101
|
Wu T, Shen M, Yu Q, Chen Y, Chen X, Yang J, Huang L, Guo X, Xie J. Cyclocarya paliurus polysaccharide improves metabolic function of gut microbiota by regulating short-chain fatty acids and gut microbiota composition. Food Res Int 2021; 141:110119. [PMID: 33641986 DOI: 10.1016/j.foodres.2021.110119] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 01/03/2021] [Accepted: 01/03/2021] [Indexed: 01/16/2023]
Abstract
The purpose of this paper was to investigate the effects of Cyclocarya paliurus polysaccharide (CP) on gut microbiota composition and predict metabolic function in healthy mice. Healthy Kunming mice were continuously gavaged with CP for 20 days, and mouse feces were collected for analysis. The results showed that CP could remarkably increase the short-chain fatty acids (SCFAs; acetic acid, propionic acid, butyric acid, and valeric acid) in the feces of healthy mice in a dose-dependent matter. 16S rRNA showed that 200 mg/kg body weight CP was effective in increasing diversity of the gut microbiota in healthy mice and affected the relative abundance of specific bacteria. Lachnospiraceae, Clostridiales, and Clostridia were identified as the phenotypic biomarkers of the CP-H group compared with the normal control group. In addition, PICRUSt2 showed that starch and sucrose metabolism, amino acid metabolism, glycerolipid metabolism, pantothenate and CoA biosynthesis, biosynthesis of unsaturated fatty acids, and C5-branched dibasic acid metabolism are the primary enriched phenotypic KEGG pathways in the CP-H group. These findings suggested that early CP intervention could enhance the metabolic function of gut microbiota by increasing the release of SCFAs and altering the composition of gut microbiota.
Collapse
Affiliation(s)
- Ting Wu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Mingyue Shen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China.
| | - Qiang Yu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Yi Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Xianxiang Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Jun Yang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Lixin Huang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Xiaomei Guo
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Jianhua Xie
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China.
| |
Collapse
|
102
|
Ramos AC, Carraso HVCJ, Bastos ELDS. Bariatric Procedures: Anatomical and Physiological Changes. MANAGEMENT OF NUTRITIONAL AND METABOLIC COMPLICATIONS OF BARIATRIC SURGERY 2021:41-67. [DOI: 10.1007/978-981-33-4702-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
103
|
Ghosh A, Firdous S, Saha S. Bioinformatics for Human Microbiome. Adv Bioinformatics 2021. [DOI: 10.1007/978-981-33-6191-1_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
104
|
Desai V, Kozyrskyj AL, Lau S, Sanni O, Dennett L, Walter J, Ospina MB. Effectiveness of Probiotic, Prebiotic, and Synbiotic Supplementation to Improve Perinatal Mental Health in Mothers: A Systematic Review and Meta-Analysis. Front Psychiatry 2021; 12:622181. [PMID: 33967849 PMCID: PMC8100186 DOI: 10.3389/fpsyt.2021.622181] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/22/2021] [Indexed: 12/16/2022] Open
Abstract
Introduction: There is an emerging interest in modulating the gut microbiota to target the gut-brain axis and improve maternal mental health in the perinatal period. This systematic review evaluated the effectiveness of prebiotics, probiotics, and synbiotics supplementation during pregnancy to reduce the risk of maternal mental health problems in the perinatal period. Methods: Electronic biomedical databases and clinical trial registries were searched from database inception through August 2020 to identify randomized controlled clinical trials (RCTs) evaluating the effect of probiotic, prebiotic, or synbiotic supplements administered to women during pregnancy on measures of perinatal depression, anxiety, and other mental health outcomes. Study selection, risk of bias appraisal, and data extraction were independently performed by two reviewers. Pooled mean differences (MD) and odds ratios (pOR) with 95% confidence intervals (CI) were calculated in random-effects meta-analyses for the outcomes of interest in the review. Results: From 3,868 studies identified through the search strategy, three RCTs of low risk of bias involving 713 participants were included, all three testing probiotics. There were no differences between probiotics and control groups in the mean depression scores (MD -0.46; 95% CI -2.16, 1.25) at end of follow-up. Although statistical significance was not achieved, probiotics showed an advantage in the proportion of participants scoring below an established cut-off for depression (pOR 0.68; 95% CI 0.43, 1.07). Compared to placebo, probiotics in pregnancy reduced anxiety symptoms (MD -0.99; 95% CI -1.80, -0.18); however, this advantage was not translated in a reduction in the proportion of participants scoring above an established cut-off for anxiety (pOR 0.65; 95% CI 0.23, 1.85). There were no differences between probiotics and control groups in global mental health scores at end of follow-up (MD 1.09; 95% CI -2.04, 4.22). Conclusion: There is limited but promising evidence about the effectiveness of probiotics during pregnancy to reduce anxiety symptoms and reduce the proportion of women scoring ABOVE a cut-off depression score. There is a lack of RCT evidence supporting prebiotics and synbiotics supplementation for similar purposes in the perinatal period. More research is needed before prebiotics, probiotics, and synbiotics are recommended to support maternal mental health and well-being in the perinatal period. Systematic Review Registration: PROSPERO, CRD42019137158.
Collapse
Affiliation(s)
- Vidhi Desai
- Department of Obstetrics & Gynecology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Anita L Kozyrskyj
- Department of Obstetrics & Gynecology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,School of Public Health, University of Alberta, Edmonton, AB, Canada
| | - Stuart Lau
- Faculty of Science, McGill University, Montreal, QC, Canada
| | - Omolara Sanni
- Department of Obstetrics & Gynecology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Liz Dennett
- John W. Scott Health Sciences Library, University of Alberta, Edmonton, AB, Canada
| | - Jens Walter
- Department of Medicine, School of Microbiology, University College Cork, National University of Ireland, Cork, Ireland.,APC Microbiome Institute Ireland, University College Cork, National University of Ireland, Cork, Ireland
| | - Maria B Ospina
- Department of Obstetrics & Gynecology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,School of Public Health, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
105
|
Jia R, Huang M, Qian L, Yan X, Lv Q, Ye H, Ye L, Wu X, Chen W, Chen Y, Jia Y, Huang Y, Wu H. The Depletion of Carbohydrate Metabolic Genes in the Gut Microbiome Contributes to the Transition From Central Obesity to Type 2 Diabetes. Front Endocrinol (Lausanne) 2021; 12:747646. [PMID: 34745012 PMCID: PMC8569854 DOI: 10.3389/fendo.2021.747646] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/21/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity, especially central obesity, is a strong risk factor for developing type 2 diabetes (T2D). However, the mechanism underlying the progression from central obesity to T2D remains unknown. Therefore, we analyzed the gut microbial profiles of central obese individuals with or without T2D from a Chinese population. Here we reported both the microbial compositional and gene functional alterations during the progression from central obesity to T2D. Several opportunistic pathogens were enriched in obese T2D patients. We also characterized thousands of genes involved in sugar and amino acid metabolism whose abundance were significantly depleted in obese T2D group. Moreover, the abundance of those genes was negatively associated with plasma glycemia level and percentage of individuals with impaired plasma glucose status. Therefore, our study indicates that the abundance of those depleted genes can be used as a potential biomarker to identify central obese individuals with high risks of developing T2D.
Collapse
MESH Headings
- Adult
- Biomarkers/metabolism
- Carbohydrate Metabolism/genetics
- Case-Control Studies
- China
- Diabetes Mellitus, Type 2/etiology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/microbiology
- Disease Progression
- Disease Susceptibility
- Female
- Gastrointestinal Microbiome/genetics
- Humans
- Male
- Metagenome/physiology
- Obesity, Abdominal/genetics
- Obesity, Abdominal/metabolism
- Obesity, Abdominal/microbiology
- Obesity, Abdominal/pathology
- RNA, Ribosomal, 16S/analysis
- RNA, Ribosomal, 16S/genetics
- Risk Factors
- Transcriptome
Collapse
Affiliation(s)
| | - Min Huang
- Department of General Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | | | - Xiaoye Yan
- Department of General Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Qing Lv
- Department of General Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Hua Ye
- GENEWIZ Inc., Suzhou, China
| | - Li Ye
- GENEWIZ Inc., Suzhou, China
| | - Xin Wu
- GENEWIZ Inc., Suzhou, China
| | | | | | | | - Yueqing Huang
- Department of General Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- *Correspondence: Yueqing Huang, ; Huihui Wu,
| | - Huihui Wu
- GENEWIZ Inc., Suzhou, China
- *Correspondence: Yueqing Huang, ; Huihui Wu,
| |
Collapse
|
106
|
Samanta S. Potential Impacts of Prebiotics and Probiotics in Cancer Prevention. Anticancer Agents Med Chem 2020; 22:605-628. [PMID: 33305713 DOI: 10.2174/1871520621999201210220442] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/15/2020] [Accepted: 10/26/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cancer is a serious problem throughout the world. The pathophysiology of cancer is multifactorial and is also related to gut microbiota. Intestinal microbes are the useful resident of the healthy human. They play various aspects of human health including nutritional biotransformation, flushing of the pathogens, toxin neutralization, immune response, and onco-suppression. Disruption in the interactions among the gut microbiota, intestinal epithelium, and the host immune system are associated with gastrointestinal disorders, neurodegenerative diseases, metabolic syndrome, and cancer. Probiotic bacteria (Lactobacillus spp., Bifidobacterium spp.) have been regarded as beneficial to health and shown to play a significant role in immunomodulation and displayed preventive role against obesity, diabetes, liver disease, inflammatory bowel disease, tumor progression, and cancer. OBJECTIVE The involvement of gut microorganisms in cancer development and prevention has been recognized as a balancing factor. The events of dysbiosis emphasize metabolic disorder and carcinogenesis. The gut flora potentiates immunomodulation and minimizes the limitations of usual chemotherapy. The significant role of prebiotics and probiotics on the improvement of immunomodulation and antitumor properties has been considered. METHODS I had reviewed the literature on the multidimensional activities of prebiotics and probiotics from the NCBI website database PubMed, Springer Nature, Science Direct (Elsevier), Google Scholar database to search relevant articles. Specifically, I had focused on the role of prebiotics and probiotics in immunomodulation and cancer prevention. RESULTS Prebiotics are the nondigestible fermentable sugars that selectively influence the growth of probiotic organisms that exert immunomodulation over the cancerous growth. The oncostatic properties of bacteria are mediated through the recruitment of cytotoxic T cells, natural killer cells, and oxidative stress-induced apoptosis in the tumor microenvironment. Moreover, approaches have also been taken to use probiotics as an adjuvant in cancer therapy. CONCLUSION The present review has indicated that dysbiosis is the crucial factor in many pathological situations including cancer. Applications of prebiotics and probiotics exhibit the immune-surveillance as oncostatic effects. These events increase the possibilities of new therapeutic strategies for cancer prevention.
Collapse
Affiliation(s)
- Saptadip Samanta
- Department of Physiology, Midnapore College, Midnapore, Paschim Medinipur, 721101, West Bengal,. India
| |
Collapse
|
107
|
Selber-Hnatiw S, Sultana T, Tse W, Abdollahi N, Abdullah S, Al Rahbani J, Alazar D, Alrumhein NJ, Aprikian S, Arshad R, Azuelos JD, Bernadotte D, Beswick N, Chazbey H, Church K, Ciubotaru E, D'Amato L, Del Corpo T, Deng J, Di Giulio BL, Diveeva D, Elahie E, Frank JGM, Furze E, Garner R, Gibbs V, Goldberg-Hall R, Goldman CJ, Goltsios FF, Gorjipour K, Grant T, Greco B, Guliyev N, Habrich A, Hyland H, Ibrahim N, Iozzo T, Jawaheer-Fenaoui A, Jaworski JJ, Jhajj MK, Jones J, Joyette R, Kaudeer S, Kelley S, Kiani S, Koayes M, Kpata AJAAL, Maingot S, Martin S, Mathers K, McCullogh S, McNamara K, Mendonca J, Mohammad K, Momtaz SA, Navaratnarajah T, Nguyen-Duong K, Omran M, Ortiz A, Patel A, Paul-Cole K, Plaisir PA, Porras Marroquin JA, Prevost A, Quach A, Rafal AJ, Ramsarun R, Rhnima S, Rili L, Safir N, Samson E, Sandiford RR, Secondi S, Shahid S, Shahroozi M, Sidibé F, Smith M, Sreng Flores AM, Suarez Ybarra A, Sénéchal R, Taifour T, Tang L, Trapid A, Tremblay Potvin M, Wainberg J, Wang DN, Weissenberg M, White A, Wilkinson G, Williams B, Wilson JR, Zoppi J, Zouboulakis K, Gamberi C. Metabolic networks of the human gut microbiota. MICROBIOLOGY-SGM 2020; 166:96-119. [PMID: 31799915 DOI: 10.1099/mic.0.000853] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The human gut microbiota controls factors that relate to human metabolism with a reach far greater than originally expected. Microbial communities and human (or animal) hosts entertain reciprocal exchanges between various inputs that are largely controlled by the host via its genetic make-up, nutrition and lifestyle. The composition of these microbial communities is fundamental to supply metabolic capabilities beyond those encoded in the host genome, and contributes to hormone and cellular signalling that support the dynamic adaptation to changes in food availability, environment and organismal development. Poor functional exchange between the microbial communities and their human host is associated with dysbiosis, metabolic dysfunction and disease. This review examines the biology of the dynamic relationship between the reciprocal metabolic state of the microbiota-host entity in balance with its environment (i.e. in healthy states), the enzymatic and metabolic changes associated with its imbalance in three well-studied diseases states such as obesity, diabetes and atherosclerosis, and the effects of bariatric surgery and exercise.
Collapse
Affiliation(s)
- Susannah Selber-Hnatiw
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Tarin Sultana
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - W Tse
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Niki Abdollahi
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Sheyar Abdullah
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Jalal Al Rahbani
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Diala Alazar
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Nekoula Jean Alrumhein
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Saro Aprikian
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Rimsha Arshad
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Jean-Daniel Azuelos
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Daphney Bernadotte
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Natalie Beswick
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Hana Chazbey
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Kelsey Church
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Emaly Ciubotaru
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Lora D'Amato
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Tavia Del Corpo
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Jasmine Deng
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Briana Laura Di Giulio
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Diana Diveeva
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Elias Elahie
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - James Gordon Marcel Frank
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Emma Furze
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Rebecca Garner
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Vanessa Gibbs
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Rachel Goldberg-Hall
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Chaim Jacob Goldman
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Fani-Fay Goltsios
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Kevin Gorjipour
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Taylor Grant
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Brittany Greco
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Nadir Guliyev
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Andrew Habrich
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Hillary Hyland
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Nabila Ibrahim
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Tania Iozzo
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Anastasia Jawaheer-Fenaoui
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Julia Jane Jaworski
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Maneet Kaur Jhajj
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Jermaine Jones
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Rodney Joyette
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Samad Kaudeer
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Shawn Kelley
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Shayesteh Kiani
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Marylin Koayes
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | | | - Shannon Maingot
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Sara Martin
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Kelly Mathers
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Sean McCullogh
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Kelly McNamara
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - James Mendonca
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Karamat Mohammad
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Sharara Arezo Momtaz
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Thiban Navaratnarajah
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Kathy Nguyen-Duong
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Mustafa Omran
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Angela Ortiz
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Anjali Patel
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Kahlila Paul-Cole
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Paul-Arthur Plaisir
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | | | - Ashlee Prevost
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Angela Quach
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Aries John Rafal
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Rewaparsad Ramsarun
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Sami Rhnima
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Lydia Rili
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Naomi Safir
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Eugenie Samson
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Rebecca Rose Sandiford
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Stefano Secondi
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Stephanie Shahid
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Mojdeh Shahroozi
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Fily Sidibé
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Megan Smith
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Alina Maria Sreng Flores
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Anabel Suarez Ybarra
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Rebecca Sénéchal
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Tarek Taifour
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Lawrence Tang
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Adam Trapid
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Maxim Tremblay Potvin
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Justin Wainberg
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Dani Ni Wang
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Mischa Weissenberg
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Allison White
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Gabrielle Wilkinson
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Brittany Williams
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Joshua Roth Wilson
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Johanna Zoppi
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Katerina Zouboulakis
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| | - Chiara Gamberi
- Biology Department, Concordia University, 7141 Sherbrooke St W, SP-375-09 Montreal, Quebec, H4B 1R6, Canada
| |
Collapse
|
108
|
Wang H, Xu R, Zhang H, Su Y, Zhu W. Swine gut microbiota and its interaction with host nutrient metabolism. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2020; 6:410-420. [PMID: 33364457 PMCID: PMC7750828 DOI: 10.1016/j.aninu.2020.10.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/09/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
Gut microbiota is generally recognized to play a crucial role in maintaining host health and metabolism. The correlation among gut microbiota, glycolipid metabolism, and metabolic diseases has been well reviewed in humans. However, the interplay between gut microbiota and host metabolism in swine remains incompletely understood. Given the limitation in conducting human experiments and the high similarity between swine and humans in terms of anatomy, physiology, polyphagy, habits, and metabolism and in terms of the composition of gut microbiota, there is a pressing need to summarize the knowledge gained regarding swine gut microbiota, its interplay with host metabolism, and the underlying mechanisms. This review aimed to outline the bidirectional regulation between gut microbiota and nutrient metabolism in swine and to emphasize the action mechanisms underlying the complex microbiome-host crosstalk via the gut microbiota-gut-brain axis. Moreover, it highlights the new advances in knowledge of the diurnal rhythmicity of gut microbiota. A better understanding of these aspects can not only shed light on healthy and efficient pork production but also promote our knowledge on the associations between gut microbiota and the microbiome-host crosstalk mechanism. More importantly, knowledge on microbiota, host health and metabolism facilitates the development of a precise intervention therapy targeting the gut microbiota.
Collapse
Affiliation(s)
- Hongyu Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Rongying Xu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - He Zhang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yong Su
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| |
Collapse
|
109
|
Demidova TY, Lobanova KG, Oinotkinova OS. [Gut microbiota is a factor of risk for obesity and type 2 diabetes]. TERAPEVT ARKH 2020; 92:97-104. [PMID: 33346486 DOI: 10.26442/00403660.2020.10.000778] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022]
Abstract
Gut microbiota (GM) is a set of bacteria which colonize the gastrointestinal tract. GM and its active metabolites take part in intestinal and hepatic gluconeogenesis, in the synthesis of incretin hormones, and affect the regulation of appetite. Thus, GM and its metabolites participate in the homeostasis of carbohydrates and fats. An imbalance in the set of the intestinal flora and a disturbance of the production of active metabolites sharply increases the risk of developing obesity and type 2 diabetes. There are conflicting data in the literature on the role of specific microorganisms in the development of metabolic disorders. Research is needed to identify specific types of bacteria and their active metabolites which affect the development of obesity and type 2 diabetes.
Collapse
Affiliation(s)
- T Y Demidova
- Pirogov Russian National Research Medical University
| | - K G Lobanova
- Pirogov Russian National Research Medical University
| | - O S Oinotkinova
- Pirogov Russian National Research Medical University.,Lomonosov Moscow State University.,Research Institute of Health Organization and Medical Management
| |
Collapse
|
110
|
Patil R, Arvindekar A. Glycation of gut proteins initiates microbial dysbiosis and can promote establishment of diabetes in experimental animals. Microb Pathog 2020; 152:104589. [PMID: 33171259 DOI: 10.1016/j.micpath.2020.104589] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022]
Abstract
Diabetes and obesity is associated with change in the gut microbiota, however, the reason for such transition is still unknown. The secondary complications in diabetes mainly stem from protein glycation, oxidative stress and inflammatory response. It is intended to study the correlation between gut proteins glycation and microbial dysbiosis and thereby progression to diabetes. The study was carried out through feeding high fructose to male Wistar rats and evaluating their gut microbiota. The rate of gut flora excretion via faecal matter was found to decrease on fructose feed for 7 days. Intestinal flora was drastically reduced and pathogenic succession observed. Intestinal fluorescence studies confirmed that there is heavy glycation of gut proteins. Microbes obtained from fructose fed animals could grow on glycated BSA. There was significant increase in level of TNF-α and IFN-γ providing evidence of inflammation. Though microbial dysbiosis was observed in diabetes, the cause for this remained elusive. In the present study we prove that high fructose feed and glycation of the gut proteins probably prevent adherence/survival of the gut microflora in control animals and promotes transition to a changed microflora which is capable of adhering/utilizing glycated proteins as well as high fructose. The changed microbiota, enhanced protein glycation and inflammation help in establishing insulin resistance.
Collapse
Affiliation(s)
- Rahul Patil
- Department of Biochemistry, Shivaji University, Kolhapur, 416 004, M.S, India
| | - Akalpita Arvindekar
- Department of Biochemistry, Shivaji University, Kolhapur, 416 004, M.S, India.
| |
Collapse
|
111
|
Gomaa EZ. Human gut microbiota/microbiome in health and diseases: a review. Antonie van Leeuwenhoek 2020; 113:2019-2040. [PMID: 33136284 DOI: 10.1007/s10482-020-01474-7] [Citation(s) in RCA: 665] [Impact Index Per Article: 133.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/12/2020] [Indexed: 12/12/2022]
Abstract
The human gut microbiota has received considerable interest in the recent years and our knowledge of the inhabitant species and their potential applications is increased particularly after the development of metagenomic studies. Gut microbiota is highly diverse and harboring trillions of microorganisms in human digestive system. The shaping and multiplication of gut microbiome starts at birth, while the modification of their composition depends mainly on various genetic, nutritional and environmental factors. The modification in the composition and function of the gut microbiota can change intestinal permeability, digestion and metabolism as well as immune responses. The pro inflammatory state caused by alternation of gut microbiota balance lead to the onset of many diseases ranging from gastrointestinal and metabolic conditions to immunological and neuropsychiatric diseases. In this context, the present review clarifies the role of gut microbiota in maintaining host health and investigates how nutritional and environmental factors affect the gut microbial structure and function. In addition, many therapeutic strategies of gut microbiota aimed at modulating and restoring of the intestinal ecosystem balance have been surveyed.
Collapse
Affiliation(s)
- Eman Zakaria Gomaa
- Department of Biological and Geological Sciences, Faculty of Education, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
112
|
Oduaran OH, Tamburini FB, Sahibdeen V, Brewster R, Gómez-Olivé FX, Kahn K, Norris SA, Tollman SM, Twine R, Wade AN, Wagner RG, Lombard Z, Bhatt AS, Hazelhurst S. Gut microbiome profiling of a rural and urban South African cohort reveals biomarkers of a population in lifestyle transition. BMC Microbiol 2020; 20:330. [PMID: 33129264 PMCID: PMC7603784 DOI: 10.1186/s12866-020-02017-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Comparisons of traditional hunter-gatherers and pre-agricultural communities in Africa with urban and suburban Western North American and European cohorts have clearly shown that diet, lifestyle and environment are associated with gut microbiome composition. Yet, little is known about the gut microbiome composition of most communities in the very diverse African continent. South Africa comprises a richly diverse ethnolinguistic population that is experiencing an ongoing epidemiological transition and concurrent spike in the prevalence of obesity, largely attributed to a shift towards more Westernized diets and increasingly inactive lifestyle practices. To characterize the microbiome of African adults living in more mainstream lifestyle settings and investigate associations between the microbiome and obesity, we conducted a pilot study, designed collaboratively with community leaders, in two South African cohorts representative of urban and transitioning rural populations. As the rate of overweight and obesity is particularly high in women, we collected single time-point stool samples from 170 HIV-negative women (51 at Soweto; 119 at Bushbuckridge), performed 16S rRNA gene sequencing on these samples and compared the data to concurrently collected anthropometric data. RESULTS We found the overall gut microbiome of our cohorts to be reflective of their ongoing epidemiological transition. Specifically, we find that geographical location was more important for sample clustering than lean/obese status and observed a relatively higher abundance of the Melainabacteria, Vampirovibrio, a predatory bacterium, in Bushbuckridge. Also, Prevotella, despite its generally high prevalence in the cohorts, showed an association with obesity. In comparisons with benchmarked datasets representative of non-Western populations, relatively higher abundance values were observed in our dataset for Barnesiella (log2fold change (FC) = 4.5), Alistipes (log2FC = 3.9), Bacteroides (log2FC = 4.2), Parabacteroides (log2FC = 3.1) and Treponema (log2FC = 1.6), with the exception of Prevotella (log2FC = - 4.7). CONCLUSIONS Altogether, this work identifies putative microbial features associated with host health in a historically understudied community undergoing an epidemiological transition. Furthermore, we note the crucial role of community engagement to the success of a study in an African setting, the importance of more population-specific studies to inform targeted interventions as well as present a basic foundation for future research.
Collapse
Affiliation(s)
- O. H. Oduaran
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa
- Division of Human Genetics, National Health Laboratory Service, and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - F. B. Tamburini
- Department of Genetics, Stanford University, Stanford, CA USA
| | - V. Sahibdeen
- Division of Human Genetics, National Health Laboratory Service, and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - R. Brewster
- School of Medicine, Stanford University, Stanford, CA USA
| | - F. X. Gómez-Olivé
- MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- INDEPTH Network, East Legon, Accra, Ghana
| | - K. Kahn
- MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- INDEPTH Network, East Legon, Accra, Ghana
| | - S. A. Norris
- SAMRC Developmental Pathways for Health Research Unit, Department of Paediatrics, University of the Witwatersrand, Johannesburg, South Africa
- School of Human Development and Health, University of Southampton, Southampton, UK
| | - S. M. Tollman
- MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- INDEPTH Network, East Legon, Accra, Ghana
| | - R. Twine
- MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - A. N. Wade
- MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - R. G. Wagner
- MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- INDEPTH Network, East Legon, Accra, Ghana
| | - Z. Lombard
- Division of Human Genetics, National Health Laboratory Service, and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - A. S. Bhatt
- Department of Genetics, Stanford University, Stanford, CA USA
- School of Medicine, Stanford University, Stanford, CA USA
- Department of Medicine (Hematology), Stanford University, Stanford, CA USA
| | - S. Hazelhurst
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa
- School of Electrical and Information Engineering, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
113
|
Ibrahim KS, Bourwis N, Dolan S, Lang S, Spencer J, Craft JA. Characterisation of gut microbiota of obesity and type 2 diabetes in a rodent model. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2020; 40:65-74. [PMID: 33520571 PMCID: PMC7817511 DOI: 10.12938/bmfh.2019-031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 10/06/2020] [Indexed: 12/20/2022]
Abstract
Various studies have suggested that the gut microbiome interacts with the host and may have a significant role in the aetiology of obesity and Type 2 Diabetes (T2D). It was hypothesised that bacterial communities in obesity and T2D differ from control and compromise normal interactions between host and microbiota. Obesity and T2D were developed in rats by feeding a high-fat diet or a high-fat diet plus a single low-dose streptozotocin administration, respectively. The microbiome profiles and their metabolic potentials were established by metagenomic 16S rRNA sequencing and bioinformatics. Taxonomy and predicted metabolism-related genes in obesity and T2D were markedly different from controls and indeed from each other. Diversity was reduced in T2D but not in Obese rats. Factors likely to compromise host intestinal, barrier integrity were found in Obese and T2D rats including predicted, decreased bacterial butyrate production. Capacity to increase energy extraction via ABC-transporters and carbohydrate metabolism were enhanced in Obese and T2D rats. T2D was characterized by increased proinflammatory molecules. While obesity and T2D show distinct differences, results suggest that in both conditions Bacteroides and Blautia species were increased indicating a possible mechanistic link.
Collapse
Affiliation(s)
- Khalid S Ibrahim
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, G4 0BA, United Kingdom.,Department of Biology, Faculty of Science, University of Zakho, Zakho International Road, Kurdistan Region-Iraq
| | - Nowara Bourwis
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, G4 0BA, United Kingdom
| | - Sharron Dolan
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, G4 0BA, United Kingdom
| | - Sue Lang
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, G4 0BA, United Kingdom.,Present address: School of Clinical and Applied Sciences, Leeds Beckett University, Portland Building, City Campus, Leeds, LS1 3HE, United Kingdom
| | - Janice Spencer
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, G4 0BA, United Kingdom
| | - John A Craft
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, G4 0BA, United Kingdom
| |
Collapse
|
114
|
Igwe EO, Roodenrys S, Probst YC, do Rosario V, Netzel ME, Hong HT, Netzel G, Phan ADT, Charlton KE. Low anthocyanin plum nectar does not impact cognition, blood pressure and gut microbiota in healthy older adults: A randomized crossover trial. Nutr Res 2020; 82:74-87. [PMID: 32977254 DOI: 10.1016/j.nutres.2020.08.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 07/22/2020] [Accepted: 08/10/2020] [Indexed: 02/07/2023]
Abstract
Queen Garnet plum (QGP), known for its high levels of anthocyanins, is a hybrid of the Japanese plum developed in Queensland, Australia. Anthocyanins provide the red, blue, and purple pigments in plants with demonstrated beneficial health effects. This study hypothesized that low-dose anthocyanin QGP intake will have a significant positive effect on cognition, blood pressure, and gut microbiota in healthy older adults. A randomized crossover trial was conducted to determine the effect and within subject variance on cognition and 24 hr. ambulatory blood pressure in older adults without cognitive impairment following daily consumption of 200 mL low-dose anthocyanin (5 mg/100 g) QGP nectar (intervention) or raspberry cordial (control). Secondary outcomes included inflammatory markers (C-reactive protein), nerve growth factor (BDNF), and gut microbiota (16S rRNA gene sequencing). Twenty-eight participants (55+ years) were recruited. Each randomized treatment arm lasted for 8 weeks with a 4-week washout period. Cognition, blood pressure, and urine samples were measured at each visit (5 total) while blood and fecal samples were collected at baseline, 8 weeks, and 20 weeks. Repeated-measures ANOVA was used to analyze the data. Across the treatments, no significant difference was observed for the different domains of cognition, blood pressure, or anti-inflammatory biomarkers. No intervention effect was found for genera or class of gut microbes. Low anthocyanin nectar derived from the QGP did not have any significant effects on cognition, blood pressure, or gut microbiota in healthy older adults.
Collapse
Affiliation(s)
- Ezinne O Igwe
- School of Medicine, University of Wollongong, Wollonggong, NSW, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Wollonggong, NSW, Australia.
| | - Steven Roodenrys
- School of Psychology, University of Wollongong, Wollonggong, NSW, Australia
| | - Yasmine C Probst
- School of Medicine, University of Wollongong, Wollonggong, NSW, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Wollonggong, NSW, Australia
| | - Vinicius do Rosario
- School of Medicine, University of Wollongong, Wollonggong, NSW, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Wollonggong, NSW, Australia
| | - Michael E Netzel
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Coopers Plains, QLD 4108, Australia
| | - Hung T Hong
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Coopers Plains, QLD 4108, Australia
| | - Gabriele Netzel
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Coopers Plains, QLD 4108, Australia
| | - Anh D T Phan
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Coopers Plains, QLD 4108, Australia
| | - Karen E Charlton
- School of Medicine, University of Wollongong, Wollonggong, NSW, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Wollonggong, NSW, Australia
| |
Collapse
|
115
|
Both maternal and newborn IgMs inhibit influenza virus-induced hemagglutination in vitro. UKRAINIAN BIOCHEMICAL JOURNAL 2020. [DOI: 10.15407/ubj92.04.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
116
|
Proffitt C, Bidkhori G, Moyes D, Shoaie S. Disease, Drugs and Dysbiosis: Understanding Microbial Signatures in Metabolic Disease and Medical Interventions. Microorganisms 2020; 8:microorganisms8091381. [PMID: 32916966 PMCID: PMC7565856 DOI: 10.3390/microorganisms8091381] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023] Open
Abstract
Since the discovery of the potential role for the gut microbiota in health and disease, many studies have gone on to report its impact in various pathologies. These studies have fuelled interest in the microbiome as a potential new target for treating disease Here, we reviewed the key metabolic diseases, obesity, type 2 diabetes and atherosclerosis and the role of the microbiome in their pathogenesis. In particular, we will discuss disease associated microbial dysbiosis; the shift in the microbiome caused by medical interventions and the altered metabolite levels between diseases and interventions. The microbial dysbiosis seen was compared between diseases including Crohn’s disease and ulcerative colitis, non-alcoholic fatty liver disease, liver cirrhosis and neurodegenerative diseases, Alzheimer’s and Parkinson’s. This review highlights the commonalities and differences in dysbiosis of the gut between diseases, along with metabolite levels in metabolic disease vs. the levels reported after an intervention. We identify the need for further analysis using systems biology approaches and discuss the potential need for treatments to consider their impact on the microbiome.
Collapse
Affiliation(s)
- Ceri Proffitt
- Centre for Host–Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (G.B.); (D.M.)
- Correspondence: (C.P.); (S.S.)
| | - Gholamreza Bidkhori
- Centre for Host–Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (G.B.); (D.M.)
| | - David Moyes
- Centre for Host–Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (G.B.); (D.M.)
| | - Saeed Shoaie
- Centre for Host–Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (G.B.); (D.M.)
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, 114 17 Stockholm, Sweden
- Correspondence: (C.P.); (S.S.)
| |
Collapse
|
117
|
Type 2 Diabetes Mellitus Associated with Obesity (Diabesity). The Central Role of Gut Microbiota and Its Translational Applications. Nutrients 2020; 12:nu12092749. [PMID: 32917030 PMCID: PMC7551493 DOI: 10.3390/nu12092749] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/31/2020] [Accepted: 09/04/2020] [Indexed: 02/07/2023] Open
Abstract
Obesity is a condition of rising prevalence worldwide, with important socioeconomic implications, being considered as a growing public health concern. Frequently, obesity brings other complications in addition to itself—like Type 2 Diabetes Mellitus (T2DM)—sharing origin, risk factors and pathophysiological mechanisms. In this context, some authors have decided to include both conditions as a unique entity known as “diabesity”. In fact, understanding diabesity as a single disease is possible to maximise the benefits from therapies received in these patients. Gut microbiota plays a key role in individual’s health, and their alterations, either in its composition or derived products are related to a wide range of metabolic disorders like T2DM and obesity. The present work aims to collect the different changes reported in gut microbiota in patients with T2DM associated with obesity and their possible role in the onset, development, and establishment of the disease. Moreover, current research lines to modulate gut microbiota and the potential clinical translation derived from the knowledge of this system will also be reviewed, which may provide support for a better clinical management of such a complex condition.
Collapse
|
118
|
Sepúlveda M, Alamo M, Astorga C, Preiss Y, Saavedra S. Histologic and microbiological findings of the defunctionalized loop in sleeve gastrectomy with jejunal bypass. Surg Obes Relat Dis 2020; 17:131-138. [PMID: 33268324 DOI: 10.1016/j.soard.2020.08.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 08/05/2020] [Accepted: 08/12/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Bariatric surgery produces anatomic changes in the digestive tract that can affect the intestinal microbiome and, in some cases, can cause small intestinal bacterial overgrowth. Since the inception of the sleeve gastrectomy with jejunal bypass (SGJB) in 2004, there has been discussion regarding the possible development of those complications associated with the now abandoned jejunoileal bypass (JIB) procedure. OBJECTIVES The primary endpoint was to characterize the bacteriologic and histopathologic findings in the defunctionalized jejunal loop after the SGJB procedure and to analyze the liver profile. The secondary endpoint was to report SGJB conversions or reversions and to review the differences between SGJB and JIB. SETTING Academic medical center. METHODS We conducted a prospective study of patients who underwent laparoscopy for any reason, having previously had an SGJB. A 5-cm segment at the proximal end of the excluded limb was resected. Luminal liquid and tissue samples were taken from this segment for aerobic and anaerobic cultures, and pathologic examination of the bowel wall was performed to evaluate trophism and signs of chronic inflammation. Other variables were liver function and pre- and postoperative status. Finally, we retrospectively reviewed the causes of revisional surgery in the prospective database. RESULTS Eleven patients underwent laparoscopy. The median time after SGJB was 14 months (range, 10-144 months). There were no complications from the procedure. Eight (72.7%) of the procedures were cholecystectomies. None of the patients showed histologic alterations or signs of chronic infection. The liquid and tissue cultures were negative. The liver tests and the laparoscopic morphology of the liver were normal in all patients, except in 1 with previously documented liver cirrhosis. The number of SGJB revisions was 19 of 1074 (1.8 %), and all of them were converted to Roux-en-Y gastric bypass for severe gastroesophageal reflux. CONCLUSIONS In this study, we were unable to demonstrate the presence of symptoms or histologic alterations that would suggest that patients undergoing SGJB develop small intestinal bacterial overgrowth in the short- and medium-term follow-up, unlike those who have undergone JIB. The study constitutes an initial step toward establishing what happens to the defunctionalized jejunal limb as a result of this surgical technique.
Collapse
Affiliation(s)
- Matías Sepúlveda
- Bariatric and Metabolic Surgery Center, Surgery Department, DIPRECA Hospital, Santiago, Chile; Center of Nutrition and Bariatric Surgery, Clínica Las Condes, Santiago, Chile; Escuela de Medicina, Universidad Diego Portales, Santiago, Chile.
| | - Munir Alamo
- Surgery Department, Hospital de Carabineros, Santiago, Chile
| | - Cristián Astorga
- Bariatric and Metabolic Surgery Center, Surgery Department, DIPRECA Hospital, Santiago, Chile; Center of Nutrition and Bariatric Surgery, Clínica Las Condes, Santiago, Chile; Escuela de Medicina, Universidad Diego Portales, Santiago, Chile
| | - Yudith Preiss
- Bariatric and Metabolic Surgery Center, Surgery Department, DIPRECA Hospital, Santiago, Chile
| | - Sebastián Saavedra
- Bariatric and Metabolic Surgery Center, Surgery Department, DIPRECA Hospital, Santiago, Chile
| |
Collapse
|
119
|
Machado AS, Oliveira JR, Lelis DDF, de Paula AMB, Guimarães ALS, Andrade JMO, Brandi IV, Santos SHS. Oral Probiotic Bifidobacterium Longum Supplementation Improves Metabolic Parameters and Alters the Expression of the Renin-Angiotensin System in Obese Mice Liver. Biol Res Nurs 2020; 23:100-108. [PMID: 32700545 DOI: 10.1177/1099800420942942] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Obesity and non-alcoholic fatty liver disease (NAFLD) have been increasing at an alarming rate worldwide. Bifidobacterium longum (BL), a common member of the human gut microbiota, has important health benefits through several mechanisms. OBJECTIVES We evaluated the BL supplementation effects on body metabolism and renin-angiotensin components hepatic expression in mice fed a high-fat diet. METHODS Thirty-two male mice were divided into four groups: standard diet + placebo (ST), standard diet + Bifidobacterium longum (ST + BL), high-fat diet + placebo (HFD) and high-fat diet + Bifidobacterium longum (HFD + BL). Following the obesity induction period, the ST + BL and HFD + BL groups were supplemented with Bifidobacterium longum for 4 weeks. Then, body, biochemical, histological and molecular parameters were evaluated. RESULTS HFD + BL mice had a significant decrease in adipose tissue mass and blood glucose levels, as well as a significant reduction in blood glucose during an intraperitoneal glucose tolerance test. The treatment also resulted in reduced levels of total cholesterol and hepatic fat accumulation. Moreover, we observed an increase in angiotensin converting enzyme 2 (ACE2) and Mas receptor (MASR) expression levels in BL-treated obese mice. CONCLUSIONS These data demonstrate that BL may have the potential to prevent obesity and NAFLD by modulating the mRNA expression of renin-angiotensin system components.
Collapse
Affiliation(s)
- Amanda S Machado
- Laboratory of Health Science, Postgraduation Program in Health Sciences, 153595Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Janaína R Oliveira
- Laboratory of Health Science, Postgraduation Program in Health Sciences, 153595Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Deborah de F Lelis
- Laboratory of Health Science, Postgraduation Program in Health Sciences, 153595Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Alfredo M B de Paula
- Laboratory of Health Science, Postgraduation Program in Health Sciences, 153595Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - André L S Guimarães
- Laboratory of Health Science, Postgraduation Program in Health Sciences, 153595Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - João M O Andrade
- Laboratory of Health Science, Postgraduation Program in Health Sciences, 153595Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Igor V Brandi
- Institute of Agricultural Sciences, Food Engineering, 28114Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil
| | - Sérgio H S Santos
- Laboratory of Health Science, Postgraduation Program in Health Sciences, 153595Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil.,Institute of Agricultural Sciences, Food Engineering, 28114Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil
| |
Collapse
|
120
|
Gut microbial composition in patients with atrial fibrillation: effects of diet and drugs. Heart Vessels 2020; 36:105-114. [PMID: 32683492 PMCID: PMC7788021 DOI: 10.1007/s00380-020-01669-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/10/2020] [Indexed: 12/12/2022]
Abstract
Atrial fibrillation (AF) reduces the quality of life by triggering stroke and heart failure. The association between AF onset and gut metabolites suggests a causal relationship between AF and gut microbiota dysbiosis; however, the relationship remains poorly understood. We prospectively enrolled 34 hospitalized patients with AF and 66 age-, sex-, and comorbidity-matched control subjects without a history of AF. Gut microbial compositions were evaluated by amplicon sequencing targeting the 16S ribosomal RNA gene. We assessed differences in dietary habits by using a brief-type self-administered diet history questionnaire (BDHQ). Gut microbial richness was lower in AF patients, although the diversity of gut microbiota did not differ between the two groups. At the genus level, Enterobacter was depleted, while Parabacteroides, Lachnoclostridium, Streptococcus, and Alistipes were enriched in AF patients compared to control subjects. The BDHQ revealed that the intake of n-3 polyunsaturated fatty acids and eicosadienoic acid was higher in AF patients. Our results suggested that AF patients had altered gut microbial composition in connection with dietary habits.
Collapse
|
121
|
Zha H, Chen Y, Wu J, Chang K, Lu Y, Zhang H, Xie J, Wang Q, Tang R, Li L. Characteristics of three microbial colonization states in the duodenum of the cirrhotic patients. Future Microbiol 2020; 15:855-868. [PMID: 32662659 DOI: 10.2217/fmb-2019-0270] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Aim: Investigation of characteristics of different duodenal microbial colonization states in patients with liver cirrhosis (LC). Materials & methods: Deep-sequencing analyses of the 16S rRNA gene V1-V3 regions were performed. Results: Both bacterial compositions and richness were different between the three-clustered LC microbiotas, in other words, Cluster_1_LC, Cluster_2_LC and Cluster_3_LC. Cluster_1_LC were more likely at severe dysbiosis status due to its lowest modified cirrhosis dysbiosis ratio. OTU12_Prevotella and OTU10_Comamonas were most associated with Cluster_1_LC and Cluster_3_LC, respectively, while OTU38_Alloprevotella was vital in Cluster_2_LC. Pyruvate-ferredoxin/flavodoxin oxidoreductase, dihydroorotate dehydrogenase and branched-chain amino acid transport system substrate-binding protein were most associated with Cluster_1_LC, Cluster_2_LC and Cluster_3_LC, respectively. Conclusion: The three duodenal microbial colonization states had distinct representative characteristics, which might reflect the health status of cirrhotic patients.
Collapse
Affiliation(s)
- Hua Zha
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis & Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,School of Biological Sciences, The University of Auckland, Auckland, New Zealand.,Institute of Marine Science, The University of Auckland, Auckland, New Zealand
| | - Yanfei Chen
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis & Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jieyun Wu
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand.,Plant Health & Environment Laboratory, Ministry for Primary Industries, Auckland, New Zealand
| | - Kevin Chang
- Department of Statistics, The University of Auckland, Auckland, New Zealand
| | - Yanmeng Lu
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis & Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Hua Zhang
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis & Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jiaojiao Xie
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis & Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Qiangqiang Wang
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis & Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ruiqi Tang
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis & Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis & Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
122
|
Targeting the Intestinal Microbiota to Prevent Type 2 Diabetes and Enhance the Effect of Metformin on Glycaemia: A Randomised Controlled Pilot Study. Nutrients 2020; 12:nu12072041. [PMID: 32660025 PMCID: PMC7400852 DOI: 10.3390/nu12072041] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 12/17/2022] Open
Abstract
Early treatment may prevent or delay the onset of type 2 diabetes mellitus (T2DM) in individuals who are at high risk. Lifestyle interventions and the hypoglycemic drug metformin have been shown to reduce T2DM incidence. The effectiveness of such interventions may be enhanced by targeting environmental factors such as the intestinal microbiota, which has been proven to predict the response to lifestyle interventions and play a part in mediating the glucose-lowering effects of metformin. Shifts in the intestinal microbiota “towards a more balanced state” may promote glucose homeostasis by regulating short-chain fatty acids’ production. This study aimed to investigate the safety and effect of a multi-strain probiotic on glycemic, inflammatory, and permeability markers in adults with prediabetes and early T2DM and to assess whether the probiotic can enhance metformin’s effect on glycaemia. A randomised controlled pilot study was conducted in 60 adults with a BMI ≥ 25 kg/m2 and with prediabetes or T2DM (within the previous 12 months). The participants were randomised to a multi-strain probiotic (L. plantarum, L. bulgaricus, L. gasseri, B. breve, B. animalis sbsp. lactis, B. bifidum, S. thermophilus, and S. boulardii) or placebo for 12 weeks. Analyses of the primary outcome (fasting plasma glucose) and secondary outcomes, including, but not limited to, circulating lipopolysaccharide, zonulin, and short chain fatty acids and a metagenomic analysis of the fecal microbiome were performed at baseline and 12 weeks post-intervention. The results showed no significant differences in the primary and secondary outcome measures between the probiotic and placebo group. An analysis of a subgroup of participants taking metformin showed a decrease in fasting plasma glucose, HbA1c, insulin resistance, and zonulin; an increase in plasma butyrate concentrations; and an enrichment of microbial butyrate-producing pathways in the probiotic group but not in the placebo group. Probiotics may act as an adjunctive to metformin by increasing the production of butyrate, which may consequently enhance glucose management.
Collapse
|
123
|
Sharma N, Navik U, Tikoo K. Unveiling the presence of epigenetic mark by Lactobacillus supplementation in high-fat diet-induced metabolic disorder in Sprague-Dawley rats. J Nutr Biochem 2020; 84:108442. [PMID: 32629239 DOI: 10.1016/j.jnutbio.2020.108442] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/15/2020] [Accepted: 05/24/2020] [Indexed: 12/26/2022]
Abstract
Gut dysbiosis, particularly bacteria from Firmicutes and Bacteroidetes phyla, plays a fundamental role in the progression of metabolic disorders. Probiotics have shown to restore the gut microbiota composition in metabolic disorders with subsequent beneficial effects. Recent studies have reported that several species of Lactobacillus as probiotic supplementation improve insulin sensitivity and glucose metabolism. Nonetheless, whether Lactobacillus could influence the epigenetic modifications that underlie insulin-resistant conditions is still unexplored. Therefore, the current study examined the therapeutic effects and underlying epigenetic mechanisms of three different species of Lactobacillus in the high-fat diet (HFD)-induced insulin-resistant rats. Three different species of Lactobacillus; Lactobacillus casei, Lactobacillus gasseri, and Lactobacillus rhamnosus were individually supplemented orally (109 CFU/mL) to insulin-resistant SD rats for 12 weeks. Lactobacillus supplementation led to a significant reduction in the hyperglycemia, hyperinsulinemia, and hyperlipidemia associated with HFD-induced insulin resistance. Histopathological examination also indicated the protective effects of Lactobacillus supplementation against the hepatic and intestinal damage caused by the high-fat diet. Lactobacillus supplementation also down-regulated the expression of FOXO1, a major transcription factor of insulin signaling. In addition, at the epigenetic level, Lactobacillus supplementation predominantly prevented methylation and demethylation of H3K79me2 and H3K27me3, respectively. Chromatin Immunoprecipitation (ChIP) coupled with quantitative PCR (ChIP-qPCR) assay revealed the presence of cross-talk between these two histone modifications at the promoter region of FOXO1. Taken together, this is the first report to observe that the effects of Lactobacillus supplementation involve alteration in FOXO1 expression via cross-talking between H3K79me2 and H3K27me3 histone modifications.
Collapse
Affiliation(s)
- Nisha Sharma
- Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab-160062, India
| | - Umashanker Navik
- Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab-160062, India
| | - Kulbhushan Tikoo
- Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab-160062, India.
| |
Collapse
|
124
|
Arrhythmic Gut Microbiome Signatures Predict Risk of Type 2 Diabetes. Cell Host Microbe 2020; 28:258-272.e6. [PMID: 32619440 DOI: 10.1016/j.chom.2020.06.004] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 05/08/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022]
Abstract
Lifestyle, obesity, and the gut microbiome are important risk factors for metabolic disorders. We demonstrate in 1,976 subjects of a German population cohort (KORA) that specific microbiota members show 24-h oscillations in their relative abundance and identified 13 taxa with disrupted rhythmicity in type 2 diabetes (T2D). Cross-validated prediction models based on this signature similarly classified T2D. In an independent cohort (FoCus), disruption of microbial oscillation and the model for T2D classification was confirmed in 1,363 subjects. This arrhythmic risk signature was able to predict T2D in 699 KORA subjects 5 years after initial sampling, being most effective in combination with BMI. Shotgun metagenomic analysis functionally linked 26 metabolic pathways to the diurnal oscillation of gut bacteria. Thus, a cohort-specific risk pattern of arrhythmic taxa enables classification and prediction of T2D, suggesting a functional link between circadian rhythms and the microbiome in metabolic diseases.
Collapse
|
125
|
Ye L, Das P, Li P, Ji B, Nielsen J. Carbohydrate active enzymes are affected by diet transition from milk to solid food in infant gut microbiota. FEMS Microbiol Ecol 2020; 95:5582603. [PMID: 31589310 DOI: 10.1093/femsec/fiz159] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 10/04/2019] [Indexed: 01/09/2023] Open
Abstract
Infants experience a dramatic change in their food in the first year after birth when they shift from breast milk to solid food. This results in a large change in presence of indigestible polysaccharides, a primary energy resource of gut microbes. How the gut microbiota adapts to this dietary shift has not been well examined. Here, by using metagenomics data, we studied carbohydrate-active enzymes (CAZymes) of gut microbiota, which are essential enzymes catalyzing the breakdown of polysaccharides, during this dietary shift. We developed a new approach to categorize CAZyme families by food intake and found CAZyme families associated with milk or solid food. We also found CAZymes with most abundance in 12 months infants that are not associated with solid food or milk but may be related to modulating carbohydrates in the mucus. Additionally, the abundance of gut CAZymes were found to be affected by many other factors, including delivery modes and life style in adults. Taken together, our findings provide novel insights into the dynamic change of gut CAZymes in early human life and provide potential markers for food interference or gut microbiota restoration.
Collapse
Affiliation(s)
- Lingqun Ye
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemigården 4, SE-412 96 Gothenburg, Sweden
| | - Promi Das
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemigården 4, SE-412 96 Gothenburg, Sweden
| | - Peishun Li
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemigården 4, SE-412 96 Gothenburg, Sweden
| | - Boyang Ji
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemigården 4, SE-412 96 Gothenburg, Sweden
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemigården 4, SE-412 96 Gothenburg, Sweden.,Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Kgs. Lyngby DK2800, Denmark.,BioInnovation Institute, Ole Maaløes Vej, DK2200 Copenhagen N, Denmark
| |
Collapse
|
126
|
Rajagopala SV, Singh H, Yu Y, Zabokrtsky KB, Torralba MG, Moncera KJ, Frank B, Pieper R, Sender L, Nelson KE. Persistent Gut Microbial Dysbiosis in Children with Acute Lymphoblastic Leukemia (ALL) During Chemotherapy. MICROBIAL ECOLOGY 2020; 79:1034-1043. [PMID: 31754744 DOI: 10.1007/s00248-019-01448-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 10/01/2019] [Indexed: 06/10/2023]
Abstract
Prophylactic or therapeutic antibiotic use along with chemotherapy treatment potentially has a long-standing adverse effect on the resident gut microbiota. We have established a case-control cohort of 32 pediatric and adolescent acute lymphoblastic leukemia (ALL) patients and 25 healthy siblings (sibling controls) to assess the effect of chemotherapy as well as antibiotic prophylaxis on the gut microbiota. We observe that the microbiota diversity and richness of the ALL group is significantly lower than that of the control group at diagnosis and during chemotherapy. The microbiota diversity is even lower in antibiotics-exposed ALL patients. Although the gut microbial diversity tends to stabilize after 1-year post-chemotherapy, their abundances were altered because of chemotherapy and prophylactic antibiotic treatments. Specifically, the abundances of mucolytic gram-positive anaerobic bacteria, including Ruminococcus gnavus and Ruminococcus torques, tended to increase during the chemotherapy regimen and continued to be elevated 1 year beyond the initiation of chemotherapy. This dysbiosis may contribute to the development of gastrointestinal complications in ALL children following chemotherapy. These findings set the stage to further understand the role of the gut microbiome dynamics in ALL patients and their potential role in alleviating some of the adverse side effects of chemotherapy and antibiotics use in immunocompromised children.
Collapse
Affiliation(s)
- Seesandra V Rajagopala
- J. Craig Venter Institute (JCVI), Rockville, MD, USA
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Yanbao Yu
- J. Craig Venter Institute (JCVI), Rockville, MD, USA
| | - Keri B Zabokrtsky
- Hyundai Cancer Genomics Center, Children's Hospital Orange County (CHOC Children's), Orange, CA, USA
- Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of California-Irvine, Orange, CA, USA
| | | | | | - Bryan Frank
- J. Craig Venter Institute (JCVI), Rockville, MD, USA
| | | | - Leonard Sender
- Hyundai Cancer Genomics Center, Children's Hospital Orange County (CHOC Children's), Orange, CA, USA
- Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of California-Irvine, Orange, CA, USA
- Division of Oncology, Hyundai Cancer Institute, CHOC Children's, Orange, CA, USA
- Department of Pediatrics, School of Medicine, University of California-Irvine, Orange, CA, USA
| | - Karen E Nelson
- J. Craig Venter Institute (JCVI), Rockville, MD, USA
- J. Craig Venter Institute (JCVI), La Jolla, CA, USA
| |
Collapse
|
127
|
Gut microbiota and human NAFLD: disentangling microbial signatures from metabolic disorders. Nat Rev Gastroenterol Hepatol 2020; 17:279-297. [PMID: 32152478 DOI: 10.1038/s41575-020-0269-9] [Citation(s) in RCA: 665] [Impact Index Per Article: 133.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/21/2020] [Indexed: 02/07/2023]
Abstract
Gut microbiota dysbiosis has been repeatedly observed in obesity and type 2 diabetes mellitus, two metabolic diseases strongly intertwined with non-alcoholic fatty liver disease (NAFLD). Animal studies have demonstrated a potential causal role of gut microbiota in NAFLD. Human studies have started to describe microbiota alterations in NAFLD and have found a few consistent microbiome signatures discriminating healthy individuals from those with NAFLD, non-alcoholic steatohepatitis or cirrhosis. However, patients with NAFLD often present with obesity and/or insulin resistance and type 2 diabetes mellitus, and these metabolic confounding factors for dysbiosis have not always been considered. Patients with different NAFLD severity stages often present with heterogeneous lesions and variable demographic characteristics (including age, sex and ethnicity), which are known to affect the gut microbiome and have been overlooked in most studies. Finally, multiple gut microbiome sequencing tools and NAFLD diagnostic methods have been used across studies that could account for discrepant microbiome signatures. This Review provides a broad insight into microbiome signatures for human NAFLD and explores issues with disentangling these signatures from underlying metabolic disorders. More advanced metagenomics and multi-omics studies using system biology approaches are needed to improve microbiome biomarkers.
Collapse
|
128
|
Torp Austvoll C, Gallo V, Montag D. Health impact of the Anthropocene: the complex relationship between gut microbiota, epigenetics, and human health, using obesity as an example. Glob Health Epidemiol Genom 2020; 5:e2. [PMID: 32363032 PMCID: PMC7176587 DOI: 10.1017/gheg.2020.2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 12/13/2019] [Accepted: 02/17/2020] [Indexed: 02/06/2023] Open
Abstract
The growing prevalence of obesity worldwide poses a public health challenge in the current geological epoch, the Anthropocene. Global changes caused by urbanisation, loss of biodiversity, industrialisation, and land-use are happening alongside microbiota dysbiosis and increasing obesity prevalence. How alterations of the gut microbiota are associated with obesity and the epigenetic mechanism mediating this and other health outcome associations are in the process of being unveiled. Epigenetics is emerging as a key mechanism mediating the interaction between human body and the environment in producing disease. Evidence suggests that the gut microbiota plays a role in obesity as it contributes to different mechanisms, such as metabolism, body weight and composition, inflammatory responses, insulin signalling, and energy extraction from food. Consistently, obese people tend to have a different epigenetic profile compared to non-obese. However, evidence is usually scattered and there is a growing need for a structured framework to conceptualise this complexity and to help shaping complex solutions. In this paper, we propose a framework to analyse the observed associations between the alterations of microbiota and health outcomes and the role of epigenetic mechanisms underlying them using obesity as an example, in the current context of global changes within the Anthropocene.
Collapse
Affiliation(s)
- Cecilie Torp Austvoll
- Centre for Primary Care and Public Health, Queen Mary University of London, London, UK
| | - Valentina Gallo
- Centre for Primary Care and Public Health, Queen Mary University of London, London, UK
- London School of Hygiene and Tropical Medicine, London, UK
- School of Public Health, Imperial College London, London, UK
| | - Doreen Montag
- Centre for Primary Care and Public Health, Queen Mary University of London, London, UK
| |
Collapse
|
129
|
Chung E, Elmassry MM, Kottapalli P, Kottapalli KR, Kaur G, Dufour JM, Wright K, Ramalingam L, Moustaid-Moussa N, Wang R, Hamood AN, Shen CL. Metabolic benefits of annatto-extracted tocotrienol on glucose homeostasis, inflammation, and gut microbiome. Nutr Res 2020; 77:97-107. [PMID: 32438021 DOI: 10.1016/j.nutres.2020.04.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 03/27/2020] [Accepted: 04/11/2020] [Indexed: 12/12/2022]
Abstract
Emerging evidence suggests that the gut microbiome plays an important role in the pathophysiology of both obesity and type 2 diabetes mellitus. We previously reported that dietary annatto-extracted tocotrienol exerts beneficial effects by modulating inflammatory responses in mice fed a high-fat diet (HFD). The purpose of this study was to test the hypothesis that tocotrienol supplementation when combined with an HFD would result in an altered gut microbiota composition. For 14 weeks, forty-eight male C57BL/6J mice were assigned to 4 groups-low-fat diet, HFD, HFD supplemented with annatto-extracted tocotrienol at 800 mg/kg diet (AT), and HFD supplemented with metformin at 200 mg/kg diet. Glucose homeostasis was assessed by glucose and insulin tolerance tests, serum and pancreas insulin levels, and histological assessments of insulin and glucagon in pancreatic tissue. The concentrations of adipokines were measured in white adipose tissues. For the gut microbiome analysis, cecal content was collected, DNA was extracted, and 16S rRNA gene sequencing was performed. AT supplementation improved glucose homeostasis and lowered resistin, leptin, and interleukin-6 levels in white adipose tissue. Relative to the HFD group, AT-supplemented mice showed a decrease in the Firmicutes to Bacteroidetes ratio and had a lower abundance of Ruminococcus lactaris, Dorea longicatena, and Lachnospiraceae family. The relative abundance of Akkermansia muciniphila was increased in the AT group compared to the low-fat diet group. The association between the metabolic improvements and the identified bacterial taxa suggests a potential metabolic modulation caused by AT supplementation through the gut microbiota composition in mice fed an HFD.
Collapse
Affiliation(s)
- Eunhee Chung
- Department of Kinesiology, Health, and Nutrition, University of Texas at San Antonio, San Antonio, TX.
| | - Moamen M Elmassry
- Department of Biological Sciences, Texas Tech University, Lubbock, TX.
| | | | | | - Gurvinder Kaur
- Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, TX; Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX; Obesity Research Institute, Texas Tech University, Lubbock, TX.
| | - Jannette M Dufour
- Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, TX; Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX; Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX; Obesity Research Institute, Texas Tech University, Lubbock, TX; Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX.
| | - Kandis Wright
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX.
| | - Latha Ramalingam
- Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX; Obesity Research Institute, Texas Tech University, Lubbock, TX; Department of Nutritional Sciences, Texas Tech University, Lubbock, TX.
| | - Naima Moustaid-Moussa
- Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX; Obesity Research Institute, Texas Tech University, Lubbock, TX; Department of Nutritional Sciences, Texas Tech University, Lubbock, TX.
| | - Rui Wang
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX.
| | - Abdul N Hamood
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX; Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX.
| | - Chwan-Li Shen
- Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX; Obesity Research Institute, Texas Tech University, Lubbock, TX; Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX.
| |
Collapse
|
130
|
Tabasi M, Ashrafian F, Khezerloo JK, Eshghjoo S, Behrouzi A, Javadinia SA, Poursadegh F, Eybpoosh S, Ahmadi S, Radmanesh A, Soroush A, Siadat SD. Changes in Gut Microbiota and Hormones After Bariatric Surgery: a Bench-to-Bedside Review. Obes Surg 2020; 29:1663-1674. [PMID: 30793228 DOI: 10.1007/s11695-019-03779-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Overweight and obesity are among the most prevalent non-communicable diseases which are generally treated successfully by bariatric or sleeve surgery. There are evidences affirming that sleeve surgery can manipulate the pH of the stomach and interact with the metabolism of fatty acids, carbohydrates, and bile acid transfer, leading to the overgrowth of gut microbiota. Therefore, this study aims to review the changes in gut microbiota and hormones after bariatric surgery.
Collapse
Affiliation(s)
- Mohsen Tabasi
- Department of Molecular Biology, Pasteur Institute of Iran, Pasteur Ave., Tehran, 13164, Iran
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Fatemeh Ashrafian
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Jamil Kheirvari Khezerloo
- Young Researchers and Elite Club, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Sahar Eshghjoo
- Microbial Pathogenesis and Immunology Department, Texas A&M University, Bryan, TX, USA
| | - Ava Behrouzi
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Alireza Javadinia
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Islamic Republic of Iran
| | - Farid Poursadegh
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Islamic Republic of Iran
| | - Sana Eybpoosh
- Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Sara Ahmadi
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Amin Radmanesh
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Ahmadreza Soroush
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
131
|
Da Silva CC, Monteil MA, Davis EM. Overweight and Obesity in Children Are Associated with an Abundance of Firmicutes and Reduction of Bifidobacterium in Their Gastrointestinal Microbiota. Child Obes 2020; 16:204-210. [PMID: 31934770 DOI: 10.1089/chi.2019.0280] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background: Obesity in children continues to be a global epidemic. Given the recent focus on the potential role gut microbiota play in the development of obesity and the continuous rise of obesity in childhood worldwide, it has become necessary that the association between gut microbes and weight is explored in previously unexplored regions such as the Caribbean island of Trinidad. As such, our objective was to characterize the composition of the gut microbiota in children with obesity/overweight and children of normal weight to determine if there are any gut microbes associated with overweight and obesity. Methods: Fifty-one children provided stool samples and their BMIs were calculated and classified. 16S amplicons from stool samples were individually barcoded and sequenced in multiplex in the NextSeq 500 platform in a 150 bp paired-end modality and a complete gut microbiome profile was generated. Results: The relative abundance and diversity of the microbiota of the lean and children with obesity/overweight were analyzed against each other and showed that children with obesity/overweight recorded significantly more (p = 0.001) Firmicutes and significantly less Bifidobacterium (p = 0.039) than the children of normal weight. Children with obesity/overweight also recorded significantly (p = 0.009) reduced alpha diversity compared with the lean children. Conclusion: This study has shown associations of Firmicutes to obesity/overweight and Bifidobacterium species with healthy weight in children.
Collapse
Affiliation(s)
- Cerano C Da Silva
- Department of Pre-Clinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad
| | - Michele A Monteil
- Augusta University-The University of Georgia Medical Partnership, Athens, GA
| | - Elaine Monica Davis
- Department of Pre-Clinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad
| |
Collapse
|
132
|
The Gut Microbial Diversity of Newly Diagnosed Diabetics but Not of Prediabetics Is Significantly Different from That of Healthy Nondiabetics. mSystems 2020; 5:5/2/e00578-19. [PMID: 32234773 PMCID: PMC7112960 DOI: 10.1128/msystems.00578-19] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Type 2 diabetes (T2D) is a complex metabolic syndrome characterized by insulin dysfunction and abnormalities in glucose and lipid metabolism. The gut microbiome has been recently identified as an important factor for development of T2D. In this study, a total of 102 subjects were recruited, and we have looked at the gut microbiota of prediabetics (PreDMs) (n = 17), newly diagnosed diabetics (NewDMs) (n = 11), and diabetics on antidiabetic treatment (KnownDMs) (n = 39) and compared them with healthy nondiabetics (ND) (n = 35). Twenty-five different serum biomarkers were measured to assess the status of diabetes and their association with gut microbiota. Our analysis revealed nine different genera as differentially abundant in four study groups. Among them, Akkermansia, Blautia, and Ruminococcus were found to be significantly (P < 0.05) decreased, while Lactobacillus was increased in NewDMs compared to ND and recovered in KnownDMs. Akkermansia was inversely correlated with HbA1c and positively correlated with total antioxidants. Compared to ND, there was increased abundance of Megasphaera, Escherichia, and Acidaminococcus and decreased abundance of Sutterella in KnownDMs. Among many taxa known to act as community drivers during disease progression, we observed genus Sutterella as a common driver taxon among all diabetic groups. On the basis of the results of random forest analysis, we found that the genera Akkermansia and Sutterella and that the serum metabolites fasting glucose, HbA1c, methionine, and total antioxidants were highly discriminative factors among studied groups. Taken together, our data revealed that gut microbial diversity of NewDMs but not of PreDMs is significantly different from that of ND. Interestingly, after antidiabetic treatment, the microbial diversity of KnownDMs tends to recover toward that of ND.IMPORTANCE Gut microbiota is considered to play a role in disease progression, and previous studies have reported an association of microbiome dysbiosis with T2D. In this study, we have attempted to investigate gut microbiota of ND, PreDMs, NewDMs, and KnownDMs. We found that the genera Akkermansia and Blautia decreased significantly (P < 0.05) in treatment-naive diabetics and were restored in KnownDMs on antidiabetic treatment. To the best of our knowledge, comparative studies on shifts in the microbial community in individuals of different diabetic states are lacking. Understanding the transition of microbiota and its association with serum biomarkers in diabetics with different disease states may pave the way for new therapeutic approaches for T2D.
Collapse
|
133
|
Chowdhury S, Fong SS. Computational Modeling of the Human Microbiome. Microorganisms 2020; 8:microorganisms8020197. [PMID: 32023941 PMCID: PMC7074762 DOI: 10.3390/microorganisms8020197] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/23/2020] [Accepted: 01/27/2020] [Indexed: 12/20/2022] Open
Abstract
The impact of microorganisms on human health has long been acknowledged and studied, but recent advances in research methodologies have enabled a new systems-level perspective on the collections of microorganisms associated with humans, the human microbiome. Large-scale collaborative efforts such as the NIH Human Microbiome Project have sought to kick-start research on the human microbiome by providing foundational information on microbial composition based upon specific sites across the human body. Here, we focus on the four main anatomical sites of the human microbiome: gut, oral, skin, and vaginal, and provide information on site-specific background, experimental data, and computational modeling. Each of the site-specific microbiomes has unique organisms and phenomena associated with them; there are also high-level commonalities. By providing an overview of different human microbiome sites, we hope to provide a perspective where detailed, site-specific research is needed to understand causal phenomena that impact human health, but there is equally a need for more generalized methodology improvements that would benefit all human microbiome research.
Collapse
Affiliation(s)
- Shomeek Chowdhury
- Integrative Life Sciences, Virginia Commonwealth University, 1000 West Cary Street, Richmond, VA 23284 USA;
| | - Stephen S. Fong
- Chemical and Life Science Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA 23284, USA
- Correspondence:
| |
Collapse
|
134
|
Chen X, Sun H, Jiang F, Shen Y, Li X, Hu X, Shen X, Wei P. Alteration of the gut microbiota associated with childhood obesity by 16S rRNA gene sequencing. PeerJ 2020; 8:e8317. [PMID: 31976177 PMCID: PMC6968493 DOI: 10.7717/peerj.8317] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 11/29/2019] [Indexed: 12/13/2022] Open
Abstract
Background Obesity is a global epidemic in the industrialized and developing world, and many children suffer from obesity-related complications. Gut microbiota dysbiosis might have significant effect on the development of obesity. The microbiota continues to develop through childhood and thus childhood may be the prime time for microbiota interventions to realize health promotion or disease prevention. Therefore, it is crucial to understand the structure and function of pediatric gut microbiota. Methods According to the inclusion criteria and exclusion criteria, twenty-three normal weight and twenty-eight obese children were recruited from Nanjing, China. Genomic DNA was extracted from fecal samples. The V4 region of the bacterial 16S rDNA was amplified by PCR, and sequencing was applied to analyze the gut microbiota diversity and composition using the Illumina HiSeq 2500 platform. Results The number of operational taxonomic units (OTUs) showed a decrease in the diversity of gut microbiota with increasing body weight. The alpha diversity indices showed that the normal weight group had higher abundance and observed species than the obese group (Chao1: P < 0.001; observed species: P < 0.001; PD whole tree: P < 0.001; Shannon index: P = 0.008). Principal coordinate analysis (PCoA) and Nonmetric multidimensional scaling (NMDS) revealed significant differences in gut microbial community structure between the normal weight group and the obese group. The liner discriminant analysis (LDA) effect size (LEfSe) analysis showed that fifty-five species of bacteria were abundant in the fecal samples of the normal weight group and forty-five species of bacteria were abundant in the obese group. In regard to phyla, the gut microbiota in the obese group had lower proportions of Bacteroidetes (51.35%) compared to the normal weight group (55.48%) (P = 0.030). There was no statistical difference in Firmicutes between the two groups (P = 0.436), and the Firmicutes/Bacteroidetes between the two groups had no statistical difference (P = 0.983). At the genus level, Faecalibacterium, Phascolarctobacterium, Lachnospira, Megamonas, and Haemophilus were significantly more abundant in the obese group than in the normal weight group (P = 0.048, P = 0.018, P < 0.001, P = 0.040, and P = 0.003, respectively). The fecal microbiota of children in the obese group had lower proportions of Oscillospira and Dialister compared to the normal weight group (P = 0.002 and P = 0.002, respectively). Conclusions Our results showed a decrease in gut microbiota abundance and diversity as the BMI increased. Variations in the bacterial community structure were associated with obesity. Gut microbiota dysbiosis might play a crucial part in the development of obesity in Chinese children.
Collapse
Affiliation(s)
- Xiaowei Chen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China.,Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| | - Haixiang Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Fei Jiang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China.,Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| | - Yan Shen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China.,Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| | - Xin Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Xueju Hu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Xiaobing Shen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China.,Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| | - Pingmin Wei
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China.,Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| |
Collapse
|
135
|
Piras C, Pintus R, Pruna D, Dessì A, Atzori L, Fanos V. Pediatric Acute-onset Neuropsychiatric Syndrome and Mycoplasma Pneumoniae Infection: A Case Report Analysis with a Metabolomics Approach. Curr Pediatr Rev 2020; 16:183-193. [PMID: 31642785 PMCID: PMC8193809 DOI: 10.2174/1573396315666191022102925] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/26/2019] [Accepted: 10/02/2019] [Indexed: 01/27/2023]
Abstract
Pediatric Acute-onset Neuropsychiatric Syndrome (PANS) is a clinical condition characterized by a sudden and dramatic obsessive-compulsive disorder with a suggested post-infectious immune-mediated etiology. This condition is accompanied by an extensive series of relatively serious neuropsychiatric symptoms. The diagnosis of PANS is made by "exclusion", as the individual PANS symptoms overlap with a multiplicity of psychiatric disorders with the onset in childhood. A number of researchers accumulated evidence to support the hypothesis that PANS was closely associated with a number of infections. In the last decade, metabolomics played an essential role in improving the knowledge of complex biological systems and identifying potential new biomarkers as indicators of pathological progressions or pharmacologic responses to therapy. The metabolome is considered the most predictive phenotype, capable of recognizing epigenetic differences, reflecting more closely the clinical reality at any given moment and thus providing extremely dynamic data. In the present work, the most recent hypothesis and suggested mechanisms of this condition are reviewed and the case of a 10 - year-old girl with PANS is described, before and after clarithromycin treatment. The main results of this case report are discussed from a metabolomics point of view. The alteration of several metabolic pathways concerning the microbial activity highlights the possible role of the microbiome in the development of PANS. Furthermore, different metabolic perturbations at the level of protein biosynthesis, energy and amino acid metabolisms are observed and discussed. Based on our observations, it is believed that metabolomics is a promising technology to unravel the mysteries of PANS in the near future.
Collapse
Affiliation(s)
- Cristina Piras
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Roberta Pintus
- Department of Surgical Sciences, Neonatal Intensive Care Unit, Puericulture Institute and Neonatal Section, Azienda Ospedaliera Universitaria, Cagliari, Italy
| | - Dario Pruna
- Pediatric Neurology and Epileptology Unit, Brotzu Hospital Trust, Cagliari, Italy
| | - Angelica Dessì
- Department of Surgical Sciences, Neonatal Intensive Care Unit, Puericulture Institute and Neonatal Section, Azienda Ospedaliera Universitaria, Cagliari, Italy
| | - Luigi Atzori
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Vassilios Fanos
- Department of Surgical Sciences, Neonatal Intensive Care Unit, Puericulture Institute and Neonatal Section, Azienda Ospedaliera Universitaria, Cagliari, Italy
| |
Collapse
|
136
|
Khan MW, Layden BT. Gestational Glucose Metabolism: Focus on the Role and Mechanisms of Insulin Resistance. MATERNAL-FETAL AND NEONATAL ENDOCRINOLOGY 2020:75-90. [DOI: 10.1016/b978-0-12-814823-5.00006-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
137
|
Nakov R, Velikova T. Chemical Metabolism of Xenobiotics by Gut Microbiota. Curr Drug Metab 2020; 21:260-269. [PMID: 32124693 DOI: 10.2174/1389200221666200303113830] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/05/2020] [Accepted: 02/12/2020] [Indexed: 02/08/2023]
Abstract
Among the gut microbiota's newly explored roles in human biology is the ability to modify the chemical structures of foreign compounds (xenobiotics). A growing body of evidence has now provided sufficient acumen on the role of the gut microbiota on xenobiotic metabolism, which could have an intense impact on the therapy for various diseases in the future. Gut microbial xenobiotic metabolites have altered bioavailability, bioactivity and toxicity and can intervene with the actions of human xenobiotic-metabolizing enzymes to affect the destiny of other ingested molecules. These modifications are diverse and could lead to physiologically important consequences. In the current manuscript we aim to review the data currently available on how the gut microbiota directly modifies drugs, dietary compounds, chemicals, pollutants, pesticides and herbal supplements.
Collapse
Affiliation(s)
- Radislav Nakov
- Clinic of Gastroenterology, Tsaritsa Yoanna University Hospital, Medical University of Sofia, Sofia, Bulgaria
| | | |
Collapse
|
138
|
Microbiota, type 2 diabetes and non-alcoholic fatty liver disease: protocol of an observational study. J Transl Med 2019; 17:408. [PMID: 31801616 PMCID: PMC6891972 DOI: 10.1186/s12967-019-02130-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/07/2019] [Indexed: 02/07/2023] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is characterized by triglyceride accumulation in the hepatocytes in the absence of alcohol overconsumption, commonly associated with insulin resistance and obesity. Both NAFLD and type 2 diabetes (T2D) are characterized by an altered microbiota composition, however the role of the microbiota in NAFLD and T2D is not well understood. To assess the relationship between alteration in the microbiota and NAFLD while dissecting the role of T2D, we established a nested study on T2D and non-T2D individuals within the Cooperative Health Research In South Tyrol (CHRIS) study, called the CHRIS-NAFLD study. Here, we present the study protocol along with baseline and follow-up characteristics of study participants. Methods Among the first 4979 CHRIS study participants, 227 individuals with T2D were identified and recalled, along with 227 age- and sex-matched non-T2D individuals. Participants underwent ultrasound and transient elastography examination to evaluate the presence of hepatic steatosis and liver stiffness. Additionally, sampling of saliva and faeces, biochemical measurements and clinical interviews were carried out. Results We recruited 173 T2D and 183 non-T2D participants (78% overall response rate). Hepatic steatosis was more common in T2D (63.7%) than non-T2D (36.3%) participants. T2D participants also had higher levels of liver stiffness (median 4.8 kPa, interquartile range (IQR) 3.7, 5.9) than non-T2D participants (median 3.9 kPa, IQR 3.3, 5.1). The non-invasive scoring systems like the NAFLD fibrosis score (NFS) suggests an increased liver fibrosis in T2D (mean − 0.55, standard deviation, SD, 1.30) than non-T2D participants (mean − 1.30, SD, 1.17). Discussion Given the comprehensive biochemical and clinical characterization of study participants, once the bioinformatics classification of the microbiota will be completed, the CHRIS-NAFLD study will become a useful resource to further our understanding of the relationship between microbiota, T2D and NAFLD.
Collapse
|
139
|
Francescangeli F, De Angelis ML, Zeuner A. Dietary Factors in the Control of Gut Homeostasis, Intestinal Stem Cells, and Colorectal Cancer. Nutrients 2019; 11:nu11122936. [PMID: 31816977 PMCID: PMC6950549 DOI: 10.3390/nu11122936] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/21/2019] [Accepted: 11/27/2019] [Indexed: 12/16/2022] Open
Abstract
Colorectal cancer (CRC) is the third commonly diagnosed cancer and the second leading cause of cancer-related deaths worldwide. Global CRC burden is expected to increase by 60% in the next decade, with low-income countries experiencing an escalation of CRC incidence and mortality in parallel to the adoption of western lifestyles. CRC incidence is also sharply increasing in individuals younger than 50 years, often presenting at advanced stages and with aggressive features. Both genetic and environmental factors have been recognized as major contributors for the development of CRC, the latter including diet-related conditions such as chronic inflammation and obesity. In particular, a diet rich in fat and sugars (Western-style diet, WSD) has been shown to induce multiple pathophysiological changes in the intestine linked to an increased risk of CRC. In this scenario, dietary factors have been recently shown to play novel unexpected roles in the regulation of intestinal stem cells (ISCs) and of the gut microbiota, which represent the two main biological systems responsible for intestinal homeostasis. Furthermore, diet is increasingly recognized to play a key role in the neoplastic transformation of ISCs and in the metabolic regulation of colorectal cancer stem cells. This review illustrates novel discoveries on the role of dietary components in regulating intestinal homeostasis and colorectal tumorigenesis. Particular focus is dedicated to new areas of research with potential clinical relevance including the effect of food components on ISCs and cancer stem cells (CSCs), the existence of CRC-specific microbial signatures and the alterations of intestinal homeostasis potentially involved in early-onset CRC. New insights on the role of dietary factors in intestinal regulation will provide new tools not only for the prevention and early diagnosis of CRC but also for improving the effectiveness of current CRC therapies.
Collapse
|
140
|
McCaffery JM. Precision behavioral medicine: Implications of genetic and genomic discoveries for behavioral weight loss treatment. ACTA ACUST UNITED AC 2019; 73:1045-1055. [PMID: 30394782 DOI: 10.1037/amp0000253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This article reviews the concept of precision behavioral medicine and the progress toward applying genetics and genomics as tools to optimize weight management intervention. We discuss genetic, epigenetic, and genomic markers, as well as interactions between genetics and the environment as they relate to obesity and behavioral weight loss to date. Recommendations for the conditions under which genetics and genomics could be incorporated to support clinical decision-making in behavioral weight loss are outlined and illustrative scenarios of how this approach could improve clinical outcomes are provided. It is concluded that there is not yet sufficient evidence to leverage genetics or genomics to aid the treatment of obesity but the foundations are being laid. (PsycINFO Database Record (c) 2018 APA, all rights reserved).
Collapse
Affiliation(s)
- Jeanne M McCaffery
- Weight Control and Diabetes Research Center, Department of Psychiatry and Human Behavior, The Miriam Hospital
| |
Collapse
|
141
|
Karusheva Y, Koessler T, Strassburger K, Markgraf D, Mastrototaro L, Jelenik T, Simon MC, Pesta D, Zaharia OP, Bódis K, Bärenz F, Schmoll D, Wolkersdorfer M, Tura A, Pacini G, Burkart V, Müssig K, Szendroedi J, Roden M. Short-term dietary reduction of branched-chain amino acids reduces meal-induced insulin secretion and modifies microbiome composition in type 2 diabetes: a randomized controlled crossover trial. Am J Clin Nutr 2019; 110:1098-1107. [PMID: 31667519 PMCID: PMC6821637 DOI: 10.1093/ajcn/nqz191] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/19/2019] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Epidemiological studies have shown that increased circulating branched-chain amino acids (BCAAs) are associated with insulin resistance and type 2 diabetes (T2D). This may result from altered energy metabolism or dietary habits. OBJECTIVE We hypothesized that a lower intake of BCAAs improves tissue-specific insulin sensitivity. METHODS This randomized, placebo-controlled, double-blinded, crossover trial examined well-controlled T2D patients receiving isocaloric diets (protein: 1 g/kg body weight) for 4 wk. Protein requirements were covered by commercially available food supplemented ≤60% by an AA mixture either containing all AAs or lacking BCAAs. The dietary intervention ensured sufficient BCAA supply above the recommended minimum daily intake. The patients underwent the mixed meal tolerance test (MMT), hyperinsulinemic-euglycemic clamps (HECs), and skeletal muscle and white adipose tissue biopsies to assess insulin signaling. RESULTS After the BCAA- diet, BCAAs were reduced by 17% during fasting (P < 0.001), by 13% during HEC (P < 0.01), and by 62% during the MMT (P < 0.001). Under clamp conditions, whole-body and hepatic insulin sensitivity did not differ between diets. After the BCAA- diet, however, the oral glucose sensitivity index was 24% (P < 0.01) and circulating fibroblast-growth factor 21 was 21% higher (P < 0.05), whereas meal-derived insulin secretion was 28% lower (P < 0.05). Adipose tissue expression of the mechanistic target of rapamycin was 13% lower, whereas the mitochondrial respiratory control ratio was 1.7-fold higher (both P < 0.05). The fecal microbiome was enriched in Bacteroidetes but depleted of Firmicutes. CONCLUSIONS Short-term dietary reduction of BCAAs decreases postprandial insulin secretion and improves white adipose tissue metabolism and gut microbiome composition. Longer-term studies will be needed to evaluate the safety and metabolic efficacy in diabetes patients.This trial was registered at clinicaltrials.gov as NCT03261362.
Collapse
Affiliation(s)
- Yanislava Karusheva
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany
| | - Theresa Koessler
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Klaus Strassburger
- German Center for Diabetes Research, München-Neuherberg, Germany,Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Daniel Markgraf
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany
| | - Lucia Mastrototaro
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany
| | - Tomas Jelenik
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany
| | - Marie-Christine Simon
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany
| | - Dominik Pesta
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany
| | - Oana-Patricia Zaharia
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany
| | - Kálmán Bódis
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Felix Bärenz
- Sanofi-Aventis Deutschland GmbH, Frankfurt, Germany
| | | | | | - Andrea Tura
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council, Padova, Italy
| | - Giovanni Pacini
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council, Padova, Italy
| | - Volker Burkart
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany
| | - Karsten Müssig
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Julia Szendroedi
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany,Address correspondence to JS (e-mail: )
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
142
|
Stedman A, Brunner K, Nigro G. Decrypting the communication between microbes and the intestinal mucosa-A brief review on Pathogénie Microbienne Moléculaire's latest research. Cell Microbiol 2019; 21:e13118. [PMID: 31634976 DOI: 10.1111/cmi.13118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 09/13/2019] [Accepted: 09/14/2019] [Indexed: 12/11/2022]
Abstract
Over the past 10 years, the "Pathogénie Microbienne Moléculaire" unit of Professor Philippe Sansonetti has studied the molecular cross talk between the intestinal microbiota and the gut epithelium, aiming to better understand how this mutualistic symbiosis delineates homoeostasis and, when perturbed, prompts pathology. To do so, the unit has manipulated both bacterial and epithelial cells, and used cutting-edge technology. More recently, the lab has turned its focus also on studying the intestinal crypt and more specifically the intestinal stem cell for their role in epithelial regeneration and long-term epithelium renewal. Here, we provide a brief review summarising recent results obtained from the lab, with particular focus on the intestinal crypt.
Collapse
Affiliation(s)
- Aline Stedman
- Molecular Microbial Pathogenesis Unit, INSERM U1202, Institut Pasteur, Paris, France.,Institut de Biologie Paris-Seine, CNRS, Sorbonne Université, Paris, France
| | - Katja Brunner
- Molecular Microbial Pathogenesis Unit, INSERM U1202, Institut Pasteur, Paris, France
| | - Giulia Nigro
- Molecular Microbial Pathogenesis Unit, INSERM U1202, Institut Pasteur, Paris, France
| |
Collapse
|
143
|
Huang YC, Huang LT, Sheen JM, Hou CY, Yeh YT, Chiang CP, Lin IC, Tiao MM, Tsai CC, Lin YJ, Chen CC, Tain YL, Yu HR. Resveratrol treatment improves the altered metabolism and related dysbiosis of gut programed by prenatal high-fat diet and postnatal high-fat diet exposure. J Nutr Biochem 2019; 75:108260. [PMID: 31707285 DOI: 10.1016/j.jnutbio.2019.108260] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 05/21/2019] [Accepted: 10/23/2019] [Indexed: 02/07/2023]
Abstract
A maternal high-fat (HF) diet sensitizes offspring to the adverse effects of postnatal HF intake and can lead to metabolic dysregulation. Resveratrol, a natural polyphenolic compound found in grapes and red wine, could help to relieve metabolic syndrome dysregulation. Since the gut microbiota is known to be closely related to metabolic homeostasis, this study aimed to investigate the impact of a combination of maternal and postweaning HF diets on the gut microbiota and whether resveratrol could relieve the gut dysbiosis associated with metabolic dysregulation. Sprague-Dawley dams were sustained on either a chow or HF diet before mating, during pregnancy and during lactation. Their offspring were randomly fed chow or a HF diet after weaning. Four experimental groups were generated: CC (maternal/postnatal chow diet), HC (maternal HF/postnatal chow diet), CH (maternal chow/postnatal high-fat diet) and HH (maternal/postnatal HF diet). A fifth group consisted of HH with resveratrol treatment. We found that both maternal and postnatal HF exposure has a distinct effect on the gut microbiota metagenome of offspring. Maternal HF diet exposure decreased plasma acetate, propionate and butyrate level, while postnatal HF diet exposure decreased plasma acetate level in adult life. The metabolic dysregulation programed by the maternal and postnatal HF diets was related to the relevant gut microbiota. Resveratrol treatment ameliorated the altered plasma propionate level related to maternal HF and postnatal HF diet treatment. Resveratrol treatment also improved most of the altered metabolic dysregulation and related dysbiosis programmed by maternal and postnatal HF diet exposure.
Collapse
Affiliation(s)
- Yi-Chuan Huang
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Li-Tung Huang
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Jiunn-Ming Sheen
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chih-Yao Hou
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung, Taiwan
| | - Yao-Tsung Yeh
- Aging and Disease Prevention Research Center, Fooyin University, Kaohsiung, Taiwan; Department of Education and Research, Fooyin University Hospital, Pingtung, Taiwan; Department of Medical Laboratory Sciences and Biotechnology, Fooyin University, Kaohsiung, Taiwan
| | - Chih-Po Chiang
- Aging and Disease Prevention Research Center, Fooyin University, Kaohsiung, Taiwan; Department of Education and Research, Fooyin University Hospital, Pingtung, Taiwan; Department of Medical Laboratory Sciences and Biotechnology, Fooyin University, Kaohsiung, Taiwan
| | - I-Chun Lin
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Mao-Meng Tiao
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ching-Chou Tsai
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital
| | - Yu-Ju Lin
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital
| | - Chih-Cheng Chen
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - You-Lin Tain
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hong-Ren Yu
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| |
Collapse
|
144
|
Medina DA, Li T, Thomson P, Artacho A, Pérez-Brocal V, Moya A. Cross-Regional View of Functional and Taxonomic Microbiota Composition in Obesity and Post-obesity Treatment Shows Country Specific Microbial Contribution. Front Microbiol 2019; 10:2346. [PMID: 31681211 PMCID: PMC6812679 DOI: 10.3389/fmicb.2019.02346] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 09/26/2019] [Indexed: 02/05/2023] Open
Abstract
Gut microbiota has been shown to have an important influence on host health. The microbial composition of the human gut microbiota is modulated by diet and other lifestyle habits and it has been reported that microbial diversity is altered in obese people. Obesity is a worldwide health problem that negatively impacts the quality of life. Currently, the widespread treatment for obesity is bariatric surgery. Interestingly, gut microbiota has been shown to be a relevant factor in effective weight loss after bariatric surgery. Since that the human gut microbiota of normal subjects differs between geographic regions, it is possible that rearrangements of the gut microbiota in dysbiosis context are also region-specific. To better understand how gut microbiota contribute to obesity, this study compared the composition of the human gut microbiota of obese and lean people from six different regions and showed that the microbiota compositions in the context of obesity were specific to each studied geographic location. Furthermore, we analyzed the functional patterns using shotgun DNA metagenomic sequencing and compared the results with other obesity-related metagenomic studies, we observed that microbial contribution to functional pathways were country-specific. Nevertheless, our study showed that although microbial composition of obese patients was country-specific, the overall metabolic functions appeared to be the same between countries, indicating that different microbiota components contribute to similar metabolic outcomes to yield functional redundancy. Furthermore, we studied the microbiota functional changes of obese patients after bariatric surgery, by shotgun metagenomics sequencing and observed that changes in functional pathways were specific to the type of obesity treatment. In all, our study provides new insights into the differences and similarities of obese gut microbiota in relation to geographic location and obesity treatments.
Collapse
Affiliation(s)
- Daniel A. Medina
- Laboratorio de Biotecnología Aplicada, Facultad de Medicina Veterinaria, Universidad San Sebastián, Puerto Montt, Chile
| | - Tianlu Li
- Chromatin and Disease Group, Cancer Epigenetics and Biology Programme (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
- Epigenetics and Immune Disease Group, Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
| | - Pamela Thomson
- Departamento de Ingeniería Química y Bioprocesos, Escuela de Ingeniería, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandro Artacho
- Genomics and Health Area, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO)-Salud Pública, Valencia, Spain
| | - Vicente Pérez-Brocal
- Genomics and Health Area, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO)-Salud Pública, Valencia, Spain
| | - Andrés Moya
- Genomics and Health Area, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO)-Salud Pública, Valencia, Spain
- Integrative Systems Biology Institute, University of Valencia, CSIC, Valencia, Spain
- Biomedical Research Centre Network for Epidemiology and Public Health (CIBEResp), Madrid, Spain
| |
Collapse
|
145
|
Gut Microbiome: A Promising Biomarker for Immunotherapy in Colorectal Cancer. Int J Mol Sci 2019; 20:ijms20174155. [PMID: 31450712 PMCID: PMC6747470 DOI: 10.3390/ijms20174155] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 02/06/2023] Open
Abstract
Research has been driven towards finding therapy predictive biomarkers for colorectal cancer (CRC) with a special interest in studying the gut microbiome. Gut microbiome acts not only as a barrier to prevent bacterial invasion and infection, but it also affects the efficacy of hematopoietic-cell transplantation, chemotherapy, and immunotherapy. Recently, immunotherapy, which potentiates the host immune system, has revolutionized cancer therapy in general and CRC treatment specifically by increasing the quality of life and the survival of a subset of patients with this disease. In immunotherapy, the gut microbiome plays an important role in cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) blockade, programmed cell death protein 1 (PD-L1) mediation, and T cell stimulation. As such, this review will cover the role of gut microbiome in CRC, summarize approved immunotherapy treatments for CRC, and focus on the potential use of gut microbiome as a biomarker for immunotherapy.
Collapse
|
146
|
Li H, Li H, Wang J, Guo L, Fan H, Zheng H, Yang Z, Huang X, Chu M, Yang F, He Z, Li N, Yang J, Wu Q, Shi H, Liu L. The altered gut virome community in rhesus monkeys is correlated with the gut bacterial microbiome and associated metabolites. Virol J 2019; 16:105. [PMID: 31426820 PMCID: PMC6700990 DOI: 10.1186/s12985-019-1211-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/05/2019] [Indexed: 12/13/2022] Open
Abstract
Background The gut microbiome is closely associated with the health of the host; although the interaction between the bacterial microbiome and the whole virome has rarely been studied, it is likely of medical importance. Examination of the interactions between the gut bacterial microbiome and virome of rhesus monkey would significantly contribute to revealing the gut microbiome composition. Methods Here, we conducted a metagenomic analysis of the gut microbiome of rhesus monkeys in a longitudinal cohort treated with an antibiotic cocktail, and we documented the interactions between the bacterial microbiome and virome. The depletion of viral populations was confirmed at the species level by real-time PCR. We also detected changes in the gut metabolome by GC-MS and LC-MS. Results A majority of bacteria were depleted after treatment with antibiotics, and the Shannon diversity index decreased from 2.95 to 0.22. Furthermore, the abundance-based coverage estimator (ACE) decreased from 104.47 to 33.84, and the abundance of eukaryotic viruses also changed substantially. In the annotation, 6 families of DNA viruses and 1 bacteriophage family were present in the normal monkeys but absent after gut bacterial microbiome depletion. Intriguingly, we discovered that changes in the gut bacterial microbiome composition may promote changes in the gut virome composition, and tryptophan, arginine, and quinone may play roles in the interaction between the bacterial microbiome and virome. Conclusion Our results indicated that the clearly altered composition of the virome was correlated with depletion in the bacterial community and that metabolites produced by bacteria possibly play important roles in the interaction. Electronic supplementary material The online version of this article (10.1186/s12985-019-1211-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Heng Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, China
| | - Hongzhe Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, China
| | - Jingjing Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, China
| | - Lei Guo
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, China
| | - Haitao Fan
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, China
| | - Huiwen Zheng
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, China
| | - Zening Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, China
| | - Xing Huang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, China
| | - Manman Chu
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, China
| | - Fengmei Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Zhanlong He
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Nan Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, China
| | - Jinxi Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, China
| | - Qiongwen Wu
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, China
| | - Haijing Shi
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China. .,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, China.
| | - Longding Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China. .,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, China.
| |
Collapse
|
147
|
Hasan N, Yang H. Factors affecting the composition of the gut microbiota, and its modulation. PeerJ 2019; 7:e7502. [PMID: 31440436 PMCID: PMC6699480 DOI: 10.7717/peerj.7502] [Citation(s) in RCA: 429] [Impact Index Per Article: 71.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/17/2019] [Indexed: 12/13/2022] Open
Abstract
Gut microbiota have important functions in the body, and imbalances in the composition and diversity of those microbiota can cause several diseases. The host fosters favorable microbiota by releasing specific factors, such as microRNAs, and nonspecific factors, such as antimicrobial peptides, mucus and immunoglobulin A that encourage the growth of specific types of bacteria and inhibit the growth of others. Diet, antibiotics, and age can change gut microbiota, and many studies have shown the relationship between disorders of the microbiota and several diseases and reported some ways to modulate that balance. In this review, we highlight how the host shapes its gut microbiota via specific and nonspecific factors, how environmental and nutritional factors affect it, and how to modulate it using prebiotics, probiotics, and fecal microbiota transplantation.
Collapse
Affiliation(s)
- Nihal Hasan
- Department of Microbiology, Northeast Forestry University, Harbin, Heilongjiang, China.,Faculty of Health Science, Al-Baath University, Homs, Syria
| | - Hongyi Yang
- Department of Microbiology, Northeast Forestry University, Harbin, Heilongjiang, China
| |
Collapse
|
148
|
Liu H, Pan LL, Lv S, Yang Q, Zhang H, Chen W, Lv Z, Sun J. Alterations of Gut Microbiota and Blood Lipidome in Gestational Diabetes Mellitus With Hyperlipidemia. Front Physiol 2019; 10:1015. [PMID: 31447702 PMCID: PMC6691352 DOI: 10.3389/fphys.2019.01015] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/23/2019] [Indexed: 12/12/2022] Open
Abstract
Clinical gestational diabetes mellitus (GDM) is frequently associated with hyperlipidemia comorbidity. Altered human gut microbiome has been linked to GDM and hyperlipidemia, respectively but not the comorbid condition. We hypothesize that the occurrence of hyperlipidemia with GDM may be characterized by distinguishable gut microbiome and blood metabolomic patterns. We presented comprehensive microbiomic coupled with lipidomics analyses to characterize gut microbiota and lipometabolism of plasma samples in women with GDM only, hyperlipidemia only and those with diabetes plus hyperlipidemia, and to explore association of the gut microbiota composition with blood lipid profiles and clinical parameters of gestational diabetes with or without commodity. We found that the relative abundance of bacterial taxa Streptococcus, Faecalibacterium, Veillonella, Prevotella, Haemophilus and Actinomyces was significantly higher in diabetes plus hyperlipidemia cohorts. Moreover, several bacteria were correlated with fasting plasma glucose and blood lipid levels of the participants with GDM and hyperlipidemia. The altered plasma lipidome in subjects with diabetes plus hyperlipidemia suggested that characteristic blood lipid profiles were associated with the pathogenesis of gestational diabetes plus hyperlipidemia. Collectively, this study provides insights on changes in fecal microbiota and plasma lipidome to predict and characterize the development of gestational diabetes with lipid metabolic abnormality.
Collapse
Affiliation(s)
- He Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Li-Long Pan
- School of Medicine, Jiangnan University, Wuxi, China
| | - Siting Lv
- Department of Obstetrics, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Qin Yang
- School of Medicine, Jiangnan University, Wuxi, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Zhuwu Lv
- Department of Obstetrics, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
149
|
Kemis JH, Linke V, Barrett KL, Boehm FJ, Traeger LL, Keller MP, Rabaglia ME, Schueler KL, Stapleton DS, Gatti DM, Churchill GA, Amador-Noguez D, Russell JD, Yandell BS, Broman KW, Coon JJ, Attie AD, Rey FE. Genetic determinants of gut microbiota composition and bile acid profiles in mice. PLoS Genet 2019; 15:e1008073. [PMID: 31465442 PMCID: PMC6715156 DOI: 10.1371/journal.pgen.1008073] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 06/14/2019] [Indexed: 02/03/2023] Open
Abstract
The microbial communities that inhabit the distal gut of humans and other mammals exhibit large inter-individual variation. While host genetics is a known factor that influences gut microbiota composition, the mechanisms underlying this variation remain largely unknown. Bile acids (BAs) are hormones that are produced by the host and chemically modified by gut bacteria. BAs serve as environmental cues and nutrients to microbes, but they can also have antibacterial effects. We hypothesized that host genetic variation in BA metabolism and homeostasis influence gut microbiota composition. To address this, we used the Diversity Outbred (DO) stock, a population of genetically distinct mice derived from eight founder strains. We characterized the fecal microbiota composition and plasma and cecal BA profiles from 400 DO mice maintained on a high-fat high-sucrose diet for ~22 weeks. Using quantitative trait locus (QTL) analysis, we identified several genomic regions associated with variations in both bacterial and BA profiles. Notably, we found overlapping QTL for Turicibacter sp. and plasma cholic acid, which mapped to a locus containing the gene for the ileal bile acid transporter, Slc10a2. Mediation analysis and subsequent follow-up validation experiments suggest that differences in Slc10a2 gene expression associated with the different strains influences levels of both traits and revealed novel interactions between Turicibacter and BAs. This work illustrates how systems genetics can be utilized to generate testable hypotheses and provide insight into host-microbe interactions.
Collapse
Affiliation(s)
- Julia H. Kemis
- Department of Bacteriology, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Vanessa Linke
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Kelsey L. Barrett
- Department of Bacteriology, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Frederick J. Boehm
- Department of Statistics, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Lindsay L. Traeger
- Department of Bacteriology, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Mark P. Keller
- Department of Biochemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Mary E. Rabaglia
- Department of Biochemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Kathryn L. Schueler
- Department of Biochemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Donald S. Stapleton
- Department of Biochemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Daniel M. Gatti
- Jackson Laboratory, Bar Harbor, Maine, United States of America
| | | | - Daniel Amador-Noguez
- Department of Bacteriology, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Jason D. Russell
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Brian S. Yandell
- Department of Statistics, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Karl W. Broman
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Joshua J. Coon
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Morgridge Institute for Research, Madison, Wisconsin, United States of America
- Department of Biomolecular Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Alan D. Attie
- Department of Biochemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Federico E. Rey
- Department of Bacteriology, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
150
|
Clos-Garcia M, Andrés-Marin N, Fernández-Eulate G, Abecia L, Lavín JL, van Liempd S, Cabrera D, Royo F, Valero A, Errazquin N, Vega MCG, Govillard L, Tackett MR, Tejada G, Gónzalez E, Anguita J, Bujanda L, Orcasitas AMC, Aransay AM, Maíz O, López de Munain A, Falcón-Pérez JM. Gut microbiome and serum metabolome analyses identify molecular biomarkers and altered glutamate metabolism in fibromyalgia. EBioMedicine 2019; 46:499-511. [PMID: 31327695 PMCID: PMC6710987 DOI: 10.1016/j.ebiom.2019.07.031] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/24/2019] [Accepted: 07/10/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Fibromyalgia is a complex, relatively unknown disease characterised by chronic, widespread musculoskeletal pain. The gut-brain axis connects the gut microbiome with the brain through the enteric nervous system (ENS); its disruption has been associated with psychiatric and gastrointestinal disorders. To gain an insight into the pathogenesis of fibromyalgia and identify diagnostic biomarkers, we combined different omics techniques to analyse microbiome and serum composition. METHODS We collected faeces and blood samples to study the microbiome, the serum metabolome and circulating cytokines and miRNAs from a cohort of 105 fibromyalgia patients and 54 age- and environment-matched healthy individuals. We sequenced the V3 and V4 regions of the 16S rDNA gene from faeces samples. UPLC-MS metabolomics and custom multiplex cytokine and miRNA analysis (FirePlex™ technology) were used to examine sera samples. Finally, we combined the different data types to search for potential biomarkers. RESULTS We found that the diversity of bacteria is reduced in fibromyalgia patients. The abundance of the Bifidobacterium and Eubacterium genera (bacteria participating in the metabolism of neurotransmitters in the host) in these patients was significantly reduced. The serum metabolome analysis revealed altered levels of glutamate and serine, suggesting changes in neurotransmitter metabolism. The combined serum metabolomics and gut microbiome datasets showed a certain degree of correlation, reflecting the effect of the microbiome on metabolic activity. We also examined the microbiome and serum metabolites, cytokines and miRNAs as potential sources of molecular biomarkers of fibromyalgia. CONCLUSIONS Our results show that the microbiome analysis provides more significant biomarkers than the other techniques employed in the work. Gut microbiome analysis combined with serum metabolomics can shed new light onto the pathogenesis of fibromyalgia. We provide a list of bacteria whose abundance changes in this disease and propose several molecules as potential biomarkers that can be used to evaluate the current diagnostic criteria.
Collapse
Affiliation(s)
- Marc Clos-Garcia
- Exosomes Laboratory, CIC bioGUNE, CIBERehd, Bizkaia Technology Park, Derio, Spain; Department of Gastroenterology, Instituto Biodonostia, Universidad del País Vasco (UPV/EHU), CIBERehd (Centro de investigación en red de enfermedades hepáticas y digestiva) San Sebastian, Spain.
| | | | - Gorka Fernández-Eulate
- Department of Neurology, Donostia University Hospital, San Sebastian, Spain; Neuroscience Area, Biodonostia Health Research Institute, San Sebastian, Spain.
| | - Leticia Abecia
- Macrophage and Tick Vaccine Laboratory, CIC bioGUNE, Bizkaia Technology Park, Derio, Spain.
| | - José L Lavín
- Bioinformatics Unit, CIC bioGUNE, Bizkaia Technology Park, Derio, Spain.
| | - Sebastiaan van Liempd
- Metabolomics Platform, CIC bioGUNE, CIBERehd, Bizkaia Technology Park, Derio, Spain.
| | - Diana Cabrera
- Metabolomics Platform, CIC bioGUNE, CIBERehd, Bizkaia Technology Park, Derio, Spain.
| | - Félix Royo
- Exosomes Laboratory, CIC bioGUNE, CIBERehd, Bizkaia Technology Park, Derio, Spain.
| | - Alejandro Valero
- Department of Rheumatology, Donostia University Hospital, San Sebastian, Spain.
| | - Nerea Errazquin
- Department of Rheumatology, Gipuzcoa Policlinic, San Sebastian, Spain.
| | | | | | | | | | - Esperanza Gónzalez
- Exosomes Laboratory, CIC bioGUNE, CIBERehd, Bizkaia Technology Park, Derio, Spain.
| | - Juan Anguita
- Macrophage and Tick Vaccine Laboratory, CIC bioGUNE, Bizkaia Technology Park, Derio, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| | - Luis Bujanda
- Department of Gastroenterology, Instituto Biodonostia, Universidad del País Vasco (UPV/EHU), CIBERehd (Centro de investigación en red de enfermedades hepáticas y digestiva) San Sebastian, Spain.
| | | | - Ana M Aransay
- Genome Analysis Platform, CIC bioGUNE, CIBERehd, Bizkaia Technology Park, Derio, Spain.
| | - Olga Maíz
- Department of Rheumatology, Donostia University Hospital, San Sebastian, Spain.
| | - Adolfo López de Munain
- Department of Neurology, Donostia University Hospital, San Sebastian, Spain; Neuroscience Area, Biodonostia Health Research Institute, San Sebastian, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Spain; Department of Neurosciences, University of Basque Country UPV/EHU, San Sebastian, Spain.
| | - Juan Manuel Falcón-Pérez
- Exosomes Laboratory, CIC bioGUNE, CIBERehd, Bizkaia Technology Park, Derio, Spain; Metabolomics Platform, CIC bioGUNE, CIBERehd, Bizkaia Technology Park, Derio, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|