151
|
Abstract
Mice transplanted with human microbiota are essential tools for studying the role of microbiota in health and disease, striving for the development of microbiota-modulating therapeutics. Traditionally, germ-free mice have been the principal option for establishing human microbiota-associated (HMA) mouse models, leading to significant insights into the composition and function of the human microbiota. However, there are limitations in using germ-free mice as recipients of human microbiota, including considerable resource allocation to establish and maintain the model and incomplete development of their immune system and physiological functions. Thus, antibiotic-treated, non-germ-free mice have been developed as an alternative to satisfy the growing demand for an accessible HMA mouse model. Several methods have been described for creating "humanized" mice. These protocols vary in their key components, mainly antibiotic conditioning and frequency of oral gavage. To address this practical challenge and formulate a simple and repeatable protocol, we established a HMA mouse model with antibiotic-treated conventional and specific-pathogen free (SPF) C57BL/6J mice, revealing that a single oral gavage allows stable engraftment of the human microbiota. In this chapter, we present our simple protocol for antibiotic conditioning to prepare mice for stable engraftment of human gut microbiota.
Collapse
|
152
|
Grander C, Grabherr F, Enrich B, Meyer M, Mayr L, Schwärzler J, Pedrini A, Effenberger M, Adolph TE, Tilg H. Hepatic Meteorin-like and Krüppel-like Factor 3 are Associated with Weight Loss and Liver Injury. Exp Clin Endocrinol Diabetes 2021; 130:406-414. [PMID: 34407548 DOI: 10.1055/a-1537-8950] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Laparoscopic adjustable gastric banding (LAGB) was found to be effective in reducing body weight and improving insulin resistance in patients with obesity and non-alcoholic fatty liver disease (NAFLD). The adipokine/myokine meteorin-like (METNRL) is an important regulator of whole-body energy expenditure. Krüppel-like factor 3 (KLF3), a regulator of METRNL expression in eosinophils, inhibits the beiging of adipose tissue in mice and therefore regulates adipose tissue development. METHODS Thirty-three obese patients undergoing LAGB were included in the study. The hepatic and adipose tissue expression of METNRL and KLF3 was determined before (t0) and 6 months after (t6) LABG. The human liver cancer cell line (HepG2) was stimulated with cytokines and fatty acids and METNRL and KLF3 expressions were analyzed. RESULTS LAGB-associated weight loss was correlated with decreased hepatic METNRL expression. The expression of METNRL and KLF3 in hepatic-and adipose tissues correlated before and after LAGB. Individuals with augmented LAGB-induced weight loss (>20 kg) showed lower hepatic METNRL and KLF3 expression before and after LAGB than patients with <20 kg weight loss. METNRL and KLF3 levels were higher in patients with higher NAFLD activity scores. HepG2 stimulation with interleukin-1β, tumor necrosis factor-α, palmitic acid but not interleukin-6, oleic acid, or lipopolysaccharide, induced the expression of one or both investigated adipokines. CONCLUSIONS The novel description of METRNL and KLF3 as hepatokines could pave the way to target their production and/or signaling in obesity, NAFLD, and related disorders. Both proteins may act as possible biomarkers to estimate weight loss after bariatric surgery.
Collapse
Affiliation(s)
- Christoph Grander
- Department of Internal Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| | - Barbara Enrich
- Department of Internal Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| | - Moritz Meyer
- Department of Internal Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lisa Mayr
- Department of Internal Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| | - Julian Schwärzler
- Department of Internal Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| | - Alisa Pedrini
- Department of Internal Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| | - Maria Effenberger
- Department of Internal Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
153
|
Shafiee S, Cegolon L, Khafaei M, Gholami N, Zhao S, Khalesi N, Moosavian H, Fathi S, Izadi M, Ghadian A, Javanbakht M, Javanbakht A, Akhavan-Sigari R. Gastrointestinal cancers, ACE-2/TMPRSS2 expression and susceptibility to COVID-19. Cancer Cell Int 2021; 21:431. [PMID: 34399734 PMCID: PMC8365127 DOI: 10.1186/s12935-021-02129-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/30/2021] [Indexed: 12/22/2022] Open
Abstract
Recent studies on the pathophysiology of COVID-19 are indicating that the Angiotensin convertase enzyme 2 (ACE-2) and transmembrane serine protease 2 (TMPRSS2) can act as a major component in the fusion of SARS-Cov-2 with target cells. It has also been observed that the expression of ACE-2 and TMPRSS2 can be altered in malignancies. Shedding light on this matter could be crucial since the COVID-19 pandemic interfered with many gastrointestinal cancer screening programs. Herein we discuss the possibility of severe forms of COVID-19 in patients with gastrointestinal cancers due to the gastrointestinal entry route of SARS-CoV-2 into the human body. The disruption of cancer screening programs caused by the current COVID-19 pandemic could therefore have massive negative health impact on patients affected by gastrointestinal malignancies.
Collapse
Affiliation(s)
- Sepehr Shafiee
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Luca Cegolon
- Public Health Department, Local Health Unit N.2 "Marca Trevigiana", 31100, Treviso, Italy
| | - Mostafa Khafaei
- Human Genetics Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Nasrin Gholami
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shi Zhao
- JC School of Public Health and Primary Care, Chinese University of Hong Kong, Hong Kong, China
| | - Nasrin Khalesi
- Department of Pediatrics, Iran University of Medical Sciences, Tehran, Iran
| | - Hamidreza Moosavian
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Saeid Fathi
- Department of Parasite Vaccine Research and Production, Razi Vaccine and Serum Research Institute, Agriculture Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Morteza Izadi
- Health Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Alireza Ghadian
- Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Javanbakht
- Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | | | | |
Collapse
|
154
|
Plasma Imidazole Propionate Is Positively Correlated with Blood Pressure in Overweight and Obese Humans. Nutrients 2021; 13:nu13082706. [PMID: 34444866 PMCID: PMC8399073 DOI: 10.3390/nu13082706] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022] Open
Abstract
Background: The gut microbiota and its metabolites are essential for host health and dysbiosis has been involved in several pathologic conditions such as type 2 diabetes (T2D) and cardiovascular disease (CVD). Recent studies have identified that plasma imidazole propionate (ImP), a microbial-produced metabolite, is increased in patients with prediabetes and T2D. More recently, ImP was found to be significantly increased in patients with overt CVD. Here, we aimed to investigate the association between ImP and CVD risk factors: blood pressure, HDL-cholesterol, LDL-cholesterol and insulin-resistance in overweight and obese subjects without T2D or use of any metabolic diseases-related medication. Methods: Plasma metabolites, including ImP, were determined in 107 male or post-menopausal women with overweight/obesity, but without T2D. Insulin-sensitivity was assessed with the gold standard method: the hyperinsulinemic-euglycemic clamp using the isotope [6,6-2H2] glucose and expressed as glucose rate of disposal (Rd) for peripheral insulin sensitivity and suppression of endogenous glucose production (EGP) for hepatic insulin sensitivity. Results: Partial correlation analysis controlled for BMI and age showed a significant correlation between ImP and diastolic blood pressure (rs = 0.285, p = 0.004) and a borderline significance with systolic blood pressure (rs = 0.187, p = 0.060); however, systolic and diastolic blood pressure did not correlate with ImP precursor histidine (rs = 0.063, p = 0.526 and r = −0.038, p = 0.712, respectively). We did not find a correlation between ImP with LDL-cholesterol or HDL-cholesterol (rs = −0.181, p = 0.064 and rs = 0.060, p = 0.546, respectively). Furthermore, there was no association between plasma ImP concentrations and Rd and EGP suppression. Conclusion: In this cohort with overweight/obese subjects without T2D, plasma ImP concentrations were positively correlated with diastolic blood pressure but not with insulin-sensitivity.
Collapse
|
155
|
Bostrom JA, Mottel B, Heffron SP. Medical and Surgical Obesity Treatments and Atherosclerosis: Mechanisms beyond Typical Risk Factors. Curr Atheroscler Rep 2021; 23:60. [PMID: 34351556 PMCID: PMC9953388 DOI: 10.1007/s11883-021-00961-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2021] [Indexed: 02/02/2023]
Abstract
PURPOSE OF REVIEW This study aims to discuss the mechanisms by which GLP-1 agonists and bariatric surgery improve cardiovascular outcomes in severely obese patients. RECENT FINDINGS Recent studies have demonstrated that both GLP-1 agonist use and bariatric surgery reduce adverse cardiovascular outcomes. Improvements in traditional atherosclerosis risk factors in association with weight loss likely contribute, but weight loss-independent mechanisms are also suggested to have roles. We review the clinical and preclinical evidence base for cardiovascular benefit of LP-1 agonists and bariatric surgery beyond traditional risk factors, including improvements in endothelial function, direct impacts on atherosclerotic plaques, and anti-inflammatory effects.
Collapse
Affiliation(s)
- John A Bostrom
- Department of Medicine, Leon H. Charney Division of Cardiology, NYU Center for the Prevention of Cardiovascular Disease, Cardiovascular Research Center, New York, NY, USA
| | - Beth Mottel
- Department of Medicine, Leon H. Charney Division of Cardiology, NYU Center for the Prevention of Cardiovascular Disease, Cardiovascular Research Center, New York, NY, USA
| | - Sean P Heffron
- Department of Medicine, Leon H. Charney Division of Cardiology, NYU Center for the Prevention of Cardiovascular Disease, Cardiovascular Research Center, New York, NY, USA.
| |
Collapse
|
156
|
Тимашева ЯР, Балхиярова ЖР, Кочетова ОВ. [Current state of the obesity research: genetic aspects, the role of microbiome, and susceptibility to COVID-19]. PROBLEMY ENDOKRINOLOGII 2021; 67:20-35. [PMID: 34533011 PMCID: PMC9753850 DOI: 10.14341/probl12775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/21/2021] [Accepted: 08/02/2021] [Indexed: 11/06/2022]
Abstract
Obesity affects over 700 million people worldwide and its prevalence keeps growing steadily. The problem is particularly relevant due to the increased risk of COVID-19 complications and mortality in obese patients. Obesity prevalence increase is often associated with the influence of environmental and behavioural factors, leading to stigmatization of people with obesity due to beliefs that their problems are caused by poor lifestyle choices. However, hereditary predisposition to obesity has been established, likely polygenic in nature. Morbid obesity can result from rare mutations having a significant effect on energy metabolism and fat deposition, but the majority of patients does not present with monogenic forms. Microbiome low diversity significantly correlates with metabolic disorders (inflammation, insulin resistance), and the success of weight loss (bariatric) surgery. However, data on the long-term consequences of bariatric surgery and changes in the microbiome composition and genetic diversity before and after surgery are currently lacking. In this review, we summarize the results of studies of the genetic characteristics of obesity patients, molecular mechanisms of obesity, contributing to the unfavourable course of coronavirus infection, and the evolution of their microbiome during bariatric surgery, elucidating the mechanisms of disease development and creating opportunities to identify potential new treatment targets and design effective personalized approaches for the diagnosis, management, and prevention of obesity.
Collapse
Affiliation(s)
- Я. Р. Тимашева
- Институт биохимии и генетики Уфимского федерального исследовательского центра Российской академии наук;
Башкирский государственный медицинский университет
| | - Ж. Р. Балхиярова
- Институт биохимии и генетики Уфимского федерального исследовательского центра Российской академии наук;
Башкирский государственный медицинский университет;
Университет Суррея
| | - О. В. Кочетова
- Институт биохимии и генетики Уфимского федерального исследовательского центра Российской академии наук
| |
Collapse
|
157
|
Bel Lassen P, Belda E, Prifti E, Dao MC, Specque F, Henegar C, Rinaldi L, Wang X, Kennedy SP, Zucker JD, Calame W, Lamarche B, Claus SP, Clément K. Protein supplementation during an energy-restricted diet induces visceral fat loss and gut microbiota amino acid metabolism activation: a randomized trial. Sci Rep 2021; 11:15620. [PMID: 34341379 PMCID: PMC8329187 DOI: 10.1038/s41598-021-94916-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/12/2021] [Indexed: 01/07/2023] Open
Abstract
Interactions between diet and gut microbiota are critical regulators of energy metabolism. The effects of fibre intake have been deeply studied but little is known about the impact of proteins. Here, we investigated the effects of high protein supplementation (Investigational Product, IP) in a double blind, randomised placebo-controled intervention study (NCT01755104) where 107 participants received the IP or an isocaloric normoproteic comparator (CP) alongside a mild caloric restriction. Gut microbiota profiles were explored in a patient subset (n = 53) using shotgun metagenomic sequencing. Visceral fat decreased in both groups (IP group: - 20.8 ± 23.2 cm2; CP group: - 14.5 ± 24.3 cm2) with a greater reduction (p < 0.05) with the IP supplementation in the Per Protocol population. Microbial diversity increased in individuals with a baseline low gene count (p < 0.05). The decrease in weight, fat mass and visceral fat mass significantly correlated with the increase in microbial diversity (p < 0.05). Protein supplementation had little effects on bacteria composition but major differences were seen at functional level. Protein supplementation stimulated bacterial amino acid metabolism (90% amino-acid synthesis functions enriched with IP versus 13% in CP group (p < 0.01)). Protein supplementation alongside a mild energy restriction induces visceral fat mass loss and an activation of gut microbiota amino-acid metabolism.Clinical trial registration: NCT01755104 (24/12/2012). https://clinicaltrials.gov/ct2/show/record/NCT01755104?term=NCT01755104&draw=2&rank=1 .
Collapse
Affiliation(s)
- Pierre Bel Lassen
- Sorbonne Université, Inserm, Nutrition and Obesity: Systemic Approaches Research Unit, NutriOmics, Sorbonne Université, 91 boulevard de l'Hôpital, 75013, Paris, France
- Assistance Publique Hôpitaux de Paris, Nutrition Department, Pitié Salpêtrière Hospital, Assistance Publique Hôpitaux de Paris, 75013, Paris, France
| | - Eugeni Belda
- Sorbonne Université, Inserm, Nutrition and Obesity: Systemic Approaches Research Unit, NutriOmics, Sorbonne Université, 91 boulevard de l'Hôpital, 75013, Paris, France
- Integrative Phenomics, Paris, France
| | - Edi Prifti
- Sorbonne Université, Inserm, Nutrition and Obesity: Systemic Approaches Research Unit, NutriOmics, Sorbonne Université, 91 boulevard de l'Hôpital, 75013, Paris, France
- Sorbonne Université, IRD, UMMISCO, Unité de Modélisation Mathématique et Informatique des Systèmes Complexes, Sorbonne Université, 93143, Bondy, France
| | - Maria Carlota Dao
- Sorbonne Université, Inserm, Nutrition and Obesity: Systemic Approaches Research Unit, NutriOmics, Sorbonne Université, 91 boulevard de l'Hôpital, 75013, Paris, France
| | - Florian Specque
- Sorbonne Université, IRD, UMMISCO, Unité de Modélisation Mathématique et Informatique des Systèmes Complexes, Sorbonne Université, 93143, Bondy, France
| | - Corneliu Henegar
- Sorbonne Université, Inserm, Nutrition and Obesity: Systemic Approaches Research Unit, NutriOmics, Sorbonne Université, 91 boulevard de l'Hôpital, 75013, Paris, France
| | - Laure Rinaldi
- YSOPIA Bioscience, 17 place de la Bourse, 33076, Bordeaux, France
| | - Xuedan Wang
- Department of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, The University of Reading, Reading, RG6 6AP, UK
| | - Sean P Kennedy
- CNRS, Institut Pasteur, Department of Computational Biology, USR 3756 , CNRS, 75015, Paris, France
| | - Jean-Daniel Zucker
- Sorbonne Université, Inserm, Nutrition and Obesity: Systemic Approaches Research Unit, NutriOmics, Sorbonne Université, 91 boulevard de l'Hôpital, 75013, Paris, France
- Sorbonne Université, IRD, UMMISCO, Unité de Modélisation Mathématique et Informatique des Systèmes Complexes, Sorbonne Université, 93143, Bondy, France
| | - Wim Calame
- StatistiCal BV, Strandwal 148, 2241 MN, Wassenaar, The Netherlands
| | - Benoît Lamarche
- Laval University, Institute On Nutrition and Functional Foods, Laval University, Québec, QC, G1V 0A6, Canada
| | - Sandrine P Claus
- YSOPIA Bioscience, 17 place de la Bourse, 33076, Bordeaux, France.
| | - Karine Clément
- Sorbonne Université, Inserm, Nutrition and Obesity: Systemic Approaches Research Unit, NutriOmics, Sorbonne Université, 91 boulevard de l'Hôpital, 75013, Paris, France.
- Assistance Publique Hôpitaux de Paris, Nutrition Department, Pitié Salpêtrière Hospital, Assistance Publique Hôpitaux de Paris, 75013, Paris, France.
| |
Collapse
|
158
|
Nayor M, Shah SH, Murthy V, Shah RV. Molecular Aspects of Lifestyle and Environmental Effects in Patients With Diabetes: JACC Focus Seminar. J Am Coll Cardiol 2021; 78:481-495. [PMID: 34325838 DOI: 10.1016/j.jacc.2021.02.070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/07/2021] [Accepted: 02/01/2021] [Indexed: 01/04/2023]
Abstract
Diabetes is characterized as an integrated condition of dysregulated metabolism across multiple tissues, with well-established consequences on the cardiovascular system. Recent advances in precision phenotyping in biofluids and tissues in large human observational and interventional studies have afforded a unique opportunity to translate seminal findings in models and cellular systems to patients at risk for diabetes and its complications. Specifically, techniques to assay metabolites, proteins, and transcripts, alongside more recent assessment of the gut microbiome, underscore the complexity of diabetes in patients, suggesting avenues for precision phenotyping of risk, response to intervention, and potentially novel therapies. In addition, the influence of external factors and inputs (eg, activity, diet, medical therapies) on each domain of molecular characterization has gained prominence toward better understanding their role in prevention. Here, the authors provide a broad overview of the role of several of these molecular domains in human translational investigation in diabetes.
Collapse
Affiliation(s)
- Matthew Nayor
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA. https://twitter.com/MattNayor
| | - Svati H Shah
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA; Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA. https://twitter.com/SvatiShah
| | - Venkatesh Murthy
- Division of Cardiovascular Medicine, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA; Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA. https://twitter.com/venkmurthy
| | - Ravi V Shah
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
159
|
Du Y, Zhang J, Chen G, Sun Z. Formulation and interpretation of the Chinese Guidelines for Surgical Treatment of Obesity and Type 2 Diabetes Mellitus. Biosci Trends 2021; 15:299-304. [PMID: 34334581 DOI: 10.5582/bst.2021.01287] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Obesity and related metabolic diseases have become one of the world's most serious public health problems. Bariatric surgery has gone through a long and difficult development process, from being rejected to gradually recognized, then widely accepted, and finally becoming the "gold standard" for the treatment of morbid obesity with metabolic diseases. Procedures have constantly been improving and evolving as the concept of bariatric surgery has been reappraised. The comparison and selection of different procedures, the emergence of new technologies and treatment methods, and the in-depth study of the mechanism of metabolic weight loss surgery are effectively promoting the rapid development of bariatric surgery. This article looks at both the 2014 and 2019 editions of the Guidelines for Diagnosis and Treatment of Obesity and Type 2 Diabetes Mellitus from the Chinese Society of Metabolic and Bariatric Surgery (CSMBS), its review the development of bariatric surgery, and it describes surgical indications and contraindications, the mechanism of weight loss, and tailored selection of the surgical procedure in order to serve as a reference.
Collapse
Affiliation(s)
- Yanmin Du
- Bariatric Surgery Center, General Surgery Department, Beijing Shijitan Hospital, Capital Medical University (Peking University Ninth School of Clinical Medicine), Beijing, China
| | - Jianlu Zhang
- Bariatric Surgery Center, General Surgery Department, Beijing Shijitan Hospital, Capital Medical University (Peking University Ninth School of Clinical Medicine), Beijing, China
| | - Guanyang Chen
- Bariatric Surgery Center, General Surgery Department, Beijing Shijitan Hospital, Capital Medical University (Peking University Ninth School of Clinical Medicine), Beijing, China
| | - Zhipeng Sun
- Bariatric Surgery Center, General Surgery Department, Beijing Shijitan Hospital, Capital Medical University (Peking University Ninth School of Clinical Medicine), Beijing, China
| |
Collapse
|
160
|
Metabolomics in Bariatric Surgery: Towards Identification of Mechanisms and Biomarkers of Metabolic Outcomes. Obes Surg 2021; 31:4564-4574. [PMID: 34318371 DOI: 10.1007/s11695-021-05566-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/17/2021] [Accepted: 06/24/2021] [Indexed: 12/27/2022]
Abstract
Bariatric surgery has been widely performed for the treatment of obesity and type 2 diabetes. Efforts have been made to investigate the mechanisms underlying the metabolic effects achieved by bariatric surgery and to identify candidates who will benefit from this surgery. Metabolomics, which includes comprehensive profiling of metabolites in biological samples, has been utilized for various disease entities to discover pathophysiological metabolic pathways and biomarkers predicting disease progression or prognosis. Over the last decade, metabolomic studies on patients undergoing bariatric surgery have identified significant biomarkers related to metabolic effects. This review describes the significance, progress, and challenges for the future of metabolomics in the area of bariatric surgery.
Collapse
|
161
|
Analyses of publicly available Hungatella hathewayi genomes revealed genetic distances indicating they belong to more than one species. Virulence 2021; 12:1950-1964. [PMID: 34304696 PMCID: PMC8312603 DOI: 10.1080/21505594.2021.1950955] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Hungatella hathewayi has been observed to be a member of the gut microbiome. Unfortunately, little is known about this organism in spite of being associated with human fatalities; it is important to understand virulence mechanisms and epidemiological prospective to cause disease. In this study, a patient with chronic neurologic symptoms presented to the clinic with subsequent isolation of a strain with phenotypic characteristics suggestive of Clostridium difficile. However, whole-genome sequence found the organism to be H. hathewayi. Analysis including publicly available Hungatella genomes found substantial genomic differences as compared to the type strain, indicating this isolate was not C. difficile. We examined the whole-genome of Hungatella species and related genera, using comparative genomics to fully examine species identification and toxin production. Orthogonal phylogenetic using the 16S rRNA gene and entire genome analyses that included genome distance analyses using Genome-to-Genome Distance (GGDC), Average Nucleotide Identity (ANI), and a pan-genome analysis with inclusion of available public genomes determined the speciation to be Hungatella. Two clearly differentiated groups were identified, one including a reference H. hathewayi genome (strain DSM-13,479) and a second group that was determined to be H. effluvii, which included our clinical isolate. Also, some genomes reported as H. hathewayi were found to belong to other genera, including Clostridium and Faecalicatena. We show that the Hungatella species have an open pan-genome reflecting high genomic diversity. This study highlights the importance of correctly assigning taxonomic identification, particularly in disease-associated strains, to better understand virulence and therapeutic options.
Collapse
|
162
|
Juárez-Fernández M, Román-Sagüillo S, Porras D, García-Mediavilla MV, Linares P, Ballesteros-Pomar MD, Urioste-Fondo A, Álvarez-Cuenllas B, González-Gallego J, Sánchez-Campos S, Jorquera F, Nistal E. Long-Term Effects of Bariatric Surgery on Gut Microbiota Composition and Faecal Metabolome Related to Obesity Remission. Nutrients 2021; 13:nu13082519. [PMID: 34444679 PMCID: PMC8397959 DOI: 10.3390/nu13082519] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/17/2022] Open
Abstract
Obesity is one of the main worldwide public health concerns whose clinical management demands new therapeutic approaches. Bariatric surgery is the most efficient treatment when other therapies have previously failed. Due to the role of gut microbiota in obesity development, the knowledge of the link between bariatric surgery and gut microbiota could elucidate new mechanistic approaches. This study aims to evaluate the long-term effects of bariatric surgery in the faecal metagenome and metabolome of patients with severe obesity. Faecal and blood samples were collected before and four years after the intervention from patients with severe obesity. Biochemical, metagenomic and metabolomic analyses were performed and faecal short-chain fatty acids were measured. Bariatric surgery improved the obesity-related status of patients and significantly reshaped gut microbiota composition. Moreover, this procedure was associated with a specific metabolome profile characterized by a reduction in energetic and amino acid metabolism. Acetate, butyrate and propionate showed a significant reduction with bariatric surgery. Finally, correlation analysis suggested the existence of a long-term compositional and functional gut microbiota profile associated with the intervention. In conclusion, bariatric surgery triggered long-lasting effects on gut microbiota composition and faecal metabolome that could be associated with the remission of obesity.
Collapse
Affiliation(s)
- María Juárez-Fernández
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24007 León, Spain; (M.J.-F.); (S.R.-S.); (D.P.); (M.V.G.-M.); (J.G.-G.); (S.S.-C.); (F.J.)
| | - Sara Román-Sagüillo
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24007 León, Spain; (M.J.-F.); (S.R.-S.); (D.P.); (M.V.G.-M.); (J.G.-G.); (S.S.-C.); (F.J.)
| | - David Porras
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24007 León, Spain; (M.J.-F.); (S.R.-S.); (D.P.); (M.V.G.-M.); (J.G.-G.); (S.S.-C.); (F.J.)
| | - María Victoria García-Mediavilla
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24007 León, Spain; (M.J.-F.); (S.R.-S.); (D.P.); (M.V.G.-M.); (J.G.-G.); (S.S.-C.); (F.J.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Pedro Linares
- Servicio de Aparato Digestivo, Complejo Asistencial Universitario de León, 24071 León, Spain; (P.L.); (B.Á.-C.)
| | - María Dolores Ballesteros-Pomar
- Departamento de Endocrinología y Nutrición, Complejo Asistencial Universitario de León, 24071 León, Spain; (M.D.B.-P.); (A.U.-F.)
| | - Ana Urioste-Fondo
- Departamento de Endocrinología y Nutrición, Complejo Asistencial Universitario de León, 24071 León, Spain; (M.D.B.-P.); (A.U.-F.)
| | - Begoña Álvarez-Cuenllas
- Servicio de Aparato Digestivo, Complejo Asistencial Universitario de León, 24071 León, Spain; (P.L.); (B.Á.-C.)
| | - Javier González-Gallego
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24007 León, Spain; (M.J.-F.); (S.R.-S.); (D.P.); (M.V.G.-M.); (J.G.-G.); (S.S.-C.); (F.J.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sonia Sánchez-Campos
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24007 León, Spain; (M.J.-F.); (S.R.-S.); (D.P.); (M.V.G.-M.); (J.G.-G.); (S.S.-C.); (F.J.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Francisco Jorquera
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24007 León, Spain; (M.J.-F.); (S.R.-S.); (D.P.); (M.V.G.-M.); (J.G.-G.); (S.S.-C.); (F.J.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Servicio de Aparato Digestivo, Complejo Asistencial Universitario de León, 24071 León, Spain; (P.L.); (B.Á.-C.)
| | - Esther Nistal
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24007 León, Spain; (M.J.-F.); (S.R.-S.); (D.P.); (M.V.G.-M.); (J.G.-G.); (S.S.-C.); (F.J.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-9-8729-1997
| |
Collapse
|
163
|
Cunningham AL, Stephens JW, Harris DA. A review on gut microbiota: a central factor in the pathophysiology of obesity. Lipids Health Dis 2021; 20:65. [PMID: 34233682 PMCID: PMC8262044 DOI: 10.1186/s12944-021-01491-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/17/2021] [Indexed: 12/13/2022] Open
Abstract
Obesity and its complications constitute a substantial burden. Considerable published research describes the novel relationships between obesity and gut microbiota communities. It is becoming evident that microbiota behave in a pivotal role in their ability to influence homeostatic mechanisms either to the benefit or detriment of host health, the extent of which is not fully understood. A greater understanding of the contribution of gut microbiota towards host pathophysiology is revealing new therapeutic avenues to tackle the global obesity epidemic. This review focuses on causal relationships and associations with obesity, proposed central mechanisms encouraging the development of obesity and promising prospective methods for microbiota manipulation.
Collapse
Affiliation(s)
- A L Cunningham
- Department of Surgery, Swansea Bay University Health Board, Swansea, SA2 8QA, UK. .,Swansea University Medical School, Swansea University, Swansea, SA2 8QA, UK.
| | - J W Stephens
- Swansea University Medical School, Swansea University, Swansea, SA2 8QA, UK
| | - D A Harris
- Department of Surgery, Swansea Bay University Health Board, Swansea, SA2 8QA, UK.,Swansea University Medical School, Swansea University, Swansea, SA2 8QA, UK
| |
Collapse
|
164
|
Schreurs MPH, de Vos van Steenwijk PJ, Romano A, Dieleman S, Werner HMJ. How the Gut Microbiome Links to Menopause and Obesity, with Possible Implications for Endometrial Cancer Development. J Clin Med 2021; 10:jcm10132916. [PMID: 34209916 PMCID: PMC8268108 DOI: 10.3390/jcm10132916] [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] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Interest is growing in the dynamic role of gut microbiome disturbances in human health and disease. No direct evidence is yet available to link gut microbiome dysbiosis to endometrial cancer. This review aims to understand any association between microbiome dysbiosis and important risk factors of endometrial cancer, high estrogen levels, postmenopause and obesity. Methods: A systematic search was performed with PubMed as primary database. Three separate searches were performed to identify all relevant studies. Results: Fifteen studies were identified as highly relevant and included in the review. Eight articles focused on the relationship with obesity and eight studies focused on the menopausal change or estrogen levels. Due to the heterogeneity in patient populations and outcome measures, no meta-analysis could be performed. Both the menopausal change and obesity were noted to enhance dysbiosis by reducing microbiome diversity and increasing the Firmicutes to Bacteroidetes ratio. Both also incurred estrobolome changes, leading to increased systemic estrogen levels, especially after menopause. Furthermore, microbiome dysbiosis was reported to be related to systemic inflammation through toll-like receptor signaling deficiencies and overexpression of pro-inflammatory cytokines. Conclusions: This review highlights that the female gut microbiome is intrinsically linked to estrogen levels, menopausal state and systemic inflammation, which indicates gut microbiome dysbiosis as a potential hallmark for risk stratification for endometrial cancer. Studies are needed to further define the role the gut microbiome plays in women at risk for endometrial cancer.
Collapse
Affiliation(s)
- Malou P. H. Schreurs
- Department of Obstetrics, Gynecology and Gynecologic Oncology, Medisch Spectrum Twente, 7512 KZ Enschede, The Netherlands
- Maastricht University Medical Centre, Department of Obstetrics and Gynecology, GROW—School for Oncology and Development Biology, 6202 AZ Maastricht, The Netherlands; (P.J.d.V.v.S.); (A.R.); (H.M.J.W.)
- Correspondence:
| | - Peggy J. de Vos van Steenwijk
- Maastricht University Medical Centre, Department of Obstetrics and Gynecology, GROW—School for Oncology and Development Biology, 6202 AZ Maastricht, The Netherlands; (P.J.d.V.v.S.); (A.R.); (H.M.J.W.)
| | - Andrea Romano
- Maastricht University Medical Centre, Department of Obstetrics and Gynecology, GROW—School for Oncology and Development Biology, 6202 AZ Maastricht, The Netherlands; (P.J.d.V.v.S.); (A.R.); (H.M.J.W.)
| | - Sabine Dieleman
- Maastricht University Medical Centre, Department of Surgery, GROW—School for Oncology and Developmental Biology, 6202 AZ Maastricht, The Netherlands;
| | - Henrica M. J. Werner
- Maastricht University Medical Centre, Department of Obstetrics and Gynecology, GROW—School for Oncology and Development Biology, 6202 AZ Maastricht, The Netherlands; (P.J.d.V.v.S.); (A.R.); (H.M.J.W.)
| |
Collapse
|
165
|
Agus A, Clément K, Sokol H. Gut microbiota-derived metabolites as central regulators in metabolic disorders. Gut 2021; 70:1174-1182. [PMID: 33272977 PMCID: PMC8108286 DOI: 10.1136/gutjnl-2020-323071] [Citation(s) in RCA: 677] [Impact Index Per Article: 169.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/17/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023]
Abstract
Metabolic disorders represent a growing worldwide health challenge due to their dramatically increasing prevalence. The gut microbiota is a crucial actor that can interact with the host by the production of a diverse reservoir of metabolites, from exogenous dietary substrates or endogenous host compounds. Metabolic disorders are associated with alterations in the composition and function of the gut microbiota. Specific classes of microbiota-derived metabolites, notably bile acids, short-chain fatty acids, branched-chain amino acids, trimethylamine N-oxide, tryptophan and indole derivatives, have been implicated in the pathogenesis of metabolic disorders. This review aims to define the key classes of microbiota-derived metabolites that are altered in metabolic diseases and their role in pathogenesis. They represent potential biomarkers for early diagnosis and prognosis as well as promising targets for the development of novel therapeutic tools for metabolic disorders.
Collapse
Affiliation(s)
- Allison Agus
- University Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, Île-de-France, France,Paris Center for Microbiome Medicine (PaCeMM) FHU, AP-HP, Paris, Île-de-France, France
| | - Karine Clément
- Paris Center for Microbiome Medicine (PaCeMM) FHU, AP-HP, Paris, Île-de-France, France,Nutrition and Obesity: systemic approach (NutriOmics) research unit, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Sorbonne Universités, INSERM, Paris, Île-de-France, France
| | - Harry Sokol
- Paris Center for Microbiome Medicine (PaCeMM) FHU, AP-HP, Paris, Île-de-France, France .,Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology department, Sorbonne Universite, INSERM, Paris, Île-de-France, France
| |
Collapse
|
166
|
Argyrakopoulou G, Dalamaga M, Spyrou N, Kokkinos A. Gender Differences in Obesity-Related Cancers. Curr Obes Rep 2021; 10:100-115. [PMID: 33523397 DOI: 10.1007/s13679-021-00426-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/11/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW In this review, we summarize the role of obesity in carcinogenesis, providing details on specific cancer sites. Special emphasis is given to gender differences in obesity-related cancers and on the effect of bariatric surgery on cancer risk. RECENT FINDINGS Accumulating evidence has highlighted the detrimental role of overweight/obesity in cancer, with almost 55% of cancers diagnosed in women and 24% diagnosed in men considered overweight- and obesity-related cancers. Sufficient data have shown that higher BMI is associated with risk of gynecologic malignancies (mainly breast and endometrial cancers) as well as cancers in sites such as the esophagus (adenocarcinoma), gastric cardia, colon, rectum, liver, gallbladder, pancreas, kidney, thyroid gland, and multiple myeloma. The main mechanisms underlying this relationship include the insulin/IGF1 system, the effect of sex hormones, and adipocytokines. Marked differences may be seen in specific cancer sites when comparing men to women. There is a higher overall incidence of obesity-related cancers among females (endometrial, ovarian, and postmenopausal female breast cancers), whereas cancers that concern both sexes show a higher incidence in males, particularly esophageal adenocarcinoma (male to female ratio: 9: 1 in the USA). Additionally, bariatric surgery has provided evidence of lowering overall cancer risk in patients with morbid obesity. Interestingly, bariatric surgery may lower overall cancer risk in women within the first 5 years after surgery due to the reduced risk of breast and endometrial cancer, and non-Hodgkin lymphoma. Obesity constitutes the base for marked metabolic, hormonal, and inflammatory alterations, including increased cancer risk in both men and women. Implementation of early obesity prevention strategies could ameliorate the continuously increasing incidence of cancer attributed to obesity.
Collapse
Affiliation(s)
- Georgia Argyrakopoulou
- Diabetes and Obesity Unit, Athens Medical Center, Distomou 5-7, Amaroussio, 15127, Athens, Greece.
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527, Athens, Greece
| | - Nikolaos Spyrou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527, Athens, Greece
| | - Alexander Kokkinos
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko Hospital, 115 27, Athens, Greece
| |
Collapse
|
167
|
Guo L, Yang K, Zhou P, Yong W. Gut microbiota in obesity and nonalcoholic fatty liver disease. SURGERY IN PRACTICE AND SCIENCE 2021. [DOI: 10.1016/j.sipas.2021.100030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
168
|
Hoozemans J, de Brauw M, Nieuwdorp M, Gerdes V. Gut Microbiome and Metabolites in Patients with NAFLD and after Bariatric Surgery: A Comprehensive Review. Metabolites 2021; 11:353. [PMID: 34072995 PMCID: PMC8227414 DOI: 10.3390/metabo11060353] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022] Open
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) is increasing, as are other manifestations of metabolic syndrome such as obesity and type 2 diabetes. NAFLD is currently the number one cause of chronic liver disease worldwide. The pathophysiology of NAFLD and disease progression is poorly understood. A potential contributing role for gut microbiome and metabolites in NAFLD is proposed. Currently, bariatric surgery is an effective therapy to prevent the progression of NAFLD and other manifestations of metabolic syndrome such as obesity and type 2 diabetes. This review provides an overview of gut microbiome composition and related metabolites in individuals with NAFLD and after bariatric surgery. Causality remains to be proven. Furthermore, the clinical effects of bariatric surgery on NAFLD are illustrated. Whether the gut microbiome and metabolites contribute to the metabolic improvement and improvement of NAFLD seen after bariatric surgery has not yet been proven. Future microbiome and metabolome research is necessary for elucidating the pathophysiology and underlying metabolic pathways and phenotypes and providing better methods for diagnostics, prognostics and surveillance to optimize clinical care.
Collapse
Affiliation(s)
- Jacqueline Hoozemans
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, AMC, 1105 AZ Amsterdam, The Netherlands; (M.N.); (V.G.)
- Department of Bariatric and General Surgery, Spaarne Hospital, 2134 TM Hoofddorp, The Netherlands;
| | - Maurits de Brauw
- Department of Bariatric and General Surgery, Spaarne Hospital, 2134 TM Hoofddorp, The Netherlands;
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, AMC, 1105 AZ Amsterdam, The Netherlands; (M.N.); (V.G.)
| | - Victor Gerdes
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, AMC, 1105 AZ Amsterdam, The Netherlands; (M.N.); (V.G.)
- Department of Internal Medicine, Spaarne Hospital, 2134 TM Hoofddorp, The Netherlands
| |
Collapse
|
169
|
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
|
170
|
Role of the Endocannabinoid System in the Adipose Tissue with Focus on Energy Metabolism. Cells 2021; 10:cells10061279. [PMID: 34064024 PMCID: PMC8224009 DOI: 10.3390/cells10061279] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/12/2021] [Accepted: 05/15/2021] [Indexed: 12/15/2022] Open
Abstract
The endocannabinoid system is involved in a wide range of processes including the control of energy acquisition and expenditure. Endocannabinoids and their receptors are present in the central nervous system but also in peripheral tissues, notably the adipose tissues. The endocannabinoid system interacts with two main hormones regulating appetite, namely leptin and ghrelin. The inhibitory effect of the cannabinoid receptor 1 (CB1) antagonist rimonabant on fat mass suggested that the endocannabinoid system can also have a peripheral action in addition to its effect on appetite reduction. Thus, several investigations have focused on the peripheral role of the endocannabinoid system in the regulation of metabolism. The white adipose tissue stores energy as triglycerides while the brown adipose tissue helps to dissipate energy as heat. The endocannabinoid system regulates several functions of the adipose tissues to favor energy accumulation. In this review we will describe the presence of the endocannabinoid system in the adipose tissue. We will survey the role of the endocannabinoid system in the regulation of white and brown adipose tissue metabolism and how the eCB system participates in obesity and metabolic diseases.
Collapse
|
171
|
Jie Z, Yu X, Liu Y, Sun L, Chen P, Ding Q, Gao Y, Zhang X, Yu M, Liu Y, Zhang Y, Kristiansen K, Jia H, Brix S, Cai K. The Baseline Gut Microbiota Directs Dieting-Induced Weight Loss Trajectories. Gastroenterology 2021; 160:2029-2042.e16. [PMID: 33482223 DOI: 10.1053/j.gastro.2021.01.029] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 12/07/2020] [Accepted: 01/09/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND & AIMS Elucidating key factors affecting personal responses to food is the first step toward implementing personalized nutrition strategies in for example weight loss programs. Here, we aimed to identify factors of importance for individual weight loss trajectories in a natural setting where participants were provided dietary advice but otherwise asked to self-manage the daily caloric intake and data reporting. METHODS A 6-month weight-reduction program with longitudinal collection of dietary, physical activity, body weight, and fecal microbiome data as well as single-nucleotide polymorphism genotypes in 83 participants was conducted, followed by integration of the high-dimensional data to define the most determining factors for weight loss in a dietician-guided, smartphone-assisted dieting program. RESULTS The baseline gut microbiota was found to outperform other factors as a predieting predictor of individual weight loss trajectories. Weight loss was also linked to the magnitude of changes in abundances of certain bacterial species during dieting. Ruminococcus gnavus (MGS0160) was significantly enriched in obese individuals and decreased during weight loss. Akkermansia muciniphila (MGS0120) and Alistipes obesi (MGS0342) were significantly enriched in lean individuals, and their abundance increased during dieting. Finally, Blautia wexlerae (MGS0575) and Bacteroides dorei (MGS0187) were the strongest predictors for weight loss when present in high abundance at baseline. CONCLUSION Altogether, the baseline gut microbiota was found to excel as a central personal factor in capturing the relationship between dietary factors and weight loss among individuals on a dieting program.
Collapse
Affiliation(s)
- Zhuye Jie
- BGI-Shenzhen, Shenzhen, China; China National Genebank, Shenzhen, China; Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, Shenzhen, China
| | - Xinlei Yu
- BGI-Shenzhen, Shenzhen, China; China National Genebank, Shenzhen, China
| | - Yinghua Liu
- Chinese PLA General Hospital, Beijing, China
| | - Lijun Sun
- BGI-Shenzhen, Shenzhen, China; China National Genebank, Shenzhen, China
| | - Peishan Chen
- BGI-Shenzhen, Shenzhen, China; China National Genebank, Shenzhen, China; Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, Shenzhen, China
| | - Qiuxia Ding
- BGI-Shenzhen, Shenzhen, China; China National Genebank, Shenzhen, China; Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, Shenzhen, China
| | - Yuan Gao
- BGI-Shenzhen, Shenzhen, China; China National Genebank, Shenzhen, China
| | | | | | | | - Yong Zhang
- Chinese PLA General Hospital, Beijing, China
| | - Karsten Kristiansen
- BGI-Shenzhen, Shenzhen, China; China National Genebank, Shenzhen, China; Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Qingdao-Europe Advanced Institute for Life Sciences, Qingdao, Shandong 266555, China
| | - Huijue Jia
- BGI-Shenzhen, Shenzhen, China; China National Genebank, Shenzhen, China; Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, Shenzhen, China
| | - Susanne Brix
- Qingdao-Europe Advanced Institute for Life Sciences, Qingdao, Shandong 266555, China; Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark.
| | - Kaiye Cai
- BGI-Shenzhen, Shenzhen, China; China National Genebank, Shenzhen, China; Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, Shenzhen, China.
| |
Collapse
|
172
|
Hrncir T, Hrncirova L, Kverka M, Hromadka R, Machova V, Trckova E, Kostovcikova K, Kralickova P, Krejsek J, Tlaskalova-Hogenova H. Gut Microbiota and NAFLD: Pathogenetic Mechanisms, Microbiota Signatures, and Therapeutic Interventions. Microorganisms 2021; 9:microorganisms9050957. [PMID: 33946843 PMCID: PMC8146698 DOI: 10.3390/microorganisms9050957] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease. Its worldwide prevalence is rapidly increasing and is currently estimated at 24%. NAFLD is highly associated with many features of the metabolic syndrome, including obesity, insulin resistance, hyperlipidaemia, and hypertension. The pathogenesis of NAFLD is complex and not fully understood, but there is increasing evidence that the gut microbiota is strongly implicated in the development of NAFLD. In this review, we discuss the major factors that induce dysbiosis of the gut microbiota and disrupt intestinal permeability, as well as possible mechanisms leading to the development of NAFLD. We also discuss the most consistent NAFLD-associated gut microbiota signatures and immunological mechanisms involved in maintaining the gut barrier and liver tolerance to gut-derived factors. Gut-derived factors, including microbial, dietary, and host-derived factors involved in NAFLD pathogenesis, are discussed in detail. Finally, we review currently available diagnostic and prognostic methods, summarise latest knowledge on promising microbiota-based biomarkers, and discuss therapeutic strategies to manipulate the microbiota, including faecal microbiota transplantation, probiotics and prebiotics, deletions of individual strains with bacteriophages, and blocking the production of harmful metabolites.
Collapse
Affiliation(s)
- Tomas Hrncir
- Czech Academy of Sciences, Institute of Microbiology, 142 20 Prague, Czech Republic; (L.H.); (M.K.); (V.M.); (E.T.); (K.K.); (H.T.-H.)
- Correspondence:
| | - Lucia Hrncirova
- Czech Academy of Sciences, Institute of Microbiology, 142 20 Prague, Czech Republic; (L.H.); (M.K.); (V.M.); (E.T.); (K.K.); (H.T.-H.)
- The Faculty of Medicine in Hradec Kralove, Charles University in Prague, 500 03 Hradec Kralove, Czech Republic; (P.K.); (J.K.)
| | - Miloslav Kverka
- Czech Academy of Sciences, Institute of Microbiology, 142 20 Prague, Czech Republic; (L.H.); (M.K.); (V.M.); (E.T.); (K.K.); (H.T.-H.)
| | - Robert Hromadka
- NEXARS (C2P), The Campus Science Park, 625 00 Brno, Czech Republic;
| | - Vladimira Machova
- Czech Academy of Sciences, Institute of Microbiology, 142 20 Prague, Czech Republic; (L.H.); (M.K.); (V.M.); (E.T.); (K.K.); (H.T.-H.)
| | - Eva Trckova
- Czech Academy of Sciences, Institute of Microbiology, 142 20 Prague, Czech Republic; (L.H.); (M.K.); (V.M.); (E.T.); (K.K.); (H.T.-H.)
| | - Klara Kostovcikova
- Czech Academy of Sciences, Institute of Microbiology, 142 20 Prague, Czech Republic; (L.H.); (M.K.); (V.M.); (E.T.); (K.K.); (H.T.-H.)
| | - Pavlina Kralickova
- The Faculty of Medicine in Hradec Kralove, Charles University in Prague, 500 03 Hradec Kralove, Czech Republic; (P.K.); (J.K.)
| | - Jan Krejsek
- The Faculty of Medicine in Hradec Kralove, Charles University in Prague, 500 03 Hradec Kralove, Czech Republic; (P.K.); (J.K.)
| | - Helena Tlaskalova-Hogenova
- Czech Academy of Sciences, Institute of Microbiology, 142 20 Prague, Czech Republic; (L.H.); (M.K.); (V.M.); (E.T.); (K.K.); (H.T.-H.)
| |
Collapse
|
173
|
Cardoneanu A, Cozma S, Rezus C, Petrariu F, Burlui AM, Rezus E. Characteristics of the intestinal microbiome in ankylosing spondylitis. Exp Ther Med 2021; 22:676. [PMID: 33986841 PMCID: PMC8112129 DOI: 10.3892/etm.2021.10108] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 03/26/2021] [Indexed: 12/16/2022] Open
Abstract
The importance of intestinal microbiota in the development of various systemic diseases has been highlighted over time. Ankylosing spondylitis (AS) is a systemic disease with a complex pathogenesis involving a particular genetic marker and distinctive environmental triggers such as a specific gut dysbiosis. We conducted a prospective case-control study which included 60 subjects from Iasi Rehabilitation Hospital: 28 AS cases and 32 healthy controls. Intestinal microbiota analysis was performed by real-time polymerase chain reaction (qPCR) in stool samples. We performed the quantitative analysis of gut microbiome, focusing both on anti-inflammatory (Bifidobacterium, Lactobacillus, Faecalibacterium prausnitzii) and pro-inflammatory (Bacteroides, Escherichia coli) species. Overall, intestinal bacterial diversity in the AS group was decreased compared to that noted in the control. A significantly decreased level of Clostridium leptum was observed, associated with an increased level of Escherichia coli. We showed correlations between laboratory tests (liver and kidney functional tests, inflammatory syndrome), the presence of HLA-B27, smoker status, the forms of AS with peripheral arthritis vs. pure axial forms and bacterial structures. No significant correlations were shown for disease activity scores, radiological stage of sacroiliitis or for body mass index. Our findings support that the intestinal microbiome in AS patients has a special signature characterized by an inflammatory status. Numerous environmental, genetical, clinical and paraclinical factors can lead to changes in gut bacterial diversity in these cases.
Collapse
Affiliation(s)
- Anca Cardoneanu
- Department of Rheumatology and Physiotherapy, Grigore T Popa University of Medicine and Pharmacy, Faculty of Medicine, 700115 Iasi, Romania
| | - Sebastian Cozma
- Department of Surgery (II), Grigore T Popa University of Medicine and Pharmacy, Faculty of Medicine, 700115 Iasi, Romania
| | - Ciprian Rezus
- Department of Internal Medicine, Grigore T Popa University of Medicine and Pharmacy, Faculty of Medicine, 700115 Iasi, Romania
| | - Florin Petrariu
- Department of Preventive Medicine and Interdisciplinarity, Grigore T Popa University of Medicine and Pharmacy, Faculty of Medicine, 700115 Iasi, Romania
| | - Alexandra Maria Burlui
- Department of Rheumatology and Physiotherapy, Grigore T Popa University of Medicine and Pharmacy, Faculty of Medicine, 700115 Iasi, Romania
| | - Elena Rezus
- Department of Rheumatology and Physiotherapy, Grigore T Popa University of Medicine and Pharmacy, Faculty of Medicine, 700115 Iasi, Romania
| |
Collapse
|
174
|
Legoux F, Salou M, Lantz O. MAIT Cell Development and Functions: the Microbial Connection. Immunity 2021; 53:710-723. [PMID: 33053329 DOI: 10.1016/j.immuni.2020.09.009] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/21/2020] [Accepted: 09/14/2020] [Indexed: 12/24/2022]
Abstract
Mucosal-associated invariant T (MAIT) cells are an evolutionarily conserved T cell subset, which reacts to most bacteria through T cell receptor (TCR)-mediated recognition of metabolites derived from the vitamin B2 biosynthetic pathway. Microbiota-derived signals affect all stages of MAIT cell biology including intra-thymic development, peripheral expansion, and functions in specific organs. In tissues, MAIT cells can integrate multiple signals and display effector functions involved in the defense against infectious pathogens. In addition to anti-bacterial activity, MAIT cells improve wound healing in the skin, suggesting a role in epithelium homeostasis through bi-directional interactions with the local microbiota. In humans, blood MAIT cell frequency is modified during several auto-immune diseases, which are often associated with microbiota dysbiosis, further emphasizing the potential interplay of MAIT cells with the microbiota. Here, we will review how microbes interact with MAIT cells, from initial intra-thymic development to tissue colonization and functions.
Collapse
Affiliation(s)
- François Legoux
- INSERM U932, PSL University, Institut Curie, Paris, 75005, France
| | - Marion Salou
- INSERM U932, PSL University, Institut Curie, Paris, 75005, France
| | - Olivier Lantz
- INSERM U932, PSL University, Institut Curie, Paris, 75005, France; Laboratoire d'immunologie clinique, Institut Curie, Paris, 75005, France; Centre d'investigation Clinique en Biothérapie, Institut Curie (CIC-BT1428), Paris, 75005, France.
| |
Collapse
|
175
|
Martín-Mateos R, Albillos A. The Role of the Gut-Liver Axis in Metabolic Dysfunction-Associated Fatty Liver Disease. Front Immunol 2021; 12:660179. [PMID: 33936094 PMCID: PMC8085382 DOI: 10.3389/fimmu.2021.660179] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
The complex interplay between the gut microbiota, the intestinal barrier, the immune system and the liver is strongly influenced by environmental and genetic factors that can disrupt the homeostasis leading to disease. Among the modulable factors, diet has been identified as a key regulator of microbiota composition in patients with metabolic syndrome and related diseases, including the metabolic dysfunction-associated fatty liver disease (MAFLD). The altered microbiota disrupts the intestinal barrier at different levels inducing functional and structural changes at the mucus lining, the intercellular junctions on the epithelial layer, or at the recently characterized vascular barrier. Barrier disruption leads to an increased gut permeability to bacteria and derived products which challenge the immune system and promote inflammation. All these alterations contribute to the pathogenesis of MAFLD, and thus, therapeutic approaches targeting the gut-liver-axis are increasingly being explored. In addition, the specific changes induced in the intestinal flora may allow to characterize distinctive microbial signatures for non-invasive diagnosis, severity stratification and disease monitoring.
Collapse
Affiliation(s)
| | - Agustín Albillos
- Department of Gastroenterology and Hepatology, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
176
|
Westerink F, Huibregtse I, De Hoog M, Bruin S, Meesters E, Brandjes D, Gerdes V. Faecal Inflammatory Biomarkers and Gastrointestinal Symptoms after Bariatric Surgery: A Longitudinal Study. Inflamm Intest Dis 2021; 6:109-116. [PMID: 34124182 DOI: 10.1159/000514576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/29/2020] [Indexed: 11/19/2022] Open
Abstract
Background Bariatric surgery induces various gastrointestinal (GI) modifications. We performed the first study longitudinally assessing the effect of bariatric surgery on faecal inflammatory biomarker levels and its relation with GI complaints. Method Faecal calprotectin, lactoferrin, and calgranulin-C levels were determined in 41 patients (34 Roux-en-Y [RYGB], 7 sleeves) before and at 6-16 weeks, 6 months, and 1 year after surgery. Changes in biomarker levels and percentage of patients above reference value were determined. Gastrointestinal symptom rating scale (GSRS) was used to assess GI complaints at corresponding time points. The postoperative relation between GSRS score and biomarker levels above reference value was investigated. Results After RYGB, median calprotectin levels are significantly higher (>188, 104-415 μg/g) than before surgery (40, 19-78 μg/g; p < 0.001), and over 90% of patients have levels above reference value 1 year after surgery. Median lactoferrin was 0.4 (0.2-1.6) μg/g before, and >5.9 (1.8-13.6) μg/g after surgery (p < 0.001). Median calgranulin-C levels remained far below the reference value and were 0.13 (0.05-0.24) μg/g before and <0.23 (0.06-0.33) μg/g after surgery. Similar results were found after sleeve gastrectomy. No difference was seen in GSRS score for patients with calprotectin and lactoferrin levels above reference values. Conclusion Faecal inflammatory biomarkers calprotectin and lactoferrin, but not calgranulin-C, rise above reference values shortly after bariatric surgery and remain elevated in the majority of patients. The discrepancy between calprotectin and calgranulin-C levels suggests no GI inflammation. Furthermore, patients after RYGB with biomarkers above the population reference value do not seem to have more GI complaints.
Collapse
Affiliation(s)
- Floris Westerink
- Department of internal medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Inge Huibregtse
- Department of gastroenterology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | | | - Sjoerd Bruin
- Department of surgery, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - Eelco Meesters
- Department of internal medicine, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - Desiderius Brandjes
- Department of internal medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Victor Gerdes
- Department of internal medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands.,Department of internal medicine, Spaarne Gasthuis, Hoofddorp, The Netherlands
| |
Collapse
|
177
|
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
|
178
|
El-Mallah CA, Beyh YS, Obeid OA. Iron Fortification and Supplementation: Fighting Anemia of Chronic Diseases or Fueling Obesity? Curr Dev Nutr 2021; 5:nzab032. [PMID: 33959691 PMCID: PMC8085477 DOI: 10.1093/cdn/nzab032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/07/2021] [Accepted: 03/31/2021] [Indexed: 12/13/2022] Open
Abstract
The significant worldwide increase in obesity has become a major health problem. Excess adiposity has been extensively linked to inflammation. Recently, studies have shown that dietary intake and microbiota dysbiosis can affect the health of the gut and lead to low-grade systemic inflammation, worsening the state of obesity and further exacerbating inflammation. The latter is shown to decrease iron status and potentially increase the risk of anemia by inhibiting iron absorption. Hence, anemia of obesity is independent of iron intake and does not properly respond to increased iron ingestion. Therefore, countries with a high rate of obesity should assess the health impact of fortification and supplementation with iron due to their potential drawbacks. This review tries to elucidate the relation between inflammation and iron status to better understand the etiology of anemia of obesity and chronic diseases and wisely design any dietary or medical interventions for the management of anemia and/or obesity.
Collapse
Affiliation(s)
- Carla A El-Mallah
- Department of Nutrition and Food Science, Faculty of Agricultural and Food Sciences, American University of Beirut, Beirut, Lebanon
| | - Yara S Beyh
- Nutrition and Health Sciences, Laney Graduate School, Emory University, Atlanta, GA, USA
| | - Omar A Obeid
- Department of Nutrition and Food Science, Faculty of Agricultural and Food Sciences, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
179
|
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
|
180
|
Tian B, Zhao J, Zhang M, Chen Z, Ma Q, Liu H, Nie C, Zhang Z, An W, Li J. Lycium ruthenicum Anthocyanins Attenuate High-Fat Diet-Induced Colonic Barrier Dysfunction and Inflammation in Mice by Modulating the Gut Microbiota. Mol Nutr Food Res 2021; 65:e2000745. [PMID: 33629483 DOI: 10.1002/mnfr.202000745] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 01/15/2021] [Indexed: 12/16/2022]
Abstract
SCOPE Gut barrier dysfunction and inflammation originating from a dysbiotic gut microbiota (GM) are strongly associated with a high-fat diet (HFD). Anthocyanins from Lycium ruthenicum (ACs) show antiobesity effects through modulating the GM. However, the mechanism linking the antiobesity effects of ACs and GM modulation remains obscure. METHODS AND RESULTS To investigate the ameliorative effects of ACs on colonic barrier dysfunction and inflammation, mice are fed an HFD with or without ACs at doses of 50, 100, and 200 mg kg-1 for 12 weeks. AC supplementation reduced weight gain, enriched short-chain fatty acid (SCFA)-producing bacteria (e.g., Ruminococcaceae, Muribaculaceae, Akkermansia, Ruminococcaceae_UCG-014, and Bacteroides) and SCFA content, depleted endotoxin-producing bacteria (e.g., Helicobacter and Desulfovibrionaceae), and decreased endotoxin (i.e., lipopolysaccharide) levels. SCFAs substantially activated G protein-coupled receptors (GPRs), inhibited histone deacetylases (HDAC), increased intestinal tight junction mRNA and protein expression levels, reduced intestinal permeability, and protected intestinal barrier integrity in HFD-induced mice. These effects mitigate intestinal inflammation by inhibiting the LPS/NF-κB/TLR4 pathway. CONCLUSION These data indicates that ACs can mitigate colonic barrier dysfunction and inflammation, induce SCFA production and inhibit endotoxin production by modulating the GM in HFD-fed mice. This finding provides a clue for understanding the antiobesity effects of ACs.
Collapse
Affiliation(s)
- Baoming Tian
- College of Food Science and Engineering, Northwest A&F University, Yangling, P. R. China
- Institute of Wolfberry Engineering Technology Research, Ningxia Academy of Agricultural and Forestry Sciences, Yinchuan, P. R. China
- National Wolfberry Engineering Research Center, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan, P. R. China
| | - Jianhua Zhao
- Institute of Wolfberry Engineering Technology Research, Ningxia Academy of Agricultural and Forestry Sciences, Yinchuan, P. R. China
- National Wolfberry Engineering Research Center, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan, P. R. China
| | - Min Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling, P. R. China
| | - Zhifei Chen
- College of Food Science and Engineering, Northwest A&F University, Yangling, P. R. China
| | - Qingyu Ma
- College of Food Science and Engineering, Northwest A&F University, Yangling, P. R. China
| | - Huicui Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, P. R. China
| | - Chenxi Nie
- College of Food Science and Engineering, Northwest A&F University, Yangling, P. R. China
| | - Ziqi Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling, P. R. China
| | - Wei An
- Institute of Wolfberry Engineering Technology Research, Ningxia Academy of Agricultural and Forestry Sciences, Yinchuan, P. R. China
- National Wolfberry Engineering Research Center, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan, P. R. China
| | - Juxiu Li
- College of Food Science and Engineering, Northwest A&F University, Yangling, P. R. China
| |
Collapse
|
181
|
Chen M, Zheng J, Zou X, Ye C, Xia H, Yang M, Gao Q, Yang Q, Liu H. Ligustrum robustum (Roxb.) blume extract modulates gut microbiota and prevents metabolic syndrome in high-fat diet-fed mice. JOURNAL OF ETHNOPHARMACOLOGY 2021; 268:113695. [PMID: 33316365 DOI: 10.1016/j.jep.2020.113695] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In Chinese folk medicine, Ligustrum robustum (Roxb.) Blume has been widely used as a healthy tea beverage for improvement in obesity and lipidemic metabolic disorders. AIM OF THE STUDY We aimed to investigate the effect of L. robustum extract (LRE) on metabolic syndrome in high-fat diet (HFD)-fed mice and to explore the underlying role of gut microbiota during the treatment. MATERIALS AND METHODS The ground dried leaves of L. robustum (Roxb.) Blume were extracted with ethanol and then purified by a resin column. The composition of L. robustum extract (LRE) was analyzed by high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). C57BL/6 J mice fed with HFD were treated with LRE for 16 weeks. RT-qPCR and morphological staining were utilized to reveal the impact of LRE on hepatic glucolipid metabolism and gut integrity. The next-generation sequencing of 16 S rDNA was applied for analyzing the gut microbial community of fecal samples. RESULTS LRE, mainly composed of ligupurpuroside A and aceteoside, alleviated insulin resistance, improved hepatic metabolism, enhanced intestinal integrity, and suppressed inflammatory responses in HFD-fed mice. Moreover, LRE treatment reshaped the gut microbiota structure by increasing the levels of genera Streptococcus, Lactobacillus, and Mucispirillum and decreasing the populations of Alistipes and Lachnospiraceae NK4A136 group in HFD-fed mice. The alteration of gut microbiota was associated with several metabolic pathways of gut bacteria. Spearman's correlation analysis further confirmed the links between the changed intestinal bacteria and multiple disease indices. CONCLUSIONS LRE prevented gut microbiota dysbiosis and metabolic disorder in HFD-fed mice, which helps to promote the application in LRE-mediated prevention from metabolic syndrome as a gut microbial regulator.
Collapse
Affiliation(s)
- Man Chen
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, PR China
| | - Junping Zheng
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, PR China
| | - Xiaojuan Zou
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, PR China
| | - Cheng Ye
- Wuhan Customs Technology Center, Qintai Avenue 588, Wuhan, 430050, PR China
| | - Hui Xia
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, PR China
| | - Ming Yang
- State Engineering Technology Institute for Karst Desertification Control, School of Karst Science, Guizhou Normal University, Guiyang, 550001, PR China
| | - Qinghua Gao
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, PR China
| | - Qingxiong Yang
- State Engineering Technology Institute for Karst Desertification Control, School of Karst Science, Guizhou Normal University, Guiyang, 550001, PR China.
| | - Hongtao Liu
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, PR China; Chongqing Academy of Chinese Materia Medica, Nanshan Road 34, Chongqing, 400065, PR China.
| |
Collapse
|
182
|
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
|
183
|
Deutsch L, Stres B. The Importance of Objective Stool Classification in Fecal 1H-NMR Metabolomics: Exponential Increase in Stool Crosslinking Is Mirrored in Systemic Inflammation and Associated to Fecal Acetate and Methionine. Metabolites 2021; 11:172. [PMID: 33809780 PMCID: PMC8002301 DOI: 10.3390/metabo11030172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/10/2021] [Accepted: 03/10/2021] [Indexed: 12/25/2022] Open
Abstract
Past studies strongly connected stool consistency-as measured by Bristol Stool Scale (BSS)-with microbial gene richness and intestinal inflammation, colonic transit time and metabolome characteristics that are of clinical relevance in numerous gastro intestinal conditions. While retention time, defecation rate, BSS but not water activity have been shown to account for BSS-associated inflammatory effects, the potential correlation with the strength of a gel in the context of intestinal forces, abrasion, mucus imprinting, fecal pore clogging remains unexplored as a shaping factor for intestinal inflammation and has yet to be determined. Our study introduced a minimal pressure approach (MP) by probe indentation as measure of stool material crosslinking in fecal samples. Results reported here were obtained from 170 samples collected in two independent projects, including males and females, covering a wide span of moisture contents and BSS. MP values increased exponentially with increasing consistency (i.e., lower BSS) and enabled stratification of samples exhibiting mixed BSS classes. A trade-off between lowest MP and highest dry matter content delineated the span of intermediate healthy density of gel crosslinks. The crossectional transects identified fecal surface layers with exceptionally high MP and of <5 mm thickness followed by internal structures with an order of magnitude lower MP, characteristic of healthy stool consistency. The MP and BSS values reported in this study were coupled to reanalysis of the PlanHab data and fecal 1H-NMR metabolomes reported before. The exponential association between stool consistency and MP determined in this study was mirrored in the elevated intestinal and also systemic inflammation and other detrimental physiological deconditioning effects observed in the PlanHab participants reported before. The MP approach described in this study can be used to better understand fecal hardness and its relationships to human health as it provides a simple, fine scale and objective stool classification approach for the characterization of the exact sampling locations in future microbiome and metabolome studies.
Collapse
Affiliation(s)
- Leon Deutsch
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, SI-1000 Ljubljana, Slovenia;
| | - Blaz Stres
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, SI-1000 Ljubljana, Slovenia;
- Faculty of Civil and Geodetic Engineering, University of Ljubljana, Jamova 2, SI-1000 Ljubljana, Slovenia
- Department of Automation, Jožef Stefan Institute, Biocybernetics and Robotics, Jamova 39, SI-1000 Ljubljana, Slovenia
- Department of Microbiology, University of Innsbruck, Technikerstrasse 25d, A-6020 Innsbruck, Austria
| |
Collapse
|
184
|
Lüll K, Arffman RK, Sola-Leyva A, Molina NM, Aasmets O, Herzig KH, Plaza-Díaz J, Franks S, Morin-Papunen L, Tapanainen JS, Salumets A, Altmäe S, Piltonen TT, Org E. The Gut Microbiome in Polycystic Ovary Syndrome and Its Association with Metabolic Traits. J Clin Endocrinol Metab 2021; 106:858-871. [PMID: 33205157 DOI: 10.1210/clinem/dgaa848] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Indexed: 12/11/2022]
Abstract
CONTEXT Despite the gut microbiome being widely studied in metabolic diseases, its role in polycystic ovary syndrome (PCOS) has been scarcely investigated. OBJECTIVE Compare the gut microbiome in late fertile age women with and without PCOS and investigate whether changes in the gut microbiome correlate with PCOS-related metabolic parameters. DESIGN Prospective, case-control study using the Northern Finland Birth Cohort 1966. SETTING General community. PARTICIPANTS A total of 102 PCOS women and 201 age- and body mass index (BMI)-matched non-PCOS control women. Clinical and biochemical characteristics of the participants were assessed at ages 31 and 46 and analyzed in the context of gut microbiome data at the age of 46. INTERVENTION (s): None. MAIN OUTCOME MEASURE(S) Bacterial diversity, relative abundance, and correlations with PCOS-related metabolic measures. RESULTS Bacterial diversity indices did not differ significantly between PCOS and controls (Shannon diversity P = .979, unweighted UniFrac P = .175). Four genera whose balance helps to differentiate between PCOS and non-PCOS were identified. In the whole cohort, the abundance of 2 genera from Clostridiales, Ruminococcaceae UCG-002, and Clostridiales Family XIII AD3011 group, were correlated with several PCOS-related markers. Prediabetic PCOS women had significantly lower alpha diversity (Shannon diversity P = .018) and markedly increased abundance of genus Dorea (false discovery rate = 0.03) compared with women with normal glucose tolerance. CONCLUSION PCOS and non-PCOS women at late fertile age with similar BMI do not significantly differ in their gut microbial profiles. However, there are significant microbial changes in PCOS individuals depending on their metabolic health.
Collapse
Affiliation(s)
- Kreete Lüll
- Institute of Genomics, Estonian Genome Centre, University of Tartu, Tartu, Estonia
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Riikka K Arffman
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Centre, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Alberto Sola-Leyva
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs GRANADA, Granada, Spain
| | - Nerea M Molina
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs GRANADA, Granada, Spain
| | - Oliver Aasmets
- Institute of Genomics, Estonian Genome Centre, University of Tartu, Tartu, Estonia
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Karl-Heinz Herzig
- Research Unit of Biomedicine, Medical Research Center, University of Oulu, Oulu University Hospital, Oulu, Finland
- Department of Paediatric Gastroenterology and Metabolic Diseases, Poznań University of Medical Sciences, Poznań, Poland
| | - Julio Plaza-Díaz
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Stephen Franks
- Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Laure Morin-Papunen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Centre, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Juha S Tapanainen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Centre, Oulu University Hospital, University of Oulu, Oulu, Finland
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Andres Salumets
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Signe Altmäe
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs GRANADA, Granada, Spain
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Terhi T Piltonen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Centre, Oulu University Hospital, University of Oulu, Oulu, Finland
- Department of Obstetrics and Gynecology, Reproductive Endocrinology and IVF Unit, Oulu, University Hospital, University of Oulu, Oulu, Finland
| | - Elin Org
- Institute of Genomics, Estonian Genome Centre, University of Tartu, Tartu, Estonia
| |
Collapse
|
185
|
Cambi MPC, Baretta GAP, Magro DDO, Boguszewski CL, Ribeiro IB, Jirapinyo P, de Moura DTH. Multidisciplinary Approach for Weight Regain-how to Manage this Challenging Condition: an Expert Review. Obes Surg 2021; 31:1290-1303. [PMID: 33392999 DOI: 10.1007/s11695-020-05164-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022]
Abstract
Weight regain is a multifactorial condition that affects many patients following bariatric surgery. The purpose of the paper is to review the multidisciplinary approach for the management of weight regain. We performed a search in current clinical evidence regarding the causes, consequences, and treatments of weight regain. The multidisciplinary approach with periodic monitoring is of fundamental importance to prevent or treat weight regain. Several therapeutic options are ranging from nutritional to surgical options, which should be tailored according to patients' anatomy, lifestyle behavior, and compliance. Specialized multidisciplinary care is the key to achieve optimal long-term weight loss and maintenance goals following bariatric surgery.
Collapse
Affiliation(s)
| | | | - Daniéla De Oliveira Magro
- Department of Surgery, Faculty of Medical Sciences, State University of Campinas UNICAMP-SP, Campinas Sao Paulo, Brazil
| | | | - Igor Braga Ribeiro
- Gastrointestinal Endoscopy Unit, Hospital das Clínicas, University of Sao Paulo School of Medicine, Av. Dr Eneas de Carvalho Aguiar, 225, 6o andar, bloco 3, Cerqueira Cesar, Sao Paulo, 05403-010, Brazil.
| | - Pichamol Jirapinyo
- Division of Gasteoenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Diogo Turiani Hourneaux de Moura
- Gastrointestinal Endoscopy Unit, Hospital das Clínicas, University of Sao Paulo School of Medicine, Av. Dr Eneas de Carvalho Aguiar, 225, 6o andar, bloco 3, Cerqueira Cesar, Sao Paulo, 05403-010, Brazil
- Division of Gasteoenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
186
|
Tan C, Zheng Z, Wan X, Cao J, Wei R, Duan J. The role of gut microbiota and amino metabolism in the effects of improvement of islet β-cell function after modified jejunoileal bypass. Sci Rep 2021; 11:4809. [PMID: 33637880 PMCID: PMC7910448 DOI: 10.1038/s41598-021-84355-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 02/15/2021] [Indexed: 12/23/2022] Open
Abstract
The change in gut microbiota is an important mechanism of the amelioration of type 2 diabetes mellitus (T2DM) after bariatric surgery. Here, we observe that the modified jejunoileal bypass effectively decreases body weight gain, fasting blood glucose, and lipids level in serum; additionally, islet β-cell function, glucose tolerance, and insulin resistance were markedly ameliorated. The hypoglycemic effect and the improvement in islet β-cell function depend on the changes in gut microbiota structure. modified jejunoileal bypass increases the abundance of gut Escherichia coli and Ruminococcus gnavus and the levels of serum glycine, histidine, and glutamine in T2DM rats; and decreases the abundance of Prevotella copri and the levels of serum branched chain amino acids, which are significantly related to the improvement of islet β-cell function in T2DM rats. Our results suggest that amino acid metabolism may contribute to the islet β-cell function in T2DM rats after modified jejunoileal bypass and that improving gut microbiota composition is a potential therapeutic strategy for T2DM.
Collapse
Affiliation(s)
- Cai Tan
- Department of Women's Health, Jiangxi Maternal and Child Health Hospital, Nanchang, 330006, China
| | - Zhihua Zheng
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xiaogang Wan
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jiaqing Cao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Ran Wei
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Jinyuan Duan
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Gastrointestinal Surgical Institute of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
187
|
Nogacka AM, de Los Reyes-Gavilán CG, Arboleya S, Ruas-Madiedo P, Martínez-Faedo C, Suarez A, He F, Harata G, Endo A, Salazar N, Gueimonde M. In vitro Selection of Probiotics for Microbiota Modulation in Normal-Weight and Severely Obese Individuals: Focus on Gas Production and Interaction With Intestinal Epithelial Cells. Front Microbiol 2021; 12:630572. [PMID: 33633711 PMCID: PMC7899977 DOI: 10.3389/fmicb.2021.630572] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/18/2021] [Indexed: 12/18/2022] Open
Abstract
The intestinal microbiota plays important roles in the maintenance of health. Strategies aiming at its modulation, such as probiotics, have received a deal of attention. Several strains have been studied in different in vitro models; however, the correlation of results obtained with the in vivo data has been limited. This questions the usefulness of such in vitro selection models, traditionally relying on over-simplified tests, not considering the influence of the accompanying microbiota or focusing on microbiota composition without considering functional traits. Here we assess the potential of six Bifidobacterium, Lactobacillus and Lacticaseibacillus strains in an in vitro model to determine their impact on the microbiota not just in terms of composition but also of functionality. Moreover, we compared the responses obtained in two different population groups: normal-weight and severely obese subjects. Fecal cultures were conducted to evaluate the impact of the strains on specific intestinal microbial groups, on the production of short-chain fatty acids, and on two functional responses: the production of gas and the interaction with human intestinal epithelial cells. The response to the different probiotics differed between both human groups. The addition of the probiotic strains did not induce major changes on the microbiota composition, with significant increases detected almost exclusively for the species added. Higher levels of gas production were observed in cultures from normal-weight subjects than in the obese population, with some strains being able to significantly reduce gas production in the latter group. Moreover, in obese subjects all the Bifidobacterium strains tested and Lacticaseibacillus rhamnosus GG were able to modify the response of the intestinal cells, restoring values similar to those obtained with the microbiotas of normal-weight subjects. Our results underline the need for the screening and selection of probiotics in a target-population specific manner by using appropriate in vitro models before enrolling in clinical intervention trials.
Collapse
Affiliation(s)
- Alicja Maria Nogacka
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Villaviciosa, Spain.,Diet, Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), Oviedo, Spain
| | - Clara G de Los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Villaviciosa, Spain.,Diet, Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), Oviedo, Spain
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Villaviciosa, Spain.,Diet, Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), Oviedo, Spain
| | - Patricia Ruas-Madiedo
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Villaviciosa, Spain.,Functionality and Ecology of Beneficial Microorganisms, Institute of Health Research of the Principality of Asturias (ISPA), Oviedo, Spain
| | - Ceferino Martínez-Faedo
- Endocrinology and Nutrition Service, Central University Hospital of Asturias (HUCA), Oviedo, Spain.,Endocrinology, Nutrition, Diabetes and Obesity Group, Institute of Health Research of the Principality of Asturias (ISPA), Oviedo, Spain
| | - Adolfo Suarez
- Diet, Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), Oviedo, Spain.,Digestive Service, Central University Hospital of Asturias (HUCA), Oviedo, Spain
| | - Fang He
- Technical Research Laboratory, Takanashi Milk Products Co., Ltd., Yokohama, Japan
| | - Gaku Harata
- Technical Research Laboratory, Takanashi Milk Products Co., Ltd., Yokohama, Japan
| | - Akihito Endo
- Department of Food and Cosmetic Science, Tokyo University of Agriculture, Abashiri, Japan
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Villaviciosa, Spain.,Diet, Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), Oviedo, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Villaviciosa, Spain.,Diet, Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), Oviedo, Spain
| |
Collapse
|
188
|
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: 406] [Impact Index Per Article: 101.5] [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
|
189
|
Schmidt L, Medawar E, Aron-Wisnewsky J, Genser L, Poitou C, Clément K, Plassmann H. Resting-state connectivity within the brain's reward system predicts weight loss and correlates with leptin. Brain Commun 2021; 3:fcab005. [PMID: 33615220 PMCID: PMC7884604 DOI: 10.1093/braincomms/fcab005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/12/2020] [Accepted: 11/30/2020] [Indexed: 12/19/2022] Open
Abstract
Weight gain is often associated with the pleasure of eating food rich in calories. This idea is based on the findings that people with obesity showed increased neural activity in the reward and motivation systems of the brain in response to food cues. Such correlations, however, overlook the possibility that obesity may be associated with a metabolic state that impacts the functioning of reward and motivation systems, which in turn could be linked to reactivity to food and eating behaviour and weight gain. In a study involving 44 female participants [14 patients with obesity, aged 20–63 years (mean: 42, SEM: 3.2 years), and 30 matched lean controls, aged 22–60 years (mean: 37, SEM: 1.8 years)], we investigated how ventromedial prefrontal cortex seed-to-voxel resting-state connectivity distinguished between lean and obese participants at baseline. We used the results of this first step of our analyses to examine whether changes in ventromedial prefrontal cortex resting-state connectivity over 8 months could formally predict weight gain or loss. It is important to note that participants with obesity underwent bariatric surgery at the beginning of our investigation period. We found that ventromedial prefrontal cortex–ventral striatum resting-state connectivity and ventromedial–dorsolateral prefrontal cortex resting-state connectivity were sensitive to obesity at baseline. However, only the ventromedial prefrontal cortex–ventral striatum resting-state connectivity predicted weight changes over time using cross-validation, out-of-sample prediction analysis. Such an out-of-sample prediction analysis uses the data of all participants of a training set to predict the actually observed data in one independent participant in the hold-out validation sample and is then repeated for all participants. In seeking to explain the reason why ventromedial pre-frontal cortex–ventral striatum resting-state connectivity as the central hub of the brain’s reward and motivational system may predict weight change over time, we linked weight loss surgery-induced changes in ventromedial prefrontal cortex–ventral striatum resting-state connectivity to surgery-induced changes in homeostatic hormone regulation. More specifically, we focussed on changes in fasting state systemic leptin, a homeostatic hormone signalling satiety, and inhibiting reward-related dopamine signalling. We found that the surgery-induced increase in ventromedial prefrontal cortex–ventral striatum resting-state connectivity was correlated with a decrease in fasting-state systemic leptin. These findings establish the first link between individual differences in brain connectivity in reward circuits in a more tonic state at rest, weight change over time and homeostatic hormone regulation.
Collapse
Affiliation(s)
- Liane Schmidt
- Control-Interoception-Attention Team, Institut du Cerveau et de la Moelle épinière (ICM), Inserm UMR 1127, CNRS UMR 7225, Sorbonne Université, 75013 Paris, France
| | - Evelyn Medawar
- Laboratoire de Neuroscience Cognitive, Ecole Normale Supérieure, Inserm U960, 75005 Paris, France
| | - Judith Aron-Wisnewsky
- Sorbonne Université, Inserm, UMRS Nutrition et Obésités; Systemic Approaches (NutriOmics), 75013 Paris, France.,Nutrition Department, CRNH Ile de France, Pitié-Salpêtrière Hospital, Assistance Publique Hôpitaux de Paris, 75013 Paris, France
| | - Laurent Genser
- Visceral Surgery Department, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, 75013 Paris, France
| | - Christine Poitou
- Sorbonne Université, Inserm, UMRS Nutrition et Obésités; Systemic Approaches (NutriOmics), 75013 Paris, France
| | - Karine Clément
- Sorbonne Université, Inserm, UMRS Nutrition et Obésités; Systemic Approaches (NutriOmics), 75013 Paris, France
| | - Hilke Plassmann
- Control-Interoception-Attention Team, Institut du Cerveau et de la Moelle épinière (ICM), Inserm UMR 1127, CNRS UMR 7225, Sorbonne Université, 75013 Paris, France.,Marketing Area, INSEAD 77305, Fontainebleau, France
| |
Collapse
|
190
|
Santos JM, Mathew MS, Shah N, Pajuelo-Vasquez R, Mistry AM, Heindl SE. Pre and Post-Operative Alterations of the Gastrointestinal Microbiome Following Bariatric Surgery. Cureus 2021; 13:e13057. [PMID: 33680599 PMCID: PMC7929544 DOI: 10.7759/cureus.13057] [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] [Indexed: 11/22/2022] Open
Abstract
Obesity in the United States is increasing at a startling rate, with more individuals turning towards bariatric surgery as treatment. A noteworthy aspect of obesity pathology is its interplay with the gastrointestinal microbiome. The gastrointestinal microbiome comprising trillions of microorganisms affects the dynamics of digestion, energy expenditure, and neurologic mechanisms that affect dietary preference. This literature review used PubMed to search for articles about obesity, gastrointestinal microbiome, and bariatric surgery. The researchers used Medical Subject Heading keywords, and then the relevant literature was selected and filtered using inclusion and exclusion criteria. This study aims to review the temporal relationship of gastrointestinal microbiome changes after bariatric surgery in association with the success and failure of treatment along with the factors that may have altered the gastrointestinal microbiome other than the anatomical aspect of bariatric surgery.
Collapse
Affiliation(s)
- James M Santos
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Meby S Mathew
- Pharmacy Practice, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Pharmacy Practice, Nirmala College of Pharmacy, Muvattupuzha, IND
| | - Nilam Shah
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Renzo Pajuelo-Vasquez
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Anuja Mahesh Mistry
- Neurology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Internal Medicine, Smt N.H.L Municipal Medical College, Ahmedabad, IND
| | - Stacey E Heindl
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Medicine, Avalon University School of Medicine, Willemstad, CUW
| |
Collapse
|
191
|
van den Broek M, de Heide LJM, Sips FLP, Koehorst M, van Zutphen T, Emous M, van Faassen M, Groen AK, van Riel NAW, de Boer JF, van Beek AP, Kuipers F. Altered bile acid kinetics contribute to postprandial hypoglycaemia after Roux-en-Y gastric bypass surgery. Int J Obes (Lond) 2021; 45:619-630. [PMID: 33452416 DOI: 10.1038/s41366-020-00726-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/16/2020] [Accepted: 12/03/2020] [Indexed: 01/13/2023]
Abstract
BACKGROUND/OBJECTIVES Bile acids (BA) act as detergents in intestinal fat absorption and as modulators of metabolic processes via activation of receptors such as FXR and TGR5. Elevated plasma BA as well as increased intestinal BA signalling to promote GLP-1 release have been implicated in beneficial health effects of Roux-en-Y gastric bypass surgery (RYGB). Whether BA also contribute to the postprandial hypoglycaemia that is frequently observed post-RYGB is unknown. METHODS Plasma BA, fibroblast growth factor 19 (FGF19), 7α-hydroxy-4-cholesten-3-one (C4), GLP-1, insulin and glucose levels were determined during 3.5 h mixed-meal tolerance tests (MMTT) in subjects after RYGB, either with (RYGB, n = 11) or without a functioning gallbladder due to cholecystectomy (RYGB-CC, n = 11). Basal values were compared to those of age, BMI and sex-matched obese controls without RYGB (n = 22). RESULTS Fasting BA as well as FGF19 levels were elevated in RYGB and RYGB-CC subjects compared to non-bariatric controls, without significant differences between RYGB and RYGB-CC. Postprandial hypoglycaemia was observed in 8/11 RYGB-CC and only in 3/11 RYGB. Subjects who developed hypoglycaemia showed higher postprandial BA levels coinciding with augmented GLP-1 and insulin responses during the MMTT. The nadir of plasma glucose concentrations after meals showed a negative relationship with postprandial BA peaks. Plasma C4 was lower during MMTT in subjects experiencing hypoglycaemia, indicating lower hepatic BA synthesis. Computer simulations revealed that altered intestinal transit underlies the occurrence of exaggerated postprandial BA responses in hypoglycaemic subjects. CONCLUSION Altered BA kinetics upon ingestion of a meal, as frequently observed in RYGB-CC subjects, appear to contribute to postprandial hypoglycaemia by stimulating intestinal GLP-1 release.
Collapse
Affiliation(s)
- Merel van den Broek
- Center for Obesity North Netherlands (CON), Department of Surgery, MCL, Leeuwarden, The Netherlands.,Faculty Campus Fryslân, University of Groningen, Leeuwarden, The Netherlands
| | - Loek J M de Heide
- Center for Obesity North Netherlands (CON), Department of Surgery, MCL, Leeuwarden, The Netherlands
| | - Fianne L P Sips
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Martijn Koehorst
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Tim van Zutphen
- Faculty Campus Fryslân, University of Groningen, Leeuwarden, The Netherlands.,Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marloes Emous
- Center for Obesity North Netherlands (CON), Department of Surgery, MCL, Leeuwarden, The Netherlands
| | - Martijn van Faassen
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Albert K Groen
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Vascular Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Natal A W van Riel
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.,Department of Vascular Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Jan F de Boer
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - André P van Beek
- Center for Obesity North Netherlands (CON), Department of Surgery, MCL, Leeuwarden, The Netherlands.,Department of Endocrinology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Folkert Kuipers
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands. .,Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
192
|
Cho KY. Lifestyle modifications result in alterations in the gut microbiota in obese children. BMC Microbiol 2021; 21:10. [PMID: 33407104 PMCID: PMC7789654 DOI: 10.1186/s12866-020-02002-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023] Open
Abstract
Background The association between the gut microbiota and pediatric obesity was analyzed in a cross-sectional study. A prospective study of obese children was conducted to assess the gut microbial alterations after a weight change. We collected fecal samples from obese children before and after a 2-month weight reduction program that consisted of individual counseling for nutritional education and physical activity, and we performed 16S rRNA gene amplicon sequencing using an Illumina MiSeq platform. Results Thirty-six participants, aged 7 to 18 years, were classified into the fat loss (n = 17) and the fat gain (n = 19) groups according to the change in total body fat (%) after the intervention. The baseline analysis of the gut microbiota in the preintervention stages showed dysbiotic features of both groups compared with those of normal-weight children. In the fat loss group, significantly decreased proportions of Bacteroidetes phylum, Bacteroidia class, Bacteroidales order, Bacteroidaceae family, and Bacteroides genus, along with increased proportions of Firmicutes phylum, Clostridia class, and Clostridiales order, were observed after intervention. The microbial richness was significantly reduced, without a change in beta diversity in the fat loss group. The fat gain group showed significantly deceased proportions of Firmicutes phylum, Clostridia class, Clostridiales order, Lachnospiraceae family, and Eubacterium hallii group genus, without a change in diversity after the intervention. According to the functional metabolic analysis by the Phylogenetic Investigation of Communities by Reconstruction of Unobserved States 2, the “Nitrate Reduction VI” and “Aspartate Superpathway” pathways were predicted to increase significantly in the fat loss group. The cooccurring networks of genera were constructed and showed the different microbes that drove the changes between the pre- and postintervention stages in the fat loss and fat gain groups. Conclusions This study demonstrated that lifestyle modifications can impact the composition, richness, and predicted functional profiles of the gut microbiota in obese children after weight changes. Trial registration ClinicalTrials.govNCT03812497, registration date January 23, 2019, retrospectively registered. Supplementary information Supplementary information accompanies this paper at 10.1186/s12866-020-02002-3.
Collapse
Affiliation(s)
- Ky Young Cho
- Department of Pediatrics, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, South Korea.
| |
Collapse
|
193
|
Martin-Nuñez GM, Cornejo-Pareja I, Clemente-Postigo M, Tinahones FJ. Gut Microbiota: The Missing Link Between Helicobacter pylori Infection and Metabolic Disorders? Front Endocrinol (Lausanne) 2021; 12:639856. [PMID: 34220702 PMCID: PMC8247771 DOI: 10.3389/fendo.2021.639856] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/17/2021] [Indexed: 12/14/2022] Open
Abstract
Helicobacter pylori (H. pylori) is a gram-negative bacterium that infects approximately 4.4 billion individuals worldwide. Although the majority of infected individuals remain asymptomatic, this bacterium colonizes the gastric mucosa causing the development of various clinical conditions as peptic ulcers, chronic gastritis and gastric adenocarcinomas and mucosa-associated lymphoid tissue lymphomas, but complications are not limited to gastric ones. Extradigestive pathologies, including metabolic disturbances such as diabetes, obesity and nonalcoholic fatty liver disease, have also been associated with H. pylori infection. However, the underlying mechanisms connecting H. pylori with extragastric metabolic diseases needs to be clarified. Notably, the latest studies on the topic have confirmed that H. pylori infection modulates gut microbiota in humans. Damage in the gut bacterial community (dysbiosis) has been widely related to metabolic dysregulation by affecting adiposity, host energy balance, carbohydrate metabolism, and hormonal modulation, among others. Taking into account that Type 2 diabetic patients are more prone to be H. pylori positive, gut microbiota emerges as putative key factor responsible for this interaction. In this regard, the therapy of choice for H. pylori eradication, based on proton pump inhibitor combined with two or more antibiotics, also alters gut microbiota composition, but consequences on metabolic health of the patients has been scarcely explored. Recent studies from our group showed that, despite decreasing gut bacterial diversity, conventional H. pylori eradication therapy is related to positive changes in glucose and lipid profiles. The mechanistic insights explaining these effects should also be addressed in future research. This review will deal with the role of gut microbiota as the linking factor between H. pylori infection and metabolic diseases, and discussed the impact that gut bacterial modulation by H. pylori eradication treatment can also have in host's metabolism. For this purpose, new evidence from the latest human studies published in more recent years will be analyzed.
Collapse
Affiliation(s)
- Gracia M. Martin-Nuñez
- Unidad de Gestión Clínica de Endocrinología y Nutrición (Hospital Universitario Virgen de la Victoria), Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Isabel Cornejo-Pareja
- Unidad de Gestión Clínica de Endocrinología y Nutrición (Hospital Universitario Virgen de la Victoria), Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Mercedes Clemente-Postigo
- Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Cell Biology, Physiology and Immunology. Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)-Reina Sofia University Hospital, University of Cordoba, Córdoba, Spain
- *Correspondence: Francisco J. Tinahones, ; Mercedes Clemente-Postigo,
| | - Francisco J. Tinahones
- Unidad de Gestión Clínica de Endocrinología y Nutrición (Hospital Universitario Virgen de la Victoria), Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- *Correspondence: Francisco J. Tinahones, ; Mercedes Clemente-Postigo,
| |
Collapse
|
194
|
Fouladi F, Carroll IM, Sharpton TJ, Bulik-Sullivan E, Heinberg L, Steffen KJ, Fodor AA. A microbial signature following bariatric surgery is robustly consistent across multiple cohorts. Gut Microbes 2021; 13:1930872. [PMID: 34159880 PMCID: PMC8224199 DOI: 10.1080/19490976.2021.1930872] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/28/2021] [Accepted: 05/05/2021] [Indexed: 02/07/2023] Open
Abstract
Bariatric surgery induces significant shifts in the gut microbiota which could potentially contribute to weight loss and metabolic benefits. The aim of this study was to characterize a microbial signature following Roux-en-Y Gastric bypass (RYGB) surgery using novel and existing gut microbiota sequence data. We generated 16S rRNA gene and metagenomic sequences from fecal samples from patients undergoing RYGB surgery (n = 61 for 16S rRNA gene and n = 135 for metagenomics) at pre-surgical baseline and one, six, and twelve-month post-surgery. We compared these data with three smaller publicly available 16S rRNA gene and one metagenomic datasets from patients who also underwent RYGB surgery. Linear mixed models and machine learning approaches were used to examine the presence of a common microbial signature across studies. Comparison of our new sequences with previous longitudinal studies revealed strikingly similar profiles in both fecal microbiota composition (r = 0.41 ± 0.10; p < .05) and metabolic pathways (r = 0.70 ± 0.05; p < .001) early after surgery across multiple datasets. Notably, Veillonella, Streptococcus, Gemella, Fusobacterium, Escherichia/Shigella, and Akkermansia increased after surgery, while Blautia decreased. Machine learning approaches revealed that the replicable gut microbiota signature associated with RYGB surgery could be used to discriminate pre- and post-surgical samples. Opportunistic pathogen abundance also increased post-surgery in a consistent manner across cohorts. Our study reveals a robust microbial signature involving many commensal and pathogenic taxa and metabolic pathways early after RYGB surgery across different studies and sites. Characterization of the effects of this robust microbial signature on outcomes of bariatric surgery could provide insights into the development of microbiome-based interventions for predicting or improving outcomes following surgery.
Collapse
Affiliation(s)
- Farnaz Fouladi
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, USA
| | - Ian M. Carroll
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, USA
- Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Thomas J. Sharpton
- Department of Microbiology, Department of Statistics, Center for Genome Research and Biocomputing, Oregon State University, Corvallis, USA
| | - Emily Bulik-Sullivan
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Leslie Heinberg
- Department of Psychiatry and Psychology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, USA
| | - Kristine J. Steffen
- School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, USA
- Director of Biomedical Research, Center for Biobehavioral Research/Sanford Research, Fargo, USA
| | - Anthony A. Fodor
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, USA
| |
Collapse
|
195
|
Nieuwdorp M, Madsen K. The Promise of Maintaining Diet-Induced Weight Loss by Swallowing One's Own Feces: Time to Provide a Do-It-Yourself Manual? Gastroenterology 2021; 160:17-19. [PMID: 33091412 DOI: 10.1053/j.gastro.2020.10.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 10/15/2020] [Indexed: 12/02/2022]
Affiliation(s)
- Max Nieuwdorp
- Amsterdam Diabetes Center, Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands.
| | - Karen Madsen
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
196
|
Jia R, Huang M, Qian L, Yan X, Lv Q, Ye H, Ye L, Wu X, Chen W, Chen Y, Jia Y, Huang Y, Wu H. The Depletion of Carbohydrate Metabolic Genes in the Gut Microbiome Contributes to the Transition From Central Obesity to Type 2 Diabetes. Front Endocrinol (Lausanne) 2021; 12:747646. [PMID: 34745012 PMCID: PMC8569854 DOI: 10.3389/fendo.2021.747646] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/21/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity, especially central obesity, is a strong risk factor for developing type 2 diabetes (T2D). However, the mechanism underlying the progression from central obesity to T2D remains unknown. Therefore, we analyzed the gut microbial profiles of central obese individuals with or without T2D from a Chinese population. Here we reported both the microbial compositional and gene functional alterations during the progression from central obesity to T2D. Several opportunistic pathogens were enriched in obese T2D patients. We also characterized thousands of genes involved in sugar and amino acid metabolism whose abundance were significantly depleted in obese T2D group. Moreover, the abundance of those genes was negatively associated with plasma glycemia level and percentage of individuals with impaired plasma glucose status. Therefore, our study indicates that the abundance of those depleted genes can be used as a potential biomarker to identify central obese individuals with high risks of developing T2D.
Collapse
MESH Headings
- Adult
- Biomarkers/metabolism
- Carbohydrate Metabolism/genetics
- Case-Control Studies
- China
- Diabetes Mellitus, Type 2/etiology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/microbiology
- Disease Progression
- Disease Susceptibility
- Female
- Gastrointestinal Microbiome/genetics
- Humans
- Male
- Metagenome/physiology
- Obesity, Abdominal/genetics
- Obesity, Abdominal/metabolism
- Obesity, Abdominal/microbiology
- Obesity, Abdominal/pathology
- RNA, Ribosomal, 16S/analysis
- RNA, Ribosomal, 16S/genetics
- Risk Factors
- Transcriptome
Collapse
Affiliation(s)
| | - Min Huang
- Department of General Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | | | - Xiaoye Yan
- Department of General Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Qing Lv
- Department of General Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Hua Ye
- GENEWIZ Inc., Suzhou, China
| | - Li Ye
- GENEWIZ Inc., Suzhou, China
| | - Xin Wu
- GENEWIZ Inc., Suzhou, China
| | | | | | | | - Yueqing Huang
- Department of General Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- *Correspondence: Yueqing Huang, ; Huihui Wu,
| | - Huihui Wu
- GENEWIZ Inc., Suzhou, China
- *Correspondence: Yueqing Huang, ; Huihui Wu,
| |
Collapse
|
197
|
A body weight loss- and health-promoting gut microbiota is established after bariatric surgery in individuals with severe obesity. J Pharm Biomed Anal 2021; 193:113747. [DOI: 10.1016/j.jpba.2020.113747] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/24/2020] [Accepted: 10/31/2020] [Indexed: 02/07/2023]
|
198
|
Aron-Wisnewsky J, Warmbrunn MV, Nieuwdorp M, Clément K. Metabolism and Metabolic Disorders and the Microbiome: The Intestinal Microbiota Associated With Obesity, Lipid Metabolism, and Metabolic Health-Pathophysiology and Therapeutic Strategies. Gastroenterology 2021; 160:573-599. [PMID: 33253685 DOI: 10.1053/j.gastro.2020.10.057] [Citation(s) in RCA: 219] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/26/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022]
Abstract
Changes in the intestinal microbiome have been associated with obesity and type 2 diabetes, in epidemiological studies and studies of the effects of fecal transfer in germ-free mice. We review the mechanisms by which alterations in the intestinal microbiome contribute to development of metabolic diseases, and recent advances, such as the effects of the microbiome on lipid metabolism. Strategies have been developed to modify the intestinal microbiome and reverse metabolic alterations, which might be used as therapies. We discuss approaches that have shown effects in mouse models of obesity and metabolic disorders, and how these might be translated to humans to improve metabolic health.
Collapse
Affiliation(s)
- Judith Aron-Wisnewsky
- Nutrition and Obesities: Systemic Approaches Research Unit (Nutriomics), Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Paris, France; Nutrition Department, Assistante Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Centres de Recherche en Nutrition Humaine Ile de France, Paris, France; Department of Vascular Medicine, Amsterdam Universitair Medische Centra, location Academisch Medisch Centrum, and VUMC, University of Amsterdam, Amsterdam, the Netherlands.
| | - Moritz V Warmbrunn
- Department of Vascular Medicine, Amsterdam Universitair Medische Centra, location Academisch Medisch Centrum, and VUMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, Amsterdam Universitair Medische Centra, location Academisch Medisch Centrum, and VUMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Karine Clément
- Nutrition and Obesities: Systemic Approaches Research Unit (Nutriomics), Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Paris, France; Nutrition Department, Assistante Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Centres de Recherche en Nutrition Humaine Ile de France, Paris, France.
| |
Collapse
|
199
|
Meijnikman AS, Aydin O, Prodan A, Tremaroli V, Herrema H, Levin E, Acherman Y, Bruin S, Gerdes VE, Backhed F, Groen AK, Nieuwdorp M. Distinct differences in gut microbial composition and functional potential from lean to morbidly obese subjects. J Intern Med 2020; 288:699-710. [PMID: 32633011 DOI: 10.1111/joim.13137] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/20/2020] [Accepted: 05/14/2020] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The gut microbiome may contribute to the development of obesity. So far, the extent of microbiome variation in people with obesity has not been determined in large cohorts and for a wide range of body mass index (BMI). Here, we aimed to investigate whether the faecal microbial metagenome can explain the variance in several clinical phenotypes associated with morbid obesity. METHODS Caucasian subjects were recruited at our hospital. Blood pressure and anthropometric measurements were taken. Dietary intake was determined using questionnaires. Shotgun metagenomic sequencing was performed on faecal samples from 177 subjects. RESULTS Subjects without obesity (n = 82, BMI 24.7 ± 2.9 kg m-2 ) and subjects with obesity (n = 95, BMI 38.6 ± 5.1 kg m-2 ) could be clearly distinguished based on microbial composition and microbial metabolic pathways. A total number of 52 bacterial species differed significantly in people with and without obesity. Independent of dietary intake, we found that microbial pathways involved in biosynthesis of amino acids were enriched in subjects with obesity, whereas pathways involved in the degradation of amino acids were depleted. Machine learning models showed that more than half of the variance in body fat composition followed by BMI could be explained by the gut microbiome, composition and microbial metabolic pathways, compared with 6% of variation explained in triglycerides and 9% in HDL. CONCLUSION Based on the faecal microbiota composition, we were able to separate subjects with and without obesity. In addition, we found strong associations between gut microbial amino acid metabolism and specific microbial species in relation to clinical features of obesity.
Collapse
Affiliation(s)
- A S Meijnikman
- From the, Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands.,Department of Surgery, Spaarne Hospital, Hoofddorp, the Netherlands
| | - O Aydin
- From the, Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands.,Department of Surgery, Spaarne Hospital, Hoofddorp, the Netherlands
| | - A Prodan
- From the, Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - V Tremaroli
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, Goteborgs Universitet, Gothenburg, Sweden
| | - H Herrema
- From the, Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - E Levin
- From the, Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - Y Acherman
- Department of Surgery, Spaarne Hospital, Hoofddorp, the Netherlands
| | - S Bruin
- Department of Surgery, Spaarne Hospital, Hoofddorp, the Netherlands
| | - V E Gerdes
- From the, Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands.,Department of Surgery, Spaarne Hospital, Hoofddorp, the Netherlands
| | - F Backhed
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, Goteborgs Universitet, Gothenburg, Sweden.,Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Physiology, Region Västtra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - A K Groen
- From the, Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - M Nieuwdorp
- From the, Department of Internal and Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
200
|
Westerink F, Beijderwellen H, Huibregtse IL, De Hoog MLA, De Brauw LM, Brandjes DPM, Gerdes VEA. Lactose after Roux-en-Y gastric bypass for morbid obesity, is it a problem? Scand J Gastroenterol 2020; 55:1398-1404. [PMID: 33096008 DOI: 10.1080/00365521.2020.1837927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Roux-en-Y gastric bypass (RYGB) can cause multiple food intolerances and gastrointestinal complaints are frequently reported after dairy consumption. We aimed to determine the prevalence of lactose malabsorption and intolerance, and complaints associated with dairy consumption in daily life, before and after RYGB. METHOD The lactose breath test (LBT) and lactose tolerance test (LTT) was performed in 84 patients awaiting RYGB surgery and 84 patients after surgery. Gastrointestinal symptoms at baseline and after testing were recorded. Lactose malabsorption was defined as a positive LBT and/or LTT. Lactose intolerance as a positive test combined with an increase of gastrointestinal complains. Dairy consumption in daily life and successive gastrointestinal complaints were registered via a questionnaire. Results of preoperative and postoperative patients were compared. RESULTS Lactose malabsorption was present in 15 (17.9%) of the preoperative patients and in 25 (29.8%) of the postoperative patients (OR 2.46; 95%CI: 1.08-5.59; p = .03). Of the preoperative patients 6 (7.1%) patients met the criteria for lactose intolerance, compared to 8 (9.5%) patients in the postoperative group (OR 1.48; 95%CI 0.48-4.57; p = .50). Twelve (14.3%) preoperative patients indicated to have gastrointestinal complaints after dairy consumption in daily life versus 45 (53.6%) postoperative patients (p < .01). CONCLUSION This study shows no increase in patients with proven lactose intolerance after RYGB compared to preoperative patients. Gastrointestinal complaints after dairy consumption in daily life were far more frequently reported by RYGB patients. It is unlikely that all reported gastrointestinal complaints are actually caused by lactose. Other ingredients in dairy, like fat, are possibly contributory.
Collapse
Affiliation(s)
- F Westerink
- Department of Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands.,Department of Internal Medicine, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - H Beijderwellen
- Department of Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - I L Huibregtse
- Department of Gastroenterology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - M L A De Hoog
- Spaarne Gasthuis Academy, Hoofddorp, The Netherlands
| | - L M De Brauw
- Department of Surgery, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - D P M Brandjes
- Department of Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - V E A Gerdes
- Department of Vascular Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands.,Department of Internal Medicine, Spaarne Gasthuis, Hoofddorp, The Netherlands
| |
Collapse
|