Musumeci, M.L.; et al. The constellation of dietary factors in adolescent acne: A semantic connectivity map
approach. J. Eur. Acad. Dermatol. Venereol. 2016, 30, 96–100. [CrossRef] [PubMed]
15. Karadag, A.S.; Balta, I.; Saricaoglu, H.; Kilic, S.; Kelekci, K.H.; Yildirim, M.; Arica, D.A.; Öztürk, S.;
Karaman, G.; Çerman, A.A.; et al. The effect of personal, familial, and environmental characteristics
on acne vulgaris: A prospective, multicenter, case controlled study from Turkey. G. Ital. Dermatol. Venereol.
2017. [CrossRef]
16. Wolkenstein, P.; Machovcova, A.; Szepietowski, J.C.; Tennstedt, D.; Veraldi, S.; Delarue, A. Acne prevalence
and associations with lifestyle: A cross-sectional online survey of adolescents/young adults in 7 European
countries. J. Eur. Acad. Dermatol. Venereol. 2017. [CrossRef] [PubMed]
17. Enattah, N.S.; Sahi, T.; Savilahti, E.; Terwilliger, J.D.; Peltonen, L.; Järvelä, I. Identification of a variant
associated with adult-type hypolactasia. Nat. Genet. 2002, 30, 233–247. [CrossRef] [PubMed]
18. Smith, G.D.; Ebrahim, S. Mendelian randomization: Prospects, potentials, and limitations. Int. J. Epidemiol.
2004, 33, 30–42. [CrossRef] [PubMed]
19. Bergholdt, H.K.M.; Bathum, L.; Kvetny, J.; Rasmussen, D.B.; Moldow, B.; Hoeg, T.; Jemec, G.B.E.;
Berner-Nielsen, H.; Nordestgaard, B.G.; Ellervik, C. Study design, participation and characteristics of
the Danish General Suburban Population Study. Dan. Med. J. 2013, 60, A4693. [PubMed]
20. Dalgard, F.; Svensson, A.; Holm, J.O.; Sundby, J. Self-reported skin complaints: validation of a questionnaire
for population surveys. Br. J. Dermatol. 2003, 149, 794–810. [CrossRef] [PubMed]
21. Bergholdt, H.K.M.; Nordestgaard, B.G.; Varbo, A.; Ellervik, C. Milk intake is not associated with ischaemic
heart disease in observational or Mendelian randomization analyses in 98 529 Danish adults. Int. J. Epidemiol.
2015, 44, 587–603. [CrossRef] [PubMed]
22. Jemec, G.B.; Linneberg, A.; Nielsen, N.H.; Frolund, L.; Madsen, F.; Jorgensen, T. Have oral contraceptives
reduced the prevalence of acne? A population-based study of acne vulgaris, tobacco smoking and oral
contraceptives. Dermatology. 2002, 204, 179–184. [CrossRef] [PubMed]
23. Alan, S.; Cenesizoglu, E. Effects of hyperandrogenism and high body mass index on acne severity in women.
Saudi Med. J. 2014, 35, 886–889. [PubMed]
124
Nutrients 2018, 10, 1041
24. Dreno, B.; Thiboutot, D.; Layton, A.M.; Berson, D.; Perez, M.; Kang, S. Large-scale international study
enhances understanding of an emerging acne population: Adult females. J. Eur. Acad. Dermatol. Venereol.
2015, 29, 1096–1106. [CrossRef] [PubMed]
25. Di Landro, A.; Cazzaniga, S.; Cusano, F.; Bonci, A.; Carla, C.; Musumeci, M.L.; Patrizi, A.; Bettoli, V.;
Pezzarossa, E.; Caproni, M.; et al. Adult female acne and associated risk factors: Results of a multicenter
case-control study in Italy. J. Am. Acad. Dermatol. 2016, 75, 1134–1141. [CrossRef] [PubMed]
26. Wolkenstein, P.; Misery, L.; Amici, J.M.; Maghia, R.; Branchoux, S.; Cazeau, C.; Voisard, J.-J.; Taïeb, C.
Smoking and dietary factors associated with moderate-to-severe acne in French adolescents and young
adults: results of a survey using a representative sample. Dermatology 2015, 230, 34–39. [CrossRef] [PubMed]
27. Di Landro, A.; Cazzaniga, S.; Parazzini, F.; Ingordo, V.; Cusano, F.; Atzori, L.; Cutrì, F.T.; Musumeci, M.L.;
Zinetti, C.; Pezzarossa, E.; et al. Family history, body mass index, selected dietary factors, menstrual history,
and risk of moderate to severe acne in adolescents and young adults. J. Am. Acad. Dermatol. 2012, 67,
1129–1135. [CrossRef] [PubMed]
28. Ulvestad, M.; Bjertness, E.; Dalgard, F.; Halvorsen, J.A. Acne and dairy products in adolescence: Results from
a Norwegian longitudinal study. J. Eur. Acad. Dermatol. Venereol. 2017, 31, 530–545. [CrossRef] [PubMed]
29. Juhl, C.R.; Berholdt, H.K.M.; Miller, I.M.; Jemec, G.B.; Kanters, J.K.; Ellervik, C. Dietary intake and acne
vulgaris: a systematic review and meta-analysis of 78,529 children, adolescents, and young adults. Nutrients
2018, in press.
30. Mirdamadi, Y.; Thielitz, A.; Wiede, A.; Goihl, A.; Papakonstantinou, E.; Hartig, R.; Zouboulis, C.C.;
Reinhold, D.; Simeoni, L.; Bommhardt, U.; et al. Insulin and insulin-like growth factor-1 can modulate the
phosphoinositide-3-kinase/Akt/FoxO1 pathway in SZ95 sebocytes in vitro. Mol. Cell Endocrinol. 2015, 415,
32–44. [CrossRef] [PubMed]
31. Jansen, T.; Janssen, O.E.; Plewig, G. [Acne tarda. Acne in adults]. Hautarzt 2013, 64, 241–251. [CrossRef]
[PubMed]
32. Massa, A.F.; Burmeister, L.; Bass, D.; Zouboulis, C.C. Acne Fulminans: Treatment Experience from 26 Patients.
Dermatology 2017, 233, 136–150. [CrossRef] [PubMed]
33. Pascoe, V.L.; Kimball, A.B. Seasonal variation of acne and psoriasis: A 3-year study using the Physician
Global Assessment severity scale. J. Am. Acad. Dermatol. 2015, 73, 523–535. [CrossRef] [PubMed]
34. Itan, Y.; Jones, B.L.; Ingram, C.J.; Swallow, D.M.; Thomas, M.G. A worldwide correlation of lactase persistence
phenotype and genotypes. BMC Evol. Biol. 2010, 10, 36. [CrossRef] [PubMed]
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
125
nutrients
Article
Dairy Intake and Acne Vulgaris: A Systematic Review
and Meta-Analysis of 78,529 Children, Adolescents,
and Young Adults
Christian R. Juhl 1 , Helle K. M. Bergholdt 2 , Iben M. Miller 3 , Gregor B. E. Jemec 3,4 ,
Jørgen K. Kanters 1, * and Christina Ellervik 2,4,5,6, *
1 Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen,
2100 Copenhagen, Denmark; [email protected]
2 Department of Production, Research, and Innovation, Region Zealand, 4180 Sorø, Denmark;
[email protected]
3 Department of Dermatology, Zealand University Hospital, 4000 Roskilde, Denmark;
[email protected] (I.M.M.); [email protected] (G.B.E.J.)
4 Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen,
2100 Copenhagen, Denmark
5 Department of Laboratory Medicine, Boston Children’s Hospital, 300 Longwood Avenue,
Boston, MA 02115, USA
6 Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
* Corresponding Authors: [email protected] (J.K.K.);
[email protected] or [email protected] (C.E.)
Abstract: A meta-analysis can help inform the debate about the epidemiological evidence on dairy
intake and development of acne. A systematic literature search of PubMed from inception to
11 December 2017 was performed to estimate the association of dairy intake and acne in children,
adolescents, and young adults in observational studies. We estimated the pooled random effects
odds ratio (OR) (95% CI), heterogeneity (I2 -statistics, Q-statistics), and publication bias. We included
14 studies (n = 78,529; 23,046 acne-cases/55,483 controls) aged 7–30 years. ORs for acne were
1.25 (95% CI: 1.15–1.36; p = 6.13 × 10−8 ) for any dairy, 1.22 (1.08–1.38; p = 1.62 × 10−3 ) for full-fat
dairy, 1.28 (1.13–1.44; p = 8.23 × 10−5 ) for any milk, 1.22 (1.06–1.41; p = 6.66 × 10−3 ) for whole
milk, 1.32 (1.16–1.52; p = 4.33 × 10−5 ) for low-fat/skim milk, 1.22 (1.00–1.50; p = 5.21 × 10−2 ) for
cheese, and 1.36 (1.05–1.77; p = 2.21 × 10−2 ) for yogurt compared to no intake. ORs per frequency
of any milk intake were 1.24 (0.95–1.62) by 2–6 glasses per week, 1.41 (1.05–1.90) by 1 glass per day,
and 1.43 (1.09–1.88) by ≥2 glasses per day compared to intake less than weekly. Adjusted results were
attenuated and compared unadjusted. There was publication bias (p = 4.71 × 10−3 ), and heterogeneity
in the meta-analyses were explained by dairy and study characteristics. In conclusion, any dairy,
such as milk, yogurt, and cheese, was associated with an increased OR for acne in individuals aged
7–30 years. However, results should be interpreted with caution due to heterogeneity and bias
across studies.
1. Introduction
Acne is a common chronic inflammatory skin disease of sebaceous follicles [1,2]. Clinically,
acne is characterized by the presence of open and closed comedones, papules, pustules, and dermal
tissue damage with eventually heavy scar formation. Follicular hyperkeratosis, modifications of the
sebofollicular microbiome, increase production of sebum with increased amounts of pro-inflammatory
monounsaturated fatty acids, and Th17-cell-mediated inflammatory responses are all involved in acne
pathogenesis. Sebum production can be induced by insulin-like growth factor-1 (IGF-1) and androgens,
whose adrenal and gonadal synthesis is stimulated by IGF-1 [3]. Although prevalence varies across
studies, acne is common in children and adolescents aged 12–24 years and is moderate to severe in
15–20% of cases [1,4–6].
Heritability of acne alone does not explain high acne prevalence rates of over 80% in western
countries [5,7]. It has long been debated if a Western diet per se or specific dietary components
contribute to the prevalence and severity of acne [4,8]. This has predominantly been investigated
in observational studies and only a few trials exist [9]. In particular, dairy products have been
incriminated. Milk-derived amino acids promote insulin secretion and induce hepatic insulin-like
growth factor-1 (IGF-1) synthesis [10]. IGF-1 has been suggested as the pivotal driver of acne and
stimulates follicular epithelial growth and keratinization [11–13]. IGF-1 gene polymorphism has been
shown to increase susceptibility to acne [14] and IGF-1 plasma levels correlate with acne severity [12].
Several worldwide observational studies have been published on dairy intake and acne in children,
adolescents, and young adults (7–30 years) in various countries [15–27]. Some narrative and systematic
reviews about dairy intake and acne have been published [4,9,28]. Recently, a meta-analysis of dairy
and acne was published [29] but with several methodological flaws, including lack of bias assessment
and inadvertent double-counting of studies due to duplicate publications [19,23,30,31] that caused
inappropriate weighting of results and skewed pooled estimates. So far, no previous meta-analysis has
statistically combined the observational studies in an attempt to estimate the effect of the association
of dairy intake and acne with the heterogeneity across studies, a bias assessment, a stratified analysis
by study characteristics, and publication bias.
The primary objective of this study was therefore to perform a meta-analysis to estimate
the association of acne in children, adolescents, and young adults consuming any dairy products.
Furthermore, our aim was to explore the association between acne and intake of varies types of dairy
(milk, yogurt, cheese), dairy subgroups (full fat, low fat, skim), and various amounts and frequencies of
dairy intake (times per week or day).
2. Methods
This systematic review and meta-analysis was undertaken according to Meta-analysis of
Observational Studies in Epidemiology (MOOSE) guidelines and according to a specified protocol
(Supplementary Materials). The search, selection of studies, full-text reading, and data extraction were
performed by CRJ and verified by CE.
127
Nutrients 2018, 10, 1049
beliefs/opinions about acne aggravating food items [32,33], semi-fat/whole milk vs. skim milk/no
milk drinkers [34], Chinese ying-yang medicine [35], no control group [36], adult acne (mean age
≥ 30 years) [37,38], milk as part of a Mediterranean diet [39], milk only as a continuous variable
in acne and non-acne groups [40], and poorly defined intake [41]. In total, we included 14 studies.
Two other studies were identified outside the search, but these studies were duplicates and published
simultaneously without a clear statement of which one was the original; therefore, we did not include
these papers [30,31]. The study selection process is shown in a flow diagram (Figure 1).
128
Nutrients 2018, 10, 1049
low-fat/skim milk, analyses of frequencies (times per week or day) were performed using studies by
Adebamowo et al. [15–17], as these studies had identical ascertainment of the frequency of milk intake.
DerSimonian and Laird (D + L) pooled random effects estimates were used. We also present inverse
variance (I-V) fixed effects in supplementary Figures. Heterogeneity was assessed by Cochrane Q
statistic test and I2 -statistical analysis. The I2 -statistical analysis assess what proportion of the observed
variance reflects variance in true effect sizes rather than sampling error [44]. Publication bias was
examined visually by funnel plots and statistically using Egger’s test (one-sided) [45] and by using the
Duval and Tweedie's Trim and Fill to simulate where potential unpublished studies would belong in
the funnel plot and to calculate a hypothetical new pooled odds ratio based on the added simulated
studies. Robustness of the meta-analysis was examined by “leaving-one-out” analysis. Publication
bias and robustness were carried out by use of the statistical program Comprehensive Meta-Analysis
(CMA) version 3 (Biostat, Englewood, NJ, USA) for any dairy intake vs. no dairy intake and any milk
intake vs. no milk intake. Four studies provided adjusted estimates for milk intake, with one study
providing them as odds ratios [46], and three studies as prevalence ratios [15–17]. In a sensitivity
analysis, we used only adjusted prevalence ratios from the studies by Adebamowo et al. [15–17].
Stratification on acne severity was not possible because of too few studies.
3. Results
3.2. Findings
Random effects pooled unadjusted odds ratios for acne were 1.25 (95% CI: 1.15–1.36;
p = 6.13 × 10−8 ) for any dairy (Figure 2), 1.22 (1.08–1.38; p = 1.62 × 10−3 ) for full-fat dairy,
1.28 (1.13–1.44; p = 8.23 × 10−5 ) for any milk, 1.22 (1.06–1.41; p = 6.66 × 10−3 ) for whole milk,
1.32 (1.16–1.52; p = 4.33 × 10−5 ) for low-fat/skim milk, 1.22 (1.00–1.50; p = 5.21 × 10−2 ) for cheese,
and 1.36 (1.05–1.77; p = 2.21 × 10−2 ) for yogurt compared to those who did not consume these food
items (Figure 3 and Supplementary Figures S1–S6).
Random effects meta-analyses for acne by frequency of any milk intake compared to an intake
of ≤1 glass of milk per week showed an odds ratio of 1.24 (0.95–1.62) by 2–6 glasses per week,
1.41 (1.05–1.90) by 1 glass per day, and 1.43 (1.09–1.88) by ≥2 glasses per day for any milk; results for
whole milk and low-fat/skim milk were close (Supplementary Figures S7–S10).
129
Table 1. Characteristics of included studies for the association of dairy intake with acne in children, adolescents, and young adults.
Age, Acne
First Author Year Study Population Design Gender Country Total n Acne n No Acne n Milk Variables Acne Diagnosis
Year (%)
Adebamowo Any milk, whole milk, low-fat
2005 Population cohort (Nurses’ Health Study II) Retrospective 13–18 F USA 46,879 3412 7.28 43,467 Q
[15] milk, skim milk
Adebamowo Population cohort (GUTS)—offspring of women in Any milk, whole milk, low-fat
2006 Follow-up 9–15 F USA 3756 2588 68.9 1168 Q
[17] the Nurses’ Health Study II milk, skim milk
Nutrients 2018, 10, 1049
Adebamowo Population cohort (GUTS)—offspring of women in Any milk, whole milk, low-fat
2008 Follow-up 9–15 M USA 2759 1856 67.3 903 Q
[16] the Nurses’ Health Study II milk, skim milk
Cerman [22] 2016 Acne patients and healthy controls Case-control 19 F/M Turkey 86 50 58.1 36 Any milk D
Whole milk, low-fat milk,
Duquia [46] 2017 Acne patients vs. healthy controls in the army Cross-sectional 18 M Brasil 2201 1960 89.1 241 P
cheese, yogurt
Acne patients and non-acne dermatology Any milk, whole milk, skim
Grossi [23] 2016 Case-control 10–24 F/M Italy 563 205 36.4 358 D
patient controls milk, cheese/yogurt combined
Ismail [18] 2012 Acne patients and healthy controls Case-control 18–30 F/M Malaysia 88 44 50 44 Any milk, yogurt, cheese D
Acne patients and non-acne dermatology
Karadag [26] 2017 Case-control 21 M Turkey 4595 3836 83.5 759 Milk/cheese combined D
patient controls
South
Jung [24] 2010 Acne patients and age-matched healthy controls Case-control 24 F/M 1285 783 60.9 502 Cheese D
Korea
Okoro [21] 2016 Population cohort Cross-sectional 11–30 F/M Nigeria 450 292 64.9 158 Any milk D
South
Park [47] 2015 Population cohort Cross-sectional 7–12 F/M 693 251 36.2 442 Any milk, cheese, yogurt D
Korea
Ulvestad [25] 2016 Population cohort Follow-up 15–19 F/M Norway 2387 331 13.9 2056 Any milk, full-fat milk Q
Wolkenstein [20] 2015 Population cohort Cross-sectional 15–24 F/M France 2266 1375 60.7 891 Any milk Q
Whole milk, semi-skimmed
Wolkenstein [27] 2017 Population cohort Cross-sectional 15–24 F/M Europe * 10521 6063 57.6 4458 Q
milk, low-fat milk, dairy
130
Age: mean or range. Q: Questionnaire. D: Dermatologist verified. GUTS: Growing Up Today Study. P: Physician verified. USA: United States of America. * 7 countries: Belgium, Czech
and Slovak Republics, France, Italy, Poland, and Spain.
Nutrients 2018, 10, 1049
Figure 2. Meta-analysis of dairy intake and acne vulgaris: individual studies. The figure shows the
individual studies and the unadjusted pooled random effect estimate from the meta-analysis of dairy
intake and acne vulgaris. I2 (%): I-square heterogeneity expressed as percentage. p-value(het): p-value
from Cochran’s Q-statistic assessing heterogeneity. D + L: DerSimonian and Laird pooled random
effects estimates. See Table 1 for references.
Dairy Adjustment Gender Studies(N) Acne(N) Controls(N) Total(N) pvalue(het) I2(%) OR (95% CI) pvalue
Any dairy No All 14 23046 55483 78529 0.05 42 1.25 (1.15, 1.36) 6.13e-08
Full-fat dairy No All 7 7792 35344 43136 0.05 52 1.22 (1.08, 1.38) 1.62e-03
Any milk No All 11 18095 52164 70259 0.01 58 1.28 (1.13, 1.44) 8.23e-05
Whole milk No All 6 7452 33248 40700 0.03 59 1.22 (1.06, 1.41) 6.66e-03
Low-fat/skim milk No All 6 10672 21931 32603 0.06 53 1.32 (1.16, 1.52) 4.33e-05
Cheese No All 4 3037 1229 4266 0.54 0 1.22 (1.00, 1.50) 5.21e-02
Yogurt No All 3 2255 727 2982 0.77 0 1.36 (1.05, 1.77) 2.21e-02
Any milk Yes All 3 7856 45538 53394 0.21 36 1.15 (1.10, 1.20) 3.95e-09
Whole milk Yes All 3 4265 31893 36158 0.04 70 1.14 (1.05, 1.23) 1.97e-03
Low-fat/skim milk Yes All 3 4174 18394 22568 0.48 0 1.18 (1.13, 1.22) 2.71e-17
.8 1 2
Odds ratio (95%CI)
Figure 3. Meta-analyses of dairy intake and acne vulgaris: summary estimates. The figure shows the
unadjusted pooled random effects estimates from each of the meta-analyses, which can be found in the
supplementary material. I2 (%): I-square heterogeneity expressed as percentage. p-value(het): p-value
from Cochran’s Q-statistic assessing heterogeneity.
131
Nutrients 2018, 10, 1049
3.3. Sensitivity Analyses, Heterogeneity, Publication Bias, and Qualitative Bias Assessment
The I2 heterogeneity ranged from 0–70% (Figure 2). To explore heterogeneity, we stratified the
analysis for any dairy intake and acne by age, gender, number of cases, continent, design, acne
diagnosis, and reference group (Supplementary Table S1, Supplementary Figures S11–S17). Stratifying
by age did not show any differences. Stratifying by gender showed similar odds ratios in males and
females, but meta-analyses of females had higher heterogeneity. Stratifying by the number of acne
cases showed that larger studies had smaller odds ratios with more narrow confidence intervals,
but higher heterogeneity compared to those of the smaller studies, but the confidence intervals were
overlapping. Stratifying analyses by continent showed that studies from Europe had the smallest
odds ratios, followed by North and South American studies, and with Asian and African studies with
the largest odds ratios. Stratifying by design removed heterogeneity and showed that prospective
studies had the largest odds ratios. Stratifying by ascertainment of acne diagnosis showed that studies
using self-reported acne as an outcome had higher heterogeneity compared to studies with physician
verified diagnoses of acne. Stratifying by reference group showed overall similar summary estimates,
but with the highest heterogeneity in studies with “less than weekly” being the reference group.
The Newcastle-Ottawa qualitative assessment scale of bias with similar items as in the statistical
heterogeneity assessments revealed scores of 2–5 in case-control studies [18,22–24,26] and 2–6 in cohort
studies [15–17,20,21,25,27,46,47] out of a potential max of 9 points (Supplementary Table S2).
Random effects pooled adjusted estimates for any milk, whole milk, and low-fat/skim milk were
similar but attenuated compared to their unadjusted estimates (Supplementary Figures S18–S20).
Leave-one-out analyses for any dairy or any milk intake did not show any gross deviations, but the
retrospective study by Adebamowo [15] influenced the summary estimates the most (Supplementary
Figures S21–S22). Funnel plot and p-value for Egger’s test revealed publication bias for any dairy
(p-Egger = 4.71 × 10−3 ) (Supplementary Figure S23); Duval and Tweedie’s Trim and Fill method
estimated that five studies were missing for “any dairy”, and the imputed point estimate would be
1.16 (1.06–1.28) had these five studies been added. Funnel plot and p-value for Egger’s test revealed
publication bias for any milk (p-Egger = 2.73 × 10−2 ) (Supplementary Figure S24); Duval and Tweedie's
Trim and Fill method estimated that one study was missing for “any milk”, and the imputed point
estimate would be 1.26 (1.11–1.44) had this study been added.
The New-Castle Ottawa qualitative assessment scale of bias revealed a scores of 2–5 in case-control
studies [18,22–24,26] and 2–6 in cohort studies [15–17,20,21,25,27,46,47].
4. Discussion
Intake of any dairy, any milk, full-fat dairy, whole milk, low-fat/skim milk, and yogurt regardless
of amount or frequency were associated with a higher odds ratio for acne compared to no intake in
individuals aged 7–30 years. Intake of cheese was associated with a borderline higher odds ratio for
acne compared to no intake. Stratifying the association of any milk by frequency of intake revealed
that intake of 1 glass of milk or more per day was associated with a higher odds ratio for acne,
whereas 2–6 glasses per week was not, compared to intake less than weekly. Stratified analyses for
any dairy intake and acne fat content demonstrated that full-fat dairy and whole milk had lower
odds ratios, whereas low-fat/skim milk had higher odds ratios than the overall summary estimates;
a likely explanation for this observation could be that the amount of milk consumed for low-fat/skim
milk is higher than that for whole milk. However, results should be interpreted with caution due to
heterogeneity and bias across studies.
The meta-analyses showed considerable heterogeneity reflecting the heterogeneous age and
gender of the participants, various study characteristics, ascertainment of information about milk
intake and acne, reporting of milk intake, and acne severity across the studies. In general, stratifying
on subgroups in sensitivity analyses revealed that heterogeneity diminished for most subgroups,
but also revealed that especially meta-analyses conducted on females, whole milk, North America,
132
Nutrients 2018, 10, 1049
and questionnaire ascertained acne diagnosis demonstrated high heterogeneity. Prospective studies
and studies with physician-verified diagnosis of acne had low heterogeneity.
Despite the stratifications, confidence intervals were overlapping.
Stratifying on age and gender demonstrated similar odds ratios; however, the gender stratified
analyses had higher odds ratios than in the gender combined analyses. Smaller studies had higher
odds ratios than large studies, African and Asian studies had higher odds ratios than other studies,
and prospective designs had higher odds ratios than other designs. A recent multinational European
online questionnaire study in adolescents showed that acne prevalence did not differ by gender but
differed by country, and acne was more prevalent in younger people and obese people [27]. Intake of
milk varies globally and is largely dependent on genetically determined lactase persistency, which is
high in people of Northern European descent, but lower in people of Southern European descent,
patchy in Africa, and low in the Middle East and Asia [48]. The weaning of the lactase enzyme activity
usually happens in childhood and early adolescent years. How the age of weaning of the lactase
enzyme activity impacts acne development is not known.
We used random effects method in all meta-analyses, which includes between-study variance and
has a higher degree of statistical uncertainty built into the model. Thus, 95% confidence intervals are
wider compared to fixed effects models. Even in these models, the results of the meta-analyses were
significant. There was evidence of publication bias with Egger’s test with an overweight of smaller
studies overestimating the odds ratio compared to the pooled summary estimate. If the meta-analyses
had captured all the relevant studies, we would expect the funnel plots to be symmetric. The selective
reporting may be explained by studies with null-findings or negative results being deliberately not
published because of authors not submitting or editors rejecting them or authors not finding enough
merit in a potential publishable study [49]. Furthermore, some studies reported only the pooled
exposures for different dairy groups rather than showing the stratified results for each of the dairy
groups and/or for each reported frequency of intake [23,25,26], and some studies had only collected
an overall dairy or milk variable with no possibility for stratification [20,22]. However, the trim and fill
method did not change the overall estimates for “any dairy” or “any milk” remarkably.
There are many limitations of the included studies [4]. Self-reported acne with lack of
a physician verified diagnosis of acne [15–17,20,25] may lead to misclassification bias as validity
of self-reported acne is at best only moderate, with sensitivity of 55%, specificity of 72%, positive
predictive value of 70%, and negative predictive value of 57% [50]. Including other dermatology
patients as controls [26] may attenuate associations, as seborrhea may play a role in several
diseases. The observational studies were cross-sectional [20,21,46,47], case-control [18,22–24,26],
retrospective [15], or longitudinal [16,17,25]; thus, in most studies we cannot rule out reverse causation.
Questionnaire ascertainment of dairy intake varied between the articles and only a few studies used
validated food frequency questionnaires [15–17,21]. Despite the food questionnaire used, participants
may deliberately over- or underestimate (information bias) or not accurately remember (recall bias)
when filling out questionnaires about dairy intake and acne. Furthermore, it was not possible to
differentiate acne development, acne triggers, and severity of acne in the meta-analyses. Only a few
studies provided adjusted results [15–17,25] so we based most of the analyses on raw numbers,
which makes it difficult to rule out confounding from other dietary factors (e.g., glycemic index or
calorie intake) or other lifestyle factors previously associated with acne [4,9,28].
Acne prevalence varied remarkably across the included studies, between 7–89%. The retrospective
study by Adebamowo in 2005 with 7.3% acne cases focused on recall data provided by subjects in
the Nurses’ Health Study II (NHS), which were aged 25–42 years old in 1989 when information on
teenage acne was collected [15]; thus, the acne prevalence is likely underestimated and the results from
this study may not be representative. Furthermore, the studies from 2006 and 2008 were offspring
studies from the NHS in girls and boys [16,17]; however, leave-one-out analyses revealed that only the
Adebamowo 2005 study was an outlier [15].
133
Nutrients 2018, 10, 1049
The observational studies may suffer from bias from confounding and reverse causation [9],
are unable to indicate causality of the relationship between dairy and acne, and unable to prove
preventive effects of abstaining from dairy. Only one study exists on milk intake and acne. The study
is uncontrolled and unblinded and is based on medical students who drank milk or consumed other
potential acne provoking foods. In addition, the total number of people with and without acne lesions
were counted for all foods combined, but with no formal statistical testing [51]. Thus, there is still
a knowledge gap with respect to whether dairy intake is causally associated with acne, acne flare, or
acne severity and to what extent. To answer this question, we would ideally need results from large
clinical randomized double-blind placebo-controlled trials (RCT); however, the question is whether
this is realistically possible ethically, clinically, and/or operationally. Another approach (which no
previous studies have yet undertaken) would be to perform a Mendelian Randomization study of
lactase persistence, dairy intake, and acne using genetic lactase persistence as a proxy for lifetime dairy
intake under the assumption that alleles are randomly distributed at conception [52,53]. Such a study
design mimics an RCT and allows for the causal estimate of dairy intake and acne.
The observational studies all assessed dairy intake as an isolated factor. However, dairy is
part of various individual and cultural specific diets and not a single factor with a single factor
prediction (“reductionist approach” [54]). Instead, other factors which can affect the bioactive
properties of nutrients in dairy and milk intake should be taken into consideration, such as macro-
and micronutrients (fat, protein, carbohydrates, vitamins, sodium, and minerals), the dairy structure
(liquid or solid), fermentation, and processing (holistic approach [55]). Only two studies in the
meta-analysis also reported the glycemic load and glycemic indices of food consumed in conjunction
with milk/dairy products [18,22], but did not report the glycemic load from the dairy consumption
specifically. Hyperglycemic carbohydrates enhance insulin signaling, which promotes insulin and
IGF-1 signaling, which in a synergistic fashion with milk stimulate mTORC1(mammalian target of
rapamycin complex 1) signal transduction [56]. There is accumulating evidence that acne belongs
to the spectrum of mTORC1-driven diseases of civilization including metabolic syndrome, obesity,
insulin resistance, and cancer [57]. A randomized trial has shown that a low-glycemic-load diet
improves symptoms in acne vulgaris patients [58]. Interestingly, no acne was observed in the Kitavan
Islanders (Pacific Ocean) and in the Ache Hunter-Gatherers from Paraguay, who live under Paleolithic
conditions without milk/dairy and hyperglycemic food, although it should be acknowledged that
many other differences exist to Western societies [59]. To present the pathological effects of milk in the
Western diet it is therefore important to provide controlled studies that consider milk consumption in
association with glycemic load and index as part of a mixed diet [60].
Recently, a meta-analysis of dairy and acne was published [29] but with several
methodological flaws, including the inadvertent double-counting of studies (Landro [19]/Grossi [23],
and Tsoy [30]/Tsoy [31]) due to duplicate publications, which caused inappropriate weighting of
results and skewed pooled estimates. Using the double-counted studies by Tsoy, the authors also only
used the most severe category of acne, which caused extremely high odds ratios of 10 and 12 to be
included in the meta-analysis, further skewing the pooled estimates. Furthermore, the meta-analysis
included a study by Agamia [41], which we decided to exclude as the intake of “milk and dairy
produce” was poorly defined as “low” and “high” intake but not defined with any frequency, type,
or amount of milk. The previous meta-analysis also did not provide evidence for the exact search
strategy to be replicated, for the bias assessment using the Newcastle Ottawa scale, for leave-one-out
analyses, or funnel plots of publication bias. As a comparison, in our meta-analysis, we included
the exact search string so it can be replicated, the heterogeneity across studies, a bias assessment
using the Newcastle-Ottawa scale presented with a table, a stratified analysis by study characteristics
presented in figures, the details of the “leave-one-out” analysis presented in figures, and the publication
bias presented in figures. Furthermore, we excluded duplicate studies, and we included four more
papers [24,26,27,47] that were not included in the previous meta-analysis but should have been as the
studies were published before the search for the previous meta-analysis was done in August 2017 [29].
134
Nutrients 2018, 10, 1049
It is of crucial importance that authors of meta-analyses have a critical judgement of the reliability
and validity of the papers they consider including in a meta-analysis, otherwise the conduct and
assessment of systematic reviews may be hampered.
5. Conclusions
In conclusion, this meta-analysis of observational studies has provided new insight into the
direction and magnitude of the association between dairy intake and acne overall and by dairy type,
amount, and frequency. It has shed light on the knowledge gaps and the limitations of the studies
included compared to previous systematic and narrative reviews with no meta-analysis, heterogeneity
assessment, or bias assessment included [4,9,28].
References
1. Williams, H.C.; Dellavalle, R.P.; Garner, S. Acne vulgaris. Lancet 2012, 379, 361–372. [CrossRef]
2. Tuchayi, S.M.; Makrantonaki, E.; Ganceviciene, R.; Dessinioti, C.; Feldman, S.R.; Zouboulis, C.C. Acne
vulgaris. Nat. Rev. Dis. Primers. 2015, 1, 15029. [CrossRef] [PubMed]
3. Melnik, B.C.; Schmitz, G. Role of insulin, insulin-like growth factor-1, hyperglycaemic food and milk
consumption in the pathogenesis of acne vulgaris. Exp. Dermatol. 2009, 18, 833–841. [CrossRef] [PubMed]
4. Bhate, K.; Williams, H.C. Epidemiology of acne vulgaris. Br. J. Dermatol. 2013, 168, 474–485. [CrossRef]
[PubMed]
5. White, G.M. Recent findings in the epidemiologic evidence, classification, and subtypes of acne vulgaris.
J. Am. Acad. Dermatol. 1998, 39, S34–S47. [CrossRef]
135
Nutrients 2018, 10, 1049
6. Rademaker, M.; Garioch, J.J.; Simpson, N.B. Acne in schoolchildren: No longer a concern for dermatologists.
BMJ 1989, 298, 1217–1229. [CrossRef] [PubMed]
7. Ghodsi, S.Z.; Orawa, H.; Zouboulis, C.C. Prevalence, severity, and severity risk factors of acne in high school
pupils: A community-based study. J. Investig. Dermatol. 2009, 129, 2136–2141. [CrossRef] [PubMed]
8. Dreno, B.; Bettoli, V.; Araviiskaia, E.; Sanchez Viera, M.; Bouloc, A. The influence of exposome on acne. J. Eur.
Acad. Dermatol. Venereol. 2018, 32, 812–829. [CrossRef] [PubMed]
9. Spencer, E.H.; Ferdowsian, H.R.; Barnard, N.D. Diet and acne: A review of the evidence. Int. J. Dermatol.
2009, 48, 339–347. [CrossRef] [PubMed]
10. Rich-Edwards, J.W.; Ganmaa, D.; Pollak, M.N.; Nakamoto, E.K.; Kleinman, K.; Tserendolgor, U.; Willett, W.C.;
Frazier, A.L. Milk consumption and the prepubertal somatotropic axis. Nutr. J. 2007, 6, 28. [CrossRef]
[PubMed]
11. Holmes, M.D.; Pollak, M.N.; Willett, W.C.; Hankinson, S.E. Dietary correlates of plasma insulin-like growth
factor I and insulin-like growth factor binding protein 3 concentrations. Cancer Epidemiol. Biomarkers Prev.
2002, 11, 852–861. [PubMed]
12. Rahaman, S.M.A.; De, D.; Handa, S.; Pal, A.; Sachdeva, N.; Ghosh, T.; Kamboj, P. Association of
insulin-like growth factor (IGF)-1 gene polymorphisms with plasma levels of IGF-1 and acne severity.
J. Am. Acad. Dermatol. 2016, 75, 768–773. [CrossRef] [PubMed]
13. Mirdamadi, Y.; Thielitz, A.; Wiede, A.; Goihl, A.; Papakonstantinou, E.; Hartig, R.; Zouboulis, C.C.;
Reinhold, D.; Simeoni, L.; Bommhardt, U.; et al. Insulin and insulin-like growth factor-1 can modulate the
phosphoinositide-3-kinase/Akt/FoxO1 pathway in SZ95 sebocytes in vitro. Mol. Cell. Endocrinol. 2015, 415,
32–44. [CrossRef] [PubMed]
14. Tasli, L.; Turgut, S.; Kacar, N.; Ayada, C.; Coban, M.; Akcilar, R.; Ergin, S. Insulin-like growth factor-I gene
polymorphism in acne vulgaris. J. Eur. Acad. Dermatol. Venereol. 2013, 27, 254–267. [CrossRef] [PubMed]
15. Adebamowo, C.A.; Spiegelman, D.; Danby, F.W.; Frazier, A.L.; Willett, W.C.; Holmes, M.D. High school
dietary dairy intake and teenage acne. J. Am. Acad. Dermatol. 2005, 52, 207–214. [CrossRef] [PubMed]
16. Adebamowo, C.A.; Spiegelman, D.; Berkey, C.S.; Danby, F.W.; Rockett, H.H.; Colditz, G.A.; Willett, W.C.;
Willett, W.C. Milk consumption and acne in teenaged boys. J. Am. Acad. Dermatol. 2008, 58, 787–793.
[CrossRef] [PubMed]
17. Adebamowo, C.A.; Spiegelman, D.; Berkey, C.S.; Danby, F.W.; Rockett, H.H.; Colditz, G.A.; Willett, W.C.;
Holmes, M.D. Milk consumption and acne in adolescent girls. Dermatol. Online J. 2006, 12, 1. [PubMed]
18. Ismail, N.H.; Manaf, Z.A.; Azizan, N.Z. High glycemic load diet, milk and ice cream consumption are related
to acne vulgaris in Malaysian young adults: A case control study. BMC Dermatol. 2012, 12, 13. [CrossRef]
[PubMed]
19. Di Landro, A.; Cazzaniga, S.; Parazzini, F.; Ingordo, V.; Cusano, F.; Atzori, L.; Cutrì, F.T.; Musumeci, M.L.;
Zinetti, C.; Pezzarossa, E.; et al. Family history, body mass index, selected dietary factors, menstrual history,
and risk of moderate to severe acne in adolescents and young adults. J. Am. Acad. Dermatol. 2012, 67,
1129–1135. [CrossRef] [PubMed]
20. Wolkenstein, P.; Misery, L.; Amici, J.M.; Maghia, R.; Branchoux, S.; Cazeau, C.; Voisard, J.-J.; Taïeb, C.
Smoking and dietary factors associated with moderate-to-severe acne in French adolescents and young
adults: Results of a survey using a representative sample. Dermatology 2015, 230, 34–49. [CrossRef] [PubMed]
21. Okoro, E.O.; Ogunbiyi, A.O.; George, A.O.; Subulade, M.O. Association of diet with acne vulgaris among
adolescents in Ibadan, southwest Nigeria. Int. J. Dermatol. 2016. [CrossRef] [PubMed]
22. Cerman, A.A.; Aktas, E.; Altunay, I.K.; Arici, J.E.; Tulunay, A.; Ozturk, F.Y. Dietary glycemic factors, insulin
resistance, and adiponectin levels in acne vulgaris. J. Am. Acad. Dermatol. 2016. [CrossRef]
23. Grossi, E.; Cazzaniga, S.; Crotti, S.; Naldi, L.; Di Landro, A.; Ingordo, V.; Cusano, F.; Atzori, L.; Cutrì, F.T.;
Musumeci, M.L.; et al. The constellation of dietary factors in adolescent acne: A semantic connectivity map
approach. J. Eur. Acad. Dermatol. Venereol. 2016, 30, 96–100. [CrossRef] [PubMed]
24. Jung, J.Y.; Yoon, M.Y.; Min, S.U.; Hong, J.S.; Choi, Y.S.; Suh, D.H. The influence of dietary patterns on acne
vulgaris in Koreans. Eur. J. Dermatol. 2010, 20, 768–772. [CrossRef] [PubMed]
25. Ulvestad, M.; Bjertness, E.; Dalgard, F.; Halvorsen, J.A. Acne and dairy products in adolescence: Results
from a Norwegian longitudinal study. J. Eur. Acad. Dermatol. Venereol. 2016. [CrossRef] [PubMed]
136
Nutrients 2018, 10, 1049
26. Karadag, A.S.; Balta, I.; Saricaoglu, H.; Kilic, S.; Kelekci, K.H.; Yildirim, M.; Arica, D.A.; Öztürk, S.; Karaman, G.;
Cerman, A.A.; et al. The effect of personal, familial, and environmental characteristics on acne vulgaris:
A prospective, multicenter, case controlled study from Turkey. G. Ital. Dermatol. Venereol. 2017. [CrossRef]
27. Wolkenstein, P.; Machovcova, A.; Szepietowski, J.C.; Tennstedt, D.; Veraldi, S.; Delarue, A. Acne prevalence
and associations with lifestyle: A cross-sectional online survey of adolescents/young adults in 7 European
countries. J. Eur. Acad. Dermatol. Venereol. 2017. [CrossRef] [PubMed]
28. Veith, W.B.; Silverberg, N.B. The association of acne vulgaris with diet. Cutis 2011, 88, 84–91. [PubMed]
29. Aghasi, M.; Golzarand, M.; Shab-Bidar, S.; Aminianfar, A.; Omidian, M.; Taheri, F. Dairy intake and acne
development: A meta-analysis of observational studies. Clin. Nutr. 2018. [CrossRef] [PubMed]
30. Tsoy, N. The Influence of Dietary Habits on Acne. World J. Med. Sci. 2013, 8, 212. [CrossRef]
31. Tsoy, N.O. Effect of Milk and Dairy Products upon Severity of Acne for Young People. World Appl. Sci. J.
2013, 24, 403–417. [CrossRef]
32. Burris, J.; Rietkerk, W.; Woolf, K. Relationships of self-reported dietary factors and perceived acne severity in
a cohort of New York young adults. J. Acad. Nutr. Diet. 2014, 114, 384–392. [CrossRef] [PubMed]
33. Poli, F.; Auffret, N.; Beylot, C.; Chivot, M.; Faure, M.; Moyse, D.; Henri, P.; Jean, R.; Brigitte, D. Acne as seen
by adolescents: Results of questionnaire study in 852 French individuals. Acta Derm. Venereol. 2011, 91,
531–546. [CrossRef] [PubMed]
34. Semedo, D.; Ladeiro, F.; Ruivo, M.; D’Oliveira, C.; De Sousa, F.; Gayo, M.; Amado, J.; Lima, C.; Magalhães, F.;
Brandão, R.; et al. Adult Acne: Prevalence and Portrayal in Primary Healthcare Patients, in the Greater Porto
Area, Portugal. Acta. Med. Port. 2016, 29, 507–513. [CrossRef] [PubMed]
35. Law, M.P.; Chuh, A.A.; Molinari, N.; Lee, A. An investigation of the association between diet and occurrence
of acne: a rational approach from a traditional Chinese medicine perspective. Clin. Exp Dermatol. 2010, 35,
31–45. [CrossRef] [PubMed]
36. Seleit, I.; Bakry, O.A.; Abdou, A.G.; Hashim, A. Body mass index, selected dietary factors, and acne severity:
Are they related to in situ expression of insulin-like growth factor-1? Anal. Quant. Cytopathol. Histpathol.
2014, 36, 267–278. [PubMed]
37. Dreno, B.; Thiboutot, D.; Layton, A.M.; Berson, D.; Perez, M.; Kang, S. Large-scale international study
enhances understanding of an emerging acne population: Adult females. J. Eur. Acad. Dermatol. Venereol.
2015, 29, 1096–1106. [CrossRef] [PubMed]
38. Di Landro, A.; Cazzaniga, S.; Cusano, F.; Bonci, A.; Carla, C.; Musumeci, M.L.; Patrizi, A.; Bettoli, V.;
Pezzarossa, E.; Caproni, M.; et al. Adult female acne and associated risk factors: Results of a multicenter
case-control study in Italy. J. Am. Acad. Dermatol. 2016, 75, 1134–1141. [CrossRef] [PubMed]
39. Skroza, N.; Tolino, E.; Semyonov, L.; Proietti, I.; Bernardini, N.; Nicolucci, F.; Viola, G.L.; Prete, G.D.; Saulle, R.;
Potenza, C.; et al. Mediterranean diet and familial dysmetabolism as factors influencing the development of
acne. Scand. J. Public Health 2012, 40, 466–474. [CrossRef] [PubMed]
40. LaRosa, C.L.; Quach, K.A.; Koons, K.; Kunselman, A.R.; Zhu, J.; Thiboutot, D.M.; Zaenglein, A.L.
Consumption of dairy in teenagers with and without acne. J. Am. Acad. Dermatol. 2016, 75, 318–322.
[CrossRef] [PubMed]
41. Agamia, N.F.; Abdallah, D.M.; Sorour, O.; Mourad, B.; Younan, D.N. Skin expression of mammalian target of
rapamycin and forkhead box transcription factor O1, and serum insulin-like growth factor-1 in patients with
acne vulgaris and their relationship with diet. Br. J. Dermatol. 2016, 174, 1299–1307. [CrossRef] [PubMed]
42. Stang, A. Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of
nonrandomized studies in meta-analyses. Eur. J. Epidemiol. 2010, 25, 603–615. [CrossRef] [PubMed]
43. Newcastle—Ottawa Quality Assessment Scale Case Control Studies. Available online: http://www.ohri.ca/
programs/clinical_epidemiology/nosgen.pdf (accessed on 12 December 2017).
44. Huedo-Medina, T.B.; Sanchez-Meca, J.; Marin-Martinez, F.; Botella, J. Assessing heterogeneity in
meta-analysis: Q statistic or I2 index? Psychol. Methods 2006, 11, 193–206. [CrossRef] [PubMed]
45. Egger, M.; Smith, G.D.; Schneider, M.; Minder, C. Bias in meta-analysis detected by a simple, graphical test.
BMJ 1997, 315, 629–634. [CrossRef] [PubMed]
46. Pereira Duquia, R.; da Silva Dos Santos, I.; de Almeida, H., Jr.; Martins Souza, P.R.; de Avelar Breunig, J.;
Zouboulis, C.C. Epidemiology of Acne Vulgaris in 18-Year-Old Male Army Conscripts in a South Brazilian
City. Dermatology 2017, 233, 145–154. [CrossRef] [PubMed]
137
Nutrients 2018, 10, 1049
47. Park, S.Y.; Kwon, H.H.; Min, S.; Yoon, J.Y.; Suh, D.H. Epidemiology and risk factors of childhood acne
in Korea: A cross-sectional community based study. Clin. Exp. Dermatol. 2015, 40, 844–850. [CrossRef]
[PubMed]
48. Itan, Y.; Jones, B.L.; Ingram, C.J.; Swallow, D.M.; Thomas, M.G. A worldwide correlation of lactase persistence
phenotype and genotypes. BMC Evol. Biol. 2010, 10, 36. [CrossRef] [PubMed]
49. Higgins, J.P.T.; Green, S. Cochrane Handbook for Systematic Reviews of Interventions. Available online:
http://handbook-5-1.cochrane.org/ (accessed on 12 December 2017).
50. Menon, C.; Gipson, K.; Bowe, W.P.; Hoffstad, O.J.; Margolis, D.J. Validity of subject self-report for acne.
Dermatology 2008, 217, 164–178. [CrossRef] [PubMed]
51. Anderson, P.C. Foods as the cause of acne. Am. Fam. Physician 1971, 3, 102–113. [PubMed]
52. Bergholdt, H.K.M.; Nordestgaard, B.G.; Varbo, A.; Ellervik, C. Milk intake is not associated with ischaemic
heart disease in observational or Mendelian randomization analyses in 98,529 Danish adults. Int. J. Epidemiol.
2015, 44, 587–603. [CrossRef] [PubMed]
53. Juhl, C.R.; Bergholdt, H.K.M.; Miller, I.M.; Jemec, G.B.; Kanters, J.K.; Ellervik, C. Lactase persistence, milk
intake and adult acne: A Mendelian randomization study of 20,416 Danish adults. Nutrients 2018, 10, 1041.
[CrossRef]
54. Fardet, A.; Rock, E. Toward a new philosophy of preventive nutrition: From a reductionist to a holistic
paradigm to improve nutritional recommendations. Adv. Nutr. 2014, 5, 430–446. [CrossRef] [PubMed]
55. Thorning, T.K.; Bertram, H.C.; Bonjour, J.P.; de Groot, L.; Dupont, D.; Feeney, E.; Ipsen, R.; Lecerf, J.M.;
Mackie, A.; McKinley, M.C.; et al. Whole dairy matrix or single nutrients in assessment of health effects:
Current evidence and knowledge gaps. Am. J. Clin. Nutr. 2017, 105, 1033–1045. [CrossRef] [PubMed]
56. Melnik, B.C. Milk—A Nutrient System of Mammalian Evolution Promoting mTORC1-Dependent Translation.
Int. J. Mol. Sci. 2015, 16, 17048–17087. [CrossRef] [PubMed]
57. Melnik, B.C. Acne vulgaris: The metabolic syndrome of the pilosebaceous follicle. Clin. Dermatol. 2018, 36,
29–40. [CrossRef] [PubMed]
58. Smith, R.N.; Mann, N.J.; Braue, A.; Makelainen, H.; Varigos, G.A. A low-glycemic-load diet improves
symptoms in acne vulgaris patients: A randomized controlled trial. Am. J. Clin. Nutr. 2007, 86, 107–115.
[CrossRef] [PubMed]
59. Cordain, L.; Lindeberg, S.; Hurtado, M.; Hill, K.; Eaton, S.B.; Brand-Miller, J. Acne vulgaris: A disease of
Western civilization. Arch. Dermatol. 2002, 138, 1584–1590. [CrossRef] [PubMed]
60. Elmstahl, H.L.; Bjorck, I. Milk as a supplement to mixed meals may elevate postprandial insulinaemia. Eur. J.
Clin. Nutr. 2001, 55, 994–1009. [CrossRef] [PubMed]
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
138
nutrients
Article
Association between Fasting Glucose Concentration,
Lipid Profile and 25(OH)D Status in Children
Aged 9–11
Lukasz Szternel 1, *, Magdalena Krintus 1 , Katarzyna Bergmann 1 , Tadeusz Derezinski 2
and Grazyna Sypniewska 1
1 Department of Laboratory Medicine, Nicolaus Copernicus University, Collegium Medicum,
85094 Bydgoszcz, Poland; [email protected] (M.K.); [email protected] (K.B.); [email protected] (G.S.)
2 Outpatient Clinic, “Esculap”, 88140 Gniewkowo, Poland; [email protected]
* Correspondence: [email protected]; Tel.: +48-525-854-023; Fax: +48-525-854-024
Abstract: Background: The aim of this study was to assess the relationship between vitamin D status
and the prevalence of dyslipidemia and impaired fasting glucose (IFG) in children. Methods and
Summary: 284 children (150 boys and 134 girls) aged 9–11 were included in the study. Children with
deficient 25(OH)D (25-hydroxycholecalciferol) levels ≤20 ng/mL (50 nmol/L) were characterized by
a more frequent occurrence of impaired fasting glucose (IFG) (Odd ratios (OR) = 1.966, 95% confidence
interval (CI): 1.055–3.663; p = 0.033) when compared to children with 25(OH)D >20 ng/mL. Serum
25(OH)D with concentration lower by 1 ng/mL (2.5 nmol/L) was linked to higher fasting glucose
(by 0.25 mg/dL, 0.013 mmol/L; p = 0.017), higher total cholesterol (TC) by almost 1 mg/dL
(0.96 mg/dL, 0.25 mmol/L; p = 0.006) and higher high-density lipoprotein cholesterol (HDL-C)
(by 0.57 mg/dL, 0.015 mmol/L; p < 0.001). Conclusion: 25(OH)D deficiency may negatively affect
fasting glucose and total cholesterol concentration in children aged 9–11. Vitamin D-deficient children
are twice as likely to develop prediabetes as reflected by impaired fasting glucose when compared to
those with a 25(OH)D level above 20 ng/mL (50 nmol/L).
1. Introduction
The prevalence of obesity and vitamin D deficiency among children makes this population
demographic especially vulnerable to the development of these two pervasive epidemics [1]. Research
suggests an unhealthy diet coupled with a sedentary lifestyle has become the main causal factor
affecting the development of obesity, which in turn is frequently accompanied by dyslipidemia.
A significant amount of data supports the hypothesis that optimal vitamin D concentration is linked to
a favorable lipid profile and has a positive impact on glucose homeostasis [2]. Numerous observational,
epidemiological, and cross-sectional studies indicate an inverse correlation between the concentration
of 25(OH)D (25-hydroxycholecalciferol) and the rate of conversion from a prediabetes state to fully
symptomatic diabetes mellitus [3]. In the USA, the prevalence of prediabetes (or according to the World
Health Organization (WHO) definition, “intermediate hyperglycemia”) among adolescents aged 12–19
reached 13.1% between 2005 and 2006 [4]. It is estimated that annually 5%–10% of adults convert from
a prediabetes state to overt diabetes, despite the fact that reversion to normoglycemia is much more
common in children and adolescents. There are hypotheses indicating potentially beneficial effects
of vitamin D in preventing transformation to fully symptomatic diabetes mellitus [3]. There is no
consistent agreement as to whether vitamin D deficiency causes lipid abnormalities, or if these are just a
consequence of excess adipose tissue mass storing 25(OH)D molecules. The inflammatory process links
obesity and insulin resistance to the consequences of improper glucose homeostasis [5]. Vitamin D,
being a negative marker of inflammation, seems to be a trigger molecule in the development of fully
symptomatic metabolic syndrome [6]. The effects of vitamin D on lipid and carbohydrate metabolism
may only be fully exploited with strong evidence from clinical trials of vitamin D supplementation.
The aim of this study is to assess the cross-sectional relationship between the status of vitamin D
and the indices of metabolic pathways of lipids and glucose in a pediatric population.
2. Methods
2.1. Characteristics of the Study Participants and the Panel of Laboratory Tests
This cross-sectional study involved 284 presumably healthy children aged 9–11. The recruitment
and blood collection process took place between October and November 2015. The children were
selected on the basis of age (9–11 years old) from four primary schools in the Kujawsko-Pomorskie
region of Poland. The second inclusion criterion was a fasting state (a minimum of 8 h since last meal)
before blood drawing. Whilst school nurses and specialists in internal medicine participated in the
recruitment process, the general health of the child on the day of study was subjectively evaluated
by their parents. Children with any underlying liver, kidney, or endocrine diseases, or who were
receiving drugs that affected vitamin D levels were excluded from the study. Immediately following
blood collection, the blood samples were transported to the laboratory and centrifuged. Serum was
used for further laboratory analysis. Whole blood samples were collected for HbA1c evaluation.
Vitamin D status (total 25(OH)D concentration), lipid panel (total cholesterol (TC), triglycerides (TG),
high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C)), C-reactive
protein (CRP), and glucose status (fasting glucose concentration and glycated hemoglobin) were
evaluated for all participants.
140
Nutrients 2018, 10, 1359
Risk of dyslipidemia was assessed in accord with the currently accepted cut-off values for fasting
lipids in children [11]. Vitamin D status represented by 25(OH)D concentration was also evaluated
according to currently accepted recommendations [12].
3. Results
Our study consisted of 284 children: 150 girls (52.8%) and 134 boys (47.2%) aged 9–11.
Characteristics of the study population are shown in Table 1.
141
Nutrients 2018, 10, 1359
Table 1. Cont.
Overweight and obesity levels were based on BMI percentiles according to age and sex, being
11.6% and 12.3% respectively. Vitamin D deficiency, estimated by total 25(OH)D concentration,
was identified in 37.7% of the children, whereas its insufficiency was identified in 58.8%. Children
with impaired fasting glucose (≥100 mg/dL; 5.6 mmol/L) constituted 17.6%, and an elevated
HbA1c value was seen in 11.1% (Table 1). The most commonly identified lipid abnormality was
hypercholesterolemia, which was 49.7% for TC and 35.6% for LDL-C, respectively.
Our analysis revealed that 25(OH)D concentration affected glucose metabolism as reflected by
changes in fasting glucose concentrations. As can be seen in Table 2, the risk of hyperglycemia (IFG)
was significantly higher (p = 0.033) in children with 25(OH)D deficiency (≤20 ng/mL; 50 nmol/L)
(OR = 1.966, 95% CI: 1.055–3.663).
95% CI
Variables * p Value OR
Lower Upper
Age 0.791 0.958 0.699 1.314
Sex 0.636 0.885 0.534 1.467
BMI percentiles 0.053 0.665 0.440 1.005
TC 0.169 1.688 0.800 3.562
TG 0.973 1.010 0.565 1.804
HDL-C 0.245 0.611 0.267 1.402
Non-HDL-C 0.624 1.267 0.493 3.257
142
Nutrients 2018, 10, 1359
Table 2. Cont.
95% CI
Variables * p Value OR
Lower Upper
LDL-C 0.257 0.599 0.247 1.454
hs-CRP 0.935 0.975 0.531 1.791
HBA1C 0.290 1.529 0.696 3.357
Glucose 0.069 1.828 0.955 3.499
Glucose ** 0.033 1.966 1.055 3.663
Logistic regression (multivariate): χ2 = 13.5; df = 11 (degrees of freedom); p = 0.258; r2 Cox and Snell = 0.047 (Cox and
Snell’s R squares complex samples logistic regression algorithms); r2 Nagelkerke = 0.064 (Nagelkerke R squares
complex samples logistic regression algorithms) * Statistical significance was determined in logistic regression
analysis. ** Logistic regression (univariate): χ2 = 4.513; df = 1; p = 0.034; r2 Cox and Snell=0.016; r2 Nagelkerke = 0.022.
25(OH)D: 25-hydroxycholecalciferol (calcifediol); BMI: body mass index; TC: total cholesterol; TG: triglycerides;
HDL-C: high-density lipoprotein cholesterol; non-HDL-C: non-high-density lipoprotein cholesterol (TC-(HDL-C));
LDL-C: low-density lipoprotein cholesterol (direct method); hs-CRP: high-sensitivity C-reactive protein; HbA1c :
glycated hemoglobin.
As can be seen in Figure 1, the prevalence of hyperglycemia and hypercholesterolemia was greater
in the vitamin D deficiency subgroup (24.0% and 57.3%, respectively), when compared to children
with concentrations of 25(OH)D over 50 nmol/L (20 ng/mL) (16.4% and 46.2%, respectively).
≤ 50nmol/L
24 %
25(OH)
57.3 %
ytuł osi
16.4 %
> 50nmol/L
25(OH)
46.2 %
0 10 20 30 40 50 60 70
Prevalence 漑%漒
Figure 1. Occurrence of hyperglycemia/hypercholesterolemia in vitamin D-deficient children.
TC: total cholesterol.
Pearson’s correlation coefficients across the whole study group revealed a statistically significant
weak negative correlation between 25(OH)D level and lipid variables (TG: r = −0.160; p = 0.037; HDL-C:
r = −0.209; p < 0.001), and also with glucose concentrations (r = −0.140; p = 0.018). A statistically
significant correlation between 25(OH)D and glucose was identified in the hypercholesterolemia
subgroup (r = −0.253; p = 0.002). A weak negative correlation (r = −0.215; p = 0.010) between 25(OH)D
and TC was found in the normocholesterolemic subgroup (Table 3). A significant negative correlation
of 25(OH)D with HDL-C was observed in children with hyperglycemia (r = −0.359; p = 0.010).
143
Nutrients 2018, 10, 1359
Vitamin
D/R-Pearson TC TG HDL-C Non-HDL-C LDL-C Glucose HbA1C hs-CRP
Correlation
Glucose: <100 mg/dL (<5.6 mmol/L) ((n = 234)—normoglycemia)
(R) 1 −0.136 0.001 −0.163 −0.066 −0.064 −0.122 0.131 −0.001
25(OH)D
(p) 2 0.037 0.990 0.012 0.314 0.330 0.063 0.046 0.989
Glucose: ≥100 mg/dL (≥5.6 mmol/L) ((n = 50)—hyperglycemia)
(R) 1 −0.251 −0.025 −0.359 −0.066 −0.135 −0.087 −0.008 −0.041
25(OH)D
(p) 2 0.079 0.862 0.010 0.647 0.351 0.547 0.956 0.779
Total cholesterol ≤170 mg/dL (≤4.4 mmol/L) ((n = 143)—normocholesterolemia)
(R) 1 −0.215 −0.019 −0.187 −0.066 −0.035 −0.019 0.050 −0.053
25(OH)D
(p) 2 0.010 0.820 0.026 0.436 0.677 0.821 0.554 0.531
Total cholesterol ≥170 mg/dL (≥ 4.4 mmol/L) ((n =141)—hypercholesterolemia)
(R) 1 −0.030 0.039 −0.181 0.091 0.030 −0.253 0.154 0.051
25(OH)D
(p) 2 0.724 0.644 0.032 0.282 0.722 0.002 0.070 0.546
All (n = 284)
(R) 1 −0.005 −0.160 −0.209 −0.067 −0.078 −0.140 0.091 0.001
25(OH)D
(p) 2 0.932 0.007 <0.001 0.262 0.193 0.018 0.129 0.991
1 R:Pearson correlation. 2 Statistical significance (p < 0.05), established using Pearson’s correlation analysis.
25(OH)D: 25-hydroxycholecalciferol (calcifediol); TC: total cholesterol; HDL-C: high-density lipoprotein cholesterol;
non-HDL-C: non-high-density lipoprotein cholesterol (TC-(HDL-C)); LDL-C: low-density lipoprotein cholesterol
(direct method); HbA1c : glycated hemoglobin; TG: triglycerides (logarithmic transformation); hs-CRP:
high-sensitivity C-reactive protein (logarithmic transformation).
The linear regression model (Table 4) emphasized a significant correlation between 25(OH)D and
glucose, and remained unchanged following adjustment for sex, age, and BMI percentiles.
Based on the first model presented, one can estimate that 25(OH)D reduced by 1 ng/mL
(2.5 nmol/L) was related to slight, although significant, elevation of blood glucose concentration by
144
Nutrients 2018, 10, 1359
0.238 mg/dL (0.013 mmol/L). After adjustment for sex, age, and BMI, glucose concentration elevation
was equal to 0.247 mg/dL (0.014 nmol/L) as a result of a unit decrease of 25 (OH)D concentration.
Our second model (Table 4) revealed a significant negative correlation between 25 (OH)D
(β = −0.159; p = 0.006) and TC. The model presented showed a significant elevation in TC by
0.958 mg/dL (0.025 mmol/L) linked to a 25(OH)D reduction by 1 ng/mL (2.5 nmol/L). In the adjusted
model, the TC elevation was identical.
The third linear regression model revealed an increase of HDL-C by 0.565 mg/dL (0.015 mmol/L),
as a result of the 1 ng/mL (2.5 nmol/L) decrease in 25(OH)D concentration (β = −0.203; p < 0.001).
Following adjustment, elevation of HDL-C reached 0.568 mg/dL (0.015 mmol/L) (Table 4).
4. Discussion
In the present study, the occurrence of IFG among children with vitamin D deficiency was
almost two-fold higher (OR = 1.966) when compared to children with 25(OH)D higher than
20 ng/mL (50 nmol/L). We also observed a weak negative correlation between 25(OH)D and glucose
concentration both in the whole group and in children with hypercholesterolemia.
We observed weak negative correlations between 25(OH)D and lipid components: TG and HDL-C
in the whole group, HDL-C in the subgroup with hyperglycemia or TC and HDL-C in the subgroups
with normo/hypercholesterolemia. Despite this weak correlation with lipid exponents, our study
revealed a significant association between serum 25(OH)D and TC concentration. We found that
serum 25(OH)D lower by 1 ng/mL (2.5 nmol/L) was linked to elevation in TC concentration by
0.96 mg /dL (0.025 mmol/L) and elevation of HDL-C by 0.57 mg/dL (0.015 mmol/L). The frequent
occurrence of hypercholesterolemia in our study (57.3%), being a notable trend particularly in
vitamin D-deficient children, is also worth noting. Evidence for the occurrence of dyslipidemia
in relation to vitamin D status was inconsistent. It was observed that the concentration of TC was
significantly higher in relation to vitamin D deficiency [13]. Our earlier study showed that HDL-C
concentration was higher among 25(OH)D-deficient children with newly diagnosed asthma [14].
Other cross-sectional studies conducted independently by Reis et al. and Ashraf et al. did not
find any significant association between low concentrations of vitamin D and abnormalities in lipid
parameters [15,16]. A study by Jorde et al. revealed a cross-sectional relationship between 25(OH)D
and serum lipids [17]. Meta-analysis conducted by Hao Wang et al. showed increased concentration
of TC and LDL-C with simultaneously decreased concentrations of HDL-C and TG as influenced by
vitamin D supplementation [18].
In this study we evaluated fasting glucose concentration, where abnormalities in this exponent of
carbohydrate metabolism is considered as a first stage in prediabetes development. This condition
may last several years without any additional symptoms, and depending on genetic or environmental
background, may either revert to a normoglycemic state or develop into overt diabetes [19].
The prevalence of IFG among overweight and obese Hispanic children reported by Goran et al.
ranged between 13% and 47% [20]. Our data indicated an inverse correlation between glucose and
vitamin D concentrations, consistent with an earlier NHANES III (National Health and Nutrition
Examination Survey) study where a negative relation between 25(OH)D and glycemia or diabetes
occurrence was demonstrated [21]. In another study conducted on children and teenagers aged 12–19,
it was seen that adjusted OR of fasting hyperglycemia among children with the lowest 25(OH)D
(<15 ng/mL; 37 nmol/L) was two-and-a-half fold higher (2.54, 95% CI: 1.01–6.40) when compared
to children in the highest 25(OH)D quartile (>26 ng/mL; 65 nmol/L) [15]. Our study showed that
along with the decrease of 1 ng/mL (2.5 nmol/L) of 25(OH)D, serum concentration of fasting glucose
increased by 0.25 mg/dL (0.014 mmol/L). The study conducted by Liu et al. indicated an association
between 25(OH)D and diabetes, in contrast to that presented by Pittas et al., where it was suggested
that there was insufficient data supporting hypotheses on the relation between vitamin D deficiency
and its contribution to the risk of diabetes [22,23].
145
Nutrients 2018, 10, 1359
The most significant limitation of this study was sample size, which comprised over
100 dyslipidemic and only 50 hyperglycemic subjects. A further limitation of this study was its
single-center and cross-sectional design which made it difficult to assess the influence of vitamin D
on carbohydrate and lipid metabolism, with only the relationship between variables being tested.
Therefore, our findings warrant confirmation in further studies. On the other hand, the strength of the
present study was the simultaneous evaluation of two closely related metabolic pathways of glucose
and lipids, in terms of the prevalence of vitamin D deficiency among children aged 9–11.
5. Conclusions
25(OH)D deficiency may negatively affect fasting glucose and total cholesterol concentration in
children aged 9–11. Vitamin D-deficient children are twice as likely to develop prediabetes represented
by IFG, when compared to those with 25(OH)D concentration above 20 ng/mL (50 nmol/L).
Author Contributions: Conceptualization, L.S. and G.S. Methodology, L.S., G.S. and M.K. Software, L.S. and K.B.
Validation, G.S. Visualization, L.S. Funding Acquisition, G.S. Data Curation, L.S., M.K. and K.B. Formal Analysis,
L.S. and K.B. Investigation, L.S. Project Administration, G.S. and T.D. Resources, L.S. and M.K. Supervision,
T.D. and G.S. Writing—Original Draft Preparation, L.S. Writing—Review & Editing, M.K. and G.S.
Funding: This research received no external funding.
Acknowledgments: We would like to thank all of the children and their parents for participating in our study.
Conflicts of Interest: The authors declare no conflict of interest.
Abbreviations
25(OH)D 25-Hydroxycholecalciferol
ADA American Diabetes Association
BMI Body Mass Index
CLSI Clinical and Laboratory Standards Institute
FG Fasting Glucose
HbA1c Glycated Hemoglobin
HDL-C High Density Lipoprotein Cholesterol by direct method
HS-CRP High Sensitivity C-Reactive Protein
IFG Impaired Fasting Glucose
LDL-C Low Density Lipoprotein Cholesterol by direct method
NHANES National Health and Nutrition Examination Survey
TC Total Cholesterol
TG Triglycerides
WHO World Health Organization
References
1. Peterson, C. Vitamin D deficiency and childhood obesity: Interactions, implications, and recommendations.
Nutr. Diet. Suppl. 2015, 7, 29–39. [CrossRef]
2. Rosen, C.J.; Adams, J.S.; Bikle, D.D.; Black, D.M.; Demay, M.B.; Manson, J.E.; Murad, M.H.; Kovacs, C.S.
The nonskeletal effects of vitamin D: An Endocrine Society scientific statement. Endocr. Rev. 2012, 33, 456–492.
[CrossRef] [PubMed]
3. Wimalawansa, S.J. Associations of vitamin D with insulin resistance, obesity, type 2 diabetes, and metabolic
syndrome. J. Steroid Biochem. Mol. Biol. 2018, 175, 177–189. [CrossRef] [PubMed]
4. Li, C.; Ford, E.S.; Zhao, G.; Mokdad, A.H. Prevalence of pre-diabetes and its association with clustering of
cardiometabolic risk factors and hyperinsulinemia among U.S. adolescents: National Health and Nutrition
Examination Survey 2005–2006. Diabetes Care 2009, 32, 342–347. [CrossRef] [PubMed]
5. Mutt, S.J.; Hyppönen, E.; Saarnio, J.; Järvelin, M.R.; Herzig, K.H. Vitamin D and adipose tissue-more than
storage. Front. Physiol. 2014, 5, 228. [CrossRef] [PubMed]
6. Autier, P.; Boniol, M.; Pizot, C.; Mullie, P. Vitamin D status and ill health: A systematic review. Lancet Diabetes
Endocrinol. 2014, 2, 76–89. [CrossRef]
146
Nutrients 2018, 10, 1359
7. Enko, D.; Kriegshäuser, G.; Stolba, R.; Worf, E.; Halwachs-Baumann, G. Method evaluation study of a new
generation of vitamin D assays. Biochem. Med. 2015, 25, 203–212. [CrossRef] [PubMed]
8. Manufacturers Information Product (IDS-iSYS 25-Hydroxy Vitamin Ds IS-2700S v04); Immunodiagnostic Systems:
The Boldons, UK, 2015.
9. Kułaga, Z.; Różdżyńska, A.; Palczewska, I.; Grajda, A.; Gurzkowska, B.; Napieralska, E.; Litwin, M. Percentile
charts of height, body mass and body mass index in children and adolescents in Poland—Results of the
OLAF study. Stand. Med. 2010, 7, 690–700.
10. Odrowa˛ż-Sypniewska, G. Laboratory diagnosis of prediabetes. Diagn. Lab. 2016, 52, 57–62.
11. Myśliwiec, M.; Walczak, M.; Małecka-Tendera, E.; Dobrzańska, A.; Cybulska, B.; Filipiak, K.J.; Mazur, A.;
Jarosz-Chobot, P.; Szadkowska, A.; Rynkiewicz, A.; et al. Management in familial hypercholesterolaemia in
children and adolescents. Position of the Lipid Expert Forum. Kardiol. Pol. 2013, 71, 1099–1105. [CrossRef]
[PubMed]
12. Sypniewska, G.; Krintus, M.; Fulgheri, G.; Siodmiak, J.; Kuligowska-Prusinska, M.; Stepien-Jaszowska, B.;
Staszak-Kowalska, R.; Zawadzka-Krajewska, A.; Kierat, S.; Bergmann, K.; et al. 25-Hydroxyvitamin, D,
biomarkers of eosinophilic inflammation and airway remodeling in children with newly diagnosed untreated
asthma. Allergy Asthma Proc. 2017, 38, 29–36. [CrossRef] [PubMed]
13. Kumar, J.; Muntner, P.; Kaskel, F.J.; Hailpern, S.M.; Melamed, M.L. Prevalence and Associations of
25-Hydroxyvitamin D Deficiency in US Children: NHANES 2001–2004. Pediatrics 2009, 124, 362–370.
[CrossRef] [PubMed]
14. Odrowa˛ż-Sypniewska, G.; Krintus, M.; Siódmiak, J.; Jaszowska, I.; Staszak-Kowalska, R.;
Zawadzka-Krajewska, A.; Kierat, S.Z.; Demkow, U. Serum 25(OH)D status and lipid profile in
children with newly diagnosed asthma. Med. Res. J. 2015, 3, 113–116. [CrossRef]
15. Reis, J.P.; von Mühlen, D.; Miller, E.R.; Michos, E.D.; Appel, L.J. Vitamin D Status and Cardiometabolic Risk
Factors in the United States Adolescent Population. Pediatrics 2009, 124, 371–379. [CrossRef] [PubMed]
16. Ashraf, A.; Alvarez, J.; Saenz, K.; Gower, B.; McCormick, K.; Franklin, F. Threshold for Effects of Vitamin
D Deficiency on Glucose Metabolism in Obese Female African-American Adolescents. J. Clin. Endocrinol.
Metab. 2009, 94, 3200–3206. [CrossRef] [PubMed]
17. Jorde, R.; Figenschau, Y.; Hutchinson, M.; Emaus, N.; Grimnes, G. High serum 25-hydroxyvitamin D
concentrations are associated with a favorable serum lipid profile. Eur. J. Clin. Nutr. 2010, 64, 1457–1464.
[CrossRef] [PubMed]
18. Wang, H.; Xia, N.; Yang, Y.; Peng, D.Q. Influence of vitamin D supplementation on plasma lipid profiles:
A meta-analysis of randomized controlled trials. Lipids Health Dis. 2010, 11, 42. [CrossRef] [PubMed]
19. Kleber, M.; deSousa, G.; Papcke, S.; Wabitsch, M.; Reinehr, T. Impaired glucose tolerance in obese white
children and adolescents: Three to five year follow-up in untreated patients. Exp. Clin. Endocrinol. Diabetes
2011, 119, 172–176. [CrossRef] [PubMed]
20. Michael, I.G.; Lane, C.; Toledo-Corral, C.; Weigensberg, M.J. Persistence of prediabetes in overweight
and obese Hispanic children: Association with progressive insulin resistance, poor beta-cell function,
and increasing visceral fat. Diabetes 2008, 57, 3007–3012. [CrossRef]
21. Misiorowski, W. Vitamin D in type 1 and 2 diabetes in adulthood. Stand. Med. Pediatr. 2015, 9, 639–644.
22. Liu, E.; Meigs, J.B.; Pittas, A.G.; Economos, C.D.; McKeown, N.M.; Booth, S.L.; Jacques, P.F. Predicted
25-hydroxyvitamin D score and incident type 2 diabetes in the Framingham Offspring Study. Am. J.
Clin. Nutr. 2010, 91, 1627–1633. [CrossRef] [PubMed]
23. Pittas, A.G.; Chung, M.; Trikalinos, T.; Mitri, J.; Brendel, M.; Patel, K.; Lichtenstein, A.H.; Lau, J.; Balk, E.M.
Systematic review: Vitamin D and cardiometabolic outcomes. Ann. Intern. Med. 2010, 152, 307–314.
[CrossRef] [PubMed]
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
147
nutrients
Review
What is the Validity of Questionnaires Assessing
Fruit and Vegetable Consumption in Children When
Compared with Blood Biomarkers? A Meta-Analysis
Tatiana S. Collese *, Gabriela Vatavuk-Serrati, Marcus Vinicius Nascimento-Ferreira,
Augusto César Ferreira De Moraes and Heráclito Barbosa Carvalho
YCARE (Youth/Child cArdiovascular Risk and Environmental) Research Group, Department of Preventive
Medicine, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP 01246-903, Brazil;
[email protected] (G.V.-S.); [email protected] (M.V.N.-F.);
[email protected] (A.C.F.D.M.); [email protected] (H.B.C.)
* Correspondence:[email protected]; Tel.: +55-11-3061-7091; Fax: +55-11-3061-8466
Abstract: Fruit and vegetable consumption has been associated with improved health outcomes in
children. As an extensive number of questionnaires are currently used to assess fruit and vegetable
consumption, we performed a systematic review of the criterion validity of questionnaires used to
estimate fruit and vegetable consumption in children, considering blood biomarkers as the reference
method. Five electronic databases (MEDLINE, CINAHL, Scopus, PsycINFO, Web of Science) were
searched from database inception to 23 July 2018. The search strategy used the following sets of
descriptors: children; fruits and vegetables; dietary questionnaires; blood biomarkers; and validation
coefficient. The search terms were adapted for use with other databases in combination with
database-specific filters. Potentially eligible articles were selected independently by two reviewers,
separately, following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses
(PRISMA) guidelines. Two articles meeting the inclusion criteria were included. The main reason
for study exclusion was the sample age range, which included adolescents. The pooled correlation
coefficient was 0.32 (95% confidence interval: 0.24–0.40).This review provided insights into assessment
methods of fruit and vegetable consumption in children. Although further studies are required,
questionnaires for assessing fruit and vegetable consumption have fair criterion validity in children.
1. Introduction
International health associations recommend that children have a diet rich in vegetables, fruits,
whole grains, low-fat dairy products, legumes, fish, and lean meat, and low in saturated fat, trans fat,
and cholesterol, to help maintain a healthy weight and promote cardiovascular health [1]. Given the
role that the diet plays in children’s health, it is crucial to accurately determine their dietary intake and
to estimate the relationship between the foods consumed and nutrients.
Regular fruit and vegetable consumption is routinely suggested as a key component of promoting
health because they are an important source of nutrients, such as water, fiber, potassium, folic
acid, vitamins, and phytochemicals [1,2]. Several systematic reviews and meta-analyses have found
consistent associations between fruit and vegetable consumption and lower risk of cardiovascular
disease, diabetes, cancer, and other chronic diseases [3–5]. Despite the well-known benefits of
consuming fruits and vegetables, children are not meeting the recommended level of five servings
per day [6]. Consequently, increasing fruit and vegetable consumption in children is one of the major
goals of dietary interventions, worldwide [6]. Thus, valid questionnaires to assess fruit and vegetable
consumption are essential to identify eating habit changes in response to interventions, and to analyze
the impact of fruit consumption on health [7].
In nutritional epidemiology, the most used methods to estimate fruit and vegetable intake are
subjective instruments, such as food frequency questionnaires (FFQs) and dietary recalls, because they
are simple, cost-effective methods to assess long-term eating habits for large samples [8,9]. However,
these subjective instruments are susceptible to both random and systematic errors [10,11], especially
when applied to children, due to their limited skills to report their own dietary intake, depending
on parental (or whoever is responsible for their food) literacy and motivation to complete these
questionnaires [12].
As all dietary assessment methods (e.g., 24 h recalls, FFQs, smart-phone apps) have their
own strengths and limitations, dietary questionnaires are usually validated against an objective
measurement, such as blood biomarkers or doubly-labeled water [8,13]. Previous studies proposed
that plasma vitamins, such as vitamin A, E, and C, are a reliable concentration biomarker of usual fruit
and vegetable intake. Hence they are commonly used in validity studies of dietary assessment methods
as independent proxy measures of fruit and vegetable consumption [14], and to evaluate whether
sources of random error are independent of errors associated with measurement by questionnaire
and/or inaccuracies within nutrient databases [15].
However, these plasma vitamins are still relatively recent, and a significant amount of studies
are warranted in nutritional epidemiology [16]. It concerns deciphering whether these biomarkers are
markers of dietary consumption or markers of altered metabolism, as a result of the food intake [16],
or regarding the individual variability in absorption or availability [17]. Validation studies with blood
biomarkers have been carried out in adults [10,11], but relatively few studies were conducted in
children, lacking literature on key issues such as the impact of the period between fruit and vegetable
consumption and the blood biomarker analysis, or the stability of blood biomarkers in children aged
3 years to less than 10 years.
Therefore, the research question of this review was “What is the criteria validity of questionnaires
used to estimate fruit and vegetable consumption in children when compared with blood biomarkers?”
2. Methods
This systematic review was designed and conducted according to the guidelines of the
Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) checklist
(Supplementary Material 1), as described in Reference [18]. At study initiation, no similar systematic
reviews were available in PROSPERO (International Prospective Register of Systematic Reviews) or
the following databases: Biomed Central, Medical Literature Analysis and Retrieval System Online
(MEDLINE), Web of Science, Cumulative Index to Nursing and Allied Health Literature (CINAHL),
Scopus, and PsycINFO. Thus, this protocol was registered in PROSPERO (CRD42017072531).
149
Nutrients 2018, 10, 1396
Two investigators examined the articles and performed the data screening, the data extraction,
and the quality assessment independently. Inter-reviewer discrepancies were resolved by consensus,
and a third reviewer was consulted for unresolved discrepancies. Relevant articles were obtained in
full and were assessed for eligibility and exclusion criteria.
Eligibility Criteria
Children (3 years to less than 10 years of age) in a free-living environment, without any
Population
specific condition (e.g., eating disorder, obesity, diabetes) or children under medical care.
Exposure Fruit and/or vegetable consumption estimated by questionnaire.
Comparator Blood biomarkers for fruit and/or vegetable consumption.
Outcome Validation coefficients of fruit and/or vegetable consumption.
Study Original articles.
Exclusion Criteria
Studies where the participants had different diseases (or disturbances) that could interfere with
fruit and vegetable consumption were excluded. These criteria were set to increase inter-study
comparability. In addition, review articles, conference papers, or books were excluded.
Articles were saved in the EndNote Web reference manager software version 3.1 (Thomson
Research Software, Carlsbad, CA, USA), and duplicates were removed. The data extracted included
the author, title of the study, citation and contact details, and study eligibility (Table 1). When studies
did not meet the eligibility criteria, the main reason for exclusion was documented in ink. Thereafter,
a table (Supplementary Material 2) was created by each reviewer with additional details about the
study, such as the sample size, settings, exposure/outcome definition and unit of measurement,
statistical analysis, bias (funding sources, adjustments), key conclusions, authors, miscellaneous
comments from the study authors, references to other relevant studies, limitations, correspondence
required, and miscellaneous comments by the authors reviewing the included studies.
150
Nutrients 2018, 10, 1396
2.6. Statistics
We used the Stata 14 (Stata Corp., College Station, TX, USA) program for all statistical analyses.
Our outcomes were the correlation coefficients between the questionnaire and blood biomarkers,
which we calculated with their corresponding 95% confidence intervals (considering p < 0.05 as
significance level). In addition, we calculated the sample-weighted pooled correlation coefficient
for the meta-analysis with the random-effects model, due to the moderate heterogeneity found in
each study. We estimated the strength of the agreement of the Spearman rank correlation coefficient
cutoff points using the classification for dietary assessment methods: weak < 0.20, fair = 0.21 to 0.40,
moderate = 0.41 to 0.60, good = 0.61 to 0.80, and >0.80, very good [22]. Additionally, we verified
the heterogeneity of the studies using the I-squared test (percentages and p values under 0.05 were
considered significant). Heterogeneity levels of 25%, 50%, and 75% were considered low, moderate, and
high, respectively [23]. We performed the Egger’s regression test to verify potential publication bias
(positive or negative results) and small-study effects. We also generated the funnel plot (Supplementary
Material 3) to examine the potential bias graphically.
3. Results
The literature search provided 680 potentially eligible records: 4 from PsycINFO, 42 from Embase,
71 from Web of Science, 430 from Scopus, and 133 from Medline (see Appendix 1 in the Supporting
Information online). After duplicates (n = 51) were excluded, 629 studies remained and were screened
by title and, posteriorly, by abstract, according to the established inclusion criteria, resulting in
19 records for which the full-text was assessed for eligibility. Of those, the main reasons for exclusion
were the population (studies included participants over 10 years of age) and biomarker (such as
skin carotenoids), as shown in Figure 1. More details about these 19 study articles are stated
in Supplementary Material 4. Consequently, two studies were eligible and were included in this
systematic review.
151
Nutrients 2018, 10, 1396
Identification
Records identified
(n = 680)
duplicates
excluded (n = 51)
records screened by
title (n = 629)
Preliminary Selection
records excluded
(n = 561)
records screened by
abstract (n = 68)
records excluded
(n = 49)
2 articles included in
the systematic review
Figure 1. Flow chart of article identification, retrieval, and inclusion, for the systematic review.
152
Nutrients 2018, 10, 1396
The two final eligible articles included in this systematic review were published in English,
but they are from different countries, one from Europe and the other from North America (Table 2).
We found one study published in 1993 [24], but Medin et al. [25] was more recent, published in 2016.
Table 3 shows that both studies asked for the parental report; however, Byers et al. [24] used the
FFQ regarding the previous three months and considered fruit juice together with fruit and vegetable
consumption. Medin et al. [25] used a web-based food recall, for four consecutive nights, but they
assessed only vegetable intake for 4th grade children (8–9 years of age).
Nine different types of biomarkers were used to validate fruit and vegetable consumption.
Both studies analyzed carotenoids (α-carotene, β-carotene, and cryptoxanthin), but Medin et al. [25]
also included lycopene, lutein, and zeaxanthin in the sum of total carotenoids. On the other hand,
Byers et al. [24] analyzed vitamin A (retinol), vitamin E (α-tocopherol), and vitamin C (ascorbic acid).
High-performance liquid chromatography (HPLC), which is considered the gold standard analytical
technique for characterization and analysis of carotenoids in biological and food samples [26], was used
to assess plasma carotenoids in all studies (and for vitamin E in Byers et al. [24]), and spectrophotometry
was used to assess plasma vitamin C (Table 3).
The correlation coefficients between fruit and vegetable consumption and blood biomarkers are
presented in Table 4. The validity coefficients were fair to moderate, varying from 0.24 (for vitamin E)
to 0.47 (carotenoids). Both studies calculated the Spearman rank correlation coefficients and adjusted
the analyses for age, sex, ethnicity, and body mass index.
153
Table 3. Validity studies on questionnaires to assess fruit and vegetable consumption with blood biomarkers in children.
Exposure
Period of Supplements Nutritional Biochemical Fasting
Reference Assessment Report Consumption Blood Biomarkers
Report Assessed Database Method Time
Method
Median fruits,
α-Carotene
Nutrients 2018, 10, 1396
154
Table 4. Outcome of the included studies.
The heterogeneity between studies was moderate (I-squared = 62.9%, p = 0.029), and
the sample-weighted pooled correlation coefficient for biomarkers was fair (r = 0.32, 95% CI:
0.24–0.40), as shown in the Figure 2. We were not able to stratify the analysis into subgroup categories
due to the small number of eligible studies included in the systematic review.
Figure 2. Forest-plot for correlation coefficients of fruit and vegetable consumption assessed by
questionnaires and blood biomarkers.
4. Discussion
At present, there is no strong evidence regarding the validity of questionnaires for assessing fruit
and vegetable consumption in children with blood biomarkers. Thus, we conducted a comprehensive
systematic review of validity studies of questionnaires to assess fruit and vegetable consumption in
children in comparison with blood biomarkers. Our findings indicated that this criteria validity was
fair. This validity stems from the challenges inherent to dietary assessment through questionnaires,
in addition to the fact that in children, we should consider the parent report, which is the subjective
perception of the parents about their children’s feeding habits [13,27].
The limited number studies (n = 2) meeting the inclusion criteria was likely due to the lack of
validated dietary questionnaires, maybe because conducting blood collection in school children is
less feasible, for practical and financial reasons [28], and due to the sample age range, since different
age group definitions were used for children (e.g., together with infants or adolescents). For example,
the study of Biltoft-Jensen et al. [29], which fulfilled our eligibility criteria, except for the population
age range, which was between 8 and 11 years of age. It is correct that 11-year-old individuals are still
children; however, the timing of biological maturation clearly signals entry into adolescence [21], and
with more cognitive abilities that enable the researchers to apply the self-report of fruit and vegetable
consumption. Therefore, it was excluded from our review.
It is important to emphasize that the credibility of our findings does not rely on the number of
studies included in the meta-analysis (n = 2), as we conducted an exhaustive and reproducible literature
search to produce results with the highest level of quality of evidence available. Murad et al. [30] state
that a systematic review or meta-analysis should evaluate the credibility of the methods applied on the
systematic review, and that this credibility depends on whether the review addressed a specific and
objective question; included an exhaustive literature search; clearly indicated the reproducibility of the
155
Nutrients 2018, 10, 1396
selection and assessment of studies; and declared the results in a convenient way that enables future
research development. Furthermore, even though only two studies were eligible in our systematic
review, each biomarker was considered separately in the analysis; in other words, we evaluated five
different items (n = 5).
The sample-weighted pooled correlation coefficient found in our study (r = 0.32, 95% CI:
0.24–0.40) was fair, and comparisons with other validity studies carried out in children and adolescents
(9–12 years) showed similar Pearson correlation coefficients for vegetable (0.26, p = 0.13) and fruit
intake (0.49; p = 0.003) with total plasma carotenoids [31]. Studies carried out in adults showed a slight
increase in the Spearman correlation coefficients for alpha-carotene (0.42; CI: 0.09–0.96), beta-carotene
(0.40; CI: 0.09–0.99), and total plasma carotenoids (0.51; CI: 0.12–1.00) [32]. However, these correlations
are not so distant from our results, and it is relevant to note that their confidence intervals were broad.
156
Nutrients 2018, 10, 1396
pop-up reminders to reduce the problem of food omissions, and in a more friendly way for kids—this
was shown to be a promising tool for application in pediatric populations.
In addition, Medin et al. [25] included four nightly 24-h recalls. This period is more than the
literature recommendation (three recalls per person) [34], and since parents answered the questionnaire
at night (the end of the day), it was easier for them to remember their child’s fruit and vegetable
consumption on the same day, minimizing the memory bias as well [34].
However, web-based food recalls reduce the limitations in terms of misreporting. Thus, there is a
need for continued development of methods, as well as to continue to evolve statistical methods to
mitigate error. Training researchers, clinical staff, and research subjects on the use of the technology
and the method is still highly warranted and might improve results and compliance with the dietary
method [19].
To be confident about the fruit and vegetable data collected, one should combine the FFQ with food
recall methods, trying to ascertain both frequent and infrequent fruit and vegetable consumption [35].
Additionally, if possible, combining technology with both dietary assessment methods is a good
alternative to reduce the burden of data handling and to make this process more friendly and joyful.
4.4. Creativity
The integration of technology, pictures, drawings, cartoons, and puppets into traditional dietary
questionnaires, considering the main factors inherent (Figure 3) to this approach, might provide
additional insights to mitigate errors in the fruit and vegetable assessment.
Figure 3. Main factors regarding the validity of dietary assessment methods in children.
When financial resources are scarce, like in studies conducted in low and middle-income countries,
using a bit of creativity might be a good solution. For example, if it is not possible to combine the
157
Nutrients 2018, 10, 1396
technology in the process of dietary assessment, a simple art-based method like a “mascot” (drawn on
paper, as a cartoon, as a toy or a hand puppet) can make a great difference in the motivation and
involvement to answer the questionnaire, turning this process into a fun moment for kids [36,37].
Additionally, pictorial assessment, such as the assessment with the help of food photography albums,
food drawings or food replicas (three-dimensional models), might also be a cheaper and smart
alternative to enhance motivation and give visual support to estimate fruit and vegetable portion
sizes [38,39].
158
Nutrients 2018, 10, 1396
4.9. Bias
The findings of our review indicated that more validity studies of fruit and vegetable consumption
need to be carried out in school-aged children. The two eligible studies were published in English and in
high-income countries. Therefore, it indicates a potential risk of publication bias, which can practically
be explained by the expensive cost of conducting a validity study with blood biomarkers [28].
Moreover, most of the 19 remaining full-text articles assessed for eligibility used the plasma
carotenes as a reference method, but only two articles assessed vitamin E [24,47], and one article
assessed vitamin C [24]. This bias is also probably related to the costs of vitamin C, to its very sensitive
biochemical methodology, and to the many difficulties and bureaucratic barriers for achieving the
reactants necessary for this methodology.
As dietary assessment cannot be estimated without error, it is paramount to attempt to understand
their effect on the data collected to minimize the confounding effect [17].
Byers et al. studied a sample of parents from a selective group of volunteers who reported their
children’s food intake once a year. Thus, the validity of their reports was probably higher than other
parents, because their motivation, involvement, and knowledge in the next step of the study could be
different to other parents who have never been involved in a nutritional research [24].
Another relevant issue of concern is the social approval of fruits and vegetables. As they are
widely known as healthy foods [48], parents may have reported their children’s fruit and vegetable
consumption, either consciously or subconsciously, in ways that make them appear favorable to the
researchers [17].
159
Nutrients 2018, 10, 1396
heterogeneity could be partially explained by the characteristics inherent to the different types of
questionnaires used (e.g., recall period, number of items, recording bias, web-based system) and the
biomarker (e.g., type of biomarker analyzed, duration of measure, different cutoff points).
5. Conclusions
Our findings suggest that the criterion validity of questionnaires and blood biomarkers
for assessing fruit and vegetable consumption in children is fair. The assessment of food
consumption—especially fruit and vegetable—in children still faces many challenges. However,
having a thorough understanding of the main factors that may influence this assessment can be a
promising path to enhance the validity of dietary assessment methods. Since measurement error is a
key remaining difficulty, validity studies of questionnaires to assess fruit and vegetable consumption
should be conducted with caution and interpreted within the broad context of the field. For example,
careful study design and meticulous development of the dietary assessment tools can be validated
with blood biomarkers, as long as the whole process of blood collection and analysis is adequate
and rigorously controlled. Moreover, informed statistical analysis, with appropriate adjustments, can
reduce the impact of these errors.
Finally, the incorporation of creativity and the development of technology, despite the challenges,
can lead to advancements in the field of dietary assessment methods, and therefore, have an important
role in the promising future of nutritional epidemiology.
160
Nutrients 2018, 10, 1396
Funding: This research was funded by the State of São Paulo Research Foundation (FAPESP), which granted
Tatiana Sadalla Collese a doctoral scholarship (proc. 2016/13922-1), Gabriela Vatavuk-Serrati a Scientific Initiation
scholarship (proc. 2018/02452-0), Marcus Vinícius Nascimento-Ferreira a doctoral scholarship (proc. 2016/18436-8
and 2017/11732-3). Augusto César Ferreira De Moraes was awarded a post-doctoral scholarship from FAPESP
(proc. 2014/13367-2). Heráclito Barbosa Carvalho received an advanced scientist scholarship from the National
Council of Technological and Scientific Development (CNPq; proc. 300951/2015-9).
Acknowledgments: We gratefully acknowledge all of the researchers from the YCARE Research Group for their
essential contributions and suggestions for improving this work.
Conflicts of Interest: The authors declare no conflict of interest.
References
1. Van Horn, L.; Carson, J.A.; Appel, L.J.; Burke, L.E.; Economos, C.; Karmally, W.; Lancaster, K.;
Lichtenstein, A.H.; Johnson, R.K.; Thomas, R.J.; et al. Recommended dietary pattern to achieve adherence
to the American heart association/American college of cardiology (AHA/ACC) guidelines: A scientific
statement from the American heart association. Circulation 2016, 134, 505–529. [CrossRef] [PubMed]
2. Pem, D.; Jeewon, R. Fruit and vegetable intake: Benefits and progress of nutrition education interventions-
Narrative review article. Iran. J. Public Health 2015, 44, 1309–1321. [PubMed]
3. Wang, X.; Ouyang, Y.; Liu, J.; Zhu, M.; Zhao, G.; Bao, W.; Hu, F.B. Fruit and vegetable consumption
and mortality from all causes, cardiovascular disease, and cancer: Systematic review and dose-response
meta-analysis of prospective cohort studies. BMJ 2014, 349, g4490. [CrossRef] [PubMed]
4. Zhan, J.; Liu, Y.J.; Cai, L.B.; Xu, F.R.; Xie, T.; He, Q.Q. Fruit and vegetable consumption and risk of
cardiovascular disease: A meta-analysis of prospective cohort studies. Crit. Rev. Food Sci. Nutr. 2017,
57, 1650–1663. [CrossRef] [PubMed]
5. Wu, Y.; Zhang, D.; Jiang, X.; Jiang, W. Fruit and vegetable consumption and risk of type 2 diabetes mellitus:
A dose-response meta-analysis of prospective cohort studies. Nutr. Metab. Cardiovasc. Dis. 2015, 25, 140–147.
[CrossRef] [PubMed]
6. Myers, G.; Wright, S.; Blane, S.; Pratt, I.S.; Pettigrew, S. A process and outcome evaluation of an in-class
vegetable promotion program. Appetite 2018, 125, 182–189. [CrossRef] [PubMed]
7. Biltoft-Jensen, A.; Trolle, E.; Christensen, T.; Islam, N.; Andersen, L.F.; Egenfeldt-Nielsen, S.; Tetens, I.
Webdasc: A web-based dietary assessment software for 8-11-year-old Danish children. J. Hum. Nutr. Diet.
2014, 27, 43–53. [CrossRef] [PubMed]
8. Fatihah, F.; Ng, B.K.; Hazwanie, H.; Norimah, A.K.; Shanita, S.N.; Ruzita, A.T.; Poh, B.K. Development and
validation of a food frequency questionnaire for dietary intake assessment among multi-ethnic primary
school-aged children. Singap. Med. J. 2015, 56, 687–694. [CrossRef] [PubMed]
9. Molag, M.L.; de Vries, J.H.; Ocke, M.C.; Dagnelie, P.C.; van den Brandt, P.A.; Jansen, M.C.; van Staveren, W.A.;
van’t Veer, P. Design characteristics of food frequency questionnaires in relation to their validity.
Am. J. Epidemiol. 2007, 166, 1468–1478. [CrossRef] [PubMed]
10. Corella, D.; Ordovas, J.M. Biomarkers: Background, classification and guidelines for applications in
nutritional epidemiology. Nutr. Hosp. 2015, 31, 177–188. [PubMed]
11. Livingstone, M.B.; Robson, P.J.; Wallace, J.M. Issues in dietary intake assessment of children and adolescents.
Br. J. Nutr. 2004, 92, 213–222. [CrossRef]
12. Lu, A.S.; Baranowski, J.; Islam, N.; Baranowski, T. How to engage children in self-administered dietary
assessment programmes. J. Hum. Nutr. Diet. 2014, 27, 5–9. [CrossRef] [PubMed]
13. Burrows, T.L.; Martin, R.J.; Collins, C.E. A systematic review of the validity of dietary assessment methods in
children when compared with the method of doubly labeled water. J. Am. Diet. Assoc. 2010, 110, 1501–1510.
[CrossRef] [PubMed]
14. Burrows, T.L.; Hutchesson, M.J.; Rollo, M.E.; Boggess, M.M.; Guest, M.; Collins, C.E. Fruit and vegetable
intake assessed by food frequency questionnaire and plasma carotenoids: A validation study in adults.
Nutrients 2015, 7, 3240–3251. [CrossRef] [PubMed]
15. Dodd, K.W.; Midthune, D.; Kipnis, V. Re: “Application of a repeat-measure biomarker measurement
error model to 2 validation studies: Examination of the effect of within-person variation in biomarker
measurements”. Am. J. Epidemiol. 2012, 175, 84–85. [CrossRef] [PubMed]
161
Nutrients 2018, 10, 1396
16. Brennan, L. Moving toward objective biomarkers of dietary intake. J. Nutr. 2018, 148, 821–822. [CrossRef]
[PubMed]
17. Collins, C.E.; Watson, J.; Burrows, T. Measuring dietary intake in children and adolescents in the context of
overweight and obesity. Int. J. Obes. 2010, 34, 1103–1115. [CrossRef] [PubMed]
18. Shamseer, L.; Moher, D.; Clarke, M.; Ghersi, D.; Liberati, A.; Petticrew, M.; Shekelle, P.; Stewart, L.A.
Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015: Elaboration
and explanation. BMJ 2015, 350, g7647. [CrossRef] [PubMed]
19. Burrows, T.L.; Rollo, M.E.; Williams, R.; Wood, L.G.; Garg, M.L.; Jensen, M.; Collins, C.E. A systematic
review of technology-based dietary intake assessment validation studies that include carotenoid biomarkers.
Nutrients 2017, 9, 140. [CrossRef] [PubMed]
20. World Health Organization. Health Problems of Adolescents; Technical Report Series 308; World Health
Organization: Geneva, Switzerland, 1965.
21. Sawyer, S.M.; Azzopardi, P.S.; Wickremarathne, D.; Patton, G.C. The age of adolescence. Lancet Child
Adolesc. Health 2018, 2, 223–228. [CrossRef]
22. Masson, L.F.; McNeill, G.; Tomany, J.O.; Simpson, J.A.; Peace, H.S.; Wei, L.; Grubb, D.A.; Bolton-Smith, C.
Statistical approaches for assessing the relative validity of a food-frequency questionnaire: Use of correlation
coefficients and the kappa statistic. Public Health Nutr. 2003, 6, 313–321. [CrossRef] [PubMed]
23. Higgins, J.P.; Thompson, S.G.; Deeks, J.J.; Altman, D.G. Measuring inconsistency in meta-analyses. BMJ 2003,
327, 557–560. [CrossRef] [PubMed]
24. Byers, T.; Trieber, F.; Gunter, E.; Coates, R.; Sowell, A.; Leonard, S.; Mokdad, A.; Jewell, S.; Miller, D.;
Serdula, M.; et al. The accuracy of parental reports of their children’s intake of fruits and vegetables:
Validation of a food frequency questionnaire with serum levels of carotenoids and vitamins C, A, and E.
Epidemiology 1993, 4, 350–355. [CrossRef] [PubMed]
25. Medin, A.C.; Carlsen, M.H.; Andersen, L.F. Associations between reported intakes of carotenoid-rich foods
and concentrations of carotenoids in plasma: A validation study of a web-based food recall for children and
adolescents. Public Health Nutr. 2016, 19, 3265–3275. [CrossRef] [PubMed]
26. Gupta, P.; Sreelakshmi, Y.; Sharma, R. A rapid and sensitive method for determination of carotenoids in
plant tissues by high performance liquid chromatography. Plant Methods 2015, 11, 5. [CrossRef] [PubMed]
27. Rangan, A.; Allman-Farinelli, M.; Donohoe, E.; Gill, T. Misreporting of energy intake in the 2007 Australian
children’s survey: Differences in the reporting of food types between plausible, under- and over-reporters of
energy intake. J. Hum. Nutr. Diet. 2014, 27, 450–458. [CrossRef] [PubMed]
28. Jenab, M.; Slimani, N.; Bictash, M.; Ferrari, P.; Bingham, S.A. Biomarkers in nutritional epidemiology:
Applications, needs and new horizons. Hum. Genet. 2009, 125, 507–525. [CrossRef] [PubMed]
29. Biltoft-Jensen, A.; Bysted, A.; Trolle, E.; Christensen, T.; Knuthsen, P.; Damsgaard, C.T.; Andersen, L.F.;
Brockhoff, P.; Tetens, I. Evaluation of web-based dietary assessment software for children: Comparing
reported fruit, juice and vegetable intakes with plasma carotenoid concentration and school lunch
observations. Br. J. Nutr. 2013, 110, 186–195. [CrossRef] [PubMed]
30. Murad, M.H.; Montori, V.M.; Ioannidis, J.P.; Jaeschke, R.; Devereaux, P.J.; Prasad, K.; Neumann, I.;
Carrasco-Labra, A.; Agoritsas, T.; Hatala, R.; et al. How to read a systematic review and meta-analysis and
apply the results to patient care: Users’ guides to the medical literature. JAMA 2014, 312, 171–179. [CrossRef]
[PubMed]
31. Nguyen, L.M.; Scherr, R.E.; Ermakov, I.V.; Gellermann, W.; Jahns, L.; Keen, C.L.; Miyamoto, S.; Steinberg, F.M.;
Young, H.M.; Zidenberg-Cherr, S. Evaluating the relationship between plasma and skin carotenoids and reported
dietary intake in elementary school children to assess fruit and vegetable intake. Arch. Biochem. Biophys. 2015, 572,
73–80. [CrossRef] [PubMed]
32. McNaughton, S.A.; Marks, G.C.; Gaffney, P.; Williams, G.; Green, A. Validation of a food-frequency
questionnaire assessment of carotenoid and vitamin e intake using weighed food records and plasma
biomarkers: The method of triads model. Eur. J. Clin. Nutr. 2005, 59, 211–218. [CrossRef] [PubMed]
162
Nutrients 2018, 10, 1396
33. Vandevijvere, S.; Geelen, A.; Gonzalez-Gross, M.; van’t Veer, P.; Dallongeville, J.; Mouratidou, T.; Dekkers, A.;
Bornhorst, C.; Breidenassel, C.; Crispim, S.P.; et al. Evaluation of food and nutrient intake assessment
using concentration biomarkers in European adolescents from the healthy lifestyle in Europe by nutrition in
adolescence study. Br. J. Nutr. 2013, 109, 736–747. [CrossRef] [PubMed]
34. Emmett, P. Assessing diet in longitudinal birth cohort studies. Paediatr. Perinat. Epidemiol. 2009, 23, 154–173.
[CrossRef] [PubMed]
35. Crispim, S.P.; Geelen, A.; Souverein, O.W.; Hulshof, P.J.; Ruprich, J.; Dofkova, M.; Huybrechts, I.; De
Keyzer, W.; Lillegaard, I.T.; Andersen, L.F.; et al. Biomarker-based evaluation of two 24-h recalls for
comparing usual fish, fruit and vegetable intakes across European centers in the Efcoval study. Eur. J.
Clin. Nutr. 2011, 65, 38–47. [CrossRef] [PubMed]
36. Androutsos, O.; Apostolidou, E.; Iotova, V.; Socha, P.; Birnbaum, J.; Moreno, L.; De Bourdeaudhuij, I.;
Koletzko, B.; Manios, Y. Process evaluation design and tools used in a kindergarten-based, family-involved
intervention to prevent obesity in early childhood. The toybox-study. Obes. Rev. 2014, 15, 74–80. [CrossRef]
[PubMed]
37. Birkin, S. Monkey mascot used to gather children’s opinions. Nurs. Child Young People 2017, 29, 7.
38. Townsend, M.S.; Shifts, M.K.; Styne, D.M.; Drake, C.; Lanoue, L.; Woodhouse, L.; Allen, L.H. Vegetable
behavioral tool demonstrates validity with my plate vegetable cups and carotenoid and inflammatory
biomarkers. Appetite 2016, 107, 628–638. [CrossRef] [PubMed]
39. Bernal-Orozco, M.F.; Vizmanos-Lamotte, B.; Rodriguez-Rocha, N.P.; Macedo-Ojeda, G.; Orozco-Valerio, M.;
Roville-Sausse, F.; Leon-Estrada, S.; Marquez-Sandoval, F.; Fernandez-Ballart, J.D. Validation of a Mexican
food photograph album as a tool to visually estimate food amounts in adolescents. Br. J. Nutr. 2013, 109,
944–952. [CrossRef] [PubMed]
40. McGloin, A.F.; Livingstone, M.B.; Greene, L.C.; Webb, S.E.; Gibson, J.M.; Jebb, S.A.; Cole, T.J.; Coward, W.A.;
Wright, A.; Prentice, A.M. Energy and fat intake in obese and lean children at varying risk of obesity. Int. J.
Obes. Relat. Metab. Disord. 2002, 26, 200–207. [CrossRef] [PubMed]
41. Saini, R.K.; Nile, S.H.; Park, S.W. Carotenoids from fruits and vegetables: Chemistry, analysis, occurrence,
bioavailability and biological activities. Food Res. Int. 2015, 76, 735–750. [CrossRef] [PubMed]
42. Paiva, S.A.; Russell, R.M. Beta-carotene and other carotenoids as antioxidants. J. Am. Coll. Nutr. 1999, 18,
426–433. [CrossRef] [PubMed]
43. Burri, B.J.; Neidlinger, T.R.; Clifford, A.J. Serum carotenoid depletion follows first-order kinetics in healthy
adult women fed naturally low carotenoid diets. J. Nutr. 2001, 131, 2096–2100. [CrossRef] [PubMed]
44. Collins, C.E.; Boggess, M.M.; Watson, J.F.; Guest, M.; Duncanson, K.; Pezdirc, K.; Rollo, M.; Hutchesson, M.J.;
Burrows, T.L. Reproducibility and comparative validity of a food frequency questionnaire for Australian
adults. Clin. Nutr. 2014, 33, 906–914. [CrossRef] [PubMed]
45. Lee, P.H.; Burstyn, I. Identification of confounder in epidemiologic data contaminated by measurement error
in covariates. BMC Med. Res. Methodol. 2016, 16, 54. [CrossRef] [PubMed]
46. Nascimento-Ferreira, M.V.; De Moraes, A.C.F.; Toazza Oliveira, P.V.; Rendo-Urteaga, T.; Gracia-Marco, L.;
Forjaz, C.L.M.; Moreno, L.A.; Carvalho, H.B. Assessment of physical activity intensity and duration in the
paediatric population: Evidence to support an a priori hypothesis and sample size in the agreement between
subjective and objective methods. Obes. Rev. 2018, 19, 810–824. [CrossRef] [PubMed]
47. Royo-Bordonada, M.A.; Gorgojo, L.; Ortega, H.; Martin-Moreno, J.M.; Lasuncion, M.A.; Garces, C.; Gil, A.;
Rodriguez-Artalejo, F.; de Oya, M. Investigators of the Four Provinces Study. Greater dietary variety is
associated with better biochemical nutritional status in Spanish children: The four provinces study. Nutr.
Metab. Cardiovasc. Dis. 2003, 13, 357–364. [CrossRef]
48. Lichtenstein, A.H. Fruits and vegetables get a golden halo once again: Is there more to the story? Circulation
2015, 132, 1946–1948. [CrossRef] [PubMed]
49. Pereira, M. Heterogeneidade e viés de publicação em revisões sistemáticas. Epidemiol. Serv. Saúde 2014, 23,
775–778. [CrossRef]
163
Nutrients 2018, 10, 1396
50. Freedman, L.S.; Commins, J.M.; Moler, J.E.; Arab, L.; Baer, D.J.; Kipnis, V.; Midthune, D.; Moshfegh, A.J.;
Neuhouser, M.L.; Prentice, R.L.; et al. Pooled results from 5 validation studies of dietary self-report
instruments using recovery biomarkers for energy and protein intake. Am. J. Epidemiol. 2014, 180, 172–188.
[CrossRef] [PubMed]
51. Riordan, F.; Ryan, K.; Perry, I.J.; Schulze, M.B.; Andersen, L.F.; Geelen, A.; Van’t Veer, P.; Eussen, S.;
Dagnelie, P.; Wijckmans-Duysens, N.; et al. A systematic review of methods to assess intake of fruits and
vegetables among healthy European adults and children: A DEDIPAC (DEterminants of DIet and Physical
Activity) study. Public Health Nutr. 2017, 20, 417–448. [CrossRef] [PubMed]
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
164
nutrients
Article
Cardiovascular Biomarkers in Association with
Dietary Intake in a Longitudinal Study of Youth with
Type 1 Diabetes
Namrata Sanjeevi *, Leah M. Lipsky and Tonja R. Nansel
Social and Behavioral Sciences Branch, Division of Intramural Population Health Research, Eunice Kennedy
Shriver National Institute of Child Health and Human Development, Bethesda, MD 20817, USA;
[email protected] (L.M.L.); [email protected] (T.R.N.)
* Correspondence: [email protected]; Tel.: +1-301-827-0831
Abstract: Despite cardioprotective effects of a healthy diet in the general population, few studies
have investigated this relationship in individuals with type 1 diabetes, who are at elevated risks of
cardiovascular disease (CVD) due to hyperglycemia. The objective of this study was to examine
the association of CVD biomarkers with overall diet quality, as measured by the Healthy Eating
Index-2015 (HEI-2015), and its dietary components in youth with type 1 diabetes. Youth with type 1
diabetes (n = 136, 8–16.9 years) were enrolled in an 18-month behavioral nutrition intervention trial.
Dietary intake from three-day diet records, CVD biomarkers (total cholesterol (TC), high-density
lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C); triglycerides (TG),
C-reactive protein (CRP), 8-iso-prostaglandin-F2alpha (8-iso-PGF2α ), systolic and diastolic blood
pressure (SBP and DBP, respectively), and glycated hemoglobin (HbA1c) were assessed at baseline,
6, 12 and 18 months. Linear mixed-effects models estimated associations of dietary intake with
CVD biomarkers, adjusting for HbA1c and other covariates. Separate models estimated associations
of time-varying change in dietary intake with time-varying change in CVD biomarkers. HEI-2015
was not associated with CVD biomarkers, but whole grain intake was inversely associated with TC,
HDL-C and DBP, and a greater increase in whole fruit intake was associated with lower DBP. Added
sugar, saturated fat and polyunsaturated fat were positively related to serum TG, HDL-C, and DBP,
respectively. Findings suggest that the intake of specific dietary components, including whole grains,
whole fruits, added sugar and PUFA, may influence cardiometabolic health in youth with type 1
diabetes, independent of glycemic control.
Keywords: type 1 diabetes; cardiovascular disease; whole grains; glycemic control; serum lipids
1. Introduction
Cardiovascular disease (CVD) is the major cause of mortality and morbidity in patients with type
1 diabetes, whose risk is several-fold higher than the general population [1,2]. This increased risk starts
early in life; 15–45% of youth with type 1 diabetes have at least two CVD risk factors, and subclinical
CVD abnormalities are observed within the first decade of diagnosis [3]. While glycemic control is
central to CVD prevention in type 1 diabetes [4], identifying additional risk modifiers would inform
timely interventions.
Several studies in the general population have indicated associations of dietary intake with
CVD indicators; however, few studies have examined these relationships in individuals with type
1 diabetes. Better cardiometabolic health in the general population is consistently associated with
better overall diet quality [5,6], greater intake of whole plant foods [7–17], and lower intakes of added
sugar [18,19], saturated fat [20,21] and sodium [22,23]. Associations of dietary intake with CVD risk in
individuals with type 1 diabetes may differ from that of the general population due to the characteristic
glucose fluctuations and hyperglycemia that trigger inflammatory responses, thereby increasing CVD
risk [24,25]. However, only two studies have examined associations of overall diet quality with CVD
risk factors in individuals with type 1 diabetes, finding an inverse association of overall diet quality
with blood pressure in adults [26] and dyslipidemia in youth [27,28]. Furthermore, associations of
CVD biomarkers with the intake of individual food groups and nutrients other than fiber [29–31],
saturated and total fat [31] in adults, and sugar-sweetened beverages [32] in youth have not been
examined. Although diet quality indices reflect overall dietary patterns, a composite score based
on several dietary components may mask associations of individual food groups or nutrients with
cardiovascular health. Understanding associations with individual dietary components also is critical
for developing behavioral targets for reducing CVD risk in patients with type 1 diabetes. Additionally,
although oxidative stress and inflammation are known CVD risk factors [24], only one study has
investigated dietary associations with inflammation in patients with type 1 diabetes, suggesting an
inverse relationship of fiber intake with inflammation. No study has examined associations of diet
with oxidative stress. Thus, the objective of this research was to investigate relationships of CVD
biomarkers (serum lipids, blood pressure, inflammation and oxidative stress) with overall diet quality,
as measured by Healthy Eating Index-2015 (HEI-2015), and its dietary components in youth with type
1 diabetes followed prospectively for 18 months.
2.1. Measures
166
Nutrients 2018, 10, 1552
cholesterol (LDL-C) were assessed using enzyme linked immunosorbent assay (ELISA). Systolic and
diastolic blood pressure (SBP and DBP) were abstracted from medical records at each visit. A high
sensitivity enzyme-linked immunosorbent assay (Architect c8000, Abbott) with a lower detection
limit of 0.2 mg/L was used to quantify CRP in the serum samples, and 8-iso-prostaglandin F2α
(8-iso-PGF2α ), an indicator of oxidative stress, was determined from urine frozen at −80 ◦ C using
enzyme linked immunosorbent assay (ELISA).
167
Nutrients 2018, 10, 1552
representing the subject-specific baseline variation in the outcome. Covariates included baseline sex,
treatment assignment, and diabetes duration, and time-varying age, body mass index, Tanner stage,
insulin regimen, physical activity, use of cardiac medications, and HbA1c. Adjustments for multiple
comparisons were not performed due to the exploratory nature of the secondary data analysis [35,36].
Since 42.6%, 85.5%, 76.7% and 54.7% of participants reported zero intake of dark green vegetables,
beans, seafood, and nuts and seeds, respectively, these variables were dichotomized as having either
zero or greater than zero intake. Additional models estimated associations of each of the dichotomized
food group variable with CVD risk factors. Further, to enable within-subject analysis of the association
of change in diet with change in CVD biomarkers, the change from baseline in each dietary intake
variable and CVD biomarkers was calculated for each assessment period. Separate models estimated
associations of time-varying change in dietary intake with time-varying change in CVD biomarkers.
Models were controlled for baseline age, sex, treatment assignment, diabetes duration, Tanner stage,
insulin regimen, HbA1c, physical activity, body mass index and use of cardiac medication for the
respective assessment period. SPSS version 21 were used for all analyses.
3. Results
Baseline demographic and diet characteristics for study participants are shown in Tables 1 and 2,
respectively. At baseline, 19.4%, 11.2%, 28.4%, and 3.7% of participants were not within the
recommended concentrations [37,38] for TG (<150 mg/dL), TC (<200 mg/dL), LDL-C (<100 mg/dL)
and HDL-C (≥35 mg/dL), respectively.
Table 1. Baseline demographic characteristics of youth with type 1 diabetes overall and by
treatment assignment.
168
Nutrients 2018, 10, 1552
Table 2. Baseline dietary characteristics of youth with type 1 diabetes overall and by treatment assignment.
Associations between dietary intake and serum lipids after adjustments of HbA1c and other
covariates are indicated in Table 3. In the adjusted model, serum lipids were not associated with
overall diet quality. However, whole grain intake was inversely associated with TC and HDL-C. Added
sugar and saturated fat intakes were positively associated with TG and HDL-C, respectively. Other
dietary components (including dark green vegetables, beans, seafood, and nuts and seeds) were not
associated with serum lipids. Findings were unchanged when the food groups, dark green vegetables,
beans, seafood, and nuts and seeds, were treated as categorical variables (Table S1).
Associations of dietary intake with inflammation, oxidative stress and blood pressure are shown
in Table 4. These CVD biomarkers were not associated with overall diet quality; however, 8-iso-PGF2α
was inversely associated with refined grain intake, and lower DBP was related to greater whole grain
intake and greater (PUFA + MUFA): SFA. To determine the specific fatty acids associated with DBP,
we further examined the association of the intake of individual fatty acid with DBP. Although MUFA
intake (% kcal) was not associated with DBP (β = 0.08, SE = 0.10, p = 0.38), PUFA intake (% kcal) was
positively related to DBP (β = 0.23, SE = 0.09, p = 0.008). Dark green vegetables, beans, seafood and
nuts and seeds were not associated with inflammation, oxidative stress and blood pressure (Table 4),
and findings were unchanged when these food groups were treated as categorical variables (Table S2).
Associations between time-varying changes in dietary intake with time-varying changes in CVD
biomarkers are indicated in Tables S3 and S4 Greater increase in whole grain intake was associated
with a decrease in LDL-C and HDL-C. Increase in whole fruit intake was inversely associated with
DBP, whereas increase in fatty acid ratio was positively associated with DBP. Increase in time-varying
total vegetable, whole grain and added sugar intake was positively associated with TG, but increase in
time-varying refined grain intake was inversely associated with TG and 8-iso-PGF2α . Further, increase
in time-varying dairy intake was positively associated with SBP.
169
Table 3. Association of diet quality and food group and nutrient intake with serum lipids in youth with type 1 diabetes 1 .
Whole fruits 3 0.01 ± 0.04 0.70 3.22 ± 3.96 0.42 0.67 ± 1.86 0.72 1.59 ± 3.68 0.67
Total vegetables 3 0.03 ± 0.03 0.28 2.44 ± 2.83 0.39 0.05 ± 1.36 0.97 −2.74 ± 2.66 0.30
Dark green vegetables 3 −0.02 ± 0.06 0.75 −9.18 ± 6.31 0.15 −2.63 ± 2.85 0.36 −8.50 ± 5.89 0.15
Beans 3 −0.14 ± 0.10 0.18 −17.17 ± 10.77 0.11 −2.81 ± 5.06 0.58 −5.16 ± 10.22 0.61
Whole grains 3 0.01 ± 0.02 0.61 −4.60 ± 2.05 0.03 −1.98 ± 0.99 0.046 −3.42 ± 1.92 0.08
Dairy 3 0.01 ± 0.03 0.79 −1.97 ± 3.78 0.60 −0.64 ± 1.81 0.72 −1.61 ± 3.44 0.64
Protein foods 3 −0.01 ± 0.01 0.30 0.05 ± 0.88 0.95 0.25 ± 0.41 0.55 0.39 ± 0.83 0.63
Seafood 3 0.01 ± 0.03 0.61 0.09 ± 2.66 0.97 −0.90 ± 1.27 0.48 1.77 ± 2.47 0.47
Nuts and seeds 3 0.004 ± 0.02 0.84 0.15 ± 1.74 0.93 0.12 ± 0.85 0.89 0.06 ± 1.64 0.97
Refined grains 3 −0.02 ± 0.01 0.08 0.54 ± 1.05 0.61 0.31 ± 0.51 0.54 1.72 ± 0.98 0.08
Sodium 4 −0.05 ± 0.03 0.10 2.13 ± 3.33 0.52 0.21 ± 1.52 0.89 5.38 ± 3.10 0.08
Added sugars, % kcal 0.004 ± 0.002 0.04 0.22 ± 0.21 0.30 −0.10 ± 0.10 0.32 0.09 ± 0.20 0.64
Saturated fat, % kcal 0.61 0.35 0.04 0.58
170
−0.002 ± 0.003 0.35 ± 0.37 0.34 ± 0.17 0.19 ± 0.35
Fatty acid 5 −0.002 ± 0.02 0.90 −0.50 ± 2.10 0.81 −0.74 ± 0.96 0.45 −1.12 ± 1.93 0.56
TG, triglycerides; TC, total cholesterol; HDL-C, high-density lipoprotein cholesterol; LDL-C low-density lipoprotein cholesterol; HEI-2015, healthy eating index-2015. 1 Models were
controlled for baseline treatment assignment, sex, diabetes duration, and time-varying age, body mass index, Tanner stage, insulin regimen, physical activity, use of cardiac medication,
and glycemic control (HbA1c); 2 log-transformed to meet the normality assumption; 3 expressed as total number of cup- or ounce-equivalents per 1000 kcal; 4 expressed as grams per
1000 kcal; 5 calculated as (polyunsaturated fat + monounsaturated fat)/saturated fat.
Table 4. Association of diet quality and food group and nutrient intake with inflammation, oxidative stress and blood pressure in youth with type 1 diabetes 1 .
Whole fruits 3 0.07 ± 0.08 0.39 −0.17 ± 0.28 0.54 −0.73 ± 0.86 0.40 −0.11 ± 0.88 0.90
Total vegetables 3 −0.02 ± 0.05 0.67 0.06 ± 0.21 0.77 −0.02 ± 0.69 0.97 0.69 ± 0.63 0.28
Dark green vegetables 3 −0.04 ± 0.12 0.72 −0.42 ± 0.50 0.40 −0.52 ± 1.46 0.72 −1.10 ± 1.38 0.42
Beans 3 −0.30 ± 0.21 0.15 0.85 ± 1.05 0.42 −0.64 ± 2.67 0.81 −0.93 ± 2.43 0.70
Whole grains 3 0.04 ± 0.04 0.26 −0.09 ± 0.16 0.60 0.03 ± 0.47 0.96 −0.98 ± 0.46 0.04
Dairy 3 −0.004 ± 0.07 0.95 −0.47 ± 0.27 0.08 0.89 ± 0.81 0.27 −0.99 ± 0.73 0.18
Protein foods 3 −0.01 ± 0.02 0.55 0.09 ± 0.07 0.22 −0.11 ± 0.20 0.59 0.20 ± 0.17 0.23
Seafood 3 0.02 ± 0.05 0.65 −0.09 ± 0.21 0.69 −0.18 ± 0.63 0.78 0.78 ± 0.61 0.20
Nuts and seeds 3 0.02 ± 0.04 0.61 0.25 ± 0.14 0.08 0.29 ± 0.45 0.53 0.21 ± 0.43 0.62
Refined grains 3 −0.02 ± 0.02 0.42 −0.18 ± 0.08 0.04 0.05 ± 0.25 0.85 0.04 ± 0.23 0.86
Sodium 4 0.02 ± 0.06 0.79 −0.42 ± 0.27 0.12 −0.53 ± 0.78 0.50 0.19 ± 0.78 0.80
Added sugars, % kcal −0.00003 ± 0.004 0.99 0.01 ± 0.02 0.58 −0.04 ± 0.05 0.34 −0.01 ± 0.04 0.89
Saturated fat, % kcal 0.43 0.57 0.87 0.13
171
0.01 ± 0.007 0.02 ± 0.03 0.01 ± 0.08 −0.12 ± 0.08
Fatty acid 5 0.02 ± 0.04 0.59 0.19 ± 0.15 0.20 0.19 ± 0.45 0.67 0.98 ± 0.42 0.02
CRP, C-reactive protein; 8-iso-PGF2α , 8-iso-prostaglandin F2α ; SBP, systolic blood pressure; DBP, diastolic blood pressure; HEI-2015, healthy eating index-2015. 1 Models were controlled
for baseline treatment assignment, sex, diabetes duration, time-varying age, body mass index, Tanner stage, insulin regimen, physical activity, use of cardiac medication and glycemic
control (HbA1c); 2 log-transformed to meet the normality assumption; 3 expressed as total number of cup- or ounce-equivalents per 1000 kcal; 4 expressed as grams per 1000 kcal;
5 calculated as (polyunsaturated fat + monounsaturated fat)/saturated fat.
Nutrients 2018, 10, 1552
4. Discussion
In contrast to the general population [5,39], CVD biomarkers were not associated with greater
adherence to DGA 2015, as measured by HEI, in this sample of youth with type 1 diabetes. However,
greater intake of whole grains and whole fruits, and lower added sugar and PUFA were associated
with more favorable CVD biomarkers. Much of the current study findings regarding the association of
intake of specific dietary components with CVD biomarkers are comparable to the general population.
However, CVD etiology in type 1 diabetes differs from that of the general population, as hyperglycemia
is known to increase CVD risk. Findings suggest that these dietary components may be associated
with CVD risk in persons with type 1 diabetes independent of glycemic control.
Several studies in the general population have indicated the protective role of whole grains in
reducing CVD risk factors, including blood pressure [40] and TC [7,8,10]. In contrast, no study has
examined this relationship in individuals with type 1 diabetes. Improved endothelial function due
to whole grain consumption [41] could explain the association of greater intake with lower blood
pressure in this sample. The inverse association of whole grain intake with TC, independent of HbA1c
could be attributed to increased clearance or decreased biosynthesis of cholesterol [42]. This pathway
also is consistent with the inverse relationship between whole grain consumption and HDL-C found
in this study. Although this result is in contrast to previous research in the general population [8,10],
it is comparable to another study finding that a whole grain diet significantly lowered HDL-C in
overweight and obese adults [43]. The inverse association of whole grain intake with HDL-C is not
considered atherogenic in this sample, given the negligible number of participants (3.7%) with low
HDL-C (<35 mg/dL) at baseline. However, it is important to consider the impact of whole grain
consumption on HDL-C in future CVD-related research in patients with type 1 diabetes. Associations
of whole grain intake with cholesterol are further supported by within-subject analyses, where increase
in whole grain intake was inversely related to LDL-C and HDL-C. Further, improved endothelial
function could explain the inverse association of whole fruits with DBP, which is consistent to previous
research in the general population [44]. However, other relationships of time-varying changes in
whole grain, refined grain and dairy intake with that of CVD biomarkers are not supported by known
biological mechanisms. Findings for within-subject analyses should be interpreted with caution, given
the modest degree of individual change over time, resulting in increased chances of spurious findings.
The positive association of added sugar intake with serum TG is consistent with previous literature
in the general population [18,19]. This result could be attributed to hepatic de novo lipogenesis
induction by common sugars [45], resulting in increased serum TG, independent of glycemic control.
This finding is also somewhat comparable to another study in youth with type 1 diabetes, whereby
increased sugar-sweetened beverage intake was related to greater serum lipids [46]. The positive
association of saturated fat intake with HDL-C is comparable to previous research [47], and may be
explained by increased transport rates of HDL-C ester by dietary fat [47]. While additional studies are
needed, these findings indicate some consistency in the relationship of food group and nutrient intake
with serum lipids in individuals with type 1 diabetes as compared with the general population.
The inverse relationship of refined grain intake with oxidative stress contrasts previous research
indicating the detrimental effects of refined grain intake on CVD biomarkers [9]. This may be attributed
to the use of urinary 8-iso-PGF2α as an oxidative stress marker, which is a less sensitive measure than
its metabolite, 2,3-dinor-5,6-dihydro-15-F2t-isoprostane [48]. Future research using more sensitive
markers of oxidative stress could better elucidate the association of refined grain intake with oxidative
stress. Finally, greater DBP was associated with increased PUFA intake. Given PUFA includes omega-6
and omega-3 PUFA, a high intake could be obtained for an individual consuming excess omega-6 and
minimal omega-3 PUFA. Although total omega-6 PUFA intake could not be obtained for the current
study, stimulation of vasoconstriction by omega-6 PUFA-derived eicosanoids [49] could explain the
relationship of PUFA intake with DBP reported in this study.
The study findings are subject to certain limitations. These findings are observational, precluding
inferences on causality. The eligibility criteria for the primary study, recruitment from a single clinic,
172
Nutrients 2018, 10, 1552
22% recruitment rate and predominantly white sample limit generalizability of the results to all youth
with type 1 diabetes. Since youth with poor glycemic control (most recent HbA1c prior to recruitment
>10.0%) were not recruited, the study may have excluded patients most affected by adverse CVD
biomarkers. The lack of adjustment for multiple comparisons could have increased the family-wise
error rate. Nevertheless, the detected associations of diet with CVD biomarkers are supported by
biological mechanisms. Although food records are among the most valid method of assessing dietary
intake, they are susceptible to reporting and response biases [50]. However, this study is strengthened
by the prospective design utilizing repeated measures of exposures and outcomes, assessment of
several CVD risk factors, and adjustment of potentially important covariates in the analyses.
5. Conclusions
Nutrition recommendations for youth with type 1 diabetes include general healthful eating for
reducing risk of complications, and there are limited disease-specific nutrition guidelines. However,
characteristic hyperglycemia in individuals with type 1 diabetes may attenuate dietary influences on
CVD biomarkers. The current study suggests that overall diet quality was not associated with CVD
biomarkers in youth with type 1 diabetes. However, specific dietary components were associated with
CVD biomarkers, independent of glycemic control. Additional studies are needed to corroborate if
intake of greater whole grains, lower added sugar and PUFA may favorably influence CVD biomarkers
in this population. Examining whether these dietary associations differ by glycemic control could
inform efforts that promote cardiovascular health in patients with type 1 diabetes.
References
1. Margeirsdottir, H.D.; Larsen, J.R.; Brunborg, C.; Overby, N.C.; Dahl-Jorgensen, K.; Norwegian Study Group
for Childhood, D. High prevalence of cardiovascular risk factors in children and adolescents with type 1
diabetes: A population-based study. Diabetologia 2008, 51, 554–561. [CrossRef] [PubMed]
2. Rawshani, A.; Sattar, N.; Franzén, S.; Rawshani, A.; Hattersley, A.T.; Svensson, A.M.; Eliasson, B.;
Gudbjörnsdottir, S. Excess mortality and cardiovascular disease in young adults with type 1 diabetes
in relation to age at onset: A nationwide, register-based cohort study. Lancet 2018, 392, 477–486. [CrossRef]
3. de Ferranti, S.D.; de Boer, I.H.; Fonseca, V.; Fox, C.S.; Golden, S.H.; Lavie, C.J.; Magge, S.N.; Marx, N.;
McGuire, D.K.; Orchard, T.J.; et al. Type 1 diabetes mellitus and cardiovascular disease: A scientific
statement from the American Heart Association and American Diabetes Association. Circulation 2014,
130, 1110–1130. [CrossRef] [PubMed]
4. Mannucci, E.; Dicembrini, I.; Lauria, A.; Pozzilli, P. Is glucose control important for prevention of
cardiovascular disease in diabetes? Diabetes Care 2013, 36 (Suppl. 2), S259–S263. [CrossRef]
5. Nicklas, T.A.; O’Neil, C.E.; Fulgoni, V.L., 3rd. Diet quality is inversely related to cardiovascular risk factors
in adults. J. Nutr. 2012, 142, 2112–2118. [CrossRef] [PubMed]
6. Kuczmarski, M.F.; Mason, M.A.; Allegro, D.; Zonderman, A.B.; Evans, M.K. Diet quality is inversely
associated with C-reactive protein levels in urban, low-income African-American and white adults. J. Acad.
Nutr. Diet. 2013, 113, 1620–1631. [CrossRef] [PubMed]
7. Newby, P.K.; Maras, J.; Bakun, P.; Muller, D.; Ferrucci, L.; Tucker, K.L. Intake of whole grains, refined grains,
and cereal fiber measured with 7-d diet records and associations with risk factors for chronic disease. Am. J.
Clin. Nutr. 2007, 86, 1745–1753. [CrossRef] [PubMed]
173
Nutrients 2018, 10, 1552
8. McKeown, N.M.; Meigs, J.B.; Liu, S.; Wilson, P.W.; Jacques, P.F. Whole-grain intake is favorably associated
with metabolic risk factors for type 2 diabetes and cardiovascular disease in the Framingham Offspring
Study. Am. J. Clin. Nutr. 2002, 76, 390–398. [CrossRef] [PubMed]
9. Masters, R.C.; Liese, A.D.; Haffner, S.M.; Wagenknecht, L.E.; Hanley, A.J. Whole and refined grain intakes
are related to inflammatory protein concentrations in human plasma. J. Nutr. 2010, 140, 587–594. [CrossRef]
[PubMed]
10. Damsgaard, C.T.; Biltoft-Jensen, A.; Tetens, I.; Michaelsen, K.F.; Lind, M.V.; Astrup, A.; Landberg, R.
Whole-Grain Intake, Reflected by Dietary Records and Biomarkers, Is Inversely Associated with Circulating
Insulin and Other Cardiometabolic Markers in 8- to 11-Year-Old Children. J. Nutr. 2017, 147, 816–824.
[CrossRef] [PubMed]
11. Mellendick, K.; Shanahan, L.; Wideman, L.; Calkins, S.; Keane, S.; Lovelady, C. Diets Rich in Fruits and
Vegetables Are Associated with Lower Cardiovascular Disease Risk in Adolescents. Nutrients 2018, 10, 136.
[CrossRef] [PubMed]
12. Djousse, L.; Arnett, D.K.; Coon, H.; Province, M.A.; Moore, L.L.; Ellison, R.C. Fruit and vegetable
consumption and LDL cholesterol: The National Heart, Lung, and Blood Institute Family Heart Study.
Am. J. Clin. Nutr. 2004, 79, 213–217. [CrossRef] [PubMed]
13. Holt, E.M.; Steffen, L.M.; Moran, A.; Basu, S.; Steinberger, J.; Ross, J.A.; Hong, C.P.; Sinaiko, A.R. Fruit and
vegetable consumption and its relation to markers of inflammation and oxidative stress in adolescents. J. Am.
Diet. Assoc. 2009, 109, 414–421. [CrossRef] [PubMed]
14. Bazzano, L.A.; Thompson, A.M.; Tees, M.T.; Nguyen, C.H.; Winham, D.M. Non-soy legume consumption
lowers cholesterol levels: A meta-analysis of randomized controlled trials. Nutr. Metab. Cardiovasc. Dis. 2011,
21, 94–103. [CrossRef] [PubMed]
15. Salehi-Abargouei, A.; Saraf-Bank, S.; Bellissimo, N.; Azadbakht, L. Effects of non-soy legume consumption
on C-reactive protein: A systematic review and meta-analysis. Nutrition 2015, 31, 631–639. [CrossRef]
[PubMed]
16. Sabate, J.; Oda, K.; Ros, E. Nut consumption and blood lipid levels: A pooled analysis of 25 intervention
trials. Arch. Intern. Med. 2010, 170, 821–827. [CrossRef] [PubMed]
17. Yu, Z.; Malik, V.S.; Keum, N.; Hu, F.B.; Giovannucci, E.L.; Stampfer, M.J.; Willett, W.C.; Fuchs, C.S.; Bao, Y.
Associations between nut consumption and inflammatory biomarkers. Am. J. Clin. Nutr. 2016, 104, 722–728.
[CrossRef] [PubMed]
18. Kell, K.P.; Cardel, M.I.; Bohan Brown, M.M.; Fernandez, J.R. Added sugars in the diet are positively associated
with diastolic blood pressure and triglycerides in children. Am. J. Clin. Nutr. 2014, 100, 46–52. [CrossRef]
[PubMed]
19. Welsh, J.A.; Sharma, A.; Cunningham, S.A.; Vos, M.B. Consumption of added sugars and indicators of
cardiovascular disease risk among US adolescents. Circulation 2011, 123, 249–257. [CrossRef] [PubMed]
20. Te Morenga, L.; Montez, J.M. Health effects of saturated and trans-fatty acid intake in children and
adolescents: Systematic review and meta-analysis. PLoS ONE 2017, 12, e0186672. [CrossRef] [PubMed]
21. King, D.E.; Egan, B.M.; Geesey, M.E. Relation of dietary fat and fiber to elevation of C-reactive protein. Am. J.
Cardiol. 2003, 92, 1335–1339. [CrossRef] [PubMed]
22. Aburto, N.J.; Ziolkovska, A.; Hooper, L.; Elliott, P.; Cappuccio, F.P.; Meerpohl, J.J. Effect of lower sodium
intake on health: Systematic review and meta-analyses. BMJ 2013, 346, f1326. [CrossRef] [PubMed]
23. Zhu, H.; Pollock, N.K.; Kotak, I.; Gutin, B.; Wang, X.; Bhagatwala, J.; Parikh, S.; Harshfield, G.A.; Dong, Y.
Dietary sodium, adiposity, and inflammation in healthy adolescents. Pediatrics 2014, 133, e635. [CrossRef]
[PubMed]
24. Pistrosch, F.; Natali, A.; Hanefeld, M. Is hyperglycemia a cardiovascular risk factor? Diabetes Care 2011,
34 (Suppl. 2), S128–S131. [CrossRef]
25. Goldberg, I.J. Clinical review 124: Diabetic dyslipidemia: Causes and consequences. J. Clin. Endocrinol. Metab.
2001, 86, 965–971. [CrossRef] [PubMed]
26. Gingras, V.; Leroux, C.; Desjardins, K.; Savard, V.; Lemieux, S.; Rabasa-Lhoret, R.; Strychar, I. Association
between Cardiometabolic Profile and Dietary Characteristics among Adults with Type 1 Diabetes Mellitus.
J. Acad. Nutr. Diet. 2015, 115, 1965–1974. [CrossRef] [PubMed]
174
Nutrients 2018, 10, 1552
27. Liese, A.D.; Bortsov, A.; Gunther, A.L.; Dabelea, D.; Reynolds, K.; Standiford, D.A.; Liu, L.; Williams, D.E.;
Mayer-Davis, E.J.; D’Agostino, R.B., Jr.; et al. Association of DASH diet with cardiovascular risk factors in
youth with diabetes mellitus: The SEARCH for Diabetes in Youth study. Circulation 2011, 123, 1410–1417.
[CrossRef] [PubMed]
28. Zhong, V.W.; Lamichhane, A.P.; Crandell, J.L.; Couch, S.C.; Liese, A.D.; The, N.S.; Tzeel, B.A.; Dabelea, D.;
Lawrence, J.M.; Marcovina, S.M.; et al. Association of adherence to a Mediterranean diet with glycemic
control and cardiovascular risk factors in youth with type I diabetes: The SEARCH Nutrition Ancillary
Study. Eur. J. Clin. Nutr. 2016, 70, 802–807. [CrossRef] [PubMed]
29. Toeller, M.; Buyken, A.E.; Heitkamp, G.; de Pergola, G.; Giorgino, F.; Fuller, J.H. Fiber intake, serum
cholesterol levels, and cardiovascular disease in European individuals with type 1 diabetes. EURODIAB
IDDM Complications Study Group. Diabetes Care 1999, 22 (Suppl. 2), B21.
30. Bernaud, F.S.; Beretta, M.V.; do Nascimento, C.; Escobar, F.; Gross, J.L.; Azevedo, M.J.; Rodrigues, T.C. Fiber
intake and inflammation in type 1 diabetes. Diabetol. Metab. Syndr. 2014, 6, 66. [CrossRef] [PubMed]
31. Toeller, M.; Buyken, A.E.; Heitkamp, G.; Scherbaum, W.A.; Krans, H.M.; Fuller, J.H. Associations of fat and
cholesterol intake with serum lipid levels and cardiovascular disease: The EURODIAB IDDM Complications
Study. Exp. Clin. Endocrinol. Diabetes 1999, 107, 512–521. [CrossRef] [PubMed]
32. Liese, A.D.; Crandell, J.L.; Tooze, J.A.; Kipnis, V.; Bell, R.; Couch, S.C.; Dabelea, D.; Crume, T.L.;
Mayer-Davis, E.J. Sugar-sweetened beverage intake and cardiovascular risk factor profile in youth with
type 1 diabetes: Application of measurement error methodology in the SEARCH Nutrition Ancillary Study.
Br. J. Nutr. 2015, 114, 430–438. [CrossRef] [PubMed]
33. Nansel, T.R.; Laffel, L.M.; Haynie, D.L.; Mehta, S.N.; Lipsky, L.M.; Volkening, L.K.; Butler, D.A.; Higgins, L.A.;
Liu, A. Improving dietary quality in youth with type 1 diabetes: Randomized clinical trial of a family-based
behavioral intervention. Int. J. Behav. Nutr. Phys. Act. 2015, 12, 58. [CrossRef] [PubMed]
34. Centers for Disease Control and Prevention. Behavioral Risk Factor Surveillance System Survey Questionnaire;
Centers for Disease Control and Prevention: Atlanta, GA, USA, 2001.
35. Bender, R.; Lange, S. Multiple test procedures other than Bonferroni’s deserve wider use. BMJ 1999, 318, 600.
[CrossRef] [PubMed]
36. Althouse, A.D. Adjust for Multiple Comparisons? It’s Not That Simple. Ann. Thorac. Surg. 2016,
101, 1644–1645. [CrossRef] [PubMed]
37. Cox, R.A.; Garcia-Palmieri, M.R. Cholesterol, Triglycerides, and Associated Lipoproteins. In Clinical Methods:
The History, Physical, and Laboratory Examinations; Walker, H.K., Hall, W.D., Hurst, J.W., Eds.; Butterworth
Publishers: Boston, MA, USA, 1990.
38. Gooding, H.C.; de Ferranti, S.D. Cardiovascular risk assessment and cholesterol management in adolescents:
Getting to the heart of the matter. Curr. Opin. Pediatr. 2010, 22, 398–404. [CrossRef] [PubMed]
39. Shah, B.S.; Freeland-Graves, J.H.; Cahill, J.M.; Lu, H.; Graves, G.R. Diet quality as measured by the healthy
eating index and the association with lipid profile in low-income women in early postpartum. J. Am.
Diet. Assoc. 2010, 110, 274–279. [CrossRef] [PubMed]
40. Flint, A.J.; Hu, F.B.; Glynn, R.J.; Jensen, M.K.; Franz, M.; Sampson, L.; Rimm, E.B. Whole grains and incident
hypertension in men. Am. J. Clin. Nutr. 2009, 90, 493–498. [CrossRef] [PubMed]
41. Katz, D.L.; Nawaz, H.; Boukhalil, J.; Chan, W.; Ahmadi, R.; Giannamore, V.; Sarrel, P.M. Effects of oat and
wheat cereals on endothelial responses. Prev. Med. 2001, 33, 476–484. [CrossRef] [PubMed]
42. Develaraja, S.; Reddy, A.; Yadav, M.; Jain, S.; Yadav, H. Whole Grains in Amelioration of Metabolic
Derangements. J. Nutr. Health Food Sci. 2016, 4, 1–11.
43. Kirwan, J.P.; Malin, S.K.; Scelsi, A.R.; Kullman, E.L.; Navaneethan, S.D.; Pagadala, M.R.; Haus, J.M.; Filion, J.;
Godin, J.P.; Kochhar, S.; et al. A Whole-Grain Diet Reduces Cardiovascular Risk Factors in Overweight and
Obese Adults: A Randomized Controlled Trial. J. Nutr. 2016, 146, 2244–2251. [CrossRef] [PubMed]
44. Borgi, L.; Muraki, I.; Satija, A.; Willett, W.C.; Rimm, E.B.; Forman, J.P. Fruit and Vegetable Consumption
and the Incidence of Hypertension in Three Prospective Cohort Studies. Hypertension 2016, 67, 288–293.
[CrossRef] [PubMed]
45. Schwarz, J.M.; Clearfield, M.; Mulligan, K. Conversion of Sugar to Fat: Is Hepatic de Novo Lipogenesis
Leading to Metabolic Syndrome and Associated Chronic Diseases? J. Am. Osteopath. Assoc. 2017, 117, 520–527.
[CrossRef] [PubMed]
175
Nutrients 2018, 10, 1552
46. Bortsov, A.V.; Liese, A.D.; Bell, R.A.; Dabelea, D.; D’Agostino, R.B., Jr.; Hamman, R.F.; Klingensmith, G.J.;
Lawrence, J.M.; Maahs, D.M.; McKeown, R.; et al. Sugar-sweetened and diet beverage consumption
is associated with cardiovascular risk factor profile in youth with type 1 diabetes. Acta Diabetol. 2011,
48, 275–282. [CrossRef] [PubMed]
47. Hayek, T.; Ito, Y.; Azrolan, N.; Verdery, R.B.; Aalto-Setala, K.; Walsh, A.; Breslow, J.L. Dietary fat increases
high density lipoprotein (HDL) levels both by increasing the transport rates and decreasing the fractional
catabolic rates of HDL cholesterol ester and apolipoprotein (Apo) A-I. Presentation of a new animal model
and mechanistic studies in human Apo A-I transgenic and control mice. J. Clin. Investig. 1993, 91, 1665–1671.
[PubMed]
48. Dorjgochoo, T.; Gao, Y.T.; Chow, W.H.; Shu, X.O.; Yang, G.; Cai, Q.; Rothman, N.; Cai, H.; Li, H.; Deng, X.;
et al. Major metabolite of F2-isoprostane in urine may be a more sensitive biomarker of oxidative stress than
isoprostane itself. Am. J. Clin. Nutr. 2012, 96, 405–414. [CrossRef] [PubMed]
49. Patterson, E.; Wall, R.; Fitzgerald, G.F.; Ross, R.P.; Stanton, C. Health implications of high dietary omega-6
polyunsaturated Fatty acids. J. Nutr. Metab. 2012, 2012, 539426. [CrossRef] [PubMed]
50. Ortega, R.M.; Perez-Rodrigo, C.; Lopez-Sobaler, A.M. Dietary assessment methods: Dietary records.
Nutr. Hosp. 2015, 31 (Suppl. 3), 38–45.
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
176
nutrients
Article
Food Perceptions and Dietary Changes for Chronic
Condition Management in Rural Peru: Insights for
Health Promotion
Silvana Perez-Leon 1, *, M. Amalia Pesantes 1 , Nathaly Aya Pastrana 2 , Shivani Raman 3 ,
Jaime Miranda 1 and L. Suzanne Suggs 2
1 CRONICAS Center of Excellence in Chronic Diseases, Universidad Peruana Cayetano Heredia, Lima 15074,
Peru; [email protected] (M.A.P.); [email protected] (J.M.)
2 BeCHANGE Research Group, Institute of Public Communication, Università della Svizzera italiana,
6900 Lugano, Switzerland; [email protected] (N.A.P.); [email protected] (L.S.S.)
3 Department of Sociology, Rice University, Houston, TX 77005, USA; [email protected]
* Correspondence: [email protected]; Tel.: +51-1-241-6978
Abstract: Peru is undergoing a nutrition transition and, at the country level, it faces a double
burden of disease where several different conditions require dietary changes to maintain a healthy
life and prevent complications. Through semistructured interviews in rural Peru with people
affected by three infectious and noninfectious chronic conditions (type 2 diabetes, hypertension,
and neurocysticercosis), their relatives, and focus group discussions with community members,
we analyzed their perspectives on the value of food and the challenges of dietary changes due to
medical diagnosis. The findings show the various ways in which people from rural northern Peru
conceptualize good (buena alimentación) and bad (mala alimentación) food, and that food choices are
based on life-long learning, experience, exposure, and availability. In the context of poverty, required
changes are not only related to what people recognize as healthy food, such as fruits and vegetables,
but also of work, family, trust, taste, as well as affordability and accessibility of foods. In this paper we
discuss the complexity of introducing dietary changes in poor rural communities whose perspectives
on food are poorly understood and rarely taken into consideration by health professionals when
promoting behavior change.
Keywords: dietary changes; health promotion; health behavior; Peru; chronic conditions
1. Introduction
Over the past several decades, low- and middle-income countries (LMICs) have experienced a
nutrition transition. Countries that have faced hunger and malnutrition as their main concerns are
now confronting an additional problem of overweight and obesity and are experiencing increasing
rates of noncommunicable diseases (NCD) such as diabetes and hypertension [1]. Additionally, LMICs
are still struggling to reduce the incidence of infectious diseases that are often caused by contaminated
food or poor hygiene practices. One example is neurocysticercosis (NCC), a neglected tropical disease
(NTD) characterized by parasitic infection of the brain that primarily affects the poor [2].
Peru is undergoing a nutrition transition. The Ministry of Health of Peru has estimated that 20%
of the burden of disease in 2011 was associated with overweight and obesity [3]. NCDs are the main
cause of mortality in Peru [3], while NTDs continue to affect vulnerable groups of the population such
as those living in rural areas with limited access to water and sanitation facilities. In the north of Peru,
the prevalence of type 2 diabetes (T2D) and hypertension (HT) is high (10% and 26%, respectively) [4,5],
and NCC is considered to be endemic in this part of the country [2].
Type 2 diabetes (T2D) and hypertension (HT) require dietary changes to manage the conditions
and though patients with NCC do not require specific changes in their diet, its prevention has a
close relation with the consumption of uncooked pork and poor sanitary practices. The control of
NCC requires breaking the life cycle of the Taenia solium [6], where the main intervention to stop
transmission in LMICs is improving sanitary practices [7,8]. The prevention of both NCDs and NCC
share the need to promote changes in dietary habits in the population.
Most studies in LMICs have addressed the problem of nutrition transition from the perspective
of accessibility and affordability [9–12] but nutrition is a complex issue since what people eat is not
only about nutritional value, as food choices and preferences are influenced by many factors [13].
The cultural and social contexts in which individuals are brought up, live, and work have a strong
influence on the food choices made, as they affect their views of foods and eating behaviors [13].
Diet is also an important aspect of social life and is related with sharing, belonging to a group, and
celebration [14]. Thus, promoting and supporting dietary changes requires a thorough understanding
of local cultural norms, values, beliefs, and practices that underlie certain unhealthy habits [15].
In this study, we examine the perspectives on the value of food and the challenges of dietary
changes among people living with T2D, HT, and/or NCC from four rural communities in Northern
Peru. We contrast and compare local perspectives of what and why a particular food item is perceived
as “good” or “bad”. Our findings are particularly relevant for culturally appropriate design of
health promotion aiming to achieve the U.N. Sustainable Development Goals for ending all forms of
malnutrition and ensuring healthy lives.
178
Nutrients 2018, 10, 1563
Interviews allowed the discovery of opinions and personal experiences on the selected chronic
conditions of the caregivers, heads of households, and patients with T2D, HT, and NCC. The rationale
for interviewing caregivers, heads of household, and patients was to have complementary views of the
condition from each family. All caregivers and head of households were relatives of those affected by
T2D, HT, or NCC. Due to the difficulties to find patients diagnosed with NCC in these communities,
we included persons with epilepsy. Epilepsy is one of the main symptoms for those with NCC, which
is endemic in Ayabaca [2].
Focus groups examined the common perceptions of community members on T2D, HT, and NCC
as well as other health problems, and focus group participants were homogeneous in age and sex.
In order to have the perspective of a vulnerable group within the selected communities, additional
focus groups were conducted with women beneficiaries of JUNTOS, the national cash transfer program
targeting poor women with children under five years old. Some focus groups did not take place
because no participants showed up. Each “failed” focus group was replaced with four individual
interviews. Additionally, when the focus groups had less than six participants, the research team
conducted a “small” focus group and complimented it with additional interviews.
2.4. Recruitment
For semistructured interviews, fieldworkers used word of mouth to find people with the
conditions of T2D, HT, and NCC, their caregiver and head of households, followed by a snowball
sampling methodology.
For the recruitment of focus groups, fieldworkers invited people gathered in different parts of
the communities to participate, e.g., church, marketplace, main square, etc., and asked them to invite
friends of the same age and sex.
179
Nutrients 2018, 10, 1563
and were then uploaded into ATLAS.ti 8 (Scientific Software Development GmbH, Berlin, Germany),
including the field notes.
A codebook was created (see Table S4: Codebook) and entered into ATLAS.ti 8. To ensure quality
of coding, weekly discussions were held between the lead researcher, two research assistants, and a
doctoral student, in which the codes were analyzed and concerns were addressed.
2.8. Ethics
The study was approved by the Comité Institucional de Ética (Institutional Review Board) at
Universidad Peruana Cayetano Heredia in Lima, Peru (code 393-22-16), and by the Institutional
Review Board at the University of Geneva in Geneva, Switzerland (IRB No. 2016-01242).
3. Results
A total of 138 individuals participated in the study. In total, 16 semistructured interviews
were conducted with individuals living with T2D (n = 4), HT (n = 7), and NCC or epilepsy (n = 5),
the caregivers (n = 8), and head of household (n = 5). Additionally, 12 focus group discussions were
arranged where 82 community dwellers participated as well as 27 individual interviews that replaced
or complimented some focus groups.
180
Nutrients 2018, 10, 1563
Based on their answers, it appeared that most people have three meals, breakfast, lunch, and dinner,
and that the quantity of food consumed at each meal was usually big in comparison to the amounts
people from urban areas eat:
People are used to eat a lot, not like in the city where for example in the morning they have coffee
and bread. (Female, 70 years old)
We eat a lot. Here is not like the city where they eat bread, here if there is no rice or cassava it is not
a meal. ( . . . ) Here people eat a lot, the big dish and double portion. (Male, 18–34 years group)
For breakfast, participants said they could have rice and fried chicken or rice and fish or egg.
At noon, some people have a snack and later they will have lunch which often consists of rice
and another starchy local food such as potato, yucca, and plantains with legumes (menestras) (see
Figures S2–S4). At dinner, participants stated they would have a soup with noodles, some vegetables,
chicken or with egg. Red meat and pork were not commonly consumed, while chicken and fish (salted
fish) were mentioned as part of the regular diet. Though pork is not consumed in their day-to-day
diets, people often mentioned it as a traditional meal, something easily found in local restaurants.
Vegetables do not appear to be a common ingredient. According to participants, the consumption of
vegetables is related to their availability according to seasonal changes, as one participant explained:
“during the rainy seasons (January, February and March) we eat more vegetables like lettuce and carrot. In April
and May we eat more tamalitos, more corn.”(Man, 56 years old). It is hard to find vegetables in the local
bodegas and if they are available, they are expensive. We observed that noodles were a staple food
in the communities, as they are affordable. Though the interview and focus group participants did
not frequently report this consumption, the observational notes that complemented this information
showed that noodles and cookies are regularly sold products in the small shops of the communities
and noodles are a common ingredient seen in the meals of local people.
Overall, participants described a high consumption of carbohydrates and legumes that they
usually grow. Meat consumption (especially chicken and salted fish) was frequent but in smaller
quantities. Few participants mentioned fruits except for bananas which are part of the local diet.
3.2. What Do People Consider to Be a “Good” and “Bad” Food in These Communities?
As expected, informants responded to the question “¿Para usted cómo es una buena alimentación?
(What is good meal/food/nutrition for you?)”, in different ways. In most cases, they mentioned
specific types of foods they consider to be a buena alimentación (good meal/food/nutrition) or, on
the contrary, foods they view as a “mala alimentacion” (bad meal/food/nutrition). In other cases,
informants mentioned characteristics of their meals (whether meals are “balanced”), certain methods
of food preparation, or aspects of the food production process.
Informants most often responded by mentioning specific foods that they consider to be a buena
alimentación or mala alimentación. In the following tables (Tables 1 and 2) we summarize the most
frequently mentioned food categories and the number of transcripts where each type of food was
mentioned. These tables include responses from both interviews and focus groups, not all the
informants answered this question. Out of 67 transcripts, 62 had a response.
181
Nutrients 2018, 10, 1563
Of those informants who identified meat as a mala alimentación (n = 18), most identified pork
(n = 9) and/or chicken (n = 6) as the most harmful meats. Informants characterized pork as mala
alimentación due to its high fat content and its link to NCC. For those diagnosed with NCC or epilepsy,
almost all (4/5) mentioned pork as a mala alimentación.
Informants also characterized a buena alimentación by mentioning specific methods of food
preparation. Several mentioned that “comidas sancochadas”, or foods that have been cooked in boiling
water, are preferable to fried foods: “(I think a good meal is) usually boiled food, because most of the people
eat fried, or making steamed fish or ceviche. Because if you eat fried food it has fat. For me healthier food is
steamed.” (Woman, 38 years old).
One participant mentioned that buena alimentación consists of maintaining good hygiene in the
process of preparing food: “We need to have a proper hygiene to be able to feed us. If we don’t have the cookware
well cleaned, the hands washed and the nails cut, then we are not going to have a good nutrition”. (Woman,
18–34 years group). The importance of hygienical preparation was also mentioned in a conversation
with a local restaurant owner.
In addition to mentioning aspects of food preparation, informants also characterized buena
alimentación by describing the food production process. Informants considered the foods grown
in their communities or in the “olden days” to be buena alimentación because they are free of
hormones, pesticides, herbicides, fertilizers, and other chemicals. On the other hand, they viewed
foods from the city or “modern” time as mala alimentación because they are produced using the
abovementioned chemicals:
The food here is healthy because the food is natural. Here you can still find natural food, mainly
what comes from the farms because the products do not have fertilizers, so the food is natural. (Man,
70 years old)
They inject them too much, those chicken don’t have nutrients. What is good is milk, lentil, beef,
mutton, pork, which is also nutritious as they eat corn. (Woman, 35 years or older group)
The truth is that we do not know from where they bring the vegetables that are sold in the market, we
do not have trust. ( . . . ) When you grow vegetables in your garden you have trust. But those plants
that come from other places are not recommended to eat because they irrigate them with wastewater.
(Women, 35 years or older group)
The food now is not like before. (Before) it was natural ( . . . ) now it has a lot of chemical, the plant
absorbs the chemical. (Women, 35 years old)
Several informants’ characterized rice (see Table 2) as mala alimentación because they consider that
it requires a greater use of chemicals to grow: “The rice is more chemical, the rice is to fill the stomach.”
(Man, 35 or older years group).
Several also described buena alimentación as eating “balanced” food/meals. While most did not specify
what they meant by “balanced” food/meals, two informants provided the following descriptions:
You have to eat meat, salad and when there is no meat it can be legumes and to drink it can be chicha
morada (typical beverage made of purple corn). (Woman with HT, 48 years old)
182
Nutrients 2018, 10, 1563
I think the meals have to be balanced. Eating cereals, fruits, vegetables in large amounts, fish and
eating more white than red meat. Red meat is a little more harmful. From time to time you can eat a
desert, like yogurt. (Man, 24 years old)
The reasons provided to the question “¿Por qué es importante tener una buena alimentación? (Why
is it important to have a good meal/food/nutrition?)” are summarized in Table 3. In many cases,
informants mentioned more than one of the listed reasons. Therefore, the categories below are not
independent of one another, rather, they are often interrelated (e.g., “to maintain good health” co-occurs
with “to prevent diseases”). This table includes responses from both interviews and focus groups, not
all the informants answered this question. Out of the 67 transcripts, 36 had a response.
Most often, informants stated that it is important to have buena alimentación in order to maintain
good health in general. Many also expressed that it is important to have buena alimentación in order
to prevent diseases. They often mentioned maintaining good health in conjunction with preventing
diseases: “To not get sick also, have a good health.” (Women with HT, 48 years old). One female informant
specifically mentioned that buena alimentación is important for preventing anemia; however, no one
mentioned T2D, HT, or NCC.
The second most commonly mentioned reason for maintaining buena alimentación was important
for having sufficient strength and endurance to carry out day-to-day labor and activities. Responses
often corresponded to their roles within their family and in the community. Men commonly mentioned
working in agriculture, while women most often mentioned performing domestic tasks and caring
for children.
Because if we are weak we cannot do anything, what are we supposed to do? You cannot work, not
even with your wife you can work. (Man with NCC, 36 years old)
To have strength to work, for example we are weaving and it’s time to have lunch, we make some
food and then again we weave with strength. (Woman with T2D, 68 years old)
In response to a question about what foods give strength to perform agricultural work, responses
commonly mentioned included mote (Peruvian corn), sango (food prepared with corn), and sweet
potato as foods that provide strength and endurance. “The sweet potato is also good, it lasts all day, from
8:00 am when you eat, until 19:00.” (Man with NCC, 36 years) Participants described mala alimentación as
foods that do not provide sufficient strength and endurance to perform daily labors:
The bread does not make your stomach full, for example here you cannot eat bread because the work
is hard. If you eat bread at breakfast it will only last until 10:00 am and you have no strength.”
(Man, 41 years old)
Some participants also mentioned that buena alimentación is important for attending school
and studying.
183
Nutrients 2018, 10, 1563
4. Discussion
Our study shows the various ways in which people from rural northern Peru value food and
the multiplicity of factors that play a role in identifying food as good/healthy or not. Understanding
food- and diet-related contexts like the one in Ayabaca, and similar rural settings facing a nutritional
184
Nutrients 2018, 10, 1563
transition, and the complexity attached to it, is fundamental to end all forms of malnutrition
(encompassing both undernutrition and overnutrition) and promoting healthy lifestyles and well-being
for all and throughout the life course, as proposed by the sustainable development goals 2 and 3 [22].
Our study highlights the challenges of dietary changes in patients with chronic conditions in
a rural area and the relevance of understanding the local context. In the communities where we
conducted this study, perceptions of buena and mala alimentación are strongly linked to the extent
to which foods enable individuals to perform day-to-day tasks and activities, and this finding is
consistent with other studies in Peru [23]. For this reason, individuals are accustomed to consuming
high-carbohydrate foods such as rice, potato, corn, and grains, often in large portion sizes. On the
other hand, there is conflict between the people recognizing vegetables and fruits as “good” food,
and the insufficient consumption of these fruits and vegetables because they are not available or
are not affordable. As other studies have reported [24,25], we also found that when foods are not
available within the community, participants do not buy them outside due to negative perceptions
about consuming food from an unknown source. Yet, there is an increasing consumption of cheap
processed foods, such as noodles.
Our findings show the complexity of introducing dietary changes in rural communities where
the perspectives on food are poorly understood and rarely taken into consideration when promoting
food- or diet-related behavior change. T2D and HT are diseases for which much of their management
can occur in the community, at the primary care level, and where the prevention of NCC needs
adequate control at the community as well. So the food people intake will have consequences in the
management of their disease or in the prevention of new diseases.
For patients with T2D, HT, and NCC, following dietary recommendations received by health
professionals is challenging. On the one hand, people have difficulties not being able to eat food they
like, the food they grew up with, the food that they perceive as nutritious or tasteful, nor eat certain
foods in the quantities they are used to. On the other hand, the food recommended by the health
professionals are things they do not like to eat. Furthermore, diet modifications involve negotiation or
arrangements with the family, which is key to successfully adapt to the recommended diet [26]. In this
sense, involving the family is a key element for dietary modifications. As Vanstone and colleagues
show in their review, “support at home is universally described as an essential component of successful
dietary modification” [27,28]. Patients in our study know and say they make dietary modifications, but
the compliance of healthy dietary practices seems more complex to accomplish. As what is reported in
other studies [27,28], making dietary changes recommended by health professionals is challenging.
This study helps understand how dietary recommendations for chronic conditions in rural agrarian
communities need to be reconciled with working conditions, access to food, perceptions of food,
and family.
Our results align with some crosscutting themes such as food availability, introducing dietary
changes, and health promotion. What people eat, especially what poor people eat, is not only about
what people identify as healthy, but the availability of such foods. Food availability is one of the four
pillars of food security [29]. Thus, a poor person’s socioeconomic status can exert a strong influence on
food choice behaviors [30–32]. It is necessary to transform food systems into sustainable and sensitive
nutrition systems that will provide a variety of healthy foods, devoting special attention to the most
vulnerable [9].
The availability of cheap unhealthy choices is a growing problem in many LMICs [33–35] and it is
also happening in rural Peru [36]. Understanding local eating habits must take into account the role of
structural factors on food choices [37]. Thus, our study supports the finding that individual choices,
especially those of the poor, are influenced by broader social and environmental factors [32,38].
Our findings are consistent with studies across the globe that show that knowledge is not enough
to influence behavior [39–41]. Theories of human behaviors, such as eating behavior, suggest that a
combination of environmental and personal factors influence behavior [42]. Environmental factors
include cultural determinants (e.g., nationality, ethnicity, identity), physical (e.g., policies, availability,
185
Nutrients 2018, 10, 1563
accessibility), and social (e.g., parental behavior, peer influence, advertisement exposure) [43]. Personal
determinants, such as liking and preference, are the strongest determinants of diet habits [44–46].
Introducing, achieving, and sustaining healthier dietary changes will require a combination of all those
factors, beyond availability and affordability.
This study highlights that patients, caregivers, and community members living in rural Peru have
reasonable knowledge of what a healthy diet consists of, yet making food choices is a complex issue
influenced by work, family, trust, taste, as well as affordability and accessibility. It also highlights
that perceptions of the value of certain foods are not necessarily consistent with the advice received
about healthy diet from health educators and health workers. Health promotion efforts therefore
must include not only factual information about nutrients, but also provide opportunities to taste
new foods, cook in new ways that are sustainable, and learn about what foods and vendors from
outside the community are prepared in ways that respect local preferences. For example, it would be
beneficial for health professionals to have a good idea of what foods are produced locally in order
to recommend these items in the diet, but also to provide a clear understanding of the relationship
between what a person needs versus how much is needed, in accordance with their daily activities
(i.e., caloric expenditure).
Some limitations are worth noting. During the recruitment process, it was not always possible to
achieve the desired sample size of patient, caregiver, and head of household. The participation in focus
groups was low due to the rainy season and there were difficulties recruiting young men in the focus
groups. Additional interviews with community dwellers were conducted so as to reach data saturation.
Additionally, our study did not include the perspective of health workers’ views, which would be
helpful in better understanding their challenges in promoting healthy eating. Furthermore, we did not
collect information about the availability of food at local stores and their prices in a systematic way
that would enable us to fully understand accessibility and affordability of food choices in our sample.
Finally, objective measures of the reality in the homes of participants were not collected and so it was
not possible to know with certainty if perceptions matched the reality in the home. Nonetheless, the
methodology and sampling procedure utilized allow for an overview of the situation regarding food
perceptions in these communities.
5. Conclusions
This study shows the important influence of culture and social conditions on food perceptions
and dietary changes in rural Peru. The findings highlight that poor people in rural northern Peru,
just like others across the globe, make decisions and have knowledge about food that are based on
life-long learning, experience, exposure, and availability. The study also stresses that knowledge
alone does not influence eating behavior and that making dietary change is difficult. Thus, promoting
healthy diets without contextualizing information and recommendations is sure to produce suboptimal
results. Health promotion focused on nutrition for rural, poor communities must take into account the
activities of daily life, access and affordability of foods, but also food taste preferences and traditions.
186
Nutrients 2018, 10, 1563
Acknowledgments: We want to thank David Beran, who provided critical feedback on earlier versions of the
manuscript and whose encouragement was key to complete the manuscript.
Conflicts of Interest: The authors declare no conflict of interest. The funders had no role in the design of the
study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, and in the decision to
publish the results.
References
1. Popkin, B.M.; Adair, L.S.; Ng, S.W. Global nutrition transition and the pandemic of obesity in developing
countries. Nutr. Rev. 2012, 70, 3–21. [CrossRef] [PubMed]
2. Moyano, L.M.; O’Neal, S.E.; Ayvar, V.; Gonzalvez, G.; Gamboa, R.; Vilchez, P.; Rodriguez, S.; Reistetter, J.;
Tsang, V.C.W.; Gilman, R.H.; et al. High Prevalence of Asymptomatic Neurocysticercosis in an Endemic
Rural Community in Peru. PLoS Negl. Trop. Dis. 2016, 10, e0005130. [CrossRef] [PubMed]
3. Diez-Canseco, F.; Saavedra-Garcia, L. Programas sociales y reducción de la obesidad en el Perú: Reflexiones
desde la investigación. Revista Peruana De Medicina Experimental Y Salud Pública 2017, 34, 105–112. [CrossRef]
[PubMed]
4. Bernabé-Ortiz, A.; Carrillo-Larco, R.M.; Gilman, R.H.; Checkley, W.; Smeeth, L.; Miranda, J.J. Contribution of
modifiable risk factors for hypertension and type-2 diabetes in Peruvian resource-limited settings. J. Epidemiol.
Community Health 2016, 70, 49–55. [CrossRef] [PubMed]
5. Bernabe-Ortiz, A.; Perel, P.; Miranda, J.J.; Smeeth, L. Diagnostic accuracy of the Finnish Diabetes Risk Score
(FINDRISC) for undiagnosed T2DM in Peruvian population. Prim. Care Diabetes 2018, in press. [CrossRef]
[PubMed]
6. Maurice, J. Of pigs and people—WHO prepares to battle cysticercosis. Lancet 2014, 384, 571–572. [CrossRef]
7. Summary of the Twenty-First Meeting of the International Task Force for Disease Eradication (II). Available
online: http://www.cartercenter.org/resources/pdfs/news/health_publications/itfde/ITFDE-summary-
071013.pdf (accessed on 7 February 2018).
8. Wielinga, P.R.; Schlundt, J. Food Safety: At the center of a One Health approach for combating zoonoses.
Curr. Top. Microbiol. Immunol. 2013, 366, 3–17. [CrossRef] [PubMed]
9. Panorama de la Seguridad Alimentaria y Nutricional en América Latina y el Caribe. Available online:
http://www.fao.org/3/a-i7914s.pdf2017 (accessed on 6 September 2018).
10. Ortiz-Hernández, L.; Delgado-Sánchez, G.; Hernández-Briones, A. Cambios en factores relacionados con la
transición alimentaria y nutricional en México. Gaceta Médica De México 2006, 142, 181–193. [PubMed]
11. Sánchez, L.S.I.; Ibarra, L.S.V.; Bernal, V.G.; Guerrero, F.H. Transición Alimentaria en México. Razón Y Palabra
2016, 58, 568–573.
12. Peña, M.; Bacallao, J. La Obesidad en la Pobreza: Un Nuevo Reto para la Salud Pública; PAO: Washington, DC,
USA, 2000; ISBN 978-92-75-31576-7.
13. Shepherd, R. Social determinants of food choice. Proc. Nutr. Soc. 1999, 58, 807–812. [CrossRef] [PubMed]
14. Mintz, S.W.; Bois, C.M.D. The Anthropology of Food and Eating. Annu. Rev. Anthropol. 2002, 31, 99–119.
[CrossRef]
15. Dickey, M.K.; John, R.; Carabin, H.; Zhou, X.-N. Focus group discussions among the Bai in China to inform a
social marketing campaign for sanitation promotion. J. Water Sanit. Hyg. Dev. 2016, 6, 121–131. [CrossRef]
16. Tackling NCDs and NTDs: The COHESION Approach to Addressing the SDGs. Health Network Shareweb
of the Swiss Agency for Development and Cooperation SDC. Available online: https://www.shareweb.ch/
site/Health/aboutus/Pages/Contributions-January-2018/Tackling-NCDs-and-NTDs.aspx (accessed on
4 September 2018).
17. PERÚ Instituto Nacional de Estadística e Informática 2015. Available online: https://www.inei.gob.pe/
(accessed on 4 July 2018).
18. World Health Organization. Gender Mainstreaming for Health Managers: A Practical Approach; WHO: Geneva,
Switzerland, 2011.
19. Kamuya, D.M.; Catherine, S.M.; Theobald, S. Gendered negotiations for research participation in
community-based studies: Implications for health research policy and practice. BMJ Glob. Health 2017,
2, e000320. [CrossRef] [PubMed]
187
Nutrients 2018, 10, 1563
20. Padfield, M.; Procter, I. The Effect of Interviewer’s Gender on the Interviewing Process: A Comparative
Enquiry. Sociology 1996, 30, 355–366. [CrossRef]
21. IV Censo Nacional Agropecuario 2012—Base de Datos REDATAM. Available online: http://censos.inei.gob.
pe/Cenagro/redatam/ (accessed on 16 October 2018).
22. Transforming our World: The 2030 Agenda for Sustainable Development. Available online: http://undocs.
org/A/RES/70/1 (accessed on 4 September 2018).
23. Pesantes, M.A.; Diez-Canseco, F.; Bernabé-Ortiz, A.; Ponce-Lucero, V.; Miranda, J.J. Taste, Salt Consumption,
and Local Explanations around Hypertension in a Rural Population in Northern Peru. Nutrients 2017, 9.
[CrossRef] [PubMed]
24. Lipus, A.C.; Leon, J.S.; Calle, S.C.; Andes, K.L. “It Is Not Natural Anymore”: Nutrition, Urbanization, and
Indigenous Identity on Bolivia’s Andean Plateau. Qual. Health Res. 2018. [CrossRef] [PubMed]
25. Garro, L.C. Intracultural variation in causal accounts of diabetes: A comparison of three Canadian
Anishinaabe (ojibway) communities. Cult. Med. Psych. 1996, 20, 381–420. [CrossRef]
26. Pesantes, M.A.; Del Valle, A.; Diez-Canseco, F.; Bernabé-Ortiz, A.; Portocarrero, J.; Trujillo, A.; Cornejo, P.;
Manrique, K.; Miranda, J.J. Family Support and Diabetes: Patient’s Experiences From a Public Hospital in
Peru. Qual. Health. Res. 2018, 28. [CrossRef] [PubMed]
27. Vanstone, M.; Rewegan, A.; Brundisini, F.; Giacomini, M.; Kandasamy, S.; DeJean, D. Diet modification
challenges faced by marginalized and nonmarginalized adults with type 2 diabetes: A systematic review
and qualitative meta-synthesis. Chronic Illn. 2017, 13, 217–235. [CrossRef] [PubMed]
28. Vanstone, M.; Giacomini, M.; Smith, A.; Brundisini, F.; DeJean, D.; Winsor, S. How diet modification
challenges are magnified in vulnerable or marginalized people with diabetes and heart disease: A systematic
review and qualitative meta-synthesis. Ont. Health Technol Assess. Ser. 2013, 13, 1–40. [PubMed]
29. Power, E.M. Conceptualizing Food Security for Aboriginal People in Canada. Can. J. Public Health 2008, 99,
95–97. [PubMed]
30. Drewnowski, A.; Rolls, B.J. Obesity Treatment and Prevention: New Directions; Karger Publishers: Basel,
Switzerland, 2012.
31. O’Neill, M.; Rebane, D.; Lester, C. Barriers to healthier eating in a disadvantaged community. Health Educ. J.
2004, 63, 220–228. [CrossRef]
32. Irala-Estévez, J.D.; Groth, M.; Johansson, L.; Oltersdorf, U.; Prättälä, R.; Martínez-González, M.A. A
systematic review of socio-economic differences in food habits in Europe: Consumption of fruit and
vegetables. Eur. J. Clin. Nutr. 2000, 54, 706–714. [CrossRef] [PubMed]
33. Stuckler, D.; McKee, M.; Ebrahim, S.; Basu, S. Manufacturing epidemics: The role of global producers
in increased consumption of unhealthy commodities including processed foods, alcohol, and tobacco.
PLoS Med. 2012, 9, e1001235. [CrossRef] [PubMed]
34. Doak, C.M.; Adair, L.S.; Monteiro, C.; Popkin, B.M. Overweight and Underweight Coexist within Households
in Brazil, China and Russia. J. Nutr. 2000, 130, 2965–2971. [CrossRef] [PubMed]
35. Monteiro, C.A.; Conde, W.L.; Popkin, B.M. Income-Specific Trends in Obesity in Brazil: 1975–2003. Am. J.
Public Health 2007, 97, 1808–1812. [CrossRef] [PubMed]
36. Díaz, V.R. Análisis Económico de la Ingesta de Alimentos en el Perú: Informe Final; Instituto de Estudios Peruanos:
Lima, Peru, 2010.
37. Attree, P. Low-income mothers, nutrition and health: A systematic review of qualitative evidence.
Matern. Child Nutr. 2005, 1, 227–240. [CrossRef] [PubMed]
38. Committee on Preventing the Global Epidemic of Cardiovascular Disease: Meeting the Challenges in
Developing Countries. Promoting Cardiovascular Health in the Developing World: A Critical Challenge to Achieve
Global Health; National Academies Press: Washington, DC, USA, 2010; ISBN 978-0-309-14774-3.
39. Rothman, A.J.; Gollwitzer, P.M.; Grant, A.M.; Neal, D.T.; Sheeran, P.; Wood, W. Hale and Hearty Policies:How
Psychological Science Can Create and Maintain Healthy Habits. Perspect. Psychol. Sci. 2015, 10, 701–705.
[CrossRef] [PubMed]
40. Gupta, A.; Smithers, L.G.; Harford, J.; Merlin, T.; Braunack-Mayer, A. Determinants of knowledge and
attitudes about sugar and the association of knowledge and attitudes with sugar intake among adults:
A systematic review. Appetite 2018, 126, 185–194. [CrossRef] [PubMed]
41. Wardle, J.; Parmenter, K.; Waller, J. Nutrition knowledge and food intake. Appetite 2000, 34, 269–275.
[CrossRef] [PubMed]
188
Nutrients 2018, 10, 1563
42. Rasmussen, M.; Krølner, R.; Klepp, K.-I.; Lytle, L.; Brug, J.; Bere, E.; Due, P. Determinants of fruit and
vegetable consumption among children and adolescents: A review of the literature. Part I: Quantitative
studies. Int. J. Behav. Nutr. Phys. Act. 2006, 3, 22. [CrossRef] [PubMed]
43. Sorokowska, A.; Pellegrino, R.; Butovskaya, M.; Marczak, M.; Niemczyk, A.; Huanca, T.; Sorokowski, P.
Dietary customs and food availability shape the preferences for basic tastes: A cross-cultural study among
Polish, Tsimane’ and Hadza societies. Appetite 2017, 116, 291–296. [CrossRef] [PubMed]
44. Drewnowski, A. Taste Preferences and Food Intake. Annu. Rev. Nutr. 1997, 17, 237–253. [CrossRef] [PubMed]
45. Blanchette, L.; Brug, J. Determinants of fruit and vegetable consumption among 6–12-year-old children and
effective interventions to increase consumption. J. Hum. Nutr. Diet. 2005, 18, 431–443. [CrossRef] [PubMed]
46. Haß, J.; Hartmann, M. What determines the fruit and vegetables intake of primary school children?—An
analysis of personal and social determinants. Appetite 2018, 120, 82–91. [CrossRef] [PubMed]
47. AgainErick. Map Ayabaca Province. Available online: https://commons.wikimedia.org/w/index.php?
curid=3229022 (accessed on 21 August 2018).
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
189
nutrients
Review
The Role of Nutrients in Reducing the Risk for
Noncommunicable Diseases during Aging
Maaike J. Bruins 1, *, Peter Van Dael 1 and Manfred Eggersdorfer 2
1 Nutrition Science & Advocacy, DSM Nutritional Products, CH-4303 Kaiseraugst, Switzerland;
[email protected]
2 University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; [email protected]
* Correspondence: [email protected]; Tel.: +41-618-158-761
Keywords: chronic disease; noncommunicable disease; nutrient inadequacies and deficiencies; nutrient
interventions; public health; musculoskeletal disorders; dementia; eye disorders; cardiovascular disease
1. Introduction
Globally, significant gains in human longevity have been made in the last couple of decades as
evidenced by an average 5.5-year increase in life expectancy between 2000 and 2016 [1]. In many
countries average life expectancy currently exceeds 80 years [1]. These longevity gains have come at a
cost, however, with the most obvious being an increase in age-related diseases [2]. Noncommunicable
diseases (NCDs) such as diabetes, musculoskeletal disorders, cardiovascular diseases, neurological
disorders, and cancers increase with age, and place a burden on individuals and healthcare systems [3].
Supporting healthy aging by preventing NCDs is a major priority for agencies such as the World
Health Organization (WHO) and United Nations [4,5].
The WHO estimates that NCDs contribute 1.6 billion disability-adjusted life-years (DALYs)
to the global burden of disease and identified unhealthy diets and physical inactivity are among
the main modifiable risk factors, together with excess alcohol and tobacco use [6]. Nutrition is
an important determinant of human health by providing the essential building blocks for growth,
development, and maintenance of a healthy status throughout life [7,8]. In this context, the co-existing
burdens of undernutrition and overnutrition represent a paradigm shift for health authorities
requiring appropriate dietary management recommendations [9]. Modern lifestyles and easy access to
high-energy, low-nutrient rich foods are considered part of the problem [3,10–12]. For example, the
economic costs of unhealthy diets and low physical activity in the EU were calculated to be €1.3 billion
per year [13].
Currently, health authorities mainly target problems associated with obesity and cardiovascular
diseases by focusing on reducing excess intake of calories, sugar, salt, and saturated fats. However,
the importance of a positive message associated with promoting adequate nutrient intake as part of a
balanced diet should not be overlooked [4]. There is considerable variation in the consumption of food
items that need to be encouraged and food items which should be limited, both between and within
different countries. This was reflected in a recent study in European countries showing suboptimal
nutrient-density of diets and significant proportions of the population consuming excess amounts
of salt, sugar and saturated fat, as well as significant proportions of the population not meeting the
required or adequate intakes for various essential nutrients (Table 1) [12].
Table 1. Percentage of adults with nutrient intakes meeting the estimated average requirement (EAR)
or adequate intake (AI) or exceeding the maximum reference value (MRV) [12].
The health consequences of poor nutrition almost certainly accumulate over the lifespan of the
individual. Table 2 presents information regarding some of the more frequently reported chronic
clinical signs associated with certain vitamin and mineral deficiencies in older adults. Clinical signs
and symptoms are mostly nonspecific and difficult to diagnose. During the aging process, a number
of changes occur, such as increased medication use, reduced food intake due to lower food appeal,
191
Nutrients 2019, 11, 85
and compromised nutrient absorption. These complex changes prevent elderly persons from meeting
their nutritional requirements. This consequently leads to increased risk of malnutrition, frailty, and
reduced quality of life (QoL) [14–17].
Health policies and interventions to improve dietary intake at the population level are essential
to reverse the global trend towards unhealthy dietary patterns and physical inactivity. However,
more individualized approaches may be needed to address persistent nutritional gaps and prevent
future morbidity in high-risk groups such as the older population [19,20]. An estimated 5% to 10%
of community-dwelling adults >70 years of age are undernourished; this proportion rises to 30% to
65% among institutionalized elderly patients. In the older adult population, nutrients of concern
include, among others, calcium, vitamin D, and vitamin B6 and B12 [15,20,21]. Vitamin D deficiency
was found not only to be a problem in the elderly, but to be a global problem common across all
age ranges [22]. Genetic variations play a role in dietary response and genetic variations also play a
role in determining nutrient status and requirements [23]. By understanding the genome that affects
the individual requirements for and response to nutrition, diseases of aging that have a nutritional
component can be addressed in a targeted way.
In general, activities endorsing lifestyles that include healthy diets have usually focused on
limiting the consumption of salt, sugar, and saturated fat. However, focus on the need to meet
adequate dietary intake of essential nutrients through a healthy diet is considered equally important.
This review focuses on the role of nutrients in the risk reduction of NCDs in disorders prevalent
in the aging population and for which the societal costs are substantial [24]. The evidence for a
connection between NCDs and inadequate intake or status of specific nutrients such as vitamins,
192
Nutrients 2019, 11, 85
carotenoids, omega-3 fatty acids, and other bioactive substances is reviewed. Furthermore, the impact
of interventions aimed at correcting these inadequacies will be discussed.
193
Nutrients 2019, 11, 85
Figure 1. Europe map of vitamin D deficiency in older adults (mean 25(OH)D status (nmol/L) in adults
aged ≥50 years) (based on: [39]).
The role of vitamin D and related analogues, with or without calcium, for preventing bone
fractures in post-menopausal women and older men was the subject of a Cochrane review [25].
This systematic review included 53 trials and 91,791 older women or men aged over 65 years from
community, hospital, and nursing-home settings, and assessed the impact of vitamin D for the
prevention of hip or other types of fracture. In this analysis vitamin D alone did not appear to have a
significant effect on fracture prevention, whereas vitamin D in combination with calcium significantly
reduced the likelihood of hip fractures (P = 0.01), non-vertebral fractures and any type of fracture.
Hip fracture incidence was particularly reduced in institutionalized residents with a risk reduction of
25%. In a separate systematic review (30 randomized controlled trials (RCTs) involving 5615 subjects
of mean age 61 years), vitamin D supplementation was shown to produce a small but statistically
significant improvement in global muscle strength. The most benefit was observed in individuals
who presented with a 25-hydroxyvitamin D level below 30 nmol/L (compared with those with a level
≥30 nmol/L), and in subjects aged 65 years or older [29].
The economic value of vitamin D supplementation has been the subject of several health economic
evaluations showing that increasing vitamin D status through supplementation or fortification can
prevent fractures and improve QoL in older adults and associated health care costs [40–44].
194
Nutrients 2019, 11, 85
play critical roles in extracellular bone matrix mineralization. Less is known about vitamin K and
health, but there is growing evidence suggesting a synergistic effect between vitamins K and D in
bone [40]. A number of studies reported that vitamin K is essential for optimization of bone health
with benefits in preventing bone loss [41]. Vitamin K2 supplementation combined with vitamin D
and calcium for 2 years in a randomized placebo-controlled trial resulted in a significant increase
in bone-mineral density and content in older women [42]. In another recent RCT it was found that
combined vitamin K2 , vitamin D and calcium supplementation for 6 months increased the bone mineral
density of lumbar 3 spine vertebra compared to vitamin D and calcium alone in postmenopausal
Korean women [43].
Current research investigating the effect of vitamin D alone or in combination with other nutrients
on fractures, cardiovascular disease, diabetes, cognitive function, immunity, and other benefits
is ongoing in two large scale studies in older adults (DO-HEALTH in Europe, FIND in Finland).
In addition, many research groups engage in basic science to study the combined action of vitamin
K2, vitamin D, and calcium, and their function on the molecular level. More studies are required that
target vitamin D supplementation in combination with other nutrients such as calcium and vitamin K
where it is needed, in people with vitamin D deficiency or older people, who are more likely to be frail
in institutionalized residents.
3. Cognitive Disorders
Dementia is a term that describes a decline in cognitive abilities including memory, and reduction
in a person’s ability to perform everyday activities [44]. Dementia prevalence is forecast to increase
dramatically in future years [45]. At present about 50 million people have dementia worldwide, and
this is projected to reach 80 million by 2030 and 150 million by 2050 [46]. Alzheimer’s disease (AD)
is the most common form of dementia in people aged >60 years, accounting for 60–70% of the total
number of cases and is the major focus of this section [46]. Vascular dementia is the second most
common cause of dementia with at least 20% of dementia cases.
Alzheimer’s disease is a complex, progressive, multifactorial, neurodegenerative disease [24,45].
The presentation generally involves progressive memory loss, impaired thinking, disorientation, and
changes in personality and mood. As the disease advances there is a marked reduction in cognitive
and physical functioning [47,48]. Genetic factors account for about 70% of the risk contributing to AD,
while modifiable factors related to general health and lifestyle may also be involved [48]. Risk factors
for vascular dementia are predominantly modifiable and of vascular origin (including hypertension,
diabetes mellitus, dyslipidemia, and the metabolic syndrome). Managing non-genetic risk factors
effectively may provide opportunity to prevent and treat the progressive cognitive decline associated
with AD [47]. The focus of this section of the review is on nutritional status and its potential role in AD.
195
Nutrients 2019, 11, 85
little to no effect with respect to preventing cognitive decline [51]. Notably, individuals with high
homocysteine levels had significant cognitive decline and B-vitamins were found to improve memory
only in this subgroup [52]. Also, evidence exists that in elderly subjects with an increased risk of
dementia, B-vitamins can slow brain shrinkage over two years by up to 30% [53]. At present, the
evidence is insufficiently compelling to support B-vitamin supplementation to prevent cognitive
decline and dementia.
Dehydroascorbic acid, a metabolite of vitamin C, is a potent antioxidant, an essential cofactor
in many enzymatic reactions, and has a role in metabolizing cholesterol. Large dietary surveys
undertaken in Germany, the Netherlands, the UK, and the US indicated inadequate vitamin C intake in
up to half of respective populations [54]. As with vitamin E, however, studies of vitamin C in patients
with AD have been equivocal. The overall conclusion of the Team of Alzheimer Drug Discovery
Foundation is that maintaining adequate levels of vitamin C through diet may offer more benefit
than supplementation.
The metabolically active form of vitamin D, 1,25-dihydroxy vitamin D, binds to vitamin D
receptors that are present in brain regions involved in cognition. Proposed mechanisms for the
protective effects of vitamin D against cognitive decline include clearing Aβ peptide, regulating
intraneuronal calcium, anti-inflammatory activity, antioxidative activity, preventing and reducing
ischemia, and regulating choline acetyltransferase neurotrophic agents. There is strong evidence that
patients with AD have lower vitamin D status than healthy controls and that lower vitamin D status is
associated with increased risk of developing dementia. Although vitamin D supplementation alone
was insufficient to improve cognition in a study of patients with newly diagnosed AD, the Vitamin D
Council recommends that middle-aged and older adults maintain vitamin D blood levels in the higher
range of normal (175–200 nmol/L; 70–80 ng/mL).
Vitamin E possesses antioxidant properties which may prevent hyperphosphorylated tau protein
dysfunction and has been shown to reduce the rate of Aβ protein-induced death in cultures of
hippocampal and cortical cells [55]. The γ-tocopherol isomer is particularly effective in scavenging
free radicals that cause inflammation [55]. By scavenging Aβ protein-associated free radicals, vitamin
E may have a neuroprotective effect during oxidative stress. However, while promising in principle,
studies of α-tocopherol supplementation in patients with AD have not been convincing and dietary
vitamin E may provide greater protection against age-related neurodegenerative conditions.
Brain membranes are composed mainly of phospholipids, predominantly the LCPUFAs DHA and
arachidonic acid (ARA). DHA has multiple actions in maintaining neurological function. Lower plasma
DHA has been associated with cognitive decline in both healthy elderly people and AD patients [56].
Investigations to date of the therapeutic potential of supplementation or higher dietary intake of DHA
in patients with AD have produced conflicting results, although it is possible that cognitive impairment
in the study populations was already resistant to intervention. Given the essential role of DHA in the
human brain, a general recommendation to maintain an adequate dietary intake of DHA throughout
adulthood appears to be a reasonable approach to prevent cognitive decline.
Due to a high concentration of oxygen free radicals relative to antioxidative defenses in the brain,
it may be especially vulnerable to oxidative stress and consequent damage to lipids and proteins [57].
AD is also associated with lower levels of acetylcholine in the hippocampal and cortical regions,
resulting in memory impairment. Fruits, vegetables, coffee, and cereal grains contain high levels of
polyphenols. In vitro and animal studies of specific dietary flavonoids and plant extracts have shown
reduction of oxidative stress and inhibition of acetylcholinesterase, suggesting a dual protective role
for polyphenols against cognitive decline and dementia [57]. Although no conclusions can be drawn
about the relative benefits of any particular plant polyphenol over another, the findings emphasize the
importance of life-long consumption of foods with high content of these antioxidants.
Trials that have reported no effect of nutrients generally included older adults who were unlikely
to have a marked decline in cognitive function [52]. Trial design should consider including older
individuals with deficiencies that increases their risk of cognitive decline, and who may benefit from
196
Nutrients 2019, 11, 85
nutrition intervention. Sensitive assessment tools and surrogate markers are needed that examine
specific aspects of brain structure and function such as neuroimaging techniques to advance the
understanding of nutrition interventions that could reduce the risk of dementia.
4. Eye Disorders
Impairments of the essential senses of vision and hearing are the second-leading cause of years of
lived with disability [58]. The most common causes of vision loss among the elderly are age-related
macular degeneration, glaucoma, cataracts, and diabetic retinopathy [59]. Aging is the greatest risk
factor associated with the development of age-related macular degeneration, but also environmental
and lifestyle factors such as smoking, oxidative stress, and diet may significantly affect the risk [60].
Recent studies suggest that increasing exposure to blue light emitted by electronics and energy-efficient
lightbulbs over time could lead to damaged retinal cells which on the long-term can cause vision
problems like age-related macular degeneration [61]. Eye health problems in the ever-increasing aging
generation, and “exposure to blue light” may result in a new NCD.
Carotenoids have a range of functions in human health and, in particular, there is evidence that
they have beneficial effects on eye health [62]. Two dietary carotenoids, lutein and zeaxanthin are
macular pigments found in the human retina [63]. Macular pigment has local antioxidant properties
and absorbs high energy, short wavelength blue light protecting the retina from photochemical
damage [64]. Macular pigment can neutralize ROS, protect against UV-induced peroxidation, and
reduce the formation of lipofuscin and associated oxidative-stress induced damage [63]. Thus, the
carotenoids provide potential benefits for ocular function and health.
Individuals who have low macular pigment optical density levels (0.2 or lower) may benefit from
supplementation with lutein/zeaxanthin which can help increase macular pigment optical density
levels [65–72]. For retinal protection, macular pigment optical density values of 0.4 to 0.6 are desirable,
especially in older adults [73]. Dietary intake of lutein and zeaxanthin may differ with age, sex,
and ethnicity. Across all age groups the intake of lutein is higher than for zeaxanthin and this is
independent of sex and ethnicity. In addition, lower zeaxanthin to lutein ratios are reported for groups
at risk of age-related macular degeneration (e.g., the elderly and females) [74]. A number of studies,
including some in healthy subjects, have demonstrated that lutein/zeaxanthin supplementation
can improve visual performance, including contrast sensitivity, glare tolerance and photo stress
recovery [65–72,75,76].
Age-related macular degeneration is an increasing problem among the elderly and studies of the
effects of lutein/zeaxanthin supplementation have produced mixed results. However, important data
were provided by secondary analyses of the large Age-Related Eye Disease Study 2 (AREDS2) [77,78].
This randomized trial investigated the effect of adding lutein/zeaxanthin 10/2 mg, DHA (350 mg)
+ EPA (650 mg), or both to the original AREDS2 formulation (vitamin C, vitamin E, β-carotene,
zinc, and copper) or to variations of this formulation (excluding β-carotene and/or with reduced
zinc). Participants (n = 4203) were followed for a median 5 years. The primary analysis found
no additional beneficial or harmful effect for lutein/zeaxanthin and/or omega-3 fatty acids on
progression to late age-related macular degeneration compared with the original AREDS1 formula
using β-carotene instead of lutein/zeaxanthin. However, a prespecified secondary analysis found a
significant 26% risk reduction for progression to advanced age-related macular degeneration when
comparing lutein/zeaxanthin supplementation with no lutein/zeaxanthin supplementation in the
quintile with the lowest dietary intake of these two carotenoids (median 0.7 mg/day), as indicated
by a hazard ratio of 0.74 (95% confidence interval 0.59–0.94, p = 0.01). In addition, a post hoc analysis
showed that lutein/zeaxanthin (excluding β-carotene) was more effective than the original AREDS
formulation containing β-carotene but no lutein/zeaxanthin for reducing progression to advanced
age-related macular degeneration (hazard ratio 0.82, 95% CI 0.69–0.96, p = 0.02) [77].
There is also some evidence suggesting there is a relationship between lutein/zeaxanthin
status and the risk of developing nuclear cataracts [79], and in the AREDS2 trial the addition of
197
Nutrients 2019, 11, 85
lutein/zeaxanthin supplementation reduced the risk of cataract surgery in the quintile with the lowest
dietary intake of these carotenoids (hazard ratio 0.68, 95% CI 0.48–0.96, p = 0.03) [80].
If the AREDS2 complex (i.e., vitamin C and E, zinc, copper, lutein/zeaxanthin and omega-3 fatty
acids) was used by all adults aged >55 years, it has been estimated this would result in an average of
about 1 million avoided age-related macular degeneration and cataract events per year in the USA
(based on a risk reduction of 23.6% for age-related macular degeneration and 16.2% for cataracts).
This would result in a net annual cost saving of US$1.2 billion, mostly as a consequence of reduced
healthcare expenditure [81]. Establishing intake recommendations for lutein is an important step
forward to support optimal visual performance and reduce the risk of age-related macular eye disease
in the general population. This would be a relevant contribution to public health in the face of a
globally aging population.
Future studies may include additional assessments of the relationship between macular pigment
and different genotypic and phenotypic forms of age-related macular degeneration, the optimum
dosages of lutein, zeaxanthin, and the possible effects when combined with other nutrients.
5. Cardiovascular Disease
Despite the global decline in cardiovascular mortality, cardiovascular diseases remain the leading
cause of morbidity and mortality, contributing to escalating health care cost [82]. Cardiovascular aging
progresses over decades, influenced by risk factors such as tobacco use, poor physical activity and diet,
resulting in hypertension, dyslipidemia (high triglycerides and lower HDL), elevated fasting blood
glucose, and central obesity [83]. Cardiovascular disease is the major clinical problem in the older
population, with 68% of adults 60–79 years having cardiovascular disease and this increases to 85%
after the age of 80 years [84].
Good nutrition plays an important role in delaying the progression of cardiovascular
disease [85,86]. The adverse effects of excess intakes of saturated and trans fats, cholesterol, added
sugars, and salt in relation to cardiovascular disease progression has been relatively well-established
whereas the effect of addressing inadequate essential nutrients is less well-known. Older adults are
highly susceptible to undernutrition due to the various physiological and socioeconomic factors [87].
In contrast to overnutrition, the potential of addressing undernutrition to optimize cardiovascular
health in older adults has received inadequate attention [88]. Evidence for nutrition in reducing the risk
for cardiovascular aging mostly derives from epidemiological studies, whereas fewer interventions
studies have been performed. The RCTs addressing cardiovascular disease generally have included, but
not exclusively, older adults, not allowing generalizability of results to typical older adults. The authors
have therefore focused on nutrition interventions addressing cardiovascular aging progress, not
restricted to elderly.
5.1.1. Diets
Lifestyle changes, including dietary modifications, are recommended as part of the management
strategy to improve lipid profiles and reduce the risk of cardiovascular disease [89–91]. The primary
emphasis of dietary interventions has been on changing dietary macronutrient and salt composition.
The effect of improving micronutrient-richness of the diet in cardiovascular disease control has been
less-well studied. A diet rich in fruits, vegetables, wholegrains, legumes, nuts, fish, poultry, and low-fat
dairy products, and limited consumption of red meat, saturated fat, and added sugar is advocated,
mostly based on positive associations with cardiovascular health [89–91]. Dietary patterns that follow
these principles include the Dietary Approaches to Stop Hypertension (DASH) diet, a diet rich in fiber,
protein, magnesium, calcium, and potassium, and low in total and saturated fats, which has been
shown to reduce low-density lipoprotein (LDL)-cholesterol levels [91], and the Mediterranean diet,
which has been shown to reduce the risk for cardiovascular disease in both primary and secondary
198
Nutrients 2019, 11, 85
settings [92,93]. Regression of coronary artery atherosclerosis has been demonstrated with a program
of intensive lifestyle changes that included a vegetarian diet, exercise, and smoking cessation [94].
In addition to dietary interventions, there has been research into the effects of individual nutrients.
While the evidence for some of these is limited, several interesting findings have been published.
5.1.2. Vitamin D
Low vitamin D has been associated with cardiovascular disease in a number of studies [95].
Few studies have been targeting low vitamin D specifically in the older population. In one study with
post-menopausal women randomized to Vitamin D3 2500 IU or placebo, daily for 4 months, vitamin
D supplementation had no effect on endothelial function, arterial stiffness, or inflammation [96].
Results of a meta-analysis of RCT with older adult participants (≥60 years) suggested that vitamin D
supplementation might protect against cardiac failure but not against MI or stroke [97]. The recent
results of the VITAL trial indicate that daily supplementation of 2000 IU vitamin D did not reduce the
occurrence of cardiovascular events in adults aged ≥50 years [98].
5.1.3. B-Vitamins
B-vitamins have been the subject of substantial research because of their established effects on
normalizing homocysteine levels, an important risk factor for cardiovascular disease. Figure 2 shows
the risk factors including B-vitamin shortages and pathogenetic mechanisms for the effect of high
homocysteine on cardiovascular disease.
↑ +RPRF\VWHLQH
(QGRWKHOLDOFHOO %ORRG
9DVFXODU VPRRWK
PXVFOHFHOO
3UROLIHUDWLRQ
0DWUL[ GHJUDGDWLRQ
↑ 5LVNDWKHURVFOHURVLV
↑ 5LVNDWKHURWKURPERVLV
Figure 2. Risk factors and mechanisms for high homocysteine in cardiovascular disease. MTHFR:
methylenetetrahydrofolate reductase, CBS: cystathionine beta-synthase.
199
Nutrients 2019, 11, 85
Particularly the B-vitamins have been investigated for their potential cardiovascular benefits due
to their established lowering effect on homocysteine levels, a marker for cardiovascular disease risk,
including ischemic stroke. A meta-analysis of 19 RCTs of B vitamins (including folic acid, vitamin B6,
vitamin B12, and B-complex vitamins) found significant reductions in homocysteine levels, however,
no significant effect of vitamin B supplementation on rates of cardiovascular disease, coronary heart
disease, myocardial infarction, cardiovascular death, or all-cause mortality whereas vitamin B reduced
the risk of stroke by 12% [99]. Another meta-analysis of 26 RCTs found that folic acid supplementation
significantly reduced the risk of stroke 7% [100].
There are various reasons for elevated blood homocysteine levels; most people have mild to
moderately elevated serum homocysteine levels due to inadequate intake of folate, vitamin B6 , or
vitamin B12 from the diet, which is reversible when intake of these vitamins is increased. Another
cause are genetic variants of methylenetetrahydrofolate reductase (MTHFR) and methionine synthase
reductase (MTRR) that are associated with elevated homocysteine levels. Elevated homocysteine
levels are a risk factor for developing blood clots in the vasculature and have been implicated
in the pathogenesis of atherosclerosis and deep vein thrombosis [101]. Given that vitamin B
supplementation is associated with normalization of elevated plasma homocysteine levels, many
studies have investigated whether these vitamins may decrease the risk of cardiovascular diseases.
Huang and colleagues undertook a meta-analysis (19 RCTs and 47,921 participants) evaluating the
effects of B vitamin supplementation (search terms: folic acid, folate, vitamin B6 , vitamin B12 , and B
vitamins) on plasma homocysteine levels and cardiovascular and all-cause mortality [99]. The overall
relative risk of a clinical outcome, versus placebo, was 0.98 for cardiovascular disease, 0.98 for CHD,
0.97 for MI, 0.97 for cardiovascular death and 0.88 for stroke; and homocysteine levels were decreased
in all RCTs. Thus, B vitamin supplementation had a significant protective effect for stroke, but not for
any other cardiovascular risk. A more recent meta-analysis of folic acid supplementation (30 RCTs,
82,334 participants) estimated a 10% lower risk of stroke and a 4% lower risk of overall cardiovascular
disease compared with controls [102]. The greatest benefit for cardiovascular disease was observed in
individuals with lower plasma folate levels at baseline and without pre-existing cardiovascular disease
(p = 0.006 for both). While patients with a cardiovascular disease history responded to B-vitamins with
normalization of homocysteine levels, those with the MTHFR 677C > T genotype were less responsive
and may have greater folate requirements than do their counterparts [103].
5.1.4. Vitamin K
Vitamin K plays an important role in anticoagulation and may overcome the detrimental side
effects associated with vitamin K antagonists such as warfarin. Vitamin K may also help to prevent
vascular calcifications, especially in patients on warfarin [104].
200
Nutrients 2019, 11, 85
reduction in cardiovascular risk only among higher risk populations, such as those with elevated
triglyceride levels (relative risk: 0.84, 95% CI 0.72–0.98) or elevated LDL-cholesterol levels (relative
risk 0.86, 95% CI 0.76–0.98) [108]. Another recent Cochrane meta-analysis of RCTs found that omega3
LCPUFAs reduced cardiovascular events in the main analysis (relative risk: 0.93, 95% CI 0.88–0.97), but
the result was not maintained in sensitivity analyses [109]. The failure of some trials to show effects of
omega-3 LCPUFA on cardiovascular disease was explained by an insufficiently high omega-3 LCPUFA
dose and/or too high omega-3 LCPUFA baseline status to demonstrate effects [110]. RCTs evaluating
the effects of omega-3 LCPUFAs on cardiovascular morbidity and mortality generally enrolled a broad
range of ages while only few RCTs have focused specifically on older adults. The Alpha Omega Trial
that included 60–80-year-olds with previous MI and at least 50% on medication found no significant
effect of approximately 400 mg of omega-3 LCPUFA on cardiovascular events [111]. In the AREDS2
study, 1 g omega-3 LCPUFA given in addition to a standard Vitamin C, Vitamin E, beta-carotene,
zinc oxide, and cupric oxide supplement for 6 months to participants between 50 and 85 years had
no effect on cardiovascular outcomes [112]. The recent results of the VITAL trial showed that in
adults aged ≥50 y daily consuming 840 mg of omega-3 LCPUFA lowered the risk of heart attack by
28%, of fatal heart attack by 50% without significant effect on stroke or cardiovascular deaths [98].
The most pronounced benefits on major cardiovascular event reduction were found in participants
who reported low fish intake at baseline. A recent meta-analysis of RCTs found that omega-3 LCPUFA
supplementation caused a small, but significant, reduction in heart rate (−2.23 bpm, 95% CI −3.07 to
−1.40) [113], which is considered a risk factor for cardiovascular morbidity and mortality [114].
5.1.6. Antioxidants
Inflammation and oxidative stress appear to be key drivers for a number of cardiovascular
diseases and the metabolic syndrome [115,116]. Whereas observations studies suggest that antioxidant
nutrient such as β-carotene and vitamin E are associated with lower cardiovascular disease, the
data of RCTs on antioxidant supplements failed to confirm a significant benefit of antioxidants on
atherosclerotic cardiovascular disease. For instance, supplementation with the antioxidant nutrients
vitamin E, β-carotene, and vitamin C, had no significant effects on cardiovascular outcomes [117].
5.1.7. Vitamin E
A key attribute of vitamin E (a combination of 8 distinct tocopherol/tocotrienol isoforms) is its
antioxidant activity and, as a consequence, its ability to protect poly-unsaturated fatty acids (PUFAs),
lipoproteins, and cell membranes from oxidative damage [118]. Vitamin E has been extensively
investigated for its potential to prevent cardiovascular disease events. Nevertheless, RCTs with
vitamin E had mixed results on various cardiovascular disease endpoints. In the Women’s Health
Study, intake of 600 IU of vitamin E on alternate days in apparently healthy women non-significantly
reduced the risk for cardiovascular events by 7% and significantly reduced the risk for cardiovascular
death by 24% [119]. And among women ages 65 and older, vitamin E supplementation reduced the
risk of major cardiac events by 26% [119]. Data from the same Women’s Health Study suggested
that supplementation with vitamin E may reduce the risk of venous thromboembolism in women,
particularly in those with a prior history or genetic predisposition [120]. RCTs that retrospectively
analyzed the data for the effect of vitamin in E in subgroups of patients with this these genotypes
sometimes showed that these patients are more responsive to vitamin E supplementation [121,122].
5.1.8. Phenolics
Phenolic compounds are bioactive compounds found in plants, and there is evidence that some
may be helpful for reducing cardiovascular risk factors [116]. Flavonoids are polyphenolic compounds
found in fruits, vegetables, tea, and red wine [116]. Amongst the flavonoids, there is some evidence
that flavonols (specifically quercetin) may be effective at reducing blood pressure in hypertensive
patients; however, no effects on other cardiovascular disease risk markers such as endothelin, oxidative
201
Nutrients 2019, 11, 85
stress, or lipid profiles were found [116]. Although an early meta-analysis found that consumption of
flavonols was associated with a lower rate of cardiovascular disease [123], a more recent meta-analysis
and a systematic review do not support such an effect [116,124]. Amongst other phenolic compounds
which might have beneficial cardiovascular effects, resveratrol is a stilbene found in grape skin, red
wine, and peanuts [116]. A systematic review found that resveratrol was associated with reductions
in total cholesterol, LDL-C, triglycerides and apolipoprotein B in a range of patients, including those
with ischemic heart disease [116]. Resveratrol also reduced inflammatory and fibrinolytic biomarkers
in patients with ischemic heart disease [116].
Nutrients have been investigated for their effect on cardiovascular disease progress and as such,
outcomes. B-Vitamins reduced homocysteine levels, a risk factor of cardiovascular disease, without
significant effects on cardiovascular disease events except for the reduced risk for stroke, which was also
reduced by folic acid supplementation. Flavonoids and omega-3 LCPUFA also reduce cardiovascular
disease risk factors although evidence on cardiovascular outcomes is mixed. Possible explanations
include that patients enrolled in the RCTs were already at high risk of cardiovascular disease and
on concomitant medications, with little opportunity for nutrition to reverse the progress. Individual
nutrients like vitamin D, vitamin E and omega-3 LCPUFA on cardiovascular disease prevention
have shown mixed effects. Nutrition interventions have focused mostly on primary prevention of
cardiovascular aging in broad age groups and less on older adults. Recruited participants in the
RCTs were often at high risk of cardiovascular risk factors or preexisting disease, a modest effect of in
patients that already have heart disease or are at high risk of heart disease may be masked by effects
of medication.
5.2. Hypertension
Hypertension is a major public health concern given its link to serious cardiovascular events
such as stroke and ischemic heart disease, the leading causes of worldwide mortality [6]. It has been
estimated that hypertension is responsible for approximately 40% of cardiovascular deaths. By the
year 2025 almost 30% of the global population will be diagnosed with high blood pressure, with 25%
of these cases occurring in developing countries [125]. Hypertension rises dramatically with aging due
to longer exposure to age-associated alterations in vascular function and structure and cardiovascular
risk factors [126].
Hypertension is a multifactorial disease with lifestyle factors such as physical activity, smoking
and drinking habits, diet, bodyweight, and anxiety playing a predominant role. Management of
these is the first step to achieving adequate blood pressure control. Indeed, it has been reported
that two lifestyle modifications can help improve blood pressure control and decrease the number of
cardiovascular outcomes [127].
5.2.1. Diets
In the current healthcare environment, lifestyle changes involving a healthy diet and increased
physical activity are considered pivotal in the management of hypertension. Diets with a high
nutritional value, such as the traditional Mediterranean diet, DASH and the OmniHeart (a variation
of DASH with increased levels of protein) diets, can be important steps on the path to weight loss,
lowering blood pressure, and prevention of hypertension [125]. The benefits of the DASH diet on
blood pressure were reported in a RCT with all participants receiving graded amounts of sodium
(high, intermediate, low). There were dose-response decreases in systolic and diastolic blood pressures,
and age-related increases in blood pressure were blunted [128]. Both the DASH diet and low sodium
markedly decreased blood pressure, and the combined effect was even greater. Findings of the DASH
study also provided additional support that the sodium-to-potassium ratio is stronger associated
with blood pressure outcomes than either nutrient alone among prehypertensive and hypertensive
adults combined. These findings were later confirmed by a systematic review showing that the
202
Nutrients 2019, 11, 85
sodium-to-potassium ratio appears to be more strongly associated with blood pressure outcomes than
either nutrient alone in hypertensive adults [129].
In addition to dietary control there has been research into the effects of other nutrients, including
vitamins, on blood pressure and hypertension. While the evidence for some of these is limited a
number of interesting findings have been published.
5.2.4. Vitamin C
In short-term studies, vitamin C supplementation reduced systolic and diastolic blood pressure.
Long-term trials on the effects of vitamin C supplementation on BP and clinical events are needed
longer-term trials assessing the effects of vitamin C supplementation on blood pressure and clinical
events in patients with hypertension would seem to be worthwhile [133]
5.2.5. Vitamin D
In a study involving 283 hypertensive patients, vitamin D3 (cholecalciferol) produced a modest but
statistically significant reduction in systolic blood pressure compared with placebo after 3 months [134].
There was no significant effect on diastolic blood pressure.
5.2.6. Flavonols
Flavanols have also been found to lower blood pressure, and there is some evidence suggesting
that they improve endothelial function in patients with ischemic heart disease, but additional studies
are needed [116].
The evidence for nutrients and blood pressure is convincing for lowering sodium and
sodium-to-potassium ratio. Flavanols vitamin C and D may have modest significant effects on blood
pressure lowering.
5.3. Diabetes
Type 2 diabetes has become a global health-related pandemic which is forecast to rise from 425 to
almost 630 million by 2045 [135]. In developing countries, the forecasted increase is more alarming,
particularly in regions which are more rapidly adopting a Western lifestyle. The direct financial burden
on healthcare systems and society is huge, as are the indirect costs from loss of work attendance.
Intensive lifestyle modification, e.g., personalized nutrition and physical activity programs, with
the goal of improving glycaemia and losing excess body weight should be the mainstay of initial
management in individuals with prediabetes [136].
203
Nutrients 2019, 11, 85
5.3.1. Vitamin D
Observational studies have highlighted a link between vitamin D deficiency and type 2 diabetes,
as well as possible future cardiovascular events, whereas results from interventional studies have not
been so conclusive [137]. A recent meta-analysis [137] including a total of 20 RCTs and 2703 participants,
found that vitamin D supplementation was associated with elevated serum vitamin D levels and
significantly decreased insulin resistance. Changes in other parameters such as fasting blood glucose
and hemoglobin A1c (HbA1c) were relatively small and did not achieve statistical significance [137].
In a pilot study in 60 patients with co-existing type 2 diabetes and hypovitaminosis D, vitamin D
improved vitamin D status and several parameters associated with glycemic control such as HbA1c,
mean fasting plasma glucose, and mean post-prandial plasma glucose [138]. In addition, vitamin D in
the study lowered LDL cholesterol levels, systolic blood pressure and diastolic blood pressure.
5.3.2. Vitamin E
Diabetes patients with the haptoglobin 2-2 genotype have elevated risk of cardiovascular disease
events. The haptoglobin 2-2 genotype has inferior antioxidant properties as compared with other
haptoglobin types resulting in elevated levels of oxidative stress, an atherogenic profile and an
increased risk of cardiovascular disease events compared with other Hp genotypes [139]. The RCTs
in diabetes patients that retrospectively analyzed the data for the effect of vitamin in E found
that administration of vitamin E lowered the risk of cardiovascular disease events by 34% and
cardiovascular-related mortality by 53% among patients with the haptoglobin 2-2 genotype [140].
5.3.4. Vitamin K
To assess whether vitamin K is a risk factor for the development of type 2 diabetes mellitus,
Beulens and colleagues analyzed a cohort of 38,094 Dutch men and women over a 10-year period [142].
The study showed that both vitamin K1 and vitamin K2 intake were associated with a reduced risk
of type 2 diabetes mellitus. For vitamin K1 the risk reduction occurred at the higher levels of intake,
whereas for vitamin K2 a linear inverse association was established. In older men with diabetes
receiving vitamin K1 supplementation for 36 months, vitamin K1 significantly improved insulin
sensitivity [143].
5.3.5. Chromium
Chromium plays a role in insulin metabolism by activating oligopeptide low-molecular-weight
chromium (LMWCr)-binding substance and activating insulin-dependent kinase activity. A meta-analysis
of the efficacy of chromium supplementation suggest that there is available evidence for chromium on
glycemic control in patients with diabetes [144].
204
Nutrients 2019, 11, 85
Studies in diabetes patients showed that vitamin D supplementation can improve serum vitamin
D levels and significantly decrease insulin resistance. Currently, a large multicenter RCT is ongoing in
the US (Vitamin D and Type 2 Diabetes Study; D2d), hypothesizing that vitamin D will enhance insulin
production, glucose processing and glycemic profiles. Subgroup analyses show that vitamin E may be
promising in reducing the rate of cardiovascular events among diabetes patients with haptoglobin
2-2 genotype who are at increased risk of cardiovascular events. The evidence for omega3 LCPUFA
supplementation on fasting plasma glucose or HbA1C is less conclusive but omega-3 LCPUFA have
promising effects for reduction of triglycerides. The evidence for chromium in glycemic control
is emerging.
6. Conclusions
Inadequate or even deficient nutrient intake and status is still widely prevalent at global level
and, although generally underacknowledged, is a main risk factor for NCDs [20]. Nutrient surveys
indicate that the aging population is at particular risk for poor nutrient intake and status, which may
result in increased risk for chronic fatigue, and cardiovascular, cognitive, and neuromuscular disorders
in older adults. The present paper reviews the evidence for the role of various nutrients in modifying
the risk of development of NCDs throughout aging.
Inadequate vitamin D, calcium and vitamin K intake and status are generally reported in the
aging population and have been associated with musculoskeletal disorders, such as increased bone
fracture risks. Increased vitamin D in combination with increased calcium and possibly also vitamin K
may reduce the risk for hip fractures, thus beneficially impacting musculoskeletal health.
Inadequate B vitamins intake and status, in particular folic acid, vitamins B6 and B12 , have been
associated with age-related cognitive decline, while supplementation has been reported to improve
cognitive performance. Similarly, evidence has been reported for vitamin C, D, and E, as well as
omega-3 LCPUFAs (e.g., DHA) to slow down dementia progression.
Increased intake of lutein and zeaxanthin has been demonstrated to improve macular pigment
optical density measures, a marker of age-related macular degeneration.
Various nutrients have been reported to play a role in reducing the risk for ischemic heart disease,
stroke, myocardial infarction, heart failure, hypertension, and diabetes with varying levels of effect size
and evidence. B-Vitamins reduced homocysteine levels and reduced the risk for stroke. Some but not
all studies reported that higher omega-3 LCPUFAs intakes resulted in reduced risk of cardiovascular
events; most pronounced effects being shown in subjects with low intake or status. Vitamin C and D
may reduce hypertension, omega-3 LCPUFAs may have positive effect on blood lipid profiles, and
omega-3 LCPUFAs, vitamin D, and chromium may reduce diabetes risk factors.
Most pronounced benefits of nutrient interventions were sometimes found in subgroups which
had low baseline intake or status of the nutrient. Genetic factors can affect the status of certain nutrients,
as well as contribute to increased risk for NCDs and raise the needs for certain nutrients [139,145].
Targeted supplementation with nutrients of concern to genetically predisposed subgroups has been
shown to confer benefits as shown by some examples in this review. More research is needed to unravel
the benefits of optimizing nutrition where it is needed, for instance by targeting those at increased risk
for NCDs linked to low nutrition status or genetic profile.
Due to a growing aging global population, related NCDs including musculoskeletal disorders,
dementia, loss of vision, and cardiovascular diseases will place an increasing burden on health systems
and costs. Adequate nutrient status may help to improve health and wellbeing in older populations
and slow the progression of NCDs. Implementing a long-term preventative strategy to promote healthy
aging and break down the barriers to adequate nutrition for older adults could result in significant
healthcare cost savings. Nutrition is increasingly acknowledged and integrated into public health
policies and programs to manage healthy aging. Promoting nutrient-rich diets and adequate nutrient
intakes for healthy aging should be considered part of an integral approach to address NCDs in health
policies. There is a need for public and/or private partnerships where governments, health authorities,
205
Nutrients 2019, 11, 85
academics, and the food sector jointly promote the benefits of healthy nutrient-rich diets and lifestyle
to manage NCDs.
In conclusion, data indicate that inadequate nutrient intake and status is common in older
aged adults and represents a risk for the development of NCDs during aging. Studies for the aging
population have demonstrated that optimizing nutrition can reduce the risk and progress of NCDs.
Although the scientific evidence is not conclusive for all health benefits, it should not prevent health
authorities from promoting balanced and adequate nutrient intakes as integral part of nutrition
strategies to reduce the burden of NCDs associated with inadequate nutrition.
References
1. World Health Organization (WHO). Global Health Observatory (GHO) Data, Life Expectancy. Available
online: http://www.who.int/gho/mortality_burden_disease/life_tables/situation_trends/en/ (accessed
on 20 September 2018).
2. Figueira, I.; Fernandes, A.; Mladenovic Djordjevic, A.; Lopez-Contreras, A.; Henriques, C.M.; Selman, C.;
Ferreiro, E.; Gonos, E.S.; Trejo, J.L.; Misra, J.; et al. Interventions for age-related diseases: Shifting the
paradigm. Mech. Ageing Dev. 2016, 160, 69–92. [CrossRef]
3. Troesch, B.; Biesalski, H.K.; Bos, R.; Buskens, E.; Calder, P.C.; Saris, W.H.; Spieldenner, J.; Verkade, H.J.;
Weber, P.; Eggersdorfer, M. Increased Intake of Foods with High Nutrient Density Can Help to Break the
Intergenerational Cycle of Malnutrition and Obesity. Nutrients 2015, 7, 6016–6037. [CrossRef]
4. World Health Organization (WHO). Global Action Plan for the Prevention and Control of NCDs 2013–2020.
2018. Available online: https://www.who.int/nmh/publications/ncd-action-plan/en/ (accessed on
3 January 2019).
5. United Nations Economic and Social Council (ECOSOC). ECOSOC 2018 Task Force Resolution Urges
Partners to Mobilize Resources for the Work of the Task Force. Available online: http://www.who.int/ncds/
un-task-force/events/ecosoc-report-2018/en/ (accessed on 20 September 2018).
6. World Health Organization (WHO). Health Statistics and Information Systems. Disease Burden and Mortality
Estimates. Global Health Estimates 2016: DALYs by Age, Sex and Cause. Available online: http://www.
who.int/healthinfo/global_burden_disease/estimates/en/index1.html (accessed on 20 September 2018).
7. Eggersdorfer, M.; Walter, P. Emerging nutrition gaps in a world of affluence—Micronutrient intake and
status globally. Int. J. Vitam. Nutr. Res. 2011, 81, 238–239. [CrossRef]
8. Hoeft, B.; Weber, P.; Eggersdorfer, M. Micronutrients—A global perspective on intake, health benefits and
economics. Int. J. Vitam. Nutr. Res. 2012, 82, 316–320. [CrossRef]
9. Global Nutrition Report 2017, Nourishing the SDGs. Available online: http://165.227.233.32/wp-content/
uploads/2017/11/Report_2017-2.pdf (accessed on 20 September 2018).
10. Drewnowski, A. Nutrient density: Addressing the challenge of obesity. Br. J. Nutr. 2018, 120, S8–S14.
[CrossRef]
11. Mozaffarian, D.; Rosenberg, I.; Uauy, R. History of modern nutrition science-implications for current research,
dietary guidelines, and food policy. BMJ 2018, 361, k2392. [CrossRef]
12. Mertens, E.; Kuijsten, A.; Dofkova, M.; Mistura, L.; D’Addezio, L.; Turrini, A.; Dubuisson, C.; Favret, S.;
Havard, S.; Trolle, E.; et al. Geographic and socioeconomic diversity of food and nutrient intakes: A
comparison of four European countries. Eur. J. Nutr. 2018. [CrossRef]
13. Candari, C.J.; Cylus, J.; Nolte, E. Assessing the Economic Costs of Unhealthy Diets and low
Physical Activity: An Evidence Review and Proposed Framework. Available online: http:
//www.euro.who.int/en/publications/abstracts/assessing-the-economic-costs-of-unhealthy-diets-and-low-
physical-activity-an-evidence-review-and-proposed-framework-2017 (accessed on 3 January 2019).
206
Nutrients 2019, 11, 85
14. Peter, S.; Saris, W.H.; Mathers, J.C.; Feskens, E.; Schols, A.; Navis, G.; Kuipers, F.; Weber, P.; Eggersdorfer, M.
Nutrient Status Assessment in Individuals and Populations for Healthy Aging-Statement from an Expert
Workshop. Nutrients 2015, 7, 10491–10500. [CrossRef]
15. De Groot, L.C. Nutritional issues for older adults: Addressing degenerative ageing with long-term studies.
Proc. Nutr. Soc. 2016, 75, 169–173. [CrossRef]
16. Siddique, N.; O’Donoghue, M.; Casey, M.C.; Walsh, J.B. Malnutrition in the elderly and its effects on bone
health—A review. Clin. Nutr. ESPEN 2017, 21, 31–39. [CrossRef]
17. Leslie, W.; Hankey, C. Aging, Nutritional Status and Health. Healthcare (Basel) 2015, 3, 648–658. [CrossRef]
18. Arnim, C.A.F. Nutrition security in older adults: Status quo and future development. In Sustainable Nutrition
in a Changing World; Biesalski, H.K., Drewnowski, A., Dwyer, J.T., Strain, J.J., Weber, P., Eggersdorfer, M.,
Eds.; Springer International Publishing: Basel, Switzerland, 2017; pp. 61–73.
19. Kimokoti, R.W.; Millen, B.E. Nutrition for the Prevention of Chronic Diseases. Med. Clin. N. Am. 2016, 100,
1185–1198. [CrossRef]
20. Bruins, M.J.; Bird, J.K.; Aebischer, C.P.; Eggersdorfer, M. Considerations for Secondary Prevention of
Nutritional Deficiencies in High-Risk Groups in High-Income Countries. Nutrients 2018, 10, 47. [CrossRef]
21. Wong, C.W. Vitamin B12 deficiency in the elderly: Is it worth screening? Hong Kong Med. J. 2015, 21, 155–164.
[CrossRef]
22. Bendik, I.; Friedel, A.; Roos, F.F.; Weber, P.; Eggersdorfer, M. Vitamin D: A critical and essential micronutrient
for human health. Front. Physiol. 2014, 5, 248. [CrossRef]
23. Stover, P.J. Influence of human genetic variation on nutritional requirements. Am. J. Clin. Nutr. 2006, 83,
436S–442S. [CrossRef]
24. Prince, M.J.; Wu, F.; Guo, Y.; Gutierrez Robledo, L.M.; O’Donnell, M.; Sullivan, R.; Yusuf, S. The burden of
disease in older people and implications for health policy and practice. Lancet 2015, 385, 549–562. [CrossRef]
25. Avenell, A.; Mak, J.C.; O’Connell, D. Vitamin D and vitamin D analogues for preventing fractures in
post-menopausal women and older men. Cochrane Database Syst. Rev. 2014, 14, CD000227. [CrossRef]
26. Johnell, O.; Kanis, J.A. An estimate of the worldwide prevalence and disability associated with osteoporotic
fractures. Osteoporos. Int. 2006, 17, 1726–1733. [CrossRef]
27. World Health Organization (WHO). WHO Scientific Group on the Assessment of Osteoporosis at Primary Health
Care Level; Summary Meeting Report, Brussels, Belgium, 5–7 May 2004; WHO: Geneva, Switzerland, 2004.
28. Hernlund, E.; Svedbom, A.; Ivergard, M.; Compston, J.; Cooper, C.; Stenmark, J.; McCloskey, E.V.; Jonsson, B.;
Kanis, J.A. Osteoporosis in the European Union: Medical management, epidemiology and economic burden.
A report prepared in collaboration with the International Osteoporosis Foundation (IOF) and the European
Federation of Pharmaceutical Industry Associations (EFPIA). Arch. Osteoporos. 2013, 8, 136. [CrossRef]
29. Beaudart, C.; Buckinx, F.; Rabenda, V.; Gillain, S.; Cavalier, E.; Slomian, J.; Petermans, J.; Reginster, J.Y.;
Bruyere, O. The effects of vitamin D on skeletal muscle strength, muscle mass, and muscle power: A
systematic review and meta-analysis of randomized controlled trials. J. Clin. Endocrinol. Metab. 2014, 99,
4336–4345. [CrossRef]
30. Thiering, E.; Bruske, I.; Kratzsch, J.; Hofbauer, L.C.; Berdel, D.; von Berg, A.; Lehmann, I.; Hoffmann, B.;
Bauer, C.P.; Koletzko, S.; et al. Associations between serum 25-hydroxyvitamin D and bone turnover markers
in a population based sample of German children. Sci. Rep. 2015, 5, 18138. [CrossRef]
31. Quesada-Gomez, J.M.; Bouillon, R. Is calcifediol better than cholecalciferol for vitamin D supplementation?
Osteoporos. Int. 2018, 29, 1697–1711. [CrossRef]
32. Sai, A.J.; Walters, R.W.; Fang, X.; Gallagher, J.C. Relationship between vitamin D, parathyroid hormone, and
bone health. J. Clin. Endocrinol. Metab. 2011, 96, E436–E446. [CrossRef]
33. Heaney, R.P. Functional indices of vitamin D status and ramifications of vitamin D deficiency. Am. J. Clin.
Nutr. 2004, 80, 1706S–1709S. [CrossRef]
34. Cauley, J.A.; Parimi, N.; Ensrud, K.E.; Bauer, D.C.; Cawthon, P.M.; Cummings, S.R.; Hoffman, A.R.;
Shikany, J.M.; Barrett-Connor, E.; Orwoll, E.; et al. Serum 25-hydroxyvitamin D and the risk of hip and
nonspine fractures in older men. J. Bone Miner. Res. 2010, 25, 545–553. [CrossRef]
35. Looker, A.C.; Mussolino, M.E. Serum 25-hydroxyvitamin D and hip fracture risk in older U.S. white adults.
J. Bone Miner. Res. 2008, 23, 143–150. [CrossRef]
207
Nutrients 2019, 11, 85
36. Cashman, K.D.; Dowling, K.G.; Skrabakova, Z.; Gonzalez-Gross, M.; Valtuena, J.; De Henauw, S.; Moreno, L.;
Damsgaard, C.T.; Michaelsen, K.F.; Molgaard, C.; et al. Vitamin D deficiency in Europe: Pandemic? Am. J.
Clin. Nutr. 2016, 103, 1033–1044. [CrossRef]
37. Hilger, J.; Friedel, A.; Herr, R.; Rausch, T.; Roos, F.; Wahl, D.A.; Pierroz, D.D.; Weber, P.; Hoffmann, K. A
systematic review of vitamin D status in populations worldwide. Br. J. Nutr. 2014, 111, 23–45. [CrossRef]
38. Wahl, D.A.; Cooper, C.; Ebeling, P.R.; Eggersdorfer, M.; Hilger, J.; Hoffmann, K.; Josse, R.; Kanis, J.A.;
Mithal, A.; Pierroz, D.D.; et al. A global representation of vitamin D status in healthy populations: Reply to
comment by Saadi. Arch. Osteoporos. 2013, 8, 122. [CrossRef]
39. Spiro, A.; Buttriss, J.L. Vitamin D: An overview of vitamin D status and intake in Europe. Nutr. Bull. 2014,
39, 322–350. [CrossRef]
40. Van Ballegooijen, A.J.; Pilz, S.; Tomaschitz, A.; Grubler, M.R.; Verheyen, N. The Synergistic Interplay between
Vitamins D and K for Bone and Cardiovascular Health: A Narrative Review. Int. J. Endocrinol. 2017, 2017,
7454376. [CrossRef]
41. Cockayne, S.; Adamson, J.; Lanham-New, S.; Shearer, M.J.; Gilbody, S.; Torgerson, D.J. Vitamin K and the
prevention of fractures: Systematic review and meta-analysis of randomized controlled trials. Arch. Intern.
Med. 2006, 166, 1256–1261. [CrossRef]
42. Bolton-Smith, C.; McMurdo, M.E.; Paterson, C.R.; Mole, P.A.; Harvey, J.M.; Fenton, S.T.; Prynne, C.J.;
Mishra, G.D.; Shearer, M.J. Two-year randomized controlled trial of vitamin K1 (phylloquinone) and vitamin
D3 plus calcium on the bone health of older women. J. Bone Miner. Res. 2007, 22, 509–519. [CrossRef]
43. Je, S.H.; Joo, N.S.; Choi, B.H.; Kim, K.M.; Kim, B.T.; Park, S.B.; Cho, D.Y.; Kim, K.N.; Lee, D.J. Vitamin
K supplement along with vitamin D and calcium reduced serum concentration of undercarboxylated
osteocalcin while increasing bone mineral density in Korean postmenopausal women over sixty-years-old.
J. Korean Med. Sci. 2011, 26, 1093–1098. [CrossRef]
44. Moore, K.; Hughes, C.F.; Ward, M.; Hoey, L.; McNulty, H. Diet, nutrition and the ageing brain: Current
evidence and new directions. Proc. Nutr. Soc. 2018, 77, 152–163. [CrossRef]
45. Prince, M.; Bryce, R.; Albanese, E.; Wimo, A.; Ribeiro, W.; Ferri, C.P. The global prevalence of dementia: A
systematic review and metaanalysis. Alzheimers Dement. 2013, 9, 63–75. [CrossRef]
46. World Health Organization (WHO). Fact Sheets: Dementia. Available online: http://www.who.int/news-
room/fact-sheets/detail/dementia (accessed on 12 December 2017).
47. Jiang, T.; Yu, J.T.; Tian, Y.; Tan, L. Epidemiology and etiology of Alzheimer’s disease: From genetic to
non-genetic factors. Curr. Alzheimer Res. 2013, 10, 852–867. [CrossRef]
48. Antal, M.; Péter, S.; Eggersdorfer, M. Alzheimer’s Disease: An epidemiologic disaster from nutritional
perspective. Nutr. Health Food Sci. 2017, 5, 1–4.
49. Reynolds, E. Vitamin B12, folic acid, and the nervous system. Lancet Neurol. 2006, 5, 949–960. [CrossRef]
50. Reynolds, E.H. Folic acid, ageing, depression, and dementia. BMJ 2002, 324, 1512–1515. [CrossRef]
51. McCleery, J.; Abraham, R.P.; Denton, D.A.; Rutjes, A.W.; Chong, L.Y.; Al-Assaf, A.S.; Griffith, D.J.; Rafeeq, S.;
Yaman, H.; Malik, M.A.; et al. Vitamin and mineral supplementation for preventing dementia or delaying
cognitive decline in people with mild cognitive impairment. Cochrane Database Syst. Rev. 2018, 11, CD011905.
[CrossRef]
52. Smith, A.D.; Refsum, H.; Bottiglieri, T.; Fenech, M.; Hooshmand, B.; McCaddon, A.; Miller, J.W.;
Rosenberg, I.H.; Obeid, R. Homocysteine and Dementia: An International Consensus Statement. J. Alzheimers
Dis. 2018, 62, 561–570. [CrossRef]
53. Douaud, G.; Refsum, H.; de Jager, C.A.; Jacoby, R.; Nichols, T.E.; Smith, S.M.; Smith, A.D. Preventing
Alzheimer’s disease-related gray matter atrophy by B-vitamin treatment. Proc. Natl. Acad. Sci. USA 2013,
110, 9523–9528. [CrossRef]
54. Troesch, B.; Hoeft, B.; McBurney, M.; Eggersdorfer, M.; Weber, P. Dietary surveys indicate vitamin intakes
below recommendations are common in representative Western countries. Br. J. Nutr. 2012, 108, 692–698.
[CrossRef]
55. Gugliandolo, A.; Bramanti, P.; Mazzon, E. Role of Vitamin E in the Treatment of Alzheimer’s Disease:
Evidence from Animal Models. Int. J. Mol. Sci. 2017, 18, 2504. [CrossRef]
56. Salem, N., Jr.; Vandal, M.; Calon, F. The benefit of docosahexaenoic acid for the adult brain in aging and
dementia. Prostaglandins Leukot. Essent. Fatty Acids 2015, 92, 15–22. [CrossRef]
208
Nutrients 2019, 11, 85
57. Zhang, Y.J.; Gan, R.Y.; Li, S.; Zhou, Y.; Li, A.N.; Xu, D.P.; Li, H.B. Antioxidant Phytochemicals for the
Prevention and Treatment of Chronic Diseases. Molecules 2015, 20, 21138–21156. [CrossRef]
58. GBD 2015 DALYs and Hale Collaborators. Global, regional, and national disability-adjusted life-years
(DALYs) for 315 diseases and injuries and healthy life expectancy (HALE), 1990–2015: A systematic analysis
for the Global Burden of Disease Study 2015. Lancet 2016, 388, 1603–1658. [CrossRef]
59. Quillen, D.A. Common causes of vision loss in elderly patients. Am. Fam. Physician 1999, 60, 99–108.
60. Chen, Y.; Bedell, M.; Zhang, K. Age-related macular degeneration: Genetic and environmental factors of
disease. Mol. Interv. 2010, 10, 271–281. [CrossRef]
61. Tosini, G.; Ferguson, I.; Tsubota, K. Effects of blue light on the circadian system and eye physiology. Mol. Vis.
2016, 22, 61–72.
62. Demmig-Adams, B.; Adams, R.B. Eye nutrition in context: Mechanisms, implementation, and future
directions. Nutrients 2013, 5, 2483–2501. [CrossRef]
63. Bernstein, P.S.; Li, B.; Vachali, P.P.; Gorusupudi, A.; Shyam, R.; Henriksen, B.S.; Nolan, J.M. Lutein, zeaxanthin,
and meso-zeaxanthin: The basic and clinical science underlying carotenoid-based nutritional interventions
against ocular disease. Prog. Retin. Eye Res. 2016, 50, 34–66. [CrossRef]
64. Barker, F.M., II; Snodderly, D.M.; Johnson, E.J.; Schalch, W.; Koepcke, W.; Gerss, J.; Neuringer, M. Nutritional
manipulation of primate retinas, V: Effects of lutein, zeaxanthin, and n-3 fatty acids on retinal sensitivity to
blue-light-induced damage. Investig. Ophthalmol. Vis. Sci. 2011, 52, 3934–3942. [CrossRef]
65. Hammond, B.R.; Fletcher, L.M.; Roos, F.; Wittwer, J.; Schalch, W. A double-blind, placebo-controlled study
on the effects of lutein and zeaxanthin on photostress recovery, glare disability, and chromatic contrast.
Investig. Ophthalmol. Vis. Sci. 2014, 55, 8583–8589. [CrossRef]
66. Yao, Y.; Qiu, Q.H.; Wu, X.W.; Cai, Z.Y.; Xu, S.; Liang, X.Q. Lutein supplementation improves visual
performance in Chinese drivers: 1-year randomized, double-blind, placebo-controlled study. Nutrition
2013, 29, 958–964. [CrossRef]
67. Loughman, J.; Nolan, J.M.; Howard, A.N.; Connolly, E.; Meagher, K.; Beatty, S. The impact of macular
pigment augmentation on visual performance using different carotenoid formulations. Investig. Ophthalmol.
Vis. Sci. 2012, 53, 7871–7880. [CrossRef]
68. Nolan, J.M.; Loughman, J.; Akkali, M.C.; Stack, J.; Scanlon, G.; Davison, P.; Beatty, S. The impact of macular
pigment augmentation on visual performance in normal subjects: COMPASS. Vision Res. 2011, 51, 459–469.
[CrossRef]
69. Richer, S.P.; Stiles, W.; Graham-Hoffman, K.; Levin, M.; Ruskin, D.; Wrobel, J.; Park, D.W.; Thomas, C.
Randomized, double-blind, placebo-controlled study of zeaxanthin and visual function in patients with
atrophic age-related macular degeneration: The Zeaxanthin and Visual Function Study (ZVF) FDA IND #78,
973. Optometry 2011, 82, 667–680.
70. Stringham, J.M.; Hammond, B.R. Macular pigment and visual performance under glare conditions.
Optom. Vis. Sci. 2008, 85, 82–88. [CrossRef]
71. Wenzel, A.J.; Fuld, K.; Stringham, J.M.; Curran-Celentano, J. Macular pigment optical density and
photophobia light threshold. Vision Res. 2006, 46, 4615–4622. [CrossRef]
72. Kvansakul, J.; Rodriguez-Carmona, M.; Edgar, D.F.; Barker, F.M.; Kopcke, W.; Schalch, W.; Barbur, J.L.
Supplementation with the carotenoids lutein or zeaxanthin improves human visual performance. Ophthalmic.
Physiol. Opt. 2006, 26, 362–371. [CrossRef]
73. Feeney, J.; Finucane, C.; Savva, G.M.; Cronin, H.; Beatty, S.; Nolan, J.M.; Kenny, R.A. Low macular pigment
optical density is associated with lower cognitive performance in a large, population-based sample of older
adults. Neurobiol. Aging 2013, 34, 2449–2456. [CrossRef]
74. Johnson, E.J.; Maras, J.E.; Rasmussen, H.M.; Tucker, K.L. Intake of lutein and zeaxanthin differ with age, sex,
and ethnicity. J. Am. Diet. Assoc. 2010, 110, 1357–1362. [CrossRef]
75. Nolan, J.M.; Power, R.; Stringham, J.; Dennison, J.; Stack, J.; Kelly, D.; Moran, R.; Akuffo, K.O.; Corcoran, L.;
Beatty, S. Author Response: Comments on Enrichment of Macular Pigment Enhances Contrast Sensitivity in
Subjects Free of Retinal Disease: CREST—Report 1. Investig. Ophthalmol. Vis. Sci. 2016, 57, 5416. [CrossRef]
76. Olmedilla, B.; Granado, F.; Blanco, I.; Vaquero, M. Lutein, but not alpha-tocopherol, supplementation
improves visual function in patients with age-related cataracts: A 2-y double-blind, placebo-controlled pilot
study. Nutrition 2003, 19, 21–24. [CrossRef]
209
Nutrients 2019, 11, 85
77. Age-Related Eye Disease Study 2 Research Group. Lutein + zeaxanthin and omega-3 fatty acids for
age-related macular degeneration: The Age-Related Eye Disease Study 2 (AREDS2) randomized clinical trial.
JAMA 2013, 309, 2005–2015. [CrossRef]
78. Age-Related Eye Disease Study 2 Research Group; Chew, E.Y.; Clemons, T.E.; Sangiovanni, J.P.; Danis, R.P.;
Ferris, F.L., III; Elman, M.J.; Antoszyk, A.N.; Ruby, A.J.; Orth, D.; et al. Secondary analyses of the effects of
lutein/zeaxanthin on age-related macular degeneration progression: AREDS2 report No. 3. JAMA Ophthalmol.
2014, 132, 142–149. [CrossRef]
79. Liu, X.H.; Yu, R.B.; Liu, R.; Hao, Z.X.; Han, C.C.; Zhu, Z.H.; Ma, L. Association between lutein and zeaxanthin
status and the risk of cataract: A meta-analysis. Nutrients 2014, 6, 452–465. [CrossRef]
80. Age-Related Eye Disease Study 2 Research Group; Chew, E.Y.; SanGiovanni, J.P.; Ferris, F.L.; Wong, W.T.;
Agron, E.; Clemons, T.E.; Sperduto, R.; Danis, R.; Chandra, S.R.; et al. Lutein/zeaxanthin for the treatment of
age-related cataract: AREDS2 randomized trial report no. 4. JAMA Ophthalmol. 2013, 131, 843–850. [CrossRef]
81. Shanahan, C. The Economic Benefits of Using Lutein and Zeaxanthin Food Supplements in the European
Union. Available online: https://ww2.frost.com/files/7015/0772/2735/HCCS_Lutein_AMD.2017.10.12.pdf
(accessed on 3 January 2019).
82. GBD Causes of Death Collaborators. Global, regional, and national age-sex specific mortality for 264 causes
of death, 1980–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet 2017, 390,
1151–1210. [CrossRef]
83. Mozaffarian, D. Dietary and Policy Priorities for Cardiovascular Disease, Diabetes, and Obesity: A
Comprehensive Review. Circulation 2016, 133, 187–225. [CrossRef]
84. Leening, M.J.; Ferket, B.S.; Steyerberg, E.W.; Kavousi, M.; Deckers, J.W.; Nieboer, D.; Heeringa, J.;
Portegies, M.L.; Hofman, A.; Ikram, M.A.; et al. Sex differences in lifetime risk and first manifestation
of cardiovascular disease: Prospective population based cohort study. BMJ 2014, 349, g5992. [CrossRef]
85. The Institute for Health Metrics and Evaluation (IHME). Global Burden of Disease (GBD). GBD Data
Visualizations. Available online: http://www.healthdata.org/gbd (accessed on 20 September 2018).
86. Hankey, G.J. The Role of Nutrition in the Risk and Burden of Stroke: An Update of the Evidence. Stroke J.
Cereb. Circ. 2017, 48, 3168–3174. [CrossRef]
87. Van Staveren, W.A.; de Groot, L.C. Evidence-based dietary guidance and the role of dairy products for
appropriate nutrition in the elderly. J. Am. Coll. Nutr. 2011, 30, 429S–437S. [CrossRef]
88. Fleg, J.L.; Forman, D.E.; Berra, K.; Bittner, V.; Blumenthal, J.A.; Chen, M.A.; Cheng, S.; Kitzman, D.W.;
Maurer, M.S.; Rich, M.W.; et al. Secondary prevention of atherosclerotic cardiovascular disease in older adults:
A scientific statement from the American Heart Association. Circulation 2013, 128, 2422–2446. [CrossRef]
89. Catapano, A.L.; Graham, I.; De Backer, G.; Wiklund, O.; Chapman, M.J.; Drexel, H.; Hoes, A.W.; Jennings, C.S.;
Landmesser, U.; Pedersen, T.R.; et al. 2016 ESC/EAS Guidelines for the Management of Dyslipidaemias.
Rev. Esp. Cardiol. (Engl. Ed.) 2017, 70, 115.
90. Jellinger, P.S. American Association of Clinical Endocrinologists/American College of Endocrinology
Management of Dyslipidemia and Prevention of Cardiovascular Disease Clinical Practice Guidelines.
Diabetes Spectr. 2018, 31, 234–245. [CrossRef]
91. Eckel, R.H.; Jakicic, J.M.; Ard, J.D.; de Jesus, J.M.; Houston Miller, N.; Hubbard, V.S.; Lee, I.M.;
Lichtenstein, A.H.; Loria, C.M.; Millen, B.E.; et al. 2013 AHA/ACC guideline on lifestyle management to
reduce cardiovascular risk: A report of the American College of Cardiology/American Heart Association
Task Force on Practice Guidelines. Circulation 2014, 129, S76–S99. [CrossRef]
92. Estruch, R.; Ros, E.; Salas-Salvado, J.; Covas, M.I.; Corella, D.; Aros, F.; Gomez-Gracia, E.; Ruiz-Gutierrez, V.;
Fiol, M.; Lapetra, J.; et al. Primary Prevention of Cardiovascular Disease with a Mediterranean Diet
Supplemented with Extra-Virgin Olive Oil or Nuts. N. Engl. J. Med. 2018, 378, e34. [CrossRef]
93. De Lorgeril, M.; Salen, P.; Martin, J.L.; Monjaud, I.; Delaye, J.; Mamelle, N. Mediterranean diet, traditional
risk factors, and the rate of cardiovascular complications after myocardial infarction: Final report of the Lyon
Diet Heart Study. Circulation 1999, 99, 779–785. [CrossRef]
94. Ornish, D.; Scherwitz, L.W.; Billings, J.H.; Brown, S.E.; Gould, K.L.; Merritt, T.A.; Sparler, S.; Armstrong, W.T.;
Ports, T.A.; Kirkeeide, R.L.; et al. Intensive lifestyle changes for reversal of coronary heart disease. JAMA
1998, 280, 2001–2007. [CrossRef]
95. Kheiri, B.; Abdalla, A.; Osman, M.; Ahmed, S.; Hassan, M.; Bachuwa, G. Vitamin D deficiency and risk of
cardiovascular diseases: A narrative review. Clin. Hypertens. 2018, 24, 9. [CrossRef]
210
Nutrients 2019, 11, 85
96. Gepner, A.D.; Ramamurthy, R.; Krueger, D.C.; Korcarz, C.E.; Binkley, N.; Stein, J.H. A prospective
randomized controlled trial of the effects of vitamin D supplementation on cardiovascular disease risk.
PLoS ONE 2012, 7, e36617. [CrossRef]
97. Ford, J.A.; MacLennan, G.S.; Avenell, A.; Bolland, M.; Grey, A.; Witham, M.; Group, R.T. Cardiovascular
disease and vitamin D supplementation: Trial analysis, systematic review, and meta-analysis. Am. J. Clin.
Nutr. 2014, 100, 746–755. [CrossRef]
98. Vitamin D and Omega-3 Trial (VITAL). Available online: https://www.vitalstudy.org/findings.html
(accessed on 20 December 2018).
99. Huang, T.; Chen, Y.; Yang, B.; Yang, J.; Wahlqvist, M.L.; Li, D. Meta-analysis of B vitamin supplementation
on plasma homocysteine, cardiovascular and all-cause mortality. Clin. Nutr. 2012, 31, 448–454. [CrossRef]
100. Yang, H.T.; Lee, M.; Hong, K.S.; Ovbiagele, B.; Saver, J.L. Efficacy of folic acid supplementation in
cardiovascular disease prevention: An updated meta-analysis of randomized controlled trials. Eur. J.
Intern. Med. 2012, 23, 745–754. [CrossRef]
101. Wald, D.S.; Law, M.; Morris, J.K. Homocysteine and cardiovascular disease: Evidence on causality from a
meta-analysis. BMJ 2002, 325, 1202. [CrossRef]
102. Li, Y.; Huang, T.; Zheng, Y.; Muka, T.; Troup, J.; Hu, F.B. Folic Acid Supplementation and the Risk of
Cardiovascular Diseases: A Meta-Analysis of Randomized Controlled Trials. J. Am. Heart Assoc. 2016, 5,
e003768. [CrossRef]
103. Fezeu, L.K.; Ducros, V.; Gueant, J.L.; Guilland, J.C.; Andreeva, V.A.; Hercberg, S.; Galan, P. MTHFR 677C→T
genotype modulates the effect of a 5-year supplementation with B-vitamins on homocysteine concentration:
The SU.FOL.OM3 randomized controlled trial. PLoS ONE 2018, 13, e0193352. [CrossRef]
104. DiNicolantonio, J.J.; Bhutani, J.; O’Keefe, J.H. The health benefits of vitamin K. Open Heart 2015, 2, e000300.
[CrossRef]
105. Innes, J.K.; Calder, P.C. The Differential Effects of Eicosapentaenoic Acid and Docosahexaenoic Acid on
Cardiometabolic Risk Factors: A Systematic Review. Int. J. Mol. Sci. 2018, 19, 532. [CrossRef]
106. Thota, R.N.; Ferguson, J.J.A.; Abbott, K.A.; Dias, C.B.; Garg, M.L. Science behind the cardio-metabolic
benefits of omega-3 polyunsaturated fatty acids: Biochemical effects vs. clinical outcomes. Food Funct. 2018,
9, 3576–3596. [CrossRef]
107. Karalis, D.G. A Review of Clinical Practice Guidelines for the Management of Hypertriglyceridemia: A
Focus on High Dose Omega-3 Fatty Acids. Adv. Ther. 2017, 34, 300–323. [CrossRef]
108. Alexander, D.D.; Miller, P.E.; Van Elswyk, M.E.; Kuratko, C.N.; Bylsma, L.C. A Meta-Analysis of Randomized
Controlled Trials and Prospective Cohort Studies of Eicosapentaenoic and Docosahexaenoic Long-Chain
Omega-3 Fatty Acids and Coronary Heart Disease Risk. Mayo Clin. Proc. 2017, 92, 15–29. [CrossRef]
109. Abdelhamid, A.S.; Brown, T.J.; Brainard, J.S.; Biswas, P.; Thorpe, G.C.; Moore, H.J.; Deane, K.H.;
AlAbdulghafoor, F.K.; Summerbell, C.D.; Worthington, H.V.; et al. Omega-3 fatty acids for the primary and
secondary prevention of cardiovascular disease. Cochrane Database Syst. Rev. 2018, 7, CD003177.
110. Meyer, B.J.; Groot, R.H.M. Effects of Omega-3 Long Chain Polyunsaturated Fatty Acid Supplementation on
Cardiovascular Mortality: The Importance of the Dose of DHA. Nutrients 2017, 9, 1305. [CrossRef]
111. Kromhout, D.; Giltay, E.J.; Geleijnse, J.M.; Alpha Omega Trial, G. n-3 fatty acids and cardiovascular events
after myocardial infarction. N. Engl. J. Med. 2010, 363, 2015–2026. [CrossRef]
112. Writing Group for the AREDS2 Research Group; Bonds, D.E.; Harrington, M.; Worrall, B.B.; Bertoni, A.G.;
Eaton, C.B.; Hsia, J.; Robinson, J.; Clemons, T.E.; Fine, L.J.; et al. Effect of long-chain omega-3 fatty acids and
lutein + zeaxanthin supplements on cardiovascular outcomes: Results of the Age-Related Eye Disease Study
2 (AREDS2) randomized clinical trial. JAMA Intern. Med. 2014, 174, 763–771.
113. Hidayat, K.; Yang, J.; Zhang, Z.; Chen, G.C.; Qin, L.Q.; Eggersdorfer, M.; Zhang, W. Effect of omega-3
long-chain polyunsaturated fatty acid supplementation on heart rate: A meta-analysis of randomized
controlled trials. Eur. J. Clin. Nutr. 2018, 72, 805–817. [CrossRef]
114. Zhang, G.Q.; Zhang, W. Heart rate, lifespan, and mortality risk. Ageing Res. Rev. 2009, 8, 52–60. [CrossRef]
115. Chen, Y.; Michalak, M.; Agellon, L.B. Importance of Nutrients and Nutrient Metabolism on Human Health.
Yale J. Biol. Med. 2018, 91, 95–103.
116. Rangel-Huerta, O.D.; Pastor-Villaescusa, B.; Aguilera, C.M.; Gil, A. A Systematic Review of the Efficacy
of Bioactive Compounds in Cardiovascular Disease: Phenolic Compounds. Nutrients 2015, 7, 5177–5216.
[CrossRef]
211
Nutrients 2019, 11, 85
117. Ye, Y.; Li, J.; Yuan, Z. Effect of antioxidant vitamin supplementation on cardiovascular outcomes: A
meta-analysis of randomized controlled trials. PLoS ONE 2013, 8, e56803. [CrossRef]
118. Raederstorff, D.; Wyss, A.; Calder, P.C.; Weber, P.; Eggersdorfer, M. Vitamin E function and requirements in
relation to PUFA. Br. J. Nutr. 2015, 114, 1113–1122. [CrossRef]
119. Lee, I.M.; Cook, N.R.; Gaziano, J.M.; Gordon, D.; Ridker, P.M.; Manson, J.E.; Hennekens, C.H.; Buring, J.E.
Vitamin E in the primary prevention of cardiovascular disease and cancer: The Women’s Health Study: A
randomized controlled trial. JAMA 2005, 294, 56–65. [CrossRef]
120. Glynn, R.J.; Ridker, P.M.; Goldhaber, S.Z.; Zee, R.Y.; Buring, J.E. Effects of random allocation to vitamin E
supplementation on the occurrence of venous thromboembolism: Report from the Women’s Health Study.
Circulation 2007, 116, 1497–1503. [CrossRef]
121. Farbstein, D.; Levy, A.P. The genetics of vascular complications in diabetes mellitus. Cardiol. Clin. 2010, 28,
477–496. [CrossRef]
122. He, H.Y.; Liu, M.Z.; Zhang, Y.L.; Zhang, W. Vitamin Pharmacogenomics: New Insight into Individual
Differences in Diseases and Drug Responses. Genom. Proteom. Bioinform. 2017, 15, 94–100. [CrossRef]
123. Huxley, R.R.; Neil, H.A. The relation between dietary flavonol intake and coronary heart disease mortality:
A meta-analysis of prospective cohort studies. Eur. J. Clin. Nutr. 2003, 57, 904–908. [CrossRef] [PubMed]
124. Wang, Z.M.; Nie, Z.L.; Zhou, B.; Lian, X.Q.; Zhao, H.; Gao, W.; Wang, Y.S.; Jia, E.Z.; Wang, L.S.; Yang, Z.J.
Flavonols intake and the risk of coronary heart disease: A meta-analysis of cohort studies. Atherosclerosis
2012, 222, 270–273. [CrossRef] [PubMed]
125. Castro, I.; Waclawovsky, G.; Marcadenti, A. Nutrition and physical activity on hypertension: Implication of
current evidence and guidelines. Curr. Hypertens. Rev. 2015, 11, 91–99. [CrossRef] [PubMed]
126. Miller, A.P.; Navar, A.M.; Roubin, G.S.; Oparil, S. Cardiovascular care for older adults: Hypertension and
stroke in the older adult. J. Geriatr. Cardiol. 2016, 13, 373–379. [PubMed]
127. James, P.A.; Oparil, S.; Carter, B.L.; Cushman, W.C.; Dennison-Himmelfarb, C.; Handler, J.; Lackland, D.T.;
LeFevre, M.L.; MacKenzie, T.D.; Ogedegbe, O.; et al. 2014 evidence-based guideline for the management
of high blood pressure in adults: Report from the panel members appointed to the Eighth Joint National
Committee (JNC 8). JAMA 2014, 311, 507–520. [CrossRef] [PubMed]
128. Nguyen, H.; Odelola, O.A.; Rangaswami, J.; Amanullah, A. A review of nutritional factors in hypertension
management. Int. J. Hypertens. 2013, 2013, 698940. [CrossRef] [PubMed]
129. Perez, V.; Chang, E.T. Sodium-to-potassium ratio and blood pressure, hypertension, and related factors.
Adv. Nutr. 2014, 5, 712–741. [CrossRef]
130. Cicero, A.F.; Colletti, A.; Rosticci, M.; Cagnati, M.; Urso, R.; Giovannini, M.; Borghi, C.; D’Addato, S. Effect of
Lactotripeptides (Isoleucine-Proline-Proline/Valine-Proline-Proline) on Blood Pressure and Arterial Stiffness
Changes in Subjects with Suboptimal Blood Pressure Control and Metabolic Syndrome: A Double-Blind,
Randomized, Crossover Clinical Trial. Metab. Syndr. Relat. Disord. 2016, 14, 161–166. [CrossRef]
131. Miller, P.E.; Van Elswyk, M.; Alexander, D.D. Long-chain omega-3 fatty acids eicosapentaenoic acid and
docosahexaenoic acid and blood pressure: A meta-analysis of randomized controlled trials. Am. J. Hypertens.
2014, 27, 885–896. [CrossRef]
132. Geleijnse, J.M.; Giltay, E.J.; Grobbee, D.E.; Donders, A.R.; Kok, F.J. Blood pressure response to fish oil
supplementation: Metaregression analysis of randomized trials. J. Hypertens. 2002, 20, 1493–1499. [CrossRef]
133. Juraschek, S.P.; Guallar, E.; Appel, L.J.; Miller, E.R., III. Effects of vitamin C supplementation on blood
pressure: A meta-analysis of randomized controlled trials. Am. J. Clin. Nutr. 2012, 95, 1079–1088. [CrossRef]
[PubMed]
134. Forman, J.P.; Scott, J.B.; Ng, K.; Drake, B.F.; Suarez, E.G.; Hayden, D.L.; Bennett, G.G.; Chandler, P.D.;
Hollis, B.W.; Emmons, K.M.; et al. Effect of vitamin D supplementation on blood pressure in blacks.
Hypertension 2013, 61, 779–785. [CrossRef] [PubMed]
135. International Diabetes Federation (IDF). IDF Diabetes Atlas 8th Edition. Available online: http://www.
diabetesatlas.org/ (accessed on 20 September 2018).
136. Samocha-Bonet, D.; Debs, S.; Greenfield, J.R. Prevention and Treatment of Type 2 Diabetes: A Pathophysiological-
Based Approach. Trends Endocrinol. Metab. 2018, 29, 370–379. [CrossRef] [PubMed]
137. Li, X.; Liu, Y.; Zheng, Y.; Wang, P.; Zhang, Y. The Effect of Vitamin D Supplementation on Glycemic Control
in Type 2 Diabetes Patients: A Systematic Review and Meta-Analysis. Nutrients 2018, 10, 375. [PubMed]
212
Nutrients 2019, 11, 85
138. Upreti, V.; Maitri, V.; Dhull, P.; Handa, A.; Prakash, M.S.; Behl, A. Effect of oral vitamin D supplementation
on glycemic control in patients with type 2 diabetes mellitus with coexisting hypovitaminosis D: A parellel
group placebo controlled randomized controlled pilot study. Diabetes Metab. Syndr. 2018, 12, 509–512.
[CrossRef] [PubMed]
139. Milman, U.; Blum, S.; Shapira, C.; Aronson, D.; Miller-Lotan, R.; Anbinder, Y.; Alshiek, J.; Bennett, L.;
Kostenko, M.; Landau, M.; et al. Vitamin E supplementation reduces cardiovascular events in a subgroup of
middle-aged individuals with both type 2 diabetes mellitus and the haptoglobin 2-2 genotype: A prospective
double-blinded clinical trial. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 341–347. [CrossRef] [PubMed]
140. Asleh, R.; Briasoulis, A.; Berinstein, E.M.; Wiener, J.B.; Palla, M.; Kushwaha, S.S.; Levy, A.P. Meta-analysis
of the association of the haptoglobin genotype with cardiovascular outcomes and the pharmacogenomic
interactions with vitamin E supplementation. Pharmacogenom. Pers. Med. 2018, 11, 71–82. [CrossRef]
141. Chen, C.; Yu, X.; Shao, S. Effects of Omega-3 Fatty Acid Supplementation on Glucose Control and Lipid
Levels in Type 2 Diabetes: A Meta-Analysis. PLoS ONE 2015, 10, e0139565. [CrossRef]
142. Beulens, J.W.; van der, A.D.; Grobbee, D.E.; Sluijs, I.; Spijkerman, A.M.; van der Schouw, Y.T. Dietary
phylloquinone and menaquinones intakes and risk of type 2 diabetes. Diabetes Care 2010, 33, 1699–1705.
[CrossRef]
143. Yoshida, M.; Jacques, P.F.; Meigs, J.B.; Saltzman, E.; Shea, M.K.; Gundberg, C.; Dawson-Hughes, B.; Dallal, G.;
Booth, S.L. Effect of vitamin K supplementation on insulin resistance in older men and women. Diabetes Care
2008, 31, 2092–2096. [CrossRef]
144. Suksomboon, N.; Poolsup, N.; Yuwanakorn, A. Systematic review and meta-analysis of the efficacy and
safety of chromium supplementation in diabetes. J. Clin. Pharm. Ther. 2014, 39, 292–306. [CrossRef] [PubMed]
145. Blum, S.; Vardi, M.; Brown, J.B.; Russell, A.; Milman, U.; Shapira, C.; Levy, N.S.; Miller-Lotan, R.; Asleh, R.;
Levy, A.P. Vitamin E reduces cardiovascular disease in individuals with diabetes mellitus and the haptoglobin
2-2 genotype. Pharmacogenomics 2010, 11, 675–684. [CrossRef] [PubMed]
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
213
MDPI
St. Alban-Anlage 66
4052 Basel
Switzerland
Tel. +41 61 683 77 34
Fax +41 61 302 89 18
www.mdpi.com