1
|
Riegelman E, Xue KS, Wang JS, Tang L. Gut-Brain Axis in Focus: Polyphenols, Microbiota, and Their Influence on α-Synuclein in Parkinson's Disease. Nutrients 2024; 16:2041. [PMID: 38999791 PMCID: PMC11243524 DOI: 10.3390/nu16132041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
With the recognition of the importance of the gut-brain axis in Parkinson's disease (PD) etiology, there is increased interest in developing therapeutic strategies that target α-synuclein, the hallmark abhorrent protein of PD pathogenesis, which may originate in the gut. Research has demonstrated that inhibiting the aggregation, oligomerization, and fibrillation of α-synuclein are key strategies for disease modification. Polyphenols, which are rich in fruits and vegetables, are drawing attention for their potential role in this context. In this paper, we reviewed how polyphenols influence the composition and functional capabilities of the gut microbiota and how the resulting microbial metabolites of polyphenols may potentially enhance the modulation of α-synuclein aggregation. Understanding the interaction between polyphenols and gut microbiota and identifying which specific microbes may enhance the efficacy of polyphenols is crucial for developing therapeutic strategies and precision nutrition based on the microbiome.
Collapse
Affiliation(s)
| | | | | | - Lili Tang
- Department of Environmental Health Science, University of Georgia, Athens, GA 30602, USA; (E.R.); (K.S.X.); (J.-S.W.)
| |
Collapse
|
2
|
Li Y, Xu Y, Le Sayec M, Yan X, Spector TD, Steves CJ, Bell JT, Small KS, Menni C, Gibson R, Rodriguez-Mateos A. Development of a (Poly)phenol Metabolic Signature for Assessing (Poly)phenol-Rich Dietary Patterns. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:13439-13450. [PMID: 38829321 PMCID: PMC11181312 DOI: 10.1021/acs.jafc.4c00959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/14/2024] [Accepted: 04/30/2024] [Indexed: 06/05/2024]
Abstract
The objective assessment of habitual (poly)phenol-rich diets in nutritional epidemiology studies remains challenging. This study developed and evaluated the metabolic signature of a (poly)phenol-rich dietary score (PPS) using a targeted metabolomics method comprising 105 representative (poly)phenol metabolites, analyzed in 24 h of urine samples collected from healthy volunteers. The metabolites that were significantly associated with PPS after adjusting for energy intake were selected to establish a metabolic signature using a combination of linear regression followed by ridge regression to estimate penalized weights for each metabolite. A metabolic signature comprising 51 metabolites was significantly associated with adherence to PPS in 24 h urine samples, as well as with (poly)phenol intake estimated from food frequency questionnaires and diaries. Internal and external data sets were used for validation, and plasma, spot urine, and 24 h urine samples were compared. The metabolic signature proposed here has the potential to accurately reflect adherence to (poly)phenol-rich diets, and may be used as an objective tool for the assessment of (poly)phenol intake.
Collapse
Affiliation(s)
- Yong Li
- Department
of Nutritional Sciences, School of Life Course and Population Sciences,
Faculty of Life Sciences and Medicine, King’s
College London, London SE1 9NH, U.K.
| | - Yifan Xu
- Department
of Nutritional Sciences, School of Life Course and Population Sciences,
Faculty of Life Sciences and Medicine, King’s
College London, London SE1 9NH, U.K.
| | - Melanie Le Sayec
- Department
of Nutritional Sciences, School of Life Course and Population Sciences,
Faculty of Life Sciences and Medicine, King’s
College London, London SE1 9NH, U.K.
| | - Xinyu Yan
- Department
of Twin Research & Genetic Epidemiology, School of Life Course
and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 7EH, U.K.
| | - Tim D. Spector
- Department
of Twin Research & Genetic Epidemiology, School of Life Course
and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 7EH, U.K.
| | - Claire J. Steves
- Department
of Twin Research & Genetic Epidemiology, School of Life Course
and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 7EH, U.K.
| | - Jordana T. Bell
- Department
of Twin Research & Genetic Epidemiology, School of Life Course
and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 7EH, U.K.
| | - Kerrin S. Small
- Department
of Twin Research & Genetic Epidemiology, School of Life Course
and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 7EH, U.K.
| | - Cristina Menni
- Department
of Twin Research & Genetic Epidemiology, School of Life Course
and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 7EH, U.K.
| | - Rachel Gibson
- Department
of Nutritional Sciences, School of Life Course and Population Sciences,
Faculty of Life Sciences and Medicine, King’s
College London, London SE1 9NH, U.K.
| | - Ana Rodriguez-Mateos
- Department
of Nutritional Sciences, School of Life Course and Population Sciences,
Faculty of Life Sciences and Medicine, King’s
College London, London SE1 9NH, U.K.
| |
Collapse
|
3
|
Messeha SS, Agarwal M, Gendy SG, Mehboob SB, Soliman KFA. The Anti-Obesogenic Effects of Muscadine Grapes through Ciliary Neurotrophic Factor Receptor (Cntfr) and Histamine Receptor H1 (Hrh1) Genes in 3T3-L1 Differentiated Mouse Cells. Nutrients 2024; 16:1817. [PMID: 38931172 PMCID: PMC11206641 DOI: 10.3390/nu16121817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Obesity and type 2 diabetes are prevalent metabolic diseases that have significant links to several chronic diseases, including cancer, diabetes, hypertension, and cardiovascular disease. Muscadine grape extracts have shown the potential to reduce adiposity and improve insulin sensitivity and glucose control. Thus, this study was designed to determine the potential of muscadine grape berries extract (Pineapple and Southern Home) for its antiobesity properties in 3T3-L1 cells as a model for obesity research. The current study's data indicated the total phenolic content (TPC) and 2,2-diphenyl-1-picrylhydraziyl (DPPH) activity were higher in cultivar (CV) Southern Home, meanwhile, elevated the total flavonoid content (TFC) in Pineapple. Both extracts were safe across the tested range (0-5 mg/mL). A noticeable reduction in lipid accumulation was also found in extract-treated cells. In preadipocytes and adipocytes, the tested extracts showed significant alterations in various genes involved in glucose homeostasis and obesity. The most remarkable findings of the current study are the upregulation of two genes, Cntfr (+712.715-fold) and Hrh1 (+270.11-fold) in CV Pineapple extract-treated adipocytes 3T3-L1 and the high fold increase in Ramp3 induced by both Pineapple and Southern Home in pre-adipose cells. Furthermore, the tested extracts showed a potential to alter the mRNA of various genes, including Zfp91, B2m, Nr3c1, Insr, Atrn, Il6ra, Hsp90ab1, Sort1, and Npy1r. In conclusion, the data generated from the current study suggested that the two extracts under investigation are considered potential candidates for controlling insulin levels and managing obesity.
Collapse
Affiliation(s)
- Samia S. Messeha
- College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA;
- College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, New Pharmacy Building, 1415 ML King Blvd., Tallahassee, FL 32307, USA
| | - Meenakshi Agarwal
- Center for Viticulture & Small Fruit Research, Florida A&M University, Tallahassee, FL 32317, USA;
| | - Sherif G. Gendy
- School of Allied Health Sciences, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Sheikh B. Mehboob
- Center for Viticulture & Small Fruit Research, Florida A&M University, Tallahassee, FL 32317, USA;
| | - Karam F. A. Soliman
- College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, New Pharmacy Building, 1415 ML King Blvd., Tallahassee, FL 32307, USA
| |
Collapse
|
4
|
Vita AA, Roberts KM, Gundersen A, Farris Y, Zwickey H, Bradley R, Weir TL. Relationships between Habitual Polyphenol Consumption and Gut Microbiota in the INCLD Health Cohort. Nutrients 2024; 16:773. [PMID: 38542685 PMCID: PMC10974568 DOI: 10.3390/nu16060773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 04/01/2024] Open
Abstract
While polyphenol consumption is often associated with an increased abundance of beneficial microbes and decreased opportunistic pathogens, these relationships are not completely described for polyphenols consumed via habitual diet, including culinary herb and spice consumption. This analysis of the International Cohort on Lifestyle Determinants of Health (INCLD Health) cohort uses a dietary questionnaire and 16s microbiome data to examine relationships between habitual polyphenol consumption and gut microbiota in healthy adults (n = 96). In this exploratory analysis, microbial taxa, but not diversity measures, differed by levels of dietary polyphenol consumption. Taxa identified as exploratory biomarkers of daily polyphenol consumption (mg/day) included Lactobacillus, Bacteroides, Enterococcus, Eubacterium ventriosum group, Ruminococcus torques group, and Sutterella. Taxa identified as exploratory biomarkers of the frequency of polyphenol-weighted herb and spice use included Lachnospiraceae UCG-001, Lachnospiraceae UCG-004, Methanobrevibacter, Lachnoclostridium, and Lachnotalea. Several of the differentiating taxa carry out activities important for human health, although out of these taxa, those with previously described pro-inflammatory qualities in certain contexts displayed inverse relationships with polyphenol consumption. Our results suggest that higher quantities of habitual polyphenol consumption may support an intestinal environment where opportunistic and pro-inflammatory bacteria are represented in a lower relative abundance compared to those with less potentially virulent qualities.
Collapse
Affiliation(s)
- Alexandra Adorno Vita
- Helfgott Research Institute, National University of Natural Medicine, Portland, OR 97201, USA
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA
| | - Kristen M. Roberts
- School of Health and Rehabilitation Sciences, Ohio State University, Columbus, OH 43210, USA
| | - Anders Gundersen
- Helfgott Research Institute, National University of Natural Medicine, Portland, OR 97201, USA
| | - Yuliya Farris
- Pacific Northwest National Laboratory, Biological Sciences Division, Richland, WA 99352, USA
| | - Heather Zwickey
- Helfgott Research Institute, National University of Natural Medicine, Portland, OR 97201, USA
| | - Ryan Bradley
- Helfgott Research Institute, National University of Natural Medicine, Portland, OR 97201, USA
- Herbert Wertheim School of Public Health, University of California, San Diego, CA 92037, USA
| | - Tiffany L. Weir
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
5
|
Li Y, Xu Y, Le Roy C, Hu J, Steves CJ, Bell JT, Spector TD, Gibson R, Menni C, Rodriguez-Mateos A. Interplay between the (Poly)phenol Metabolome, Gut Microbiome, and Cardiovascular Health in Women: A Cross-Sectional Study from the TwinsUK Cohort. Nutrients 2023; 15:1900. [PMID: 37111123 PMCID: PMC10141398 DOI: 10.3390/nu15081900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/11/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Dietary (poly)phenol consumption is inversely associated with cardiovascular disease (CVD) risk in epidemiological studies, but little is known about the role of the gut microbiome in this relationship. METHODS In 200 healthy females, aged 62.0 ± 10.0 years, from the TwinsUK cohort, 114 individual (poly)phenol metabolites were measured from spot urine using ultra-high-performance liquid chromatography-mass spectrometry. The associations between metabolites, the gut microbiome (alpha diversity and genera), and cardiovascular scores were investigated using linear mixed models adjusting age, BMI, fibre, energy intake, family relatedness, and multiple testing (FDR < 0.1). RESULTS Significant associations were found between phenolic acid metabolites, CVD risk, and the gut microbiome. A total of 35 phenolic acid metabolites were associated with the Firmicutes phylum, while 5 metabolites were associated with alpha diversity (FDR-adjusted p < 0.05). Negative associations were observed between the atherosclerotic CVD (ASCVD) risk score and five phenolic acid metabolites, two tyrosol metabolites, and daidzein with stdBeta (95% (CI)) ranging from -0.05 (-0.09, -0.01) for 3-(2,4-dihydroxyphenyl)propanoic acid to -0.04 (-0.08, -0.003) for 2-hydroxycinnamic acid (FDR-adjusted p < 0.1). The genus 5-7N15 in the Bacteroidetes phylum was positively associated with the same metabolites, including 3-(3,5-dihydroxyphenyl)propanoic acid, 3-(2,4-dihydroxyphenyl)propanoic acid, 3-(3,4-dihydroxyphenyl)propanoic acid), 3-hydroxyphenylethanol-4-sulfate, and 4-hydroxyphenylethanol-3-sulfate)(stdBeta (95% CI): 0.23 (0.09, 0.36) to 0.28 (0.15, 0.42), FDR-adjusted p < 0.05), and negatively associated with the ASCVD score (stdBeta (95% CI): -0.05 (-0.09, -0.01), FDR-adjusted p = 0.02). Mediation analysis showed that genus 5-7N15 mediated 23.8% of the total effect of 3-(3,4-dihydroxyphenyl)propanoic acid on the ASCVD score. CONCLUSIONS Coffee, tea, red wine, and several vegetables and fruits, especially berries, are the most abundant food sources of phenolic acids that have the strongest associations with CVD risk. We found that the gut microbiome, particularly the genus 5-7N15, partially mediates the negative association between urinary (poly)phenols and cardiovascular risk, supporting a key role of the gut microbiome in the health benefits of dietary (poly)phenols.
Collapse
Affiliation(s)
- Yong Li
- Department of Nutritional Sciences, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| | - Yifan Xu
- Department of Nutritional Sciences, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| | - Caroline Le Roy
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| | - Jiaying Hu
- Department of Nutritional Sciences, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| | - Claire J. Steves
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| | - Jordana T. Bell
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| | - Rachel Gibson
- Department of Nutritional Sciences, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| | - Ana Rodriguez-Mateos
- Department of Nutritional Sciences, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| |
Collapse
|
6
|
Valles-Colomer M, Menni C, Berry SE, Valdes AM, Spector TD, Segata N. Cardiometabolic health, diet and the gut microbiome: a meta-omics perspective. Nat Med 2023; 29:551-561. [PMID: 36932240 PMCID: PMC11258867 DOI: 10.1038/s41591-023-02260-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/16/2023] [Indexed: 03/19/2023]
Abstract
Cardiometabolic diseases have become a leading cause of morbidity and mortality globally. They have been tightly linked to microbiome taxonomic and functional composition, with diet possibly mediating some of the associations described. Both the microbiome and diet are modifiable, which opens the way for novel therapeutic strategies. High-throughput omics techniques applied on microbiome samples (meta-omics) hold the unprecedented potential to shed light on the intricate links between diet, the microbiome, the metabolome and cardiometabolic health, with a top-down approach. However, effective integration of complementary meta-omic techniques is an open challenge and their application on large cohorts is still limited. Here we review meta-omics techniques and discuss their potential in this context, highlighting recent large-scale efforts and the novel insights they provided. Finally, we look to the next decade of meta-omics research and discuss various translational and clinical pathways to improving cardiometabolic health.
Collapse
Affiliation(s)
- Mireia Valles-Colomer
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Cristina Menni
- Department of Twin Research, King's College London, London, UK
| | - Sarah E Berry
- Department of Nutritional Sciences, King's College London, London, UK
| | - Ana M Valdes
- School of Medicine, University of Nottingham, Nottingham, UK
- Nottingham National Institute for Health Research Biomedical Research Centre, Nottingham, UK
| | - Tim D Spector
- Department of Twin Research, King's College London, London, UK
| | - Nicola Segata
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy.
- European Institute of Oncology, Scientific Institute for Research, Hospitalization and Healthcare, Milan, Italy.
| |
Collapse
|
7
|
The effects of Aronia berry polyphenol supplementation on arterial function and the gut microbiome in middle aged men and women: Results from a randomized controlled trial. Clin Nutr 2022; 41:2549-2561. [DOI: 10.1016/j.clnu.2022.08.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 08/13/2022] [Accepted: 08/22/2022] [Indexed: 11/20/2022]
|
8
|
Turpin W, Dong M, Sasson G, Raygoza Garay JA, Espin-Garcia O, Lee SH, Neustaeter A, Smith MI, Leibovitzh H, Guttman DS, Goethel A, Griffiths AM, Huynh HQ, Dieleman LA, Panaccione R, Steinhart AH, Silverberg MS, Aumais G, Jacobson K, Mack D, Murthy SK, Marshall JK, Bernstein CN, Abreu MT, Moayyedi P, Paterson AD, Xu W, Croitoru K. Mediterranean-Like Dietary Pattern Associations With Gut Microbiome Composition and Subclinical Gastrointestinal Inflammation. Gastroenterology 2022; 163:685-698. [PMID: 35643175 DOI: 10.1053/j.gastro.2022.05.037] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Case-control studies have shown that patients with Crohn's disease (CD) have a microbial composition different from healthy individuals. Although the causes of CD are unknown, epidemiologic studies suggest that diet is an important contributor to CD risk, potentially via modulation of bacterial composition and gut inflammation. We hypothesized that long-term dietary clusters (DCs) are associated with gut microbiome compositions and gut inflammation. Our objectives were to identify dietary patterns and assess whether they are associated with alterations in specific gut microbial compositions and subclinical levels of gut inflammation in a cohort of healthy first-degree relatives (FDRs) of patients with CD. METHODS As part of the Genetic, Environmental, Microbial (GEM) Project, we recruited a cohort of 2289 healthy FDRs of patients with CD. Individuals provided stool samples and answered a validated food frequency questionnaire reflecting their habitual diet during the year before sample collection. Unsupervised analysis identified 3 dietary and 3 microbial composition clusters. RESULTS DC3, resembling the Mediterranean diet, was strongly associated with a defined microbial composition, with an increased abundance of fiber-degrading bacteria, such as Ruminococcus, as well as taxa such as Faecalibacterium. The DC3 diet was also significantly associated with lower levels of subclinical gut inflammation, defined by fecal calprotectin, compared with other dietary patterns. No significant associations were found between individual food items and fecal calprotectin, suggesting that long-term dietary patterns rather than individual food items contribute to subclinical gut inflammation. Additionally, mediation analysis demonstrated that DC3 had a direct effect on subclinical inflammation that was partially mediated by the microbiota. CONCLUSIONS Overall, these results indicated that Mediterranean-like dietary patterns are associated with microbiome and lower intestinal inflammation. This study will help guide future dietary strategies that affect microbial composition and host gut inflammation to prevent diseases.
Collapse
Affiliation(s)
- Williams Turpin
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Mei Dong
- Department of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Gila Sasson
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Juan Antonio Raygoza Garay
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Osvaldo Espin-Garcia
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, and Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Sun-Ho Lee
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Anna Neustaeter
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Michelle I Smith
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Haim Leibovitzh
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - David S Guttman
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, Canada; Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
| | - Ashleigh Goethel
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Anne M Griffiths
- Division of Gastroenterology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Hien Q Huynh
- Division of Gastroenterology and Nutrition, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Levinus A Dieleman
- Division of Gastroenterology and the Centre of Excellence for Gastrointestinal Inflammation and Immunity Research (CEGIIR), Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Remo Panaccione
- Inflammatory Bowel Disease Clinic, Division of Gastroenterology and Hepatology, University of Calgary, Calgary, Alberta, Canada
| | - A Hillary Steinhart
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, and Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Mark S Silverberg
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Guy Aumais
- Department of Medicine, Hôpital Maisonneuve-Rosemont, Montreal University, Montreal, Quebec, Canada
| | - Kevan Jacobson
- Canadian Gastro-Intestinal Epidemiology Consortium (CanGIEC); British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - David Mack
- Division of Gastroenterology, Hepatology & Nutrition, Children's Hospital of Eastern Ontario and University of Ottawa, Ottawa, Ontario, Canada
| | - Sanjay K Murthy
- The Ottawa Hospital Inflammatory Bowel Disease Centre, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - John K Marshall
- Department of Medicine, McMaster University, Farncombe Family Digestive Health Research Institute, Hamilton, Ontario, Canada
| | - Charles N Bernstein
- Inflammatory Bowel Disease Clinical and Research Centre, and Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Maria T Abreu
- Department of Medicine, Crohn's and Colitis Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Paul Moayyedi
- Department of Medicine, McMaster University, Farncombe Family Digestive Health Research Institute, Hamilton, Ontario, Canada
| | - Andrew D Paterson
- Department of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada; Genetics and Genome Biology, The Hospital for Sick Children Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Wei Xu
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, and Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Kenneth Croitoru
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
9
|
Abstract
Epidemiological studies have shown that higher intake of flavonoid is inversely associated with CHD risk. However, which flavonoid subclass could reduce CHD risk has remained controversial. The present meta-analysis of prospective cohort studies aimed to quantitatively assess the associations between flavonoid subclasses and CHD risk. A systematic literature search was implemented from PubMed and Web of Science databases up to March 2021, and eligible studies were identified. Multivariate-adjust relative risks (RR) with corresponding 95 % CI were pooled by using a random-effects model. A restricted cubic spline regression model was performed for non-linear dose-response analysis. A total of 19 independent prospective cohort studies with 894 471 participants and 34 707 events were included. The results showed that dietary intakes of anthocyanins (RR = 0·90; 95 % CI: 0·83, 0·98), proanthocyanidins (RR = 0·78; 95 % CI: 0·65, 0·94), flavonols (RR = 0·88; 95 % CI: 0·79, 0·98), flavones (RR = 0·94; 95 % CI: 0·89, 0·99) and isoflavones (RR = 0·90; 95 % CI: 0·83, 0·98) were negatively associated with CHD risk. Dose-response analysis showed that increment of 50 mg/d anthocyanins, 100 mg/d proanthocyanidins, 25 mg/d flavonols, 5 mg/d flavones and 0·5 mg/d isoflavones were associated with 5 % reduction in CHD risk, respectively. Sensitivity and subgroup analyses were used to further support these associations. The present results indicate that dietary intakes of fruits and vegetables abundant five flavonoid subclasses, namely anthocyanins, proanthocyanidins, flavonols, flavones and isoflavones, are associated with a lower risk of CHD.
Collapse
|
10
|
Chen L, Pu Y, Xu Y, He X, Cao J, Ma Y, Jiang W. Anti-diabetic and anti-obesity: Efficacy evaluation and exploitation of polyphenols in fruits and vegetables. Food Res Int 2022; 157:111202. [DOI: 10.1016/j.foodres.2022.111202] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023]
|
11
|
Lancaster SM, Lee-McMullen B, Abbott CW, Quijada JV, Hornburg D, Park H, Perelman D, Peterson DJ, Tang M, Robinson A, Ahadi S, Contrepois K, Hung CJ, Ashland M, McLaughlin T, Boonyanit A, Horning A, Sonnenburg JL, Snyder MP. Global, distinctive, and personal changes in molecular and microbial profiles by specific fibers in humans. Cell Host Microbe 2022; 30:848-862.e7. [PMID: 35483363 PMCID: PMC9187607 DOI: 10.1016/j.chom.2022.03.036] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/19/2022] [Accepted: 03/25/2022] [Indexed: 12/11/2022]
Abstract
Dietary fibers act through the microbiome to improve cardiovascular health and prevent metabolic disorders and cancer. To understand the health benefits of dietary fiber supplementation, we investigated two popular purified fibers, arabinoxylan (AX) and long-chain inulin (LCI), and a mixture of five fibers. We present multiomic signatures of metabolomics, lipidomics, proteomics, metagenomics, a cytokine panel, and clinical measurements on healthy and insulin-resistant participants. Each fiber is associated with fiber-dependent biochemical and microbial responses. AX consumption associates with a significant reduction in LDL and an increase in bile acids, contributing to its observed cholesterol reduction. LCI is associated with an increase in Bifidobacterium. However, at the highest LCI dose, there is increased inflammation and elevation in the liver enzyme alanine aminotransferase. This study yields insights into the effects of fiber supplementation and the mechanisms behind fiber-induced cholesterol reduction, and it shows effects of individual, purified fibers on the microbiome.
Collapse
Affiliation(s)
- Samuel M Lancaster
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Brittany Lee-McMullen
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Charles Wilbur Abbott
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Jeniffer V Quijada
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Daniel Hornburg
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Heyjun Park
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Dalia Perelman
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Dylan J Peterson
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Michael Tang
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Aaron Robinson
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Sara Ahadi
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Chia-Jui Hung
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Melanie Ashland
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Tracey McLaughlin
- Division of Endocrinology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Anna Boonyanit
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Aaron Horning
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Justin L Sonnenburg
- Department of Microbiology & Immunology, Stanford School of Medicine, Stanford, CA 94305, USA; Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Michael P Snyder
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
12
|
Mladěnka P. Special Issue “Dietary (Poly)Phenols and Health”. Nutrients 2022; 14:nu14071402. [PMID: 35406015 PMCID: PMC9003021 DOI: 10.3390/nu14071402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 01/31/2022] [Indexed: 12/04/2022] Open
Affiliation(s)
- Přemysl Mladěnka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Ak. Heyrovského 1203, 50005 Hradec Králové, Czech Republic
| |
Collapse
|
13
|
Han S, Chen M, Cheng P, Zhang Z, Lu Y, Xu Y, Wang Y. A systematic review and meta-analysis of gut microbiota in diabetic kidney disease: Comparisons with diabetes mellitus, non-diabetic kidney disease, and healthy individuals. Front Endocrinol (Lausanne) 2022; 13:1018093. [PMID: 36339429 PMCID: PMC9633273 DOI: 10.3389/fendo.2022.1018093] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Gut microbiota has been reported to play an important role in diabetic kidney disease (DKD), however, the alterations of gut bacteria have not been determined. METHODS Studies comparing the differences of gut microbiome between patients with DKD and non-DKD individuals using high-throughput sequencing technology, were systematically searched and reviewed. Outcomes were set as gut bacterial diversity, microbial composition, and correlation with clinical parameters of DKD. Qualitative data were summarized and compared through a funnel R script, and quantitative data were estimated by meta-analysis. RESULTS A total of 15 studies and 1640 participants were included, the comparisons were conducted between DKD, diabetes mellitus (DM), non-diabetic kidney disease (NDKD), and healthy controls. There were no significant differences of α-diversity between DKD and DM, and between DKD and NDKD, however, significant lower microbial richness was found in DKD compared to healthy controls. Different bacterial compositions were found between DKD and non-DKD subjects. The phylum Actinobacteria were found to be enriched in DKD compared to healthy controls. At the genus level, we found the enrichment of Hungatella, Bilophila, and Escherichia in DKD compared to DM, patients with DKD showed lower abundances of Faecalibacterium compared to those with NDKD. The genera Butyricicoccus, Faecalibacterium, and Lachnospira were depleted in DKD compared to healthy controls, whereas Hungatella, Escherichia, and lactobacillus were significantly enriched. The genus Ruminococcus torques group was demonstrated to be inversely correlated with estimated glomerular filtration rate of DKD. CONCLUSIONS Gut bacterial alterations was demonstrated in DKD, characterized by the enrichment of the genera Hungatella and Escherichia, and the depletion of butyrate-producing bacteria, which might be associated with the occurrence and development of DKD. Further studies are still needed to validate these findings, due to substantial heterogeneity. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/prospero/, identifier CRD42022340870.
Collapse
Affiliation(s)
- Shisheng Han
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Min Chen
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pei Cheng
- Department of Hemodialysis, Lin’an Third People’s Hospital, Hangzhou, Zhejiang, China
| | - Zeng Zhang
- Department of Endocrine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Lu
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanqiu Xu
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Yanqiu Xu, ; Yi Wang,
| | - Yi Wang
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Yanqiu Xu, ; Yi Wang,
| |
Collapse
|
14
|
Polyphenols-Gut Microbiota Interrelationship: A Transition to a New Generation of Prebiotics. Nutrients 2021; 14:nu14010137. [PMID: 35011012 PMCID: PMC8747136 DOI: 10.3390/nu14010137] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 12/21/2021] [Accepted: 12/24/2021] [Indexed: 12/18/2022] Open
Abstract
The present review summarizes the studies carried out on this topic in the last five years. According to the new definitions, among all the compounds included in the group of prebiotics, polyphenols are probably the most important secondary metabolites produced by the plant kingdom. Many of these types of polyphenols have low bioavailability, therefore reaching the colon in unaltered form. Once in the colon, these compounds interact with the intestinal microbes bidirectionally by modulating them and, consequently, releasing metabolites. Despite much research on various metabolites, little is known about the chemistry of the metabolic routes used by different bacteria species. In this context, this review aims to investigate the prebiotic effect of polyphenols in preclinical and clinical studies, highlighting that the consumption of polyphenols leads to an increase in beneficial bacteria, as well as an increase in the production of valuable metabolites. In conclusion, there is much evidence in preclinical studies supporting the prebiotic effect of polyphenols, but further clinical studies are needed to investigate this effect in humans.
Collapse
|
15
|
Lu H, You Y, Zhou X, He Q, Wang M, Chen L, Zhou L, Sun X, Liu Y, Jiang P, Dai J, Fu X, Kwan HY, Zhao X, Lou L. Citrus reticulatae pericarpium Extract Decreases the Susceptibility to HFD-Induced Glycolipid Metabolism Disorder in Mice Exposed to Azithromycin in Early Life. Front Immunol 2021; 12:774433. [PMID: 34868039 PMCID: PMC8640250 DOI: 10.3389/fimmu.2021.774433] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 11/03/2021] [Indexed: 12/17/2022] Open
Abstract
Background Studies have shown that gut microbe disorder in mice due to early-life antibiotic exposure promotes glycolipid metabolism disorder in adulthood. However, the underlying mechanism remains unclear and there is not yet an effective intervention or treatment for this process. Purpose The study investigated whether early-life azithromycin (AZT) exposure in mice could promote high-fat diet (HFD)-induced glycolipid metabolism disorder in adulthood. Moreover, the effect of citrus reticulata pericarpium (CRP) extract on glycolipid metabolism disorder via regulation of gut microbiome in mice exposed to antibodies early in life were investigated. Methods and Results Three-week-old mice were treated with AZT (50 mg/kg/day) via drinking water for two weeks and then were fed a CRP diet (1% CRP extract) for four weeks and an HFD for five weeks. The results showed that early-life AZT exposure promoted HFD-induced glycolipid metabolism disorder, increased the levels of inflammatory factors, promoted the flora metabolism product trimethylamine N-oxide (TMAO), and induced microbial disorder in adult mice. Importantly, CRP extract mitigated these effects. Conclusion Taken together, these findings suggest that early-life AZT exposure increases the susceptibility to HFD-induced glycolipid metabolism disorder in adult mice, and CRP extract can decrease this susceptibility by regulating gut microbiome.
Collapse
Affiliation(s)
- Hanqi Lu
- Department of Traditional Chinese Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Yanting You
- Department of Traditional Chinese Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Xinghong Zhou
- Department of Traditional Chinese Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Qiuxing He
- Department of Traditional Chinese Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Ming Wang
- Department of Traditional Chinese Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Liqian Chen
- School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lin Zhou
- Endocrinology Department, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaomin Sun
- School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yanyan Liu
- School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Pingping Jiang
- School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jiaojiao Dai
- School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiuqiong Fu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Hiu Yee Kwan
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Xiaoshan Zhao
- Department of Traditional Chinese Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Linjie Lou
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
16
|
A High Protein Diet Is More Effective in Improving Insulin Resistance and Glycemic Variability Compared to a Mediterranean Diet-A Cross-Over Controlled Inpatient Dietary Study. Nutrients 2021; 13:nu13124380. [PMID: 34959931 PMCID: PMC8707429 DOI: 10.3390/nu13124380] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/26/2022] Open
Abstract
The optimal dietary pattern to improve metabolic function remains elusive. In a 21-day randomized controlled inpatient crossover feeding trial of 20 insulin-resistant obese women, we assessed the extent to which two isocaloric dietary interventions—Mediterranean (M) and high protein (HP)—improved metabolic parameters. Obese women were assigned to one of the following dietary sequences: M–HP or HP–M. Cardiometabolic parameters, body weight, glucose monitoring and gut microbiome composition were assessed. Sixteen women completed the study. Compared to the M diet, the HP diet was more effective in (i) reducing insulin resistance (insulin: Beta (95% CI) = −6.98 (−12.30, −1.65) µIU/mL, p = 0.01; HOMA-IR: −1.78 (95% CI: −3.03, −0.52), p = 9 × 10−3); and (ii) improving glycemic variability (−3.13 (−4.60, −1.67) mg/dL, p = 4 × 10−4), a risk factor for T2D development. We then identified a panel of 10 microbial genera predictive of the difference in glycemic variability between the two diets. These include the genera Coprococcus and Lachnoclostridium, previously associated with glucose homeostasis and insulin resistance. Our results suggest that morbidly obese women with insulin resistance can achieve better control of insulin resistance and glycemic variability on a high HP diet compared to an M diet.
Collapse
|
17
|
Berding K, Vlckova K, Marx W, Schellekens H, Stanton C, Clarke G, Jacka F, Dinan TG, Cryan JF. Diet and the Microbiota-Gut-Brain Axis: Sowing the Seeds of Good Mental Health. Adv Nutr 2021; 12:1239-1285. [PMID: 33693453 PMCID: PMC8321864 DOI: 10.1093/advances/nmaa181] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
Over the past decade, the gut microbiota has emerged as a key component in regulating brain processes and behavior. Diet is one of the major factors involved in shaping the gut microbiota composition across the lifespan. However, whether and how diet can affect the brain via its effects on the microbiota is only now beginning to receive attention. Several mechanisms for gut-to-brain communication have been identified, including microbial metabolites, immune, neuronal, and metabolic pathways, some of which could be prone to dietary modulation. Animal studies investigating the potential of nutritional interventions on the microbiota-gut-brain axis have led to advancements in our understanding of the role of diet in this bidirectional communication. In this review, we summarize the current state of the literature triangulating diet, microbiota, and host behavior/brain processes and discuss potential underlying mechanisms. Additionally, determinants of the responsiveness to a dietary intervention and evidence for the microbiota as an underlying modulator of the effect of diet on brain health are outlined. In particular, we emphasize the understudied use of whole-dietary approaches in this endeavor and the need for greater evidence from clinical populations. While promising results are reported, additional data, specifically from clinical cohorts, are required to provide evidence-based recommendations for the development of microbiota-targeted, whole-dietary strategies to improve brain and mental health.
Collapse
Affiliation(s)
| | | | - Wolfgang Marx
- Deakin University, iMPACT – the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, VIC,Australia
| | - Harriet Schellekens
- APC Microbiome Ireland, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Sciences, University College Cork, Cork, Ireland
| | - Felice Jacka
- Deakin University, iMPACT – the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, VIC,Australia
- Centre for Adolescent Health, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Black Dog Institute, Randwick, NSW, Australia
- College of Public Health, Medical & Veterinary Sciences, James Cook University, Douglas, QLD, Australia
| | - Timothy G Dinan
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Sciences, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
18
|
Relationship between Nutrient Intake and Human Gut Microbiota in Monozygotic Twins. ACTA ACUST UNITED AC 2021; 57:medicina57030275. [PMID: 33809761 PMCID: PMC8002349 DOI: 10.3390/medicina57030275] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/12/2021] [Accepted: 03/12/2021] [Indexed: 02/07/2023]
Abstract
Background and Objectives: The gut microbiota is associated with human health and dietary nutrition. Various studies have been reported in this regard, but it is difficult to clearly analyze human gut microbiota as individual differences are significant. The causes of these individual differences in intestinal microflora are genetic and/or environmental. In this study, we focused on differences between identical twins in Japan to clarify the effects of nutrients consumed on the entire gut microbiome, while excluding genetic differences. Materials and Methods: We selected healthy Japanese monozygotic twins for the study and confirmed their zygosity by matching 15 short tandem repeat loci. Their fecal samples were subjected to 16S rRNA sequencing and bioinformatics analyses to identify and compare the fluctuations in intestinal bacteria. Results: We identified 12 genera sensitive to environmental factors, and found that Lactobacillus was relatively unaffected by environmental factors. Moreover, we identified protein, fat, and some nutrient intake that can affect 12 genera, which have been identified to be more sensitive to environmental factors. Among the 12 genera, Bacteroides had a positive correlation with retinol equivalent intake (rs = 0.38), Lachnospira had a significantly negative correlation with protein, sodium, iron, vitamin D, vitamin B6, and vitamin B12 intake (rs = −0.38, −0.41, −0.39, −0.63, −0.42, −0.49, respectively), Lachnospiraceae ND3007 group had a positive correlation with fat intake (rs = 0.39), and Lachnospiraceae UCG-008 group had a negative correlation with the saturated fatty acid intake (rs = −0.45). Conclusions: Our study is the first to focus on the relationship between human gut microbiota and nutrient intake using samples from Japanese twins to exclude the effects of genetic factors. These findings will broaden our understanding of the more intuitive relationship between nutrient intake and the gut microbiota and can be a useful basis for finding useful biomarkers that contribute to human health.
Collapse
|
19
|
High intake of vegetables is linked to lower white blood cell profile and the effect is mediated by the gut microbiome. BMC Med 2021; 19:37. [PMID: 33568158 PMCID: PMC7875684 DOI: 10.1186/s12916-021-01913-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/14/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Chronic inflammation, which can be modulated by diet, is linked to high white blood cell counts and correlates with higher cardiometabolic risk and risk of more severe infections, as in the case of COVID-19. METHODS Here, we assessed the association between white blood cell profile (lymphocytes, basophils, eosinophils, neutrophils, monocytes and total white blood cells) as markers of chronic inflammation, habitual diet and gut microbiome composition (determined by sequencing of the 16S RNA) in 986 healthy individuals from the PREDICT-1 nutritional intervention study. We then investigated whether the gut microbiome mediates part of the benefits of vegetable intake on lymphocyte counts. RESULTS Higher levels of white blood cells, lymphocytes and basophils were all significantly correlated with lower habitual intake of vegetables, with vegetable intake explaining between 3.59 and 6.58% of variation in white blood cells after adjusting for covariates and multiple testing using false discovery rate (q < 0.1). No such association was seen with fruit intake. A mediation analysis found that 20.00% of the effect of vegetable intake on lymphocyte counts was mediated by one bacterial genus, Collinsella, known to increase with the intake of processed foods and previously associated with fatty liver disease. We further correlated white blood cells to other inflammatory markers including IL6 and GlycA, fasting and post-prandial glucose levels and found a significant relationship between inflammation and diet. CONCLUSION A habitual diet high in vegetables, but not fruits, is linked to a lower inflammatory profile for white blood cells, and a fifth of the effect is mediated by the genus Collinsella. TRIAL REGISTRATION The ClinicalTrials.gov registration identifier is NCT03479866 .
Collapse
|
20
|
Lesser Investigated Natural Ingredients for the Management of Obesity. Nutrients 2021; 13:nu13020510. [PMID: 33557185 PMCID: PMC7913945 DOI: 10.3390/nu13020510] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/30/2021] [Accepted: 01/31/2021] [Indexed: 12/13/2022] Open
Abstract
Obesity, an epidemiological disorder, is related to various complications in both the developed and developing world. It epitomizes a crucial risk factor for health, decreasing productivity and life expectancy while increasing health care costs worldwide. Conventional therapies with synthetic drugs or bariatric surgery, associated with numerous side effects, recurrence, and surgical complexity, have been restricted in their use. Lifestyle changes and dietary restrictions are the proven methods for successful weight loss, although maintaining a strict lifestyle is a challenge. Multiple natural products have been explored for weight management with varied efficacy. The current review explores less explored natural herbs, their active constituents, and their mechanisms of action against obesity.
Collapse
|