151
|
Bariatric Surgery Affects Plasma Levels of Alanine Aminotransferase Independent of Weight Loss: A Registry-Based Study. J Clin Med 2021; 10:jcm10122724. [PMID: 34203100 PMCID: PMC8234536 DOI: 10.3390/jcm10122724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 11/17/2022] Open
Abstract
Patients that undergo bariatric surgery experience weight loss and a reduction in the plasma levels of the hepatic enzymes alanine aminotransferase (ALT) and aspartate aminotransferase (AST). We used the Israeli national bariatric registry, which includes demographic, clinical, and biochemical data on 19,403 patients, of which 1335 patients had two-year follow-up data on ALT, AST, A1C, and BMI, to test the dependence of the reduction in the levels of ALT and AST on weight loss. The data were analyzed using regression models, retrospective matching, and time course analyses. Changes in liver enzymes did not correlate with change in BMI, and linear regression models did not demonstrate that the change in ALT and AST values were dependent on pre-operative levels of BMI or the extent of weight loss. ALT and AST levels were reduced two years after surgery compared with a cohort of retrospectively matched patients for ethnicity, sex, age, BMI, and A1C. Finally, patients who regained weight displayed a reduction in levels of liver enzymes. Our results suggest that bariatric surgery affects AST and ALT levels via weight loss dependent and independent mechanisms. Mechanistic studies that will identify the nature of this effect and the clinical relevance of ALT and AST levels to the post-bariatric liver function are warranted.
Collapse
|
152
|
Li JV, Ashrafian H, Sarafian M, Homola D, Rushton L, Barker G, Cabrera PM, Lewis MR, Darzi A, Lin E, Gletsu-Miller NA, Atkin SL, Sathyapalan T, Gooderham NJ, Nicholson JK, Marchesi JR, Athanasiou T, Holmes E. Roux-en-Y gastric bypass-induced bacterial perturbation contributes to altered host-bacterial co-metabolic phenotype. MICROBIOME 2021; 9:139. [PMID: 34127058 PMCID: PMC8201742 DOI: 10.1186/s40168-021-01086-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 04/27/2021] [Indexed: 05/04/2023]
Abstract
BACKGROUND Bariatric surgery, used to achieve effective weight loss in individuals with severe obesity, modifies the gut microbiota and systemic metabolism in both humans and animal models. The aim of the current study was to understand better the metabolic functions of the altered gut microbiome by conducting deep phenotyping of bariatric surgery patients and bacterial culturing to investigate causality of the metabolic observations. METHODS Three bariatric cohorts (n = 84, n = 14 and n = 9) with patients who had undergone Roux-en-Y gastric bypass (RYGB), sleeve gastrectomy (SG) or laparoscopic gastric banding (LGB), respectively, were enrolled. Metabolic and 16S rRNA bacterial profiles were compared between pre- and post-surgery. Faeces from RYGB patients and bacterial isolates were cultured to experimentally associate the observed metabolic changes in biofluids with the altered gut microbiome. RESULTS Compared to SG and LGB, RYGB induced the greatest weight loss and most profound metabolic and bacterial changes. RYGB patients showed increased aromatic amino acids-based host-bacterial co-metabolism, resulting in increased urinary excretion of 4-hydroxyphenylacetate, phenylacetylglutamine, 4-cresyl sulphate and indoxyl sulphate, and increased faecal excretion of tyramine and phenylacetate. Bacterial degradation of choline was increased as evidenced by altered urinary trimethylamine-N-oxide and dimethylamine excretion and faecal concentrations of dimethylamine. RYGB patients' bacteria had a greater capacity to produce tyramine from tyrosine, phenylalanine to phenylacetate and tryptophan to indole and tryptamine, compared to the microbiota from non-surgery, normal weight individuals. 3-Hydroxydicarboxylic acid metabolism and urinary excretion of primary bile acids, serum BCAAs and dimethyl sulfone were also perturbed following bariatric surgery. CONCLUSION Altered bacterial composition and metabolism contribute to metabolic observations in biofluids of patients following RYGB surgery. The impact of these changes on the functional clinical outcomes requires further investigation. Video abstract.
Collapse
Affiliation(s)
- Jia V Li
- Division of Digestive Disease, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, SW7 2AZ, UK
| | - Hutan Ashrafian
- Division of Surgery, Department of Surgery and Cancer, Imperial College London, London, SW7 2AZ, UK
| | - Magali Sarafian
- Division of Digestive Disease, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, SW7 2AZ, UK
| | - Daniel Homola
- Division of Digestive Disease, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, SW7 2AZ, UK
| | - Laura Rushton
- Division of Digestive Disease, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, SW7 2AZ, UK
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, SW7 2AZ, UK
| | - Grace Barker
- Division of Digestive Disease, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, SW7 2AZ, UK
| | - Paula Momo Cabrera
- Division of Digestive Disease, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, SW7 2AZ, UK
| | - Matthew R Lewis
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, SW7 2AZ, UK
| | - Ara Darzi
- Division of Surgery, Department of Surgery and Cancer, Imperial College London, London, SW7 2AZ, UK
| | - Edward Lin
- Division of General and Gastrointestinal Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | - Nana Adwoa Gletsu-Miller
- Department of Applied Health Science, School of Public Health, Indiana University Bloomington, 1025 E 7th Street, Bloomington, IN, 47405, USA
| | | | - Thozhukat Sathyapalan
- Department of Academic Endocrinology, Diabetes and Metabolism, Hull-York Medical School, Hull, UK
| | - Nigel J Gooderham
- Division of Digestive Disease, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, SW7 2AZ, UK
| | - Jeremy K Nicholson
- Centre for Computational and Systems Medicine, The Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA, 6150, Australia
| | - Julian R Marchesi
- Division of Digestive Disease, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, SW7 2AZ, UK
| | - Thanos Athanasiou
- Division of Surgery, Department of Surgery and Cancer, Imperial College London, London, SW7 2AZ, UK
| | - Elaine Holmes
- Division of Digestive Disease, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, SW7 2AZ, UK.
- Centre for Computational and Systems Medicine, The Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA, 6150, Australia.
| |
Collapse
|
153
|
Vangoitsenhoven R, Wilson R, Sharma G, Punchai S, Corcelles R, Froylich D, Mulya A, Schauer PR, Brethauer SA, Kirwan JP, Sangwan N, Brown JM, Aminian A. Metabolic effects of duodenojejunal bypass surgery in a rat model of type 1 diabetes. Surg Endosc 2021; 35:3104-3114. [PMID: 32607903 PMCID: PMC8633809 DOI: 10.1007/s00464-020-07741-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 06/12/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Metabolic surgery has beneficial metabolic effects, including remission of type 2 diabetes. We hypothesized that duodenojejunal bypass (DJB) surgery can protect against development of type 1 diabetes (T1D) by enhancing regulation of cellular and molecular pathways that control glucose homeostasis. METHODS BBDP/Wor rats, which are prone to develop spontaneous autoimmune T1D, underwent loop DJB (n = 15) or sham (n = 15) surgery at a median age of 41 days, before development of diabetes. At T1D diagnosis, a subcutaneous insulin pellet was implanted, oral glucose tolerance test was performed 21 days later, and tissues were collected 25 days after onset of T1D. Pancreas and liver tissues were assessed by histology and RT-qPCR. Fecal microbiota composition was analyzed by 16S V4 sequencing. RESULTS Postoperatively, DJB rats weighed less than sham rats (287.8 vs 329.9 g, P = 0.04). In both groups, 14 of 15 rats developed T1D, at similar age of onset (87 days in DJB vs 81 days in sham, P = 0.17). There was no difference in oral glucose tolerance, fasting and stimulated plasma insulin and c-peptide levels, and immunohistochemical analysis of insulin-positive cells in the pancreas. DJB rats needed 1.3 ± 0.4 insulin implants vs 1.9 ± 0.5 in sham rats (P = 0.002). Fasting and glucose stimulated glucagon-like peptide 1 (GLP-1) secretion was elevated after DJB surgery. DJB rats had reduced markers of metabolic stress in liver. After DJB, the fecal microbiome changed significantly, including increases in Akkermansia and Ruminococcus, while the changes were minimal in sham rats. CONCLUSION DJB does not protect against autoimmune T1D in BBDP/Wor rats, but reduces the need for exogenous insulin and facilitates other metabolic benefits including weight loss, increased GLP-1 secretion, reduced hepatic stress, and altered gut microbiome.
Collapse
Affiliation(s)
- Roman Vangoitsenhoven
- Department of General Surgery, Bariatric and Metabolic Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Rickesha Wilson
- Department of General Surgery, Bariatric and Metabolic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Gautam Sharma
- Department of General Surgery, Bariatric and Metabolic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Suriya Punchai
- Department of General Surgery, Bariatric and Metabolic Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Surgery, Khon Kaen University, Khon Kaen, Thailand
| | - Ricard Corcelles
- Department of General Surgery, Bariatric and Metabolic Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of General Surgery, Bariatric and Metabolic Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| | - Dvir Froylich
- Department of General Surgery, Bariatric and Metabolic Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of General Surgery, Carmel Medical Center, Haifa, Israel
| | - Anny Mulya
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Philip R Schauer
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Stacy A Brethauer
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - John P Kirwan
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Naseer Sangwan
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - J Mark Brown
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ali Aminian
- Department of General Surgery, Bariatric and Metabolic Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
154
|
Zeng H, Safratowich BD, Liu Z, Bukowski MR, Ishaq SL. Adequacy of calcium and vitamin D reduces inflammation, β-catenin signaling, and dysbiotic Parasutterela bacteria in the colon of C57BL/6 mice fed a western-style diet. J Nutr Biochem 2021; 92:108613. [PMID: 33705950 DOI: 10.1016/j.jnutbio.2021.108613] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/30/2020] [Accepted: 02/02/2021] [Indexed: 12/31/2022]
Abstract
Adoption of an obesogenic diet low in calcium and vitamin D (CaD) leads to increased obesity, colonic inflammation, and cancer. However, the underlying mechanisms remain to be elucidated. We tested the hypothesis that CaD supplementation (from inadequacy to adequacy) may reduce colonic inflammation, oncogenic signaling, and dysbiosis in the colon of C57BL/6 mice fed a Western diet. Male C57/BL6 mice (4-weeks old) were assigned to 3 dietary groups for 36 weeks: (1) AIN76A as a control diet (AIN); (2) a defined rodent "new Western diet" (NWD); or (3) NWD with CaD supplementation (NWD/CaD). Compared to the AIN, mice receiving the NWD or NWD/CaD exhibited more than 0.2-fold increase in the levels of plasma leptin, tumor necrosis factor α (TNF-α) and body weight. The levels of plasma interleukin 6 (IL-6), inflammatory cell infiltration, and β-catenin/Ki67 protein (oncogenic signaling) were increased more than 0.8-fold in the NWD (but not NWD/CaD) group compared to the AIN group. Consistent with the inflammatory phenotype, colonic secondary bile acid (inflammatory bacterial metabolite) levels increased more than 0.4-fold in the NWD group compared to the NWD/CaD and AIN groups. Furthermore, the abundance of colonic Proteobacteria (e.g., Parasutterela), considered signatures of dysbiosis, was increased more than four-fold; and the α diversity of colonic bacterial species, indicative of health, was decreased by 30% in the NWD group compared to the AIN and NWD/CaD groups. Collectively, CaD adequacy reduces colonic inflammation, β-catenin oncogenic signaling, secondary bile acids, and bacterial dysbiosis in mice fed with a Western diet.
Collapse
Affiliation(s)
- Huawei Zeng
- United States Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, North Dakota.
| | - Bryan D Safratowich
- United States Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, North Dakota
| | - Zhenhua Liu
- School of Public Health and Health Sciences, University of Massachusetts, Amherst, Massachusetts
| | - Michael R Bukowski
- United States Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, North Dakota
| | - Suzanne L Ishaq
- School of Food and Agriculture, University of Maine, Orono, Maine
| |
Collapse
|
155
|
Chen R, Song J, Lin L, Liu J, Yang C, Wang W. Visualizing the Growth and Division of Rat Gut Bacteria by D-Amino Acid-Based in vivo Labeling and FISH Staining. Front Mol Biosci 2021; 8:681938. [PMID: 34124162 PMCID: PMC8193097 DOI: 10.3389/fmolb.2021.681938] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/04/2021] [Indexed: 12/16/2022] Open
Abstract
Rat is a widely used mammalian model for gut microbiota research. However, due to the difficulties of individual in vitro culture of many of the gut bacteria, much information about the microbial behaviors in the rat gut remains largely unknown. Here, to characterize the in situ growth and division of rat gut bacteria, we apply a chemical strategy that integrates the use of sequential tagging with D-amino acid-based metabolic probes (STAMP) with fluorescence in situ hybridization (FISH) to rat gut microbiota. Following sequential gavages of two different fluorescent D-amino acid probes to rats, the resulting dually labeled gut bacteria provides chronological information of their in situ cell wall synthesis. After taxonomical labeling with FISH probes, most of which are newly designed in this study, we successfully identify the growth patterns of 15 bacterial species, including two that have not been cultured separately in the laboratory. Furthermore, using our labeling protocol, we record Butyrivibrio fibrisolvens cells growing at different growth stages of a complete cell division cycle, which offers a new scope for understanding basic microbial activities in the gut of mammalian hosts.
Collapse
Affiliation(s)
- Ru Chen
- Department of Digestive Diseases of Huashan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jia Song
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liyuan Lin
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Liu
- Department of Digestive Diseases of Huashan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Chaoyong Yang
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Key Laboratory for Chemical Biology of Fujian Province State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China
| | - Wei Wang
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
156
|
Lau E, Belda E, Picq P, Carvalho D, Ferreira-Magalhães M, Silva MM, Barroso I, Correia F, Vaz CP, Miranda I, Barbosa A, Clément K, Doré J, Freitas P, Prifti E. Gut microbiota changes after metabolic surgery in adult diabetic patients with mild obesity: a randomised controlled trial. Diabetol Metab Syndr 2021; 13:56. [PMID: 34020709 PMCID: PMC8139007 DOI: 10.1186/s13098-021-00672-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 04/30/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Roux-en-Y gastric bypass (RYGB) surgery is one of the most efficient procedures for the treatment of obesity, also improving metabolic and inflammatory status, in patients with mild obesity. The underlying mechanisms have not been fully understood, but gut microbiota is hypothesized to play a key role. Our aim was to evaluate the association between gut microbiota changes and anthropometric, metabolic and inflammatory profiles after metabolic surgery compared with medical therapy, in type 2 diabetic (T2DM) adults with mild obesity (BMI 30-35 kg/m2). METHODS DM2 was an open-label, randomised controlled clinical trial (RCT: ISRCTN53984585) with 2 arms: (i) surgical, and (ii) medical. The main outcome was gut microbiota changes after: metabolic surgery (Roux-en-Y gastric bypass-RYGB) versus standard medical therapy. Secondary outcomes included anthropometric, metabolic and inflammatory profiles. Clinical visits, blood workup, and stool samples were collected at baseline and months (M)1, 3, 6, 12. Gut microbiota was profiled using 16S rRNA targeted sequencing. RESULTS Twenty patients were included: 10 in surgical and 10 in medical arm. Anthropometric and metabolic comparative analysis favoured RYGB over medical arm. At M12, the percentage of weight loss was 25.5 vs. 4.9% (p < 0.001) and HbA1c was 6.2 vs. 7.7% (p < 0.001) respectively. We observed a continuous increase of genus richness after RYGB up until M12. In the medical arm, genus richness ended-up being significantly lower at M12. Composition analysis indicated significant changes of the overall microbial ecosystem (permanova p = 0.004, [R2 = 0.17]) during the follow-up period after RYGB. There was a strong association between improvement of anthropometric/metabolic/inflammatory biomarkers and increase in microbial richness and Proteobacterial lineages. CONCLUSIONS This was the first RCT studying composite clinical, analytic, and microbiome changes in T2DM patients with class 1 obesity after RYGB versus standard medical therapy. The remarkable phenotypic improvement after surgery occurred concomitantly with changes in the gut microbiome, but at a lower level. TRIAL REGISTRATION ISRCTN53984585.
Collapse
Affiliation(s)
- Eva Lau
- Department of Endocrinology and Nutrition, Centro Hospitalar de S. João, Alameda Professor Hernani Monteiro, 4200-319 Porto, Portugal
- CINTESIS - Center for Health Technologies and Information Systems Research - Faculty of Medicine, University of Porto, Porto, Portugal
| | - Eugeni Belda
- Integromics, Institute of Cardiometabolism and Nutrition, ICAN, Paris, France
| | - Paul Picq
- Integromics, Institute of Cardiometabolism and Nutrition, ICAN, Paris, France
| | - Davide Carvalho
- Department of Endocrinology and Nutrition, Centro Hospitalar de S. João, Alameda Professor Hernani Monteiro, 4200-319 Porto, Portugal
- I3S – Instituto de Investigação e Inovação em Saúde, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Manuel Ferreira-Magalhães
- CINTESIS - Center for Health Technologies and Information Systems Research - Faculty of Medicine, University of Porto, Porto, Portugal
- Health Information and Decision Sciences Department - Faculty of Medicine, Porto University, Porto, Portugal
| | - Maria Manuel Silva
- Department of Endocrinology and Nutrition, Centro Hospitalar de S. João, Alameda Professor Hernani Monteiro, 4200-319 Porto, Portugal
- I3S – Instituto de Investigação e Inovação em Saúde, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Isaac Barroso
- Department of Biochemistry, Centro Hospitalar de S. João, Porto, Portugal
- EpiUnit – Instituto de Saúde Pública, University of Porto, Porto, Portugal
| | - Flora Correia
- Department of Nutrition, Centro Hospitalar de S. João, Porto, Portugal
- Faculty of Nutrition and Food Science, Porto, Portugal
| | - Cidália Pina Vaz
- CINTESIS - Center for Health Technologies and Information Systems Research - Faculty of Medicine, University of Porto, Porto, Portugal
- Department of Pathology, Division of Microbiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Isabel Miranda
- Surgery and Physiology, Cardiovascular Research Center, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Adelino Barbosa
- Department of Surgery, Centro Hospitalar de S. João, Porto, Portugal
| | - Karine Clément
- Sorbonne Université, INSERM, NutriOmics Research Unit, Pitié-Salpêtrière Hopital, Paris, France
| | - Joel Doré
- Université Paris-Saclay, INRA, MetaGenoPolis, AgroParisTech, MICALIS, 78350 Jouy-en-Josas, France
| | - Paula Freitas
- Department of Endocrinology and Nutrition, Centro Hospitalar de S. João, Alameda Professor Hernani Monteiro, 4200-319 Porto, Portugal
- I3S – Instituto de Investigação e Inovação em Saúde, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Edi Prifti
- Integromics, Institute of Cardiometabolism and Nutrition, ICAN, Paris, France
- Unité de Modélisation Mathématique et Informatique des Systèmes Complexes, IRD, Sorbonne Université, UMMISCO, Paris, France
| |
Collapse
|
157
|
Malnick SDH, Fisher D, Somin M, Neuman MG. Treating the Metabolic Syndrome by Fecal Transplantation-Current Status. BIOLOGY 2021; 10:447. [PMID: 34065241 PMCID: PMC8161223 DOI: 10.3390/biology10050447] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/22/2021] [Accepted: 05/14/2021] [Indexed: 12/12/2022]
Abstract
The intestinal microbiome (IM) is important for normal gastrointestinal (GI) and other organ systems' functioning. An alteration in the normal IM, dysbiosis, and changes in intestinal motility result in microorganisms' overgrowth and an alteration in intestinal permeability. The gut-brain axis is also of importance in the irritable bowel syndrome (IBS) and associated bowel overgrowth. Secondary to the epidemic of obesity, the metabolic syndrome has become a major health problem. Disturbances in the fecal microbiome are associated with the metabolic syndrome. Metabolic-associated fatty liver disease (MAFLD) is now the current terminology for non-alcoholic fatty liver disease. IM alteration by fecal transplantation is an approved treatment method for recurrent Clostridioides difficile infection. Initially performed by either duodenal infusion or colonoscopy, it is now easily performed by the administration of capsules containing stools. We discuss the intestinal microbiome-its composition, as well as the qualitative changes of microbiome composition leading to inflammation. In addition, we discuss the evidence of the effect of fecal transplantation on the metabolic syndrome and MAFLD, as well as its clinical indications.
Collapse
Affiliation(s)
- Stephen D. H. Malnick
- Department of Internal Medicine Cj Kaplan Medical Center, The Hebrew University, Rehovot 76100, Israel; (S.D.H.M.); (D.F.); (M.S.)
| | - David Fisher
- Department of Internal Medicine Cj Kaplan Medical Center, The Hebrew University, Rehovot 76100, Israel; (S.D.H.M.); (D.F.); (M.S.)
| | - Marina Somin
- Department of Internal Medicine Cj Kaplan Medical Center, The Hebrew University, Rehovot 76100, Israel; (S.D.H.M.); (D.F.); (M.S.)
| | - Manuela G. Neuman
- In Vitro Drug Safety and Biotechnology, Banting Institute, University of Toronto, Toronto, ON M5G 0A3, Canada
| |
Collapse
|
158
|
Ahmed S, Spence JD. Sex differences in the intestinal microbiome: interactions with risk factors for atherosclerosis and cardiovascular disease. Biol Sex Differ 2021; 12:35. [PMID: 34001264 PMCID: PMC8130173 DOI: 10.1186/s13293-021-00378-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 04/29/2021] [Indexed: 02/06/2023] Open
Abstract
Background There are clearly sex differences in cardiovascular disease. On average, women experience cardiovascular events at an older age, and at any age, women, on average, have less atherosclerotic plaque than men. The role of the human intestinal microbiome in health and disease has garnered significant interest in recent years, and there have been indications of sex differences in the intestinal microbiome. The purpose of this narrative review was to evaluate evidence of sex differences in the interaction between the intestinal microbiome and risk factors for cardiovascular disease. Several studies have demonstrated changes in microbiota composition and metabolic profile as a function of diet, sex hormones, and host metabolism, among other factors. This dysbiosis has consequently been associated with several disease states, including atherosclerosis and cardiovascular disease. In this respect, there is a growing appreciation for the microbiota and its secreted metabolites, including trimethylamine N-oxide (TMAO), derived from intestinal bacterial metabolic pathways involving dietary choline and l-carnitine, as novel risk factors for atherosclerosis and cardiovascular outcomes. Although traditional risk factors for vascular disease have been studied broadly over the years, there exists little research to evaluate interactions of cardiovascular risk factors with a potentially sexually dimorphic intestinal microbiome. This review evaluates the role of sex differences in the composition of the intestinal microbiome, including effects of sex hormones on the microbiome, and the effects of these sex differences on cardiovascular risk factors. Diabetes and obesity exhibit sexual dimorphism, while the data concerning hypertension and dyslipidemia remain inconclusive based on the available literature. In addition, an increased proportion of gram-negative species capable of driving metabolic endotoxemia and a low-grade inflammatory response, as well as decreased numbers of butyrate-producing species, have been observed in relation to traditional vascular risk factors. In this context, circulating SCFAs and TMAO are recognized as key metabolites of the intestinal microbiome that can be readily measured in the blood for the evaluation of metabolic profile. Conclusion Novel strategies focused on resolving intestinal dysbiosis as a means to slow progression of atherosclerosis and reduce the risk of cardiovascular disease should be evaluated through a lens of sex differences.
Collapse
Affiliation(s)
- Shamon Ahmed
- University of British Columbia Faculty of Medicine, Vancouver, British Columbia, Canada
| | - J David Spence
- Stroke Prevention and Atherosclerosis Research Centre, Robarts Research Institute, Western University, 1400 Western Road, London, Ontario, N6G 2V4, Canada.
| |
Collapse
|
159
|
Wilson BC, Vatanen T, Jayasinghe TN, Leong KSW, Derraik JGB, Albert BB, Chiavaroli V, Svirskis DM, Beck KL, Conlon CA, Jiang Y, Schierding W, Holland DJ, Cutfield WS, O’Sullivan JM. Strain engraftment competition and functional augmentation in a multi-donor fecal microbiota transplantation trial for obesity. MICROBIOME 2021; 9:107. [PMID: 33985595 PMCID: PMC8120839 DOI: 10.1186/s40168-021-01060-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 03/24/2021] [Indexed: 05/15/2023]
Abstract
BACKGROUND Donor selection is an important factor influencing the engraftment and efficacy of fecal microbiota transplantation (FMT) for complex conditions associated with microbial dysbiosis. However, the degree, variation, and stability of strain engraftment have not yet been assessed in the context of multiple donors. METHODS We conducted a double-blinded randomized control trial of FMT in 87 adolescents with obesity. Participants were randomized to receive multi-donor FMT (capsules containing the fecal microbiota of four sex-matched lean donors) or placebo (saline capsules). Following a bowel cleanse, participants ingested a total of 28 capsules over two consecutive days. Capsules from individual donors and participant stool samples collected at baseline, 6, 12, and 26 weeks post-treatment were analyzed by shotgun metagenomic sequencing allowing us to track bacterial strain engraftment and its functional implications on recipients' gut microbiomes. RESULTS Multi-donor FMT sustainably altered the structure and the function of the gut microbiome. In what was effectively a microbiome competition experiment, we discovered that two donor microbiomes (one female, one male) dominated strain engraftment and were characterized by high microbial diversity and a high Prevotella to Bacteroides (P/B) ratio. Engrafted strains led to enterotype-level shifts in community composition and provided genes that altered the metabolic potential of the community. Despite our attempts to standardize FMT dose and origin, FMT recipients varied widely in their engraftment of donor strains. CONCLUSION Our study provides evidence for the existence of FMT super-donors whose microbiomes are highly effective at engrafting in the recipient gut. Dominant engrafting male and female donor microbiomes harbored diverse microbial species and genes and were characterized by a high P/B ratio. Yet, the high variability of strain engraftment among FMT recipients suggests the host environment also plays a critical role in mediating FMT receptivity. TRIAL REGISTRATION The Gut Bugs trial was registered with the Australian New Zealand Clinical Trials Registry ( ACTRN12615001351505 ). TRIAL PROTOCOL The trial protocol is available at https://bmjopen.bmj.com/content/9/4/e026174 . Video Abstract.
Collapse
Affiliation(s)
- Brooke C. Wilson
- The Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Tommi Vatanen
- The Liggins Institute, University of Auckland, Auckland, New Zealand
- The Broad Institute of MIT and Harvard, Cambridge, MA USA
| | | | - Karen S. W. Leong
- The Liggins Institute, University of Auckland, Auckland, New Zealand
- A Better Start–National Science Challenge, Auckland, New Zealand
| | - José G. B. Derraik
- The Liggins Institute, University of Auckland, Auckland, New Zealand
- A Better Start–National Science Challenge, Auckland, New Zealand
| | - Benjamin B. Albert
- The Liggins Institute, University of Auckland, Auckland, New Zealand
- A Better Start–National Science Challenge, Auckland, New Zealand
| | | | - Darren M. Svirskis
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kathryn L. Beck
- School of Sport, Exercise and Nutrition, College of Health, Massey University, Auckland, New Zealand
| | - Cathryn A. Conlon
- School of Sport, Exercise and Nutrition, College of Health, Massey University, Auckland, New Zealand
| | - Yannan Jiang
- Department of Statistics, University of Auckland, Auckland, New Zealand
| | | | - David J. Holland
- Department of Infectious Diseases, Counties Manukau District Health Board, Auckland, New Zealand
| | - Wayne S. Cutfield
- The Liggins Institute, University of Auckland, Auckland, New Zealand
- A Better Start–National Science Challenge, Auckland, New Zealand
| | - Justin M. O’Sullivan
- The Liggins Institute, University of Auckland, Auckland, New Zealand
- A Better Start–National Science Challenge, Auckland, New Zealand
| |
Collapse
|
160
|
Host-microbial interactions in the metabolism of different dietary fats. Cell Metab 2021; 33:857-872. [PMID: 33951472 DOI: 10.1016/j.cmet.2021.04.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 03/29/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023]
Abstract
Although generally presumed to be isocaloric, dietary fats can differ in their energetic contributions and metabolic effects. Here, we show how an explicit consideration of the gut microbiome and its interactions with human physiology can enrich our understanding of dietary fat metabolism. We outline how variable human metabolic responses to different dietary fats, such as altered ileal digestibility or bile acid production, have downstream effects on the gut microbiome that differentially promote energy gain and inflammation. By incorporating host-microbial interactions into energetic models of human nutrition, we can achieve greater insight into the underlying mechanisms of diet-driven metabolic disease.
Collapse
|
161
|
Hernández-Ceballos W, Cordova-Gallardo J, Mendez-Sanchez N. Gut Microbiota in Metabolic-associated Fatty Liver Disease and in Other Chronic Metabolic Diseases. J Clin Transl Hepatol 2021; 9:227-238. [PMID: 34007805 PMCID: PMC8111113 DOI: 10.14218/jcth.2020.00131] [Citation(s) in RCA: 11] [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: 11/26/2020] [Revised: 02/05/2021] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
The gut microbiome plays a key role in the health-disease balance in the human body. Although its composition is unique for each person and tends to remain stable throughout lifetime, it has been shown that certain bacterial patterns may be determining factors in the onset of certain chronic metabolic diseases, such as type 2 diabetes mellitus (T2DM), obesity, metabolic-associated fatty liver disease (MAFLD), and metabolic syndrome. The gut-liver axis embodies the close relationship between the gut and the liver; disturbance of the normal gut microbiota, also known as dysbiosis, may lead to a cascade of mechanisms that modify the epithelial properties and facilitate bacterial translocation. Regulation of gut microbiota is fundamental to maintaining gut integrity, as well as the bile acids composition. In the present review, we summarize the current knowledge regarding the microbiota, bile acids composition and their association with MAFLD, obesity, T2DM and metabolic syndrome.
Collapse
Affiliation(s)
- Winston Hernández-Ceballos
- Plan of Combined Studies in Medicine (PECEM-MD/PhD), Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Jacqueline Cordova-Gallardo
- Department of Hepatology, Service of Surgery and Obesity Clinic, General Hospital “Dr. Manuel Gea González”, Mexico City, Mexico
- Faculty of Medicine. National Autonomous University of Mexico, Mexico City, Mexico
| | - Nahum Mendez-Sanchez
- Faculty of Medicine. National Autonomous University of Mexico, Mexico City, Mexico
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
| |
Collapse
|
162
|
Cornejo-Pareja I, Molina-Vega M, Gómez-Pérez AM, Damas-Fuentes M, Tinahones FJ. Factors Related to Weight Loss Maintenance in the Medium-Long Term after Bariatric Surgery: A Review. J Clin Med 2021; 10:jcm10081739. [PMID: 33923789 PMCID: PMC8073104 DOI: 10.3390/jcm10081739] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/27/2021] [Accepted: 04/09/2021] [Indexed: 12/15/2022] Open
Abstract
Despite bariatric surgery being the most effective treatment for obesity, some individuals do not respond adequately, especially in the long term. Identifying the predictors of correct weight maintenance in the medium (from 1 to 3 years after surgery) and long term (from 3 years and above) is of vital importance to reduce failure after bariatric surgery; therefore, we summarize the evidence about certain factors, among which we highlight surgical technique, psychological factors, physical activity, adherence to diet, gastrointestinal hormones or neurological factors related to appetite control. We conducted a search in PubMed focused on the last five years (2015–2021). Main findings are as follows: despite Roux-en-Y gastric bypass being more effective in the long term, sleeve gastrectomy shows a more beneficial effectiveness–complications balance; pre-surgical psychological and behavioral evaluation along with post-surgical treatment improve long-term surgical outcomes; physical activity programs after bariatric surgery, in addition to continuous and comprehensive care interventions regarding diet habits, improve weight loss maintenance, but it is necessary to improve adherence; the impact of bariatric surgery on the gut–brain axis seems to influence weight maintenance. In conclusion, although interesting findings exist, the evidence is contradictory in some places, and long-term clinical trials are necessary to draw more robust conclusions.
Collapse
Affiliation(s)
- Isabel Cornejo-Pareja
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, 29010 Málaga, Spain; (I.C.-P.); (M.D.-F.); (F.J.T.)
- Instituto de Investigación Biomédica de Málaga (IBIMA), Virgen de la Victoria University Hospital, 29010 Málaga, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María Molina-Vega
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, 29010 Málaga, Spain; (I.C.-P.); (M.D.-F.); (F.J.T.)
- Instituto de Investigación Biomédica de Málaga (IBIMA), Virgen de la Victoria University Hospital, 29010 Málaga, Spain
- Correspondence: (M.M.-V.); (A.M.G.-P.); Tel.: +34-95-1034-044 (M.M.-V. & A.M.G.-P.)
| | - Ana María Gómez-Pérez
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, 29010 Málaga, Spain; (I.C.-P.); (M.D.-F.); (F.J.T.)
- Instituto de Investigación Biomédica de Málaga (IBIMA), Virgen de la Victoria University Hospital, 29010 Málaga, Spain
- Correspondence: (M.M.-V.); (A.M.G.-P.); Tel.: +34-95-1034-044 (M.M.-V. & A.M.G.-P.)
| | - Miguel Damas-Fuentes
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, 29010 Málaga, Spain; (I.C.-P.); (M.D.-F.); (F.J.T.)
- Instituto de Investigación Biomédica de Málaga (IBIMA), Virgen de la Victoria University Hospital, 29010 Málaga, Spain
| | - Francisco J. Tinahones
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, 29010 Málaga, Spain; (I.C.-P.); (M.D.-F.); (F.J.T.)
- Instituto de Investigación Biomédica de Málaga (IBIMA), Virgen de la Victoria University Hospital, 29010 Málaga, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
163
|
Alterations of Serum Uric Acid Level and Gut Microbiota After Roux-en-Y Gastric Bypass and Sleeve Gastrectomy in a Hyperuricemic Rat Model. Obes Surg 2021; 30:1799-1807. [PMID: 32124218 PMCID: PMC7228899 DOI: 10.1007/s11695-019-04328-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Background The objective of this study was to observe alterations of serum uric acid (SUA) level and gut microbiota after Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG) surgery in a hyperuricemic rat model. Method We performed Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG) surgery in a hyperuricemic rat model. Serum uric acid (UA), xanthine oxidase (XO) activity, IL-6, TNF-α and lipopolysaccharide (LPS) level changes, and 16S rDNA of gut microbiota were analyzed. Results After the surgery, the RYGB and SG procedures significantly reduced body weight, serum UA, IL-6, TNF-α and LPS levels, and XO activity. In addition, the RYGB and SG procedures altered the diversity and taxonomic composition of the gut microbiota. Compared with Sham group, RYGB and SG procedures were enriched in the abundance of phylum Verrucomicrobia and species Akkermansia muciniphila, while the species Escherichia coli was reduced. Discussion We here concluded that bariatric surgery-induced weight loss and resolution of inflammatory remarkers as well as changes of gut microbiota may be responsible for the reduced XO activity and SUA level. To have a better understanding of the underlying mechanism of UA metabolism following bariatric surgery, further research is needed.
Collapse
|
164
|
Effect of Obesity on the Expression of Nutrient Receptors and Satiety Hormones in the Human Colon. Nutrients 2021; 13:nu13041271. [PMID: 33924402 PMCID: PMC8070384 DOI: 10.3390/nu13041271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/07/2021] [Accepted: 04/09/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Receptors located on enteroendocrine cells (EECs) of the colon can detect nutrients in the lumen. These receptors regulate appetite through a variety of mechanisms, including hormonal and neuronal signals. We assessed the effect of obesity on the expression of these G-protein coupled receptors (GPCRs) and hormones at both mRNA and protein level. Methods: qPCR and immunohistochemistry were used to examine colonic tissue from cohorts of patients from the Netherlands (proximal and sigmoid tissue) and the United Kingdom (tissue from across the colon) and patients were grouped by body mass index (BMI) value (BMI < 25 and BMI ≥ 25). Results: The mRNA expression of the hormones/signaling molecules serotonin, glucagon, peptide YY (PYY), CCK and somatostatin were not significantly different between BMI groups. GPR40 mRNA expression was significantly increased in sigmoid colon samples in the BMI ≥ 25 group, but not proximal colon. GPR41, GPR109a, GPR43, GPR120, GPRC6A, and CaSR mRNA expression were unaltered between low and high BMI. At the protein level, serotonin and PYY containing cell numbers were similar in high and low BMI groups. Enterochromaffin cells (EC) showed high degree of co-expression with amino acid sensing receptor, CaSR while co-expression with PYY containing L-cells was limited, regardless of BMI. Conclusions: While expression of medium/long chain fatty acid receptor GPR40 was increased in the sigmoid colon of the high BMI group, expression of other nutrient sensing GPCRs, and expression profiles of EECs involved in peripheral mechanisms of appetite regulation were unchanged. Collectively, these data suggest that in human colonic tissue, EEC and nutrient-sensing receptor expression profiles are not affected despite changes to BMI.
Collapse
|
165
|
Wu WK, Chen YH, Lee PC, Yang PJ, Chang CC, Liu KL, Hsu CC, Huang CC, Chuang HL, Sheen LY, Liu CJ, Wu MS. Mining Gut Microbiota From Bariatric Surgery for MAFLD. Front Endocrinol (Lausanne) 2021; 12:612946. [PMID: 33897617 PMCID: PMC8063105 DOI: 10.3389/fendo.2021.612946] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 03/10/2021] [Indexed: 02/06/2023] Open
Abstract
The progression of metabolic dysfunction associated fatty liver disease (MAFLD) leads to steatohepatitis, liver fibrosis and hepatocellular carcinoma. Thus far, there have been no FDA-approved medications for MAFLD. Bariatric surgery (BS) has been found to improve insulin resistance, steatohepatitis and liver fibrosis but is not recommended for treating MAFLD due to its invasiveness. Recent studies suggest the improved glucose metabolism after BS is a result of, at least partly, alterations to the gut microbiota and its associated metabolites, including short chain fatty acids and bile acids. It makes sense the improved steatohepatitis and fibrosis after BS are also induced by the gut microbiota that involves in host metabolic modulation, for example, through altering bile acids composition. Given that the gut-liver axis is a path that may harbor unexplored mechanisms behind MAFLD, we review current literatures about disentangling the metabolic benefits of MAFLD after BS, with a focus on gut microbiota. Some useful research tools including the rodent BS model, the multiomics approach, and the human microbiota associated (HMA) mice are presented and discussed. We believe, by taking advantage of these modern translational tools, researchers will uncover microbiota related pathways to serve as potential therapeutic targets for treating MAFLD.
Collapse
Affiliation(s)
- Wei-Kai Wu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Hsun Chen
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Po-Chu Lee
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Po-Jen Yang
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Chin-Chen Chang
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
| | - Kao-Lang Liu
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
| | - Cheng-Chih Hsu
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Chi-Chang Huang
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan, Taiwan
| | - Hsiao-Li Chuang
- National Laboratory Animal Center, National Applied Research Laboratories Research Institute, Taipei, Taiwan
| | - Lee-Yan Sheen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Chun-Jen Liu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ming-Shiang Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
166
|
Gao Y, Zhang J, Xiao X, Ren Y, Yan X, Yue J, Wang T, Wu Z, Lv Y, Wu R. The Role of Gut Microbiota in Duodenal-Jejunal Bypass Surgery-Induced Improvement of Hepatic Steatosis in HFD-Fed Rats. Front Cell Infect Microbiol 2021; 11:640448. [PMID: 33869077 PMCID: PMC8050338 DOI: 10.3389/fcimb.2021.640448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/16/2021] [Indexed: 12/12/2022] Open
Abstract
Bariatric surgery including duodenal-jejunal bypass surgery (DJB) improves insulin sensitivity and reduces obesity-associated inflammation. However, the underlying mechanism for such an improvement is still incompletely understood. Our objective was to investigate the role of the gut microbiota in DJB-associated improvement of hepatic steatosis in high fat diet (HFD)-fed rats. To study this, hepatic steatosis was induced in male adult Sprague-Dawley rats by feeding them with a 60% HFD. At 8 weeks after HFD feeding, the rats were subjected to either DJB or sham operation. HFD was resumed 1 week after the surgery for 3 more weeks. In additional groups of animals, feces were collected from HFD-DJB rats at 2 weeks after DJB. These feces were then transplanted to HFD-fed rats without DJB at 8 weeks after HFD feeding. Hepatic steatosis and fecal microbiota were analyzed at 4 weeks after surgery or fecal transplantation. Our results showed that DJB alleviated hepatic steatosis in HFD-fed rats. Fecal microbiota analysis showed that HFD-fed and standard diet-fed rats clustered differently. DJB induced substantial compositional changes in the gut microbiota. The fecal microbiota of HFD-fed rats received fecal transplant from DJB rats overlapped with that of HFD-DJB rats. Treatment of rats with HFD-induced liver lesions by fecal transplant from DJB-operated HFD-fed rats also attenuated hepatic steatosis. Thus, alterations in the gut microbiota after DJB surgery are sufficient to attenuate hepatic steatosis in HFD-fed rats. Targeting the gut microbiota could be a promising approach for preventing or treating human NAFLD.
Collapse
Affiliation(s)
- Yi Gao
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Gastrointestinal Surgery Department, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jia Zhang
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiao Xiao
- School of Basic Medicine, Hubei University of Medicine, Shiyan, China
| | - Yifan Ren
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaopeng Yan
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jing Yue
- Gastrointestinal Surgery Department, Affiliated Hospital of Guilin Medical University, Guilin, China
- School of Basic Medicine, Hubei University of Medicine, Shiyan, China
| | - Tieyan Wang
- Department of Pathology, Shiyan Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Zheng Wu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yi Lv
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Rongqian Wu
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
167
|
Fukuda T, Bouchi R, Takeuchi T, Amo-Shiinoki K, Kudo A, Tanaka S, Tanabe M, Akashi T, Hirayama K, Odamaki T, Igarashi M, Kimura I, Tanabe K, Tanizawa Y, Yamada T, Ogawa Y. Importance of Intestinal Environment and Cellular Plasticity of Islets in the Development of Postpancreatectomy Diabetes. Diabetes Care 2021; 44:1002-1011. [PMID: 33627367 DOI: 10.2337/dc20-0864] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 01/18/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To elucidate the pathogenesis of postpancreatectomy diabetes mellitus (PPDM). RESEARCH DESIGN AND METHODS Forty-eight patients without diabetes undergoing either pancreatoduodenectomy (PD) (n = 20) or distal pancreatectomy (DP) (n = 28) were included. A 75-g oral glucose tolerance test was performed every 6 months. Microbiome composition and short-chain fatty acids (SCFAs) in feces were examined before and 6 months after surgery. The association of histological characteristics of the resected pancreas with PPDM was examined. RESULTS During follow-up (median 3.19 years), 2 of 20 PD patients and 16 of 28 DP patients developed PPDM. Proteobacteria relative abundance, plasma glucagon-like peptide 1 (GLP-1), and fecal butyrate levels increased only after PD. Postsurgical butyrate levels were correlated with postsurgical GLP-1 levels. With no significant difference in the volume of the resected pancreas between the surgical procedures, both β-cell and α-cell areas in the resected pancreas were significantly higher in DP patients than in PD patients. In DP patients, the progressors to diabetes showed preexisting insulin resistance compared with nonprogressors, and both increased α- and β-cell areas were predictors of PPDM. Furthermore, in DP patients, α-cell and β-cell areas were associated with ALDH1A3 expression in islets. CONCLUSIONS We postulate that a greater removal of β-cells contributes to the development of PPDM after DP. Islet expansion along with preexisting insulin resistance is associated with high cellular plasticity, which may predict the development of PPDM after DP. In contrast, PD is associated with alterations of gut microbiome and increases in SCFA production and GLP-1 secretion, possibly protecting against PPDM development.
Collapse
Affiliation(s)
- Tatsuya Fukuda
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryotaro Bouchi
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine, Tokyo, Japan
- Diabetes and Metabolism Information Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Takato Takeuchi
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kikuko Amo-Shiinoki
- Division of Endocrinology, Metabolism, Hematological Sciences and Therapeutics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Atsushi Kudo
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinji Tanaka
- Department of Molecular Oncology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Minoru Tanabe
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takumi Akashi
- Department of Human Pathology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazuhiro Hirayama
- Laboratory of Veterinary Public Health, Department of Veterinary Medical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Toshitaka Odamaki
- R&D Division, Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | - Miki Igarashi
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Ikuo Kimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Katsuya Tanabe
- Division of Endocrinology, Metabolism, Hematological Sciences and Therapeutics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Yukio Tanizawa
- Division of Endocrinology, Metabolism, Hematological Sciences and Therapeutics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Tetsuya Yamada
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshihiro Ogawa
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Molecular and Cellular Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- AMED-CREST, Tokyo, Japan
| |
Collapse
|
168
|
Zhang Y, Yan T, Xu C, Yang H, Zhang T, Liu Y. Probiotics Can Further Reduce Waist Circumference in Adults with Morbid Obesity after Bariatric Surgery: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:5542626. [PMID: 33859706 PMCID: PMC8032506 DOI: 10.1155/2021/5542626] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 01/09/2023]
Abstract
Whether probiotics could be used as an adjunct to bariatric surgery is controversial. This meta-analysis aimed to evaluate the effects of probiotics on body weight, body mass index (BMI), percentage of the excess weight loss (%EWL), waist circumference (WC), and C-reactive protein (CRP) in adults with obesity after bariatric surgery (BS). PUBMED, EMBASE, and the Cochrane Central Registry of Controlled Trials were searched from the earliest record to March 2020. All randomized controlled trials (RCTs) on the effects of probiotics in adults with obesity after bariatric surgery were analyzed according to the eligibility criteria. Four RCTs, including 172 participants, were analyzed. There was a statistically significant difference in probiotics in the reduction of waist circumference at 12 months after bariatric surgery. However, probiotics were not effective in weight, BMI, %EWL, WC, and CRP both within 3 months and at 12 months postoperation. Probiotics aid adults with morbid obesity in achieving further waist circumference improvement after BS, with no significant effect on weight, BMI, %EWL, and CRP. More quality clinical studies are needed to confirm the efficacy and safety of probiotics, and address a number of practical issues before the routine clinical use of probiotics in adults with obesity undergoing BS.
Collapse
Affiliation(s)
- Yu Zhang
- The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Tong Yan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, China
| | - Chenxin Xu
- The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Huawu Yang
- The Center of Gastrointestinal and Minimally Invasive Surgery, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, China
| | - Tongtong Zhang
- Medical Research Center, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, China
| | - Yanjun Liu
- The Center of Gastrointestinal and Minimally Invasive Surgery, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, China
| |
Collapse
|
169
|
Abstract
Bariatric and metabolic surgery has evolved from simple experimental procedures for a chronic problem associated with significant morbidity into a sophisticated multidisciplinary treatment modality rooted in biology and physiology. Although the complete mechanistic narrative of bariatric surgery cannot yet be written, significant advance in knowledge has been made in the past 2 decades. This article provides a brief overview of the most studied hypotheses and their supporting evidence. Ongoing research, especially in frontier areas, such as the microbiome, will continue to refine, and perhaps even revise, current mechanistic understanding.
Collapse
|
170
|
Berkovskaya MA, Sych YP, Gurova OY, Fadeev VV. Significance of intestinal microbiota in implementing metabolic effects of bariatric surgery. RUSSIAN OPEN MEDICAL JOURNAL 2021. [DOI: 10.15275/rusomj.2021.0112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Bariatric surgery is among successful methods of obesity treatment, with effects going beyond weight reduction alone, but rather involving improved glucose tolerance, along with control or remission of the type 2 diabetes mellitus. The precise mechanisms causing metabolic effects of bariatric surgery are not fully elucidated, even though substantial evidence suggest that they include changes in the gut microbiota, bile acid homeostasis, and the close interactions of these factors.
Intestinal microflora is directly involved in the energy metabolism of a host human. Obesity and type 2 diabetes mellitus are associated with certain changes in the species composition and diversity of intestinal microflora, which are considered important factors in the development and progression of these ailments. Bariatric surgery leads to significant and persistent changes in the composition of the intestinal microbiota, often bringing it closer to the characteristics of the microbiota of an average person with a normal weight. An important role in implementing the metabolic effects of bariatric surgery, primarily in the improvement of glucose metabolism, belongs to postoperative changes in homeostasis of bile acids. These changes imply close metabolism. Moreover, changes in the bile acid metabolism after bariatric surgery affect the microbiota of the host. Further study of these relationships would clarify the mechanisms underlying metabolic surgery, make it more predictable, targeted and controlled, as well as open new therapeutic targets in the treatment of obesity and associated conditions.
Collapse
Affiliation(s)
| | - Yulia P. Sych
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - Olesya Yu. Gurova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - Valentin V. Fadeev
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| |
Collapse
|
171
|
Are Faecal Microbiota Analyses on Species-Level Suitable Clinical Biomarkers? A Pilot Study in Subjects with Morbid Obesity. Microorganisms 2021; 9:microorganisms9030664. [PMID: 33806783 PMCID: PMC8005088 DOI: 10.3390/microorganisms9030664] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND An abnormal faecal microbiota could be a causal factor for disease. This study evaluated a new method for faecal microbiota analysis in subjects with obesity and irritable bowel syndrome. METHODS The study had a matched case-control design. Forty-six subjects with morbid obesity (defined as BMI > 40 or >35 kg/m2 with obesity-related complications) of whom 23 had irritable bowel syndrome (IBS), were compared with 46 healthy volunteers. The faecal microbiota was analysed with Precision Microbiome Profiling (PMP™) which quantified 104 bacteria species. The primary aim was comparisons between the cases and controls. RESULTS Two men and 44 women with a mean age of 43.6 years were included in each of the groups; BMI in the groups was (mean and SD) 41.9 (3.5) and 22.5 (1.5) kg/m2, respectively. Seventeen bacterial species showed statistically significant differences between the groups after adjusting for multiple testing. In a post hoc analysis, the sensitivity and specificity were 78%. Alpha diversity was lower in the group with obesity. In subjects with morbid obesity, no clinically significant differences were seen between subjects with and without IBS or from before to six months after bariatric surgery. CONCLUSIONS The results encourage further evaluation of the new microbiome profiling tool.
Collapse
|
172
|
Zhang J, Feng M, Pan L, Wang F, Wu P, You Y, Hua M, Zhang T, Wang Z, Zong L, Han Y, Guan W. Effects of vitamin D deficiency on the improvement of metabolic disorders in obese mice after vertical sleeve gastrectomy. Sci Rep 2021; 11:6036. [PMID: 33727603 PMCID: PMC7971024 DOI: 10.1038/s41598-021-85531-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 02/28/2021] [Indexed: 01/31/2023] Open
Abstract
Vertical sleeve gastrectomy (VSG) is one of the most commonly performed clinical bariatric surgeries for the remission of obesity and diabetes. Its effects include weight loss, improved insulin resistance, and the improvement of hepatic steatosis. Epidemiologic studies demonstrated that vitamin D deficiency (VDD) is associated with many diseases, including obesity. To explore the role of vitamin D in metabolic disorders for patients with obesity after VSG. We established a murine model of diet-induced obesity + VDD, and we performed VSGs to investigate VDD's effects on the improvement of metabolic disorders present in post-VSG obese mice. We observed that in HFD mice, the concentration of VitD3 is four fold of HFD + VDD one. In the post-VSG obese mice, VDD attenuated the improvements of hepatic steatosis, insulin resistance, intestinal inflammation and permeability, the maintenance of weight loss, the reduction of fat loss, and the restoration of intestinal flora that were weakened. Our results suggest that in post-VSG obese mice, maintaining a normal level of vitamin D plays an important role in maintaining the improvement of metabolic disorders.
Collapse
Affiliation(s)
- Jie Zhang
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Min Feng
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Lisha Pan
- Center for Growth, Metabolism and Aging, Analytical and Testing Center, Key Laboratory of Bio-Resource and Eco-Environment, College of Life Sciences, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu, 610065, China
| | - Feng Wang
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Pengfei Wu
- Center for Growth, Metabolism and Aging, Analytical and Testing Center, Key Laboratory of Bio-Resource and Eco-Environment, College of Life Sciences, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu, 610065, China
| | - Yang You
- Center for Growth, Metabolism and Aging, Analytical and Testing Center, Key Laboratory of Bio-Resource and Eco-Environment, College of Life Sciences, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu, 610065, China
| | - Meiyun Hua
- Center for Growth, Metabolism and Aging, Analytical and Testing Center, Key Laboratory of Bio-Resource and Eco-Environment, College of Life Sciences, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu, 610065, China
| | - Tianci Zhang
- Center for Growth, Metabolism and Aging, Analytical and Testing Center, Key Laboratory of Bio-Resource and Eco-Environment, College of Life Sciences, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu, 610065, China
| | - Zheng Wang
- Department of Pathology, The First Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Liang Zong
- Department of Gastrointestinal Surgery, Changzhi People's Hospital, The Affiliated Hospital of Changzhi Medical College, Changzhi, Shanxi, China.
| | - Yuanping Han
- Center for Growth, Metabolism and Aging, Analytical and Testing Center, Key Laboratory of Bio-Resource and Eco-Environment, College of Life Sciences, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu, 610065, China.
| | - Wenxian Guan
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China.
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, China.
| |
Collapse
|
173
|
Chen J, Haase N, Haange SB, Sucher R, Münzker J, Jäger E, Schischke K, Seyfried F, von Bergen M, Hankir MK, Krügel U, Fenske WK. Roux-en-Y gastric bypass contributes to weight loss-independent improvement in hypothalamic inflammation and leptin sensitivity through gut-microglia-neuron-crosstalk. Mol Metab 2021; 48:101214. [PMID: 33741533 PMCID: PMC8095174 DOI: 10.1016/j.molmet.2021.101214] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/20/2021] [Accepted: 03/12/2021] [Indexed: 12/13/2022] Open
Abstract
Objective Hypothalamic inflammation and endoplasmic reticulum (ER) stress are extensively linked to leptin resistance and overnutrition-related diseases. Surgical intervention remains the most efficient long-term weight-loss strategy for morbid obesity, but mechanisms underlying sustained feeding suppression remain largely elusive. This study investigated whether Roux-en-Y gastric bypass (RYGB) interacts with obesity-associated hypothalamic inflammation to restore central leptin signaling as a mechanistic account for post-operative appetite suppression. Methods RYGB or sham surgery was performed in high-fat diet-induced obese Wistar rats. Sham-operated rats were fed ad libitum or by weight matching to RYGB via calorie restriction (CR) before hypothalamic leptin signaling, microglia reactivity, and the inflammatory pathways were examined to be under the control of gut microbiota-derived circulating signaling. Results RYGB, other than CR-induced adiposity reduction, ameliorates hypothalamic gliosis, inflammatory signaling, and ER stress, which are linked to enhanced hypothalamic leptin signaling and responsiveness. Mechanistically, we demonstrate that RYGB interferes with hypothalamic ER stress and toll-like receptor 4 (TLR4) signaling to restore the anorexigenic action of leptin, which most likely results from modulation of a circulating factor derived from the altered gut microbial environment upon RYGB surgery. Conclusions Our data demonstrate that RYGB interferes with hypothalamic TLR4 signaling to restore the anorexigenic action of leptin, which most likely results from modulation of a circulating factor derived from the post-surgical altered gut microbial environment. RYGB surgery-related weight loss independently restores hypothalamic leptin signaling and action in diet-induced obesity. RGYB modulates hypothalamic TLR4-mediated pro-inflammatory signaling and ER stress to restore leptin's anorexigenic action. Humoral factors contribute to modulated microglia-POMC neuron interaction, which appears specific to the RYGB procedure. Altering the gut microbiota environment by antibiotics deteriorates leptin's feeding suppressive action after RYGB.
Collapse
Affiliation(s)
- Jiesi Chen
- Medical Department III, Endocrinology, Nephrology, and Rheumatology, University Hospital of Leipzig, Leipzig, Germany
| | - Nadine Haase
- Medical Department III, Endocrinology, Nephrology, and Rheumatology, University Hospital of Leipzig, Leipzig, Germany
| | - Sven-Bastiaan Haange
- Department of Molecular Systems Biology, Helmholtz Center for Environmental Research-UFZ, Permoserstraße 15, 04318 Leipzig, Germany
| | - Robert Sucher
- Division of Bariatric Surgery, Clinic of Visceral, Transplant, Thoracic, and Vascular Surgery, University Hospital, Liebigstraße 20, D-4015, Leipzig, Germany
| | - Julia Münzker
- Medical Department III, Endocrinology, Nephrology, and Rheumatology, University Hospital of Leipzig, Leipzig, Germany
| | - Elisabeth Jäger
- Medical Department III, Endocrinology, Nephrology, and Rheumatology, University Hospital of Leipzig, Leipzig, Germany
| | - Kristin Schischke
- Medical Department III, Endocrinology, Nephrology, and Rheumatology, University Hospital of Leipzig, Leipzig, Germany
| | - Florian Seyfried
- Department of General, Visceral, Transplant, Vascular, and Pediatric Surgery, University Hospital, Würzburg, Germany
| | - Martin von Bergen
- Department of Molecular Systems Biology, Helmholtz Center for Environmental Research-UFZ, Permoserstraße 15, 04318 Leipzig, Germany; Institute of Biochemistry, Faculty of Life Sciences, University of Leipzig, Talstraße 33, 04103 Leipzig, Germany
| | - Mohammed K Hankir
- Department of Experimental Surgery, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Ute Krügel
- Rudolf Boehm Institute of Pharmacology and Toxicology, University of Leipzig, Härtelstraße 16-18, 04107 Leipzig, Germany
| | - Wiebke K Fenske
- Medical Department III, Endocrinology, Nephrology, and Rheumatology, University Hospital of Leipzig, Leipzig, Germany; Division of Endocrinology, Diabetes, and Metabolism, Medical Department I, University Hospital of Bonn, Bonn, Germany.
| |
Collapse
|
174
|
Elliott JA, Reynolds JV. Visceral Obesity, Metabolic Syndrome, and Esophageal Adenocarcinoma. Front Oncol 2021; 11:627270. [PMID: 33777773 PMCID: PMC7994523 DOI: 10.3389/fonc.2021.627270] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 02/19/2021] [Indexed: 12/16/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) represents an exemplar of obesity-associated carcinogenesis, with a progressive increase in EAC risk with increased body mass index. In this context, there is increased focus on visceral adipose tissue and associated metabolic dysfunction, including hypertension, diabetes mellitus and hyperlipidemia, or combinations of these in the metabolic syndrome. Visceral obesity (VO) may promote EAC via both directly impacting on gastro-esophageal reflux disease and Barrett's esophagus, as well as via reflux-independent effects, involving adipokines, growth factors, insulin resistance, and the microbiome. In this review these pathways are explored, including the impact of VO on the tumor microenvironment, and on cancer outcomes. The current evidence-based literature regarding the role of dietary, lifestyle, pharmacologic and surgical interventions to modulate the risk of EAC is explored.
Collapse
Affiliation(s)
- Jessie A Elliott
- Trinity St. James's Cancer Institute, Trinity College Dublin and St. James's Hospital, Dublin, Ireland
| | - John V Reynolds
- Trinity St. James's Cancer Institute, Trinity College Dublin and St. James's Hospital, Dublin, Ireland
| |
Collapse
|
175
|
Hankir MK, Seyfried F, Schellinger IN, Schlegel N, Arora T. Leaky Gut as a Potential Culprit for the Paradoxical Dysglycemic Response to Gastric Bypass-Associated Ileal Microbiota. Metabolites 2021; 11:153. [PMID: 33800456 PMCID: PMC7998592 DOI: 10.3390/metabo11030153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/24/2021] [Accepted: 03/05/2021] [Indexed: 12/04/2022] Open
Abstract
Altered host-intestinal microbiota interactions are increasingly implicated in the metabolic benefits of Roux-en-Y gastric bypass (RYGB) surgery. We previously found, however, that RYGB-associated ileal microbiota can paradoxically impair host glycemic control when transferred to germ-free mice. Here we present complementary evidence suggesting that this could be due to the heightened development of systemic endotoxemia. Consistently, application of ileal content from RYGB-treated compared with sham-operated rats onto Caco-2 cell monolayers compromised barrier function and decreased expression of the barrier-stabilizing proteins claudin-4 and desmoglein-2. Our findings raise the possibility that RYGB-associated ileal microbiota produce and release soluble metabolites which locally increase intestinal permeability to promote systemic endotoxemia-induced insulin resistance, with potential implications for the treatment of RYGB patients who eventually relapse onto type 2 diabetes.
Collapse
Affiliation(s)
- Mohammed K. Hankir
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, Oberdürrbacherstraße 6, 97080 Würzburg, Germany; (F.S.); (N.S.)
| | - Florian Seyfried
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, Oberdürrbacherstraße 6, 97080 Würzburg, Germany; (F.S.); (N.S.)
| | - Isabel N. Schellinger
- Department of Endocrinology and Nephrology, University Hospital Leipzig, Liebigstraße 20, 04103 Leipzig, Germany;
| | - Nicolas Schlegel
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, Oberdürrbacherstraße 6, 97080 Würzburg, Germany; (F.S.); (N.S.)
| | - Tulika Arora
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Blegdamsvej 3B, 2200 København, Denmark
| |
Collapse
|
176
|
Dauksiene A, Ruzauskas M, Gruzauskas R, Zavistanaviciute P, Starkute V, Lele V, Klupsaite D, Klementaviciute J, Bartkiene E. A Comparison Study of the Caecum Microbial Profiles, Productivity and Production Quality of Broiler Chickens Fed Supplements Based on Medium Chain Fatty and Organic Acids. Animals (Basel) 2021; 11:ani11030610. [PMID: 33652631 PMCID: PMC7996795 DOI: 10.3390/ani11030610] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary The ban of growth promoters in poultry farming in the European Union has resulted in the development of alternatives. Among these alternatives, medium chain fatty acids (MCFAs) or organic acids (OAs) are considered to be suitable for in-feed use. However, their effect on microbiota modulation and the meat quality of broiler chickens are still under-investigated. The aim of this study was to estimate the influence of MCFAs and OAs supplements on the caecum microbial profiles, productivity and production quality characteristics of broiler chickens. The 42-days experiment was conducted using 900-day-old broiler chickens, allocated into three groups, consisting of 300 birds per group. The results indicated that the addition of OAs results in a more appropriate environment in the caecum for beneficial microorganisms rather than diets supplemented with MCFAs. These positive changes led to a higher efficiency of poultry productivity (higher body weight and lower mortality); however, for most of the analysed broilers’, technological parameters were not considerably influenced by treatments. Abstract The aim of this study was to evaluate the influence of medium chain fatty acids (MCFAs) and organic acids (OAs) supplements on the caecum microbial profiles, productivity and production quality characteristics of broiler chickens (BCs). BC (900 chicks) were attributed to three groups: (i) control; (ii) MCFAs group (BCs fed with feed supplemented with MCFAs); (iii) OAs group (BCs fed with feed supplemented with OAs). Broilers were slaughtered at the end of the trial (42 days old), and the caecum microbial profiles, productivity and production quality characteristics were analysed. Supplementation with OAs resulted in a more appropriate environment in the caecum for beneficial microorganisms than with a diet supplemented with MCFAs. This was supported by data on the presence of higher amounts and an increased species variety of probiotic bacteria (Lactobacillus and Bifidobacterium) in the caecum of birds. The above-mentioned changes of the caecum microbiota led to significantly higher villus height (p = 0.003) of the OAs broiler group and significantly lower crypt depth (p = 0.037). Notwithstanding the significant increase of acetic, propionic, isobutyric, butyric, isovaleric, and valeric acids that were established in caecum samples from the MCFAs group, better parameters of broiler production performance (higher body weight and lower mortality) and carcass traits (higher both thigh and shin muscles with skin and bone weight; both shin muscles without skin and bone weight; abdominal fat yield) were found in the OAs-treated group. For chemical, physical and technological characteristics of breast meat samples, increased yellowness and water holding capacity by 14.7% and 2.3%, respectively, were found in MCFAs group samples. A more appropriate environment in the caecum for beneficial microorganisms could be obtained when BCs were fed with OAs supplement, comparing to MCFAs, and these positive changes were associated with higher efficiency of poultry production.
Collapse
Affiliation(s)
- Agila Dauksiene
- Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Tilzes str. 18, LT-47181 Kaunas, Lithuania; (P.Z.); (V.S.); (V.L.); (D.K.); (J.K.); (E.B.)
- Department of Anatomy and Physiology, Lithuanian University of Health Sciences, Tilzes str. 18, LT-47181 Kaunas, Lithuania;
- Correspondence: ; Tel.: +370-685-25327
| | - Modestas Ruzauskas
- Department of Anatomy and Physiology, Lithuanian University of Health Sciences, Tilzes str. 18, LT-47181 Kaunas, Lithuania;
- Microbiology and Virology Institute, Lithuanian University of Health Sciences, Tilzes str. 18, LT-47181 Kaunas, Lithuania
| | - Romas Gruzauskas
- Department of Food Sciences and Technology, Kaunas University of Technology, Radvilenu str. 19, LT-50254 Kaunas, Lithuania;
| | - Paulina Zavistanaviciute
- Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Tilzes str. 18, LT-47181 Kaunas, Lithuania; (P.Z.); (V.S.); (V.L.); (D.K.); (J.K.); (E.B.)
- Department of Food Safety and Quality, Lithuanian University of Health Sciences, Tilzes str. 18, LT-47181 Kaunas, Lithuania
| | - Vytaute Starkute
- Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Tilzes str. 18, LT-47181 Kaunas, Lithuania; (P.Z.); (V.S.); (V.L.); (D.K.); (J.K.); (E.B.)
- Department of Food Safety and Quality, Lithuanian University of Health Sciences, Tilzes str. 18, LT-47181 Kaunas, Lithuania
| | - Vita Lele
- Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Tilzes str. 18, LT-47181 Kaunas, Lithuania; (P.Z.); (V.S.); (V.L.); (D.K.); (J.K.); (E.B.)
- Department of Food Safety and Quality, Lithuanian University of Health Sciences, Tilzes str. 18, LT-47181 Kaunas, Lithuania
| | - Dovile Klupsaite
- Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Tilzes str. 18, LT-47181 Kaunas, Lithuania; (P.Z.); (V.S.); (V.L.); (D.K.); (J.K.); (E.B.)
| | - Jolita Klementaviciute
- Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Tilzes str. 18, LT-47181 Kaunas, Lithuania; (P.Z.); (V.S.); (V.L.); (D.K.); (J.K.); (E.B.)
| | - Elena Bartkiene
- Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Tilzes str. 18, LT-47181 Kaunas, Lithuania; (P.Z.); (V.S.); (V.L.); (D.K.); (J.K.); (E.B.)
- Department of Food Safety and Quality, Lithuanian University of Health Sciences, Tilzes str. 18, LT-47181 Kaunas, Lithuania
| |
Collapse
|
177
|
Wang S, Li XY, Shen L. Modulation effects of Dendrobium officinale on gut microbiota of type 2 diabetes model mice. FEMS Microbiol Lett 2021; 368:6145026. [PMID: 33606020 DOI: 10.1093/femsle/fnab020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 02/17/2021] [Indexed: 12/17/2022] Open
Abstract
In recent years, the relationship between type 2 diabetes (T2D) and gut microbiota has attracted much interest. Dendrobium officinale is a valuable traditional Chinese medicine (TCM) with anti-T2D potential, while its action mechanism remains to be further studied. This study was designed to investigate the modulation effects of D. officinale on gut microbiota of T2D model mice to provide clues to its pharmacology by high-throughput sequencing techniques. It was found that D. officinale supplement could significantly reduce the fasting blood glucose levels of T2D mice. Dendrobium officinale supplement could modulate the composition of gut microbiota and increase the relative abundances of key bacterial taxa associated with T2D development, including Akkermansia and Parabacteroides. Compared with placebo group mice, several Kyoto Encyclopedia of Gene and Genomes pathways associated with T2D altered in the D. officinale treated group. These findings indicated the modulation of D. officinale on gut microbiota of T2D mice, which provide potential pharmacological implications.
Collapse
Affiliation(s)
- Sai Wang
- Institute of Biomedical Research, Shandong University of Technology, Zibo, Shandong 255000, People's Republic of China.,Shandong Provincial Research Center for Bioinformatic Engineering and Technique, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255000, People's Republic of China
| | - Xin-Yu Li
- Institute of Biomedical Research, Shandong University of Technology, Zibo, Shandong 255000, People's Republic of China.,Shandong Provincial Research Center for Bioinformatic Engineering and Technique, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255000, People's Republic of China
| | - Liang Shen
- Institute of Biomedical Research, Shandong University of Technology, Zibo, Shandong 255000, People's Republic of China.,Shandong Provincial Research Center for Bioinformatic Engineering and Technique, School of Life Sciences, Shandong University of Technology, Zibo, Shandong 255000, People's Republic of China
| |
Collapse
|
178
|
Association of Gut Hormones and Microbiota with Vascular Dysfunction in Obesity. Nutrients 2021; 13:nu13020613. [PMID: 33668627 PMCID: PMC7918888 DOI: 10.3390/nu13020613] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/25/2021] [Accepted: 02/10/2021] [Indexed: 02/08/2023] Open
Abstract
In the past few decades, obesity has reached pandemic proportions. Obesity is among the main risk factors for cardiovascular diseases, since chronic fat accumulation leads to dysfunction in vascular endothelium and to a precocious arterial stiffness. So far, not all the mechanisms linking adipose tissue and vascular reactivity have been explained. Recently, novel findings reported interesting pathological link between endothelial dysfunction with gut hormones and gut microbiota and energy homeostasis. These findings suggest an active role of gut secretome in regulating the mediators of vascular function, such as nitric oxide (NO) and endothelin-1 (ET-1) that need to be further investigated. Moreover, a central role of brain has been suggested as a main player in the regulation of the different factors and hormones beyond these complex mechanisms. The aim of the present review is to discuss the state of the art in this field, by focusing on the processes leading to endothelial dysfunction mediated by obesity and metabolic diseases, such as insulin resistance. The role of perivascular adipose tissue (PVAT), gut hormones, gut microbiota dysbiosis, and the CNS function in controlling satiety have been considered. Further understanding the crosstalk between these complex mechanisms will allow us to better design novel strategies for the prevention of obesity and its complications.
Collapse
|
179
|
Ahmed B, Sultana R, Greene MW. Adipose tissue and insulin resistance in obese. Biomed Pharmacother 2021; 137:111315. [PMID: 33561645 DOI: 10.1016/j.biopha.2021.111315] [Citation(s) in RCA: 413] [Impact Index Per Article: 103.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 02/08/2023] Open
Abstract
Currently, obesity has become a global health issue and is referred to as an epidemic. Dysfunctional obese adipose tissue plays a pivotal role in the development of insulin resistance. However, the mechanism of how dysfunctional obese-adipose tissue develops insulin-resistant circumstances remains poorly understood. Therefore, this review attempts to highlight the potential mechanisms behind obesity-associated insulin resistance. Multiple risk factors are directly or indirectly associated with the increased risk of obesity; among them, environmental factors, genetics, aging, gut microbiota, and diets are prominent. Once an individual becomes obese, adipocytes increase in their size; therefore, adipose tissues become larger and dysfunctional, recruit macrophages, and then these polarize to pro-inflammatory states. Enlarged adipose tissues release excess free fatty acids (FFAs), reactive oxygen species (ROS), and pro-inflammatory cytokines. Excess systemic FFAs and dietary lipids enter inside the cells of non-adipose organs such as the liver, muscle, and pancreas, and are deposited as ectopic fat, generating lipotoxicity. Toxic lipids dysregulate cellular organelles, e.g., mitochondria, endoplasmic reticulum, and lysosomes. Dysregulated organelles release excess ROS and pro-inflammation, resulting in systemic inflammation. Long term low-grade systemic inflammation prevents insulin from its action in the insulin signaling pathway, disrupts glucose homeostasis, and results in systemic dysregulation. Overall, long-term obesity and overnutrition develop into insulin resistance and chronic low-grade systemic inflammation through lipotoxicity, creating the circumstances to develop clinical conditions. This review also shows that the liver is the most sensitive organ undergoing insulin impairment faster than other organs, and thus, hepatic insulin resistance is the primary event that leads to the subsequent development of peripheral tissue insulin resistance.
Collapse
Affiliation(s)
- Bulbul Ahmed
- Department of Nutrition, Auburn University, Auburn, AL, 36849, United States.
| | - Rifat Sultana
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, 57007, United States
| | - Michael W Greene
- Department of Nutrition, Auburn University, Auburn, AL, 36849, United States
| |
Collapse
|
180
|
Guimarães M, Pereira SS, Monteiro MP. From Entero-Endocrine Cell Biology to Surgical Interventional Therapies for Type 2 Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1307:273-297. [PMID: 32016913 DOI: 10.1007/5584_2020_480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The physiological roles of the enteroendocrine system in relation to energy and glucose homeostasis regulation have been extensively studied in the past few decades. Considerable advances were made that enabled to disclose the potential use of gastro-intestinal (GI) hormones to target obesity and type 2 diabetes (T2D). The recognition of the clinical relevance of these discoveries has led the pharmaceutical industry to design several hormone analogues to either to mitigate physiological defects or target pharmacologically T2D.Amongst several advances, a major breakthrough in the field was the unexpected observation that enteroendocrine system modulation to T2D target could be achieved by surgically induced anatomical rearrangement of the GI tract. These findings resulted from the widespread use of bariatric surgery procedures for obesity treatment, which despite initially devised to induce weight loss by limiting the systemic availably of nutrients, are now well recognized to influence GI hormone dynamics in a manner that is highly dependent on the type of anatomical rearrangement produced.This chapter will focus on enteroendocrine system related mechanisms leading to improved glycemic control in T2D after bariatric surgery interventions.
Collapse
Affiliation(s)
- Marta Guimarães
- Endocrine, Cardiovascular & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal.,Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.,Department of General Surgery, Centro Hospitalar de Entre o Douro e Vouga, Santa Maria da Feira, Portugal
| | - Sofia S Pereira
- Endocrine, Cardiovascular & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal.,Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Mariana P Monteiro
- Endocrine, Cardiovascular & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal. .,Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.
| |
Collapse
|
181
|
Ceglarek VM, Bertasso IM, Pietrobon CB, Scomazzon SP, Leite NC, Bonfleur ML, Araújo ACF, Balbo SL, Grassiolli S. Maternal Roux-en-Y gastric bypass surgery reduces lipid deposition and increases UCP1 expression in the brown adipose tissue of male offspring. Sci Rep 2021; 11:1158. [PMID: 33441773 PMCID: PMC7806700 DOI: 10.1038/s41598-020-80104-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 12/03/2020] [Indexed: 01/22/2023] Open
Abstract
Maternal obesity induced by cafeteria diet (CAF) predisposes offspring to obesity and metabolic diseases, events that could be avoided by maternal bariatric surgery (BS). Herein we evaluated whether maternal BS is able to modulate brown adipose tissue (BAT) morphology and function in adult male rats born from obese female rats submitted to Roux-en-Y gastric bypass (RYGB). For this, adult male rat offspring were obtained from female rats that consumed standard diet (CTL), or CAF diet, and were submitted to simulated operation or RYGB. Analysis of offspring showed that, at 120 days of life, the maternal CAF diet induced adiposity and decreased the expression of mitochondrial Complex I (CI) and Complex III (CIII) in the BAT, resulting in higher accumulation of lipids than in BAT from offspring of CTL dams. Moreover, maternal RYGB increased UCP1 expression and prevented excessive deposition of lipids in the BAT of adult male offspring rats. However, maternal RYGB failed to reverse the effects of maternal diet on CI and CIII expression. Thus, maternal CAF promotes higher lipid deposition in the BAT of offspring, contributing to elevated adiposity. Maternal RYGB prevented obesity in offspring, probably by increasing the expression of UCP1.
Collapse
Affiliation(s)
- Vanessa Marieli Ceglarek
- Laboratory of Endocrine and Metabolic Physiology, Biosciences and Health, Postgraduate, University of West Parana, Cascavel, PR, Brazil. .,Institute of Basic Health Sciences. Biological Sciences: Physiology, postgraduate. Department of Physiology, Room 337-7, Laboratory of Neurophysiology of Cognition and Development of the Brain, Federal University of Rio Grande do Sul, 500, Sarmento Leite - Farroupilha, Porto Alegre, RS, 90050-170, Brazil.
| | - Iala Milene Bertasso
- Laboratory of Endocrine and Metabolic Physiology, Biosciences and Health, Postgraduate, University of West Parana, Cascavel, PR, Brazil
| | - Carla Bruna Pietrobon
- Laboratory of Endocrine and Metabolic Physiology, Biosciences and Health, Postgraduate, University of West Parana, Cascavel, PR, Brazil
| | - Sofia Pizzato Scomazzon
- Medical Sciences: Endocrinology Post Graduate Program, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Nayara Carvalho Leite
- Obesity Comorbidities and Research Center, University of Campinas, Campinas, SP, Brazil
| | - Maria Lúcia Bonfleur
- Laboratory of Endocrine and Metabolic Physiology, Biosciences and Health, Postgraduate, University of West Parana, Cascavel, PR, Brazil
| | - Allan Cezar Faria Araújo
- Laboratory of Endocrine and Metabolic Physiology, Biosciences and Health, Postgraduate, University of West Parana, Cascavel, PR, Brazil
| | - Sandra Lucinei Balbo
- Laboratory of Endocrine and Metabolic Physiology, Biosciences and Health, Postgraduate, University of West Parana, Cascavel, PR, Brazil
| | - Sabrina Grassiolli
- Laboratory of Endocrine and Metabolic Physiology, Biosciences and Health, Postgraduate, University of West Parana, Cascavel, PR, Brazil
| |
Collapse
|
182
|
Haruta H, Kasama K, Seki Y, Lefor AK. Revisional Surgery: Sleeve to DJB. LAPAROSCOPIC SLEEVE GASTRECTOMY 2021:595-603. [DOI: 10.1007/978-3-030-57373-7_58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
183
|
Wu KC, Yu EW, Schafer AL. Skeletal health after bariatric surgery. MARCUS AND FELDMAN'S OSTEOPOROSIS 2021:1261-1280. [DOI: 10.1016/b978-0-12-813073-5.00051-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
184
|
Sugama J, Moritoh Y, Yashiro H, Tsuchimori K, Watanabe M. Enteropeptidase inhibition improves obesity by modulating gut microbiota composition and enterobacterial metabolites in diet-induced obese mice. Pharmacol Res 2021; 163:105337. [PMID: 33276106 DOI: 10.1016/j.phrs.2020.105337] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 11/05/2020] [Accepted: 11/25/2020] [Indexed: 12/19/2022]
Abstract
Enteropeptidase is a transmembrane serine protease localized in the lumen of the duodenum that acts as a key enzyme for protein digestion. SCO-792 is an orally available enteropeptidase inhibitor that has been reported to have therapeutic effects on obesity and diabetes in mice. However, the mechanism underlying the therapeutic effect of SCO-792 has not yet been fully elucidated. In this study, we evaluated the role of gut microbiota on SCO-792-induced body weight (BW) reduction in high-fat diet-induced obese (DIO) mice. Chronic administration of SCO-792 substantially decreased BW and food intake in DIO mice. While the pair-fed study uncovered food intake-independent mechanisms of BW reduction by SCO-792. Interestingly, antibiotics-induced microbiota elimination in the gut canceled SCO-792-induced BW reduction by nearly half without affecting the anorectic effect, indicating the involvement of gut microbiota in the anti-obesity mechanism that is independent of food intake reduction. Microbiome analysis revealed that SCO-792 altered the gut microbiota composition in DIO mice. Notably, it was found that the abundance of Firmicutes decreased while that of Verrucomicrobia increased at the phylum level. Increased abundance of Akkermansia muciniphila, a bacterium known to be useful for host metabolism, was observed in SCO-792-treated mice. Fecal metabolome analysis revealed increased amino acid levels, indicating gut enteropeptidase inhibition. In addition, SCO-792 was found to increase the level of short-chain fatty acids, including propionate, and bile acids in the feces, which all help maintain gut health and improve metabolism. Furthermore, it was found that SCO-792 induced the elevation of colonic immunoglobulin A (IgA) concentration, which may maintain the microbiota condition, in DIO mice. In conclusion, this study demonstrates the contribution of microbiota to SCO-792-induced BW reduction. Enteropeptidase-mediated regulation of microbiota, enterobacterial metabolites, and IgA in the gut may coordinately drive the therapeutic effects of SCO-792 in obesity.
Collapse
Affiliation(s)
- Jun Sugama
- Research and Development Division, SCOHIA PHARMA Inc., Kanagawa, Japan.
| | - Yusuke Moritoh
- Research and Development Division, SCOHIA PHARMA Inc., Kanagawa, Japan.
| | - Hiroaki Yashiro
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | - Kazue Tsuchimori
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | - Masanori Watanabe
- Research and Development Division, SCOHIA PHARMA Inc., Kanagawa, Japan.
| |
Collapse
|
185
|
Gheorghe CE, Ritz NL, Martin JA, Wardill HR, Cryan JF, Clarke G. Investigating causality with fecal microbiota transplantation in rodents: applications, recommendations and pitfalls. Gut Microbes 2021; 13:1941711. [PMID: 34328058 PMCID: PMC8331043 DOI: 10.1080/19490976.2021.1941711] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 06/02/2021] [Accepted: 06/04/2021] [Indexed: 02/04/2023] Open
Abstract
In recent years, studies investigating the role of the gut microbiota in health and diseases have increased enormously - making it essential to deepen and question the research methodology employed. Fecal microbiota transplantation (FMT) in rodent studies (either from human or animal donors) allows us to better understand the causal role of the intestinal microbiota across multiple fields. However, this technique lacks standardization and requires careful experimental design in order to obtain optimal results. By comparing several studies in which rodents are the final recipients of FMT, we summarize the common practices employed. In this review, we document the limitations of this method and highlight different parameters to be considered while designing FMT Studies. Standardizing this method is challenging, as it differs according to the research topic, but avoiding common pitfalls is feasible. Several methodological questions remain unanswered to this day and we offer a discussion on issues to be explored in future studies.
Collapse
Affiliation(s)
- Cassandra E. Gheorghe
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Nathaniel L. Ritz
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jason A. Martin
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Hannah R. Wardill
- Precision Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
- Adelaide Medical School, the University of Adelaide, Adelaide, Australia
| | - John F. Cryan
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- INFANT Research Centre, University College Cork, Cork, Ireland
| |
Collapse
|
186
|
Hankir MK, Langseder T, Bankoglu EE, Ghoreishi Y, Dischinger U, Kurlbaum M, Kroiss M, Otto C, le Roux CW, Arora T, Seyfried F, Schlegel N. Simulating the Post-gastric Bypass Intestinal Microenvironment Uncovers a Barrier-Stabilizing Role for FXR. iScience 2020; 23:101777. [PMID: 33294786 PMCID: PMC7689555 DOI: 10.1016/j.isci.2020.101777] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/12/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023] Open
Abstract
Regional changes to the intestinal microenvironment brought about by Roux-en-Y gastric bypass (RYGB) surgery may contribute to some of its potent systemic metabolic benefits through favorably regulating various local cellular processes. Here, we show that the intestinal contents of RYGB-operated compared with sham-operated rats region-dependently confer superior glycemic control to recipient germ-free mice in association with suppression of endotoxemia. Correspondingly, they had direct barrier-stabilizing effects on an intestinal epithelial cell line which, bile-exposed intestinal contents, were partly farnesoid X receptor (FXR)-dependent. Further, circulating fibroblast growth factor 19 levels, a readout of intestinal FXR activation, negatively correlated with endotoxemia severity in longitudinal cohort of RYGB patients. These findings suggest that various host- and/or microbiota-derived luminal factors region-specifically and synergistically stabilize the intestinal epithelial barrier following RYGB through FXR signaling, which could potentially be leveraged to better treat endotoxemia-induced insulin resistance in obesity in a non-invasive and more targeted manner.
Collapse
Affiliation(s)
- Mohammed K. Hankir
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Center of Operative Medicine, Oberduerrbacherstrasse 6, Wuerzburg, Bavaria 97080, Germany
| | - Theresa Langseder
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Center of Operative Medicine, Oberduerrbacherstrasse 6, Wuerzburg, Bavaria 97080, Germany
| | - Ezgi Eyluel Bankoglu
- Institute of Pharmacology and Toxicology, University of Wuerzburg, Wuerzburg, Bavaria 97080, Germany
| | - Yalda Ghoreishi
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Center of Operative Medicine, Oberduerrbacherstrasse 6, Wuerzburg, Bavaria 97080, Germany
| | - Ulrich Dischinger
- Department of Endocrinology and Diabetology, University Hospital Wuerzburg, Wuerzburg, Bavaria 97080, Germany
| | - Max Kurlbaum
- Department of Endocrinology and Diabetology, University Hospital Wuerzburg, Wuerzburg, Bavaria 97080, Germany
| | - Matthias Kroiss
- Department of Endocrinology and Diabetology, University Hospital Wuerzburg, Wuerzburg, Bavaria 97080, Germany
| | - Christoph Otto
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Center of Operative Medicine, Oberduerrbacherstrasse 6, Wuerzburg, Bavaria 97080, Germany
| | - Carel W. le Roux
- Diabetes Complications Research Centre, University College Dublin, Dublin 4, Ireland
| | - Tulika Arora
- Novo Nordisk Foundation Centre for Basic Metabolic Research, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Florian Seyfried
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Center of Operative Medicine, Oberduerrbacherstrasse 6, Wuerzburg, Bavaria 97080, Germany
| | - Nicolas Schlegel
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Center of Operative Medicine, Oberduerrbacherstrasse 6, Wuerzburg, Bavaria 97080, Germany
| |
Collapse
|
187
|
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
|
188
|
Liu Z, Coales I, Penney N, McDonald JAK, Phetcharaburanin J, Seyfried F, Li JV. A Subset of Roux-en-Y Gastric Bypass Bacterial Consortium Colonizes the Gut of Nonsurgical Rats without Inducing Host-Microbe Metabolic Changes. mSystems 2020; 5:e01047-20. [PMID: 33293406 PMCID: PMC8579838 DOI: 10.1128/msystems.01047-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/19/2020] [Indexed: 01/02/2023] Open
Abstract
Roux-en-Y gastric bypass (RYGB) is an effective weight loss surgery, resulting in a characteristic increase of fecal Gammaproteobacteria The contribution of this compositional change to metabolic benefits of RYGB is currently debatable. Therefore, this study employed 16S rRNA gene sequencing and metabolic profiling to monitor the dynamic colonization of the RYGB microbial consortium and their metabolic impact on the host. Eleven Wistar rats received vancomycin and enrofloxacin, followed by fecal microbiota transplantation (FMT) of cecal slurry obtained from either RYGB- or sham-operated rats. Urine and feces from the microbiota recipients (RYGB microbiota recipients [RYGBr], n = 6; sham microbiota recipients [SHAMr], n = 5) were collected pre- and post-antibiotics and 1, 3, 6, 9, and 16 days post-FMT. No significant differences in body weight and food intake were observed between RYGBr and SHAMr. While neither group reached the community richness of that of their donors, by day 6, both groups reached the richness and diversity of that prior to antibiotic treatment. However, the typical signature of RYGB microbiome-increased Enterobacteriaceae-was not replicated in these recipients after two consecutive FMT, suggesting that the environmental changes induced by the anatomical rearrangements of RYGB could be key for sustaining such a consortium. The transplanted bacteria did not induce the same metabolic signature of urine and feces as those previously reported in RYGB-operated rats. Future work is required to explore environmental factors that shape the RYGB microbiota in order to further investigate the metabolic functions of the RYGB microbiota, thereby teasing out the mechanisms of the RYGB surgery.IMPORTANCE Roux-en-Y gastric bypass (RYGB) surgery results in a long-term gut bacterial shift toward Gammaproteobacteria in both patients and rodents. The contribution of this compositional shift, or the RYGB bacterial consortium, to the metabolic benefit of the surgery remains debatable. It is unclear how well these bacteria colonize in an anatomically normal gut. This is a fundamental question in both defining the function of the RYGB microbiota and evaluating its potential as a nonsurgical treatment for obesity. We monitored the dynamic colonization of the RYGB bacterial consortium and observed that while approximately one-third of the bacterial taxa from the RYGB donor colonized in the gut of the nonoperated recipients, Gammaproteobacteria were unable to colonize for longer than 3 days. The study highlighted that a successful long-term colonization of Gammaproteobacteria-rich RYGB microbiota in nonsurgical animals requires key environmental factors that may be dictated by the intestinal anatomical modification by the surgery itself.
Collapse
Affiliation(s)
- Zhigang Liu
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, South Kensington Campus, London, United Kingdom
| | - Isabelle Coales
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Nicholas Penney
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, St. Mary's Hospital Campus, London, United Kingdom
| | - Julie A K McDonald
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, South Kensington Campus, London, United Kingdom
| | | | - Florian Seyfried
- Department of General, Visceral, Transplant, Vascular, and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Jia V Li
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, South Kensington Campus, London, United Kingdom
| |
Collapse
|
189
|
Yu J, Sun H, Cao W, Han L, Song Y, Wan D, Jiang Z. Applications of gut microbiota in patients with hematopoietic stem-cell transplantation. Exp Hematol Oncol 2020; 9:35. [PMID: 33292670 PMCID: PMC7716583 DOI: 10.1186/s40164-020-00194-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Abstract
Studies of the gut microbiota (GM) have demonstrated the close link between human wellness and intestinal commensal bacteria, which mediate development of the host immune system. The dysbiosis, a disruption of the microbiome natural balance, can cause serious health problems. Patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) may cause significant changes in GM due to their underlying malignancies and exposure to extensive chemotherapy and systemic antibiotics, which may lead to different disorders. There are complex and multi-directional interactions among intestinal inflammation, GM and immune reactivity after HSCT. There is considerable effect of the human intestinal microbiome on clinical course following HSCT. Some bacteria in the intestinal ecosystem may be potential biomarkers or therapeutic targets for preventing relapse and improving survival rate after HSCT. Microbiota can be used as predictor of mortality in allo-HSCT. Two different strategies with targeted modulation of GM, preemptive and therapeutic, have been used for preventing or treating GM dysbiosis in patients with HSCT. Preemptive strategies include enteral nutrition (EN), prebiotic, probiotic, fecal microbiota transplantation (FMT) and antibiotic strategies, while therapeutic strategies include FMT, probiotic and lactoferrine usages. In this review, we summarize the advance of therapies targeting GM in patients with HSCT.
Collapse
Affiliation(s)
- Jifeng Yu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Academy of Medical and Pharmaceutical Sciences of Zhengzhou University, Zhengzhou, 450052, China
| | - Hao Sun
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Weijie Cao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Lijie Han
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yongping Song
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Dingming Wan
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
190
|
Badran M, Mashaqi S, Gozal D. The gut microbiome as a target for adjuvant therapy in obstructive sleep apnea. Expert Opin Ther Targets 2020; 24:1263-1282. [PMID: 33180654 PMCID: PMC9394230 DOI: 10.1080/14728222.2020.1841749] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: Gut dysbiosis is assumed to play a role in obstructive sleep apnea (OSA)-associated morbidities. Pre- and probiotics, short chain fatty acids (SCFA) and fecal matter transplantation (FMT) may offer potential as novel therapeutic strategies that target this gut dysbiosis. As more mechanisms of OSA-induced dysbiosis are being elucidated, these novel approaches are being tested in preclinical and clinical development. Areas covered: We examined the evidence linking OSA to gut dysbiosis and discuss the effects of pre- and probiotics on associated cardiometabolic, neurobehavioral and gastrointestinal disorders. The therapeutic potential of SCFA and FMT are also discussed. We reviewed the National Center for Biotechnology Information database, including PubMed and PubMed Central between 2000 - 2020. Expert opinion: To date, there are no clinical trials and only limited evidence from animal studies describing the beneficial effects of pre- and probiotic supplementation on OSA-mediated dysbiosis. Thus, more work is necessary to assess whether prebiotics, probiotics and SCFA are promising future novel strategies for targeting OSA-mediated dysbiosis.
Collapse
Affiliation(s)
- Mohammad Badran
- Department of Child Health and the Child Health Research Institute, University of Missouri School of Medicine , Columbia, MO, USA
| | - Saif Mashaqi
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Arizona School of Medicine , Tucson, AZ, USA
| | - David Gozal
- Department of Child Health and the Child Health Research Institute, University of Missouri School of Medicine , Columbia, MO, USA
| |
Collapse
|
191
|
Hartstra AV, Schüppel V, Imangaliyev S, Schrantee A, Prodan A, Collard D, Levin E, Dallinga-Thie G, Ackermans MT, Winkelmeijer M, Havik SR, Metwaly A, Lagkouvardos I, Nier A, Bergheim I, Heikenwalder M, Dunkel A, Nederveen AJ, Liebisch G, Mancano G, Claus SP, Benítez-Páez A, la Fleur SE, Bergman JJ, Gerdes V, Sanz Y, Booij J, Kemper E, Groen AK, Serlie MJ, Haller D, Nieuwdorp M. Infusion of donor feces affects the gut-brain axis in humans with metabolic syndrome. Mol Metab 2020; 42:101076. [PMID: 32916306 PMCID: PMC7536740 DOI: 10.1016/j.molmet.2020.101076] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/31/2020] [Accepted: 09/04/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Increasing evidence indicates that intestinal microbiota play a role in diverse metabolic processes via intestinal butyrate production. Human bariatric surgery data suggest that the gut-brain axis is also involved in this process, but the underlying mechanisms remain unknown. METHODS We compared the effect of fecal microbiota transfer (FMT) from post-Roux-en-Y gastric bypass (RYGB) donors vs oral butyrate supplementation on (123I-FP-CIT-determined) brain dopamine transporter (DAT) and serotonin transporter (SERT) binding as well as stable isotope-determined insulin sensitivity at baseline and after 4 weeks in 24 male and female treatment-naïve metabolic syndrome subjects. Plasma metabolites and fecal microbiota were also determined at these time points. RESULTS We observed an increase in brain DAT after donor FMT compared to oral butyrate that reduced this binding. However, no effect on body weight and insulin sensitivity was demonstrated after post-RYGB donor feces transfer in humans with metabolic syndrome. Increases in fecal levels of Bacteroides uniformis were significantly associated with an increase in DAT, whereas increases in Prevotella spp. showed an inverse association. Changes in the plasma metabolites glycine, betaine, methionine, and lysine (associated with the S-adenosylmethionine cycle) were also associated with altered striatal DAT expression. CONCLUSIONS Although more and larger studies are needed, our data suggest a potential gut microbiota-driven modulation of brain dopamine and serotonin transporters in human subjects with obese metabolic syndrome. These data also suggest the presence of a gut-brain axis in humans that can be modulated. NTR REGISTRATION 4488.
Collapse
Affiliation(s)
- Annick V Hartstra
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Valentina Schüppel
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Sultan Imangaliyev
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Anouk Schrantee
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Andrei Prodan
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Didier Collard
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Evgeni Levin
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Geesje Dallinga-Thie
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Mariette T Ackermans
- Laboratory of Endocrinology, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Maaike Winkelmeijer
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Stefan R Havik
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Amira Metwaly
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Ilias Lagkouvardos
- ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
| | - Anika Nier
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Austria
| | - Ina Bergheim
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Austria
| | - Mathias Heikenwalder
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Heidelberg, Germany
| | - Andreas Dunkel
- Leibniz-Institute for Food Systems Biology, Technical University of Munich, Freising, Germany
| | - Aart J Nederveen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Gerhard Liebisch
- Department of Laboratory Medicine, University of Regensburg, Regensburg, Germany
| | - Giulia Mancano
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Sandrine P Claus
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Alfonso Benítez-Páez
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Susanne E la Fleur
- Laboratory of Endocrinology, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Jacques J Bergman
- Department of Gastroenterology, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Victor Gerdes
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Yolanda Sanz
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Jan Booij
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Elles Kemper
- Department of Clinical Pharmacy, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Albert K Groen
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Mireille J Serlie
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Dirk Haller
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany; ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands.
| |
Collapse
|
192
|
Bishehsari F, Voigt RM, Keshavarzian A. Circadian rhythms and the gut microbiota: from the metabolic syndrome to cancer. Nat Rev Endocrinol 2020; 16:731-739. [PMID: 33106657 PMCID: PMC8085809 DOI: 10.1038/s41574-020-00427-4] [Citation(s) in RCA: 197] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/18/2020] [Indexed: 12/13/2022]
Abstract
The metabolic syndrome is prevalent in developed nations and accounts for the largest burden of non-communicable diseases worldwide. The metabolic syndrome has direct effects on health and increases the risk of developing cancer. Lifestyle factors that are known to promote the metabolic syndrome generally cause pro-inflammatory alterations in microbiota communities in the intestine. Indeed, alterations to the structure and function of intestinal microbiota are sufficient to promote the metabolic syndrome, inflammation and cancer. Among the lifestyle factors that are associated with the metabolic syndrome, disruption of the circadian system, known as circadian dysrhythmia, is increasingly common. Disruption of the circadian system can alter microbiome communities and can perturb host metabolism, energy homeostasis and inflammatory pathways, which leads to the metabolic syndrome. This Perspective discusses the role of intestinal microbiota and microbial metabolites in mediating the effects of disruption of circadian rhythms on human health.
Collapse
Affiliation(s)
- Faraz Bishehsari
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL, USA
| | - Robin M Voigt
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL, USA
| | - Ali Keshavarzian
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL, USA.
- Department of Physiology, Rush University Medical Center, Chicago, IL, USA.
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.
| |
Collapse
|
193
|
Koffert J, Lahti L, Nylund L, Salminen S, Hannukainen JC, Salminen P, de Vos WM, Nuutila P. Partial restoration of normal intestinal microbiota in morbidly obese women six months after bariatric surgery. PeerJ 2020; 8:e10442. [PMID: 33304658 PMCID: PMC7700738 DOI: 10.7717/peerj.10442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/06/2020] [Indexed: 12/18/2022] Open
Abstract
We studied the impact of bariatric surgery on the intestinal microbiota of morbidly obese study subjects. A total of 13 morbidly obese women (five of which had type 2 diabetes) and 14 healthy age- and gender-matched controls were recruited and the microbiota composition of fecal samples were determined by using a phylogenetic microarray. Sampling of the patients took place just one month before and 6 months after the operation. Within six months after bariatric surgery, the obese subjects had lost on average a quarter of their weight whereas four of the five of the diabetic subjects were in remission. Bariatric surgery was associated with an increased microbial community richness and Bacteroidetes/Firmicutes ratio. In addition, we observed an increased relative abundance of facultative anaerobes, such as Streptococcus spp., and a reduction in specific butyrate-producing Firmicutes. The observed postoperative alterations in intestinal microbiota reflect adaptation to the changing conditions in the gastrointestinal tract, such as energy restriction and the inability to process fiber-rich foods after bariatric surgery.
Collapse
Affiliation(s)
- Jukka Koffert
- Department of Gastroenterology, Turku University Hospital, Turku, Finland.,Turku PET Centre, University of Turku, Turku, Finland
| | - Leo Lahti
- Department of Mathematics and Statistics, University of Turku, Turku, Finland
| | - Lotta Nylund
- Functional Foods Forum, University of Turku, Turku, Finland.,Food Chemistry and Food Development, Department of Biochemistry, University of Turku, Turku, Finland
| | - Seppo Salminen
- Functional Foods Forum, University of Turku, Turku, Finland
| | | | - Paulina Salminen
- Division of Digestive Surgery and Urology, Turku University Hospital, Turku, Finland
| | - Willem M de Vos
- RPU Immunobiology and Human Microbiome, University of Helsinki, Helsinki, Finland.,Laboratory of Microbiology, Wageningen University and Research, Wageningen, The Netherlands
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku, Turku, Finland.,Department of Endocrinology, Turku University hospital, Turku, Finland
| |
Collapse
|
194
|
Peri-operative antibiotics acutely and significantly impact intestinal microbiota following bariatric surgery. Sci Rep 2020; 10:20340. [PMID: 33230230 PMCID: PMC7684314 DOI: 10.1038/s41598-020-77285-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023] Open
Abstract
Bariatric surgery is the most effective treatment for weight loss. Vertical sleeve gastrectomy (VSG) involves the resection of ~ 80% of the stomach and was conceived to purely restrict oral intake. However, evidence suggests more complex mechanisms, particularly postoperative changes in gut microbiota, in facilitating weight loss and resolving associated comorbidities. VSG in humans is a complex procedure and includes peri-operative antibiotics and caloric restriction in addition to the altered anatomy. The impact of each of these factors on the intestinal microbiota have not been evaluated. The aim of this study was to determine the relative contributions of each of these factors on intestinal microbiota composition following VSG prior to substantial weight loss. Thirty-two obese patients underwent one of three treatments: (1) VSG plus routine intravenous peri-operative antibiotics (n = 12), (2) VSG with intravenous vancomycin chosen for its low intestinal penetrance (n = 12), and (3) caloric restriction (n = 8). Fecal samples were evaluated for bacterial composition prior to and 7 days following each intervention. Only patients undergoing VSG with routine peri-operative antibiotics showed a significant shift in community composition. Our data support the single dose of routine peri-operative antibiotics as the most influential factor of intestinal microbial composition acutely following VSG.
Collapse
|
195
|
Obese Mice with Dyslipidemia Exhibit Meibomian Gland Hypertrophy and Alterations in Meibum Composition and Aqueous Tear Production. Int J Mol Sci 2020; 21:ijms21228772. [PMID: 33233559 PMCID: PMC7699756 DOI: 10.3390/ijms21228772] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/05/2020] [Accepted: 11/16/2020] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Dyslipidemia may be linked to meibomian gland dysfunction (MGD) and altered meibum lipid composition. The purpose was to determine if plasma and meibum cholesteryl esters (CE), triglycerides (TG), ceramides (Cer) and sphingomyelins (SM) change in a mouse model of diet-induced obesity where mice develop dyslipidemia. METHODS Male C57/BL6 mice (8/group, age = 6 wks) were fed a normal (ND; 15% kcal fat) or an obesogenic high-fat diet (HFD; 42% kcal fat) for 10 wks. Tear production was measured and meibography was performed. Body and epididymal adipose tissue (eAT) weights were determined. Nano-ESI-MS/MS and LC-ESI-MS/MS were used to detect CE, TG, Cer and SM species. Data were analyzed by principal component analysis, Pearson's correlation and unpaired t-tests adjusted for multiple comparisons; significance set at p ≤ 0.05. RESULTS Compared to ND mice, HFD mice gained more weight and showed heavier eAT and dyslipidemia with higher levels of plasma CE, TG, Cer and SM. HFD mice had hypertrophic meibomian glands, increased levels of lipid species acylated by saturated fatty acids in plasma and meibum and excessive tear production. CONCLUSIONS The majority of meibum lipid species with saturated fatty acids increased with HFD feeding with evidence of meibomian gland hypertrophy and excessive tearing. The dyslipidemia is associated with altered meibum composition, a key feature of MGD.
Collapse
|
196
|
Kanwal S, Aliya S, Xin Y. Anti-Obesity Effect of Dictyophora indusiata Mushroom Polysaccharide (DIP) in High Fat Diet-Induced Obesity via Regulating Inflammatory Cascades and Intestinal Microbiome. Front Endocrinol (Lausanne) 2020; 11:558874. [PMID: 33329380 PMCID: PMC7717937 DOI: 10.3389/fendo.2020.558874] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 10/13/2020] [Indexed: 12/16/2022] Open
Abstract
Obesity is a multifactorial metabolic disorder characterized by low-grade chronic inflammation, hyper-permeability of the gut epithelium, and perturbation of the intestinal microbiome. Despite the numerous therapeutic efficacies of Dictyophora indusiata mushroom, its biological activity in alleviating obesity through regulation of the gut microbiota and inflammatory cascades remain obscure. Henceforth, we determined the modulatory impact of D. indusiata polysaccharide (DIP) in the high-fat diet (HFD)-induced obesity mice model. The experimental subjects (BALB/C mice) were supplemented with chow diet (Control group), high-fat diet (HFD group), or HFD along with DIP at a low dose [HFD + DIP(L)] and high dose [HFD + DIP(H)]. Obesity-related parameters, including body weight gain, epididymal adipocyte size, fat accumulation, adipogenic markers, lipogenic markers, inflammatory associated markers, intestinal integrity, and intestinal microbiome, were elucidated. Our findings demonstrated that the oral administration of DIP at low dose partially and at high dose significantly reversed HFD-induced obesity parameters. Furthermore, the body weight, fat accumulation, adipocyte size, adipogenic and liver associated markers, glucose levels, inflammatory cytokines, and endotoxin (Lipopolysaccharide, LPS) levels were reduced considerably. Moreover, the study revealed that DIP treatment reversed the dynamic alterations of the gut microbiome community by decreasing the Firmicutes to Bacteroidetes ratio. These findings led us to infer the therapeutic potential of DIP in alleviating HFD-induced obesity via regulating inflammatory cascades, modulating intestinal integrity and intestinal microbiome community.
Collapse
Affiliation(s)
- Sadia Kanwal
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Shams Aliya
- Faculty of Life Sciences, Institute of Cancer Therapeutics, University of Bradford, Bradford, United Kingdom
| | - Yi Xin
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
197
|
Gut Microbiota in Patients with Morbid Obesity Before and After Bariatric Surgery: a Ten-Year Review Study (2009-2019). Obes Surg 2020; 31:317-326. [PMID: 33130944 DOI: 10.1007/s11695-020-05074-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 10/14/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023]
Abstract
The changes in the composition and function of gut microbiota affect the metabolic functions (which are mediated by microbial effects) in patients with obesity, resulting in significant physiological regulation in these patients. Most of the studies emphasize that the Western-style diet (high fat and low vegetable consumption) leads to significant changes in the intestinal microbiome in individuals with metabolic syndrome. A deeper understanding of the profiles of gut microbes will contribute to the development of new therapeutic strategies for the management of metabolic syndrome and other metabolic diseases and related disorders. The aim of this review is to evaluate recent experimental evidence outlining the alterations of gut microbiota composition and function in recovery from bariatric surgical operations with an emphasis on sleeve gastrectomy and gastric bypass.
Collapse
|
198
|
Ye Y, Abu El Haija M, Morgan DA, Guo D, Song Y, Frank A, Tian L, Riedl RA, Burnett CML, Gao Z, Zhu Z, Shahi SK, Zarei K, Couvelard A, Poté N, Ribeiro-Parenti L, Bado A, Noureddine L, Bellizzi A, Kievit P, Mangalam AK, Zingman LV, Le Gall M, Grobe JL, Kaplan LM, Clegg D, Rahmouni K, Mokadem M. Endocannabinoid Receptor-1 and Sympathetic Nervous System Mediate the Beneficial Metabolic Effects of Gastric Bypass. Cell Rep 2020; 33:108270. [PMID: 33113371 PMCID: PMC7660289 DOI: 10.1016/j.celrep.2020.108270] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 05/18/2020] [Accepted: 09/23/2020] [Indexed: 12/27/2022] Open
Abstract
The exact mechanisms underlying the metabolic effects of bariatric surgery remain unclear. Here, we demonstrate, using a combination of direct and indirect calorimetry, an increase in total resting metabolic rate (RMR) and specifically anaerobic RMR after Roux-en-Y gastric bypass (RYGB), but not sleeve gastrectomy (SG). We also show an RYGB-specific increase in splanchnic sympathetic nerve activity and "browning" of visceral mesenteric fat. Consequently, selective splanchnic denervation abolishes all beneficial metabolic outcomes of gastric bypass that involve changes in the endocannabinoid signaling within the small intestine. Furthermore, we demonstrate that administration of rimonabant, an endocannabinoid receptor-1 (CB1) inverse agonist, to obese mice mimics RYGB-specific effects on energy balance and splanchnic nerve activity. On the other hand, arachidonoylethanolamide (AEA), a CB1 agonist, attenuates the weight loss and metabolic signature of this procedure. These findings identify CB1 as a key player in energy regulation post-RYGB via a pathway involving the sympathetic nervous system.
Collapse
Affiliation(s)
- Yuanchao Ye
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Marwa Abu El Haija
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Donald A Morgan
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Deng Guo
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Yang Song
- College of Pharmacy, China Medical University, 77 Puhe Rd., Liaoning 110122, P.R. China
| | - Aaron Frank
- The Biomedical Research Department, Diabetes and Obesity Research Division, Cedars Sinai Medical Center, Beverly Hills, CA 90048, USA
| | - Liping Tian
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P.R. China
| | - Ruth A Riedl
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Colin M L Burnett
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Zhan Gao
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Zhiyong Zhu
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Shailesh K Shahi
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Kasra Zarei
- Medical Scientist Training Program, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Anne Couvelard
- INSERM U1149, Centre de Recherche sur l'Inflammation, Université de Paris, Paris 75018, France; Department of Pathology, Bichat Hospital, AP-HP, Paris 75018, France
| | - Nicolas Poté
- INSERM U1149, Centre de Recherche sur l'Inflammation, Université de Paris, Paris 75018, France; Department of Pathology, Bichat Hospital, AP-HP, Paris 75018, France
| | - Lara Ribeiro-Parenti
- INSERM U1149, Centre de Recherche sur l'Inflammation, Université de Paris, Paris 75018, France; Department of General and Digestive Surgery, Bichat Hospital, AP-HP, Paris 75018, France
| | - André Bado
- INSERM U1149, Centre de Recherche sur l'Inflammation, Université de Paris, Paris 75018, France
| | - Lama Noureddine
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Andrew Bellizzi
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Paul Kievit
- Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Ashutosh K Mangalam
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Interdisciplinary Graduate Program in Immunology and Molecular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Leonid V Zingman
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Orders of Eagles Diabetes Research Center, Iowa City, IA 52242, USA; Veterans Affairs Health Care System, Iowa City, IA 52242, USA; Obesity Research & Education Initiative, University of Iowa, Iowa City, IA 52242, USA
| | - Maude Le Gall
- INSERM U1149, Centre de Recherche sur l'Inflammation, Université de Paris, Paris 75018, France
| | - Justin L Grobe
- Departments of Physiology and Biomedical Engineering, Medical College of Wisconsin, Milwaukee, MI 53226, USA
| | - Lee M Kaplan
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Obesity, Metabolism, and Nutrition Institute, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Deborah Clegg
- College of Nursing and Health Professions, Drexel University, 1601 Cherry Street, Philadelphia, PA 19102, USA
| | - Kamal Rahmouni
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Medical Scientist Training Program, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Orders of Eagles Diabetes Research Center, Iowa City, IA 52242, USA; Veterans Affairs Health Care System, Iowa City, IA 52242, USA; Obesity Research & Education Initiative, University of Iowa, Iowa City, IA 52242, USA
| | - Mohamad Mokadem
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Orders of Eagles Diabetes Research Center, Iowa City, IA 52242, USA; Veterans Affairs Health Care System, Iowa City, IA 52242, USA; Obesity Research & Education Initiative, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
199
|
Dong TS, Luu K, Lagishetty V, Sedighian F, Woo SL, Dreskin BW, Katzka W, Chang C, Zhou Y, Arias-Jayo N, Yang J, Ahdoot A, Li Z, Pisegna JR, Jacobs JP. A High Protein Calorie Restriction Diet Alters the Gut Microbiome in Obesity. Nutrients 2020; 12:3221. [PMID: 33096810 PMCID: PMC7590138 DOI: 10.3390/nu12103221] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/15/2020] [Accepted: 10/18/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND High protein calorie restriction diets have shown clinical efficacy for obesity, but the mechanisms are not fully known. The intestinal microbiome is a mediator of obesity and preclinical data support an effect of high protein diet (HPD) on the gut microbiome of obesity, but there are few studies in humans. METHODS To address this, we conducted a dietary intervention trial of 80 overweight and obese subjects who were randomized to a calorie-restricted high protein diet (HPD) (30% calorie intake) or calorie-restricted normal protein diet (NPD) (15%) for 8 weeks. Baseline dietary intake patterns were assessed by the Diet History Questionnaire III. Longitudinal fecal sampling was performed at baseline, week 1, week 2, week 4, week 6, and week 8, for a total of 365 samples. Intestinal microbiome composition was assessed by 16S rRNA gene sequencing. RESULTS At baseline, microbial composition was associated with fiber and protein intake. Subjects on the HPD showed a significant increase in microbial diversity as measured by the Shannon index compared to those on the NPD. The HPD was also associated with significant differences in microbial composition after treatment compared to the NPD. Both diets induced taxonomic shifts compared to baseline, including enrichment of Akkermansia spp. and Bifidobacterium spp. and depletion of Prevotella spp. Conclusion: These findings provide evidence that weight loss diets alter the gut microbiome in obesity and suggest differential effects of HPDs compared to NPDs which may influence the clinical response to HPD.
Collapse
Affiliation(s)
- Tien S. Dong
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.S.D.); (K.L.); (V.L.); (F.S.); (W.K.); (C.C.); (Y.Z.); (N.A.-J.); (J.Y.); (A.A.); (J.R.P.)
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA;
- Department of Medicine, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Kayti Luu
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.S.D.); (K.L.); (V.L.); (F.S.); (W.K.); (C.C.); (Y.Z.); (N.A.-J.); (J.Y.); (A.A.); (J.R.P.)
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA;
- Department of Medicine, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Venu Lagishetty
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.S.D.); (K.L.); (V.L.); (F.S.); (W.K.); (C.C.); (Y.Z.); (N.A.-J.); (J.Y.); (A.A.); (J.R.P.)
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA;
- Department of Medicine, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Farzaneh Sedighian
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.S.D.); (K.L.); (V.L.); (F.S.); (W.K.); (C.C.); (Y.Z.); (N.A.-J.); (J.Y.); (A.A.); (J.R.P.)
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
| | - Shih-Lung Woo
- Center for Human Nutrition, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
| | - Benjamin W. Dreskin
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA;
- Department of Medicine, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - William Katzka
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.S.D.); (K.L.); (V.L.); (F.S.); (W.K.); (C.C.); (Y.Z.); (N.A.-J.); (J.Y.); (A.A.); (J.R.P.)
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
| | - Candace Chang
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.S.D.); (K.L.); (V.L.); (F.S.); (W.K.); (C.C.); (Y.Z.); (N.A.-J.); (J.Y.); (A.A.); (J.R.P.)
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
| | - Yi Zhou
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.S.D.); (K.L.); (V.L.); (F.S.); (W.K.); (C.C.); (Y.Z.); (N.A.-J.); (J.Y.); (A.A.); (J.R.P.)
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
| | - Nerea Arias-Jayo
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.S.D.); (K.L.); (V.L.); (F.S.); (W.K.); (C.C.); (Y.Z.); (N.A.-J.); (J.Y.); (A.A.); (J.R.P.)
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
| | - Julianne Yang
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.S.D.); (K.L.); (V.L.); (F.S.); (W.K.); (C.C.); (Y.Z.); (N.A.-J.); (J.Y.); (A.A.); (J.R.P.)
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
| | - Aaron Ahdoot
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.S.D.); (K.L.); (V.L.); (F.S.); (W.K.); (C.C.); (Y.Z.); (N.A.-J.); (J.Y.); (A.A.); (J.R.P.)
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
| | - Zhaoping Li
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
- Department of Medicine, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Center for Human Nutrition, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
| | - Joseph R. Pisegna
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.S.D.); (K.L.); (V.L.); (F.S.); (W.K.); (C.C.); (Y.Z.); (N.A.-J.); (J.Y.); (A.A.); (J.R.P.)
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA;
- Department of Medicine, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Jonathan P. Jacobs
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.S.D.); (K.L.); (V.L.); (F.S.); (W.K.); (C.C.); (Y.Z.); (N.A.-J.); (J.Y.); (A.A.); (J.R.P.)
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA;
- Department of Medicine, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| |
Collapse
|
200
|
Man AW, Zhou Y, Xia N, Li H. Involvement of Gut Microbiota, Microbial Metabolites and Interaction with Polyphenol in Host Immunometabolism. Nutrients 2020; 12:E3054. [PMID: 33036205 PMCID: PMC7601750 DOI: 10.3390/nu12103054] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022] Open
Abstract
Immunological and metabolic processes are inextricably linked and important for maintaining tissue and organismal health. Manipulation of cellular metabolism could be beneficial to immunity and prevent metabolic and degenerative diseases including obesity, diabetes, and cancer. Maintenance of a normal metabolism depends on symbiotic consortium of gut microbes. Gut microbiota contributes to certain xenobiotic metabolisms and bioactive metabolites production. Gut microbiota-derived metabolites have been shown to be involved in inflammatory activation of macrophages and contribute to metabolic diseases. Recent studies have focused on how nutrients affect immunometabolism. Polyphenols, the secondary metabolites of plants, are presented in many foods and beverages. Several studies have demonstrated the antioxidant and anti-inflammatory properties of polyphenols. Many clinical trials and epidemiological studies have also shown that long-term consumption of polyphenol-rich diet protects against chronic metabolic diseases. It is known that polyphenols can modulate the composition of core gut microbiota and interact with the immunometabolism. In the present article, we review the mechanisms of gut microbiota and its metabolites on immunometabolism, summarize recent findings on how the interaction between microbiota and polyphenol modulates host immunometabolism, and discuss future research directions.
Collapse
Affiliation(s)
| | | | | | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany; (A.W.C.M.); (Y.Z.); (N.X.)
| |
Collapse
|