1
|
Nie P, Hu L, Feng X, Xu H. Gut Microbiota Disorders and Metabolic Syndrome: Tales of a Crosstalk Process. Nutr Rev 2025; 83:908-924. [PMID: 39504479 DOI: 10.1093/nutrit/nuae157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024] Open
Abstract
The microbiota in humans consists of trillions of microorganisms that are involved in the regulation of the gastrointestinal tract and immune and metabolic homeostasis. The gut microbiota (GM) has a prominent impact on the pathogenesis of metabolic syndrome (MetS). This process is reciprocal, constituting a crosstalk process between the GM and MetS. In this review, GM directly or indirectly inducing MetS via the host-microbial metabolic axis has been systematically reviewed. Additionally, the specifically altered GM in MetS are detailed in this review. Moreover, short-chain fatty acids (SCFAs), as unique gut microbial metabolites, have a remarkable effect on MetS, and the role of SCFAs in MetS-related diseases is highlighted to supplement the gaps in this area. Finally, the existing therapeutics are outlined, and the superiority and shortcomings of different therapeutic approaches are discussed, in hopes that this review can contribute to the development of potential treatment strategies.
Collapse
Affiliation(s)
- Penghui Nie
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Liehai Hu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Xiaoyan Feng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
- International Institute of Food Innovation Co., Ltd, Nanchang University, Nanchang 330200, China
| |
Collapse
|
2
|
Mohammadzadeh N, Razavi S, Shahriari M, Ebrahimipour G. Impact of bariatric surgery on gut microbiota in obese patients: A systematic review. Indian J Gastroenterol 2025:10.1007/s12664-025-01763-x. [PMID: 40220249 DOI: 10.1007/s12664-025-01763-x] [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: 10/30/2024] [Accepted: 02/25/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Obesity is a multi-factorial disease linked to various metabolic disorders, including insulin resistance, type-2 diabetes (T2D) and cardiovascular diseases. Traditional treatments often show limited long-term success, while bariatric surgery has emerged as the most effective intervention for sustained weight loss and comorbidity improvement. Alterations in gut microbiota may significantly contribute to these metabolic improvements. OBJECTIVE This systematic review was aimed at evaluating changes in gut microbiota composition before and after bariatric surgery and their association with clinical outcomes, including weight loss, insulin sensitivity and lipid metabolism. METHODS Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, a comprehensive search of PubMed, Scopus, Web of Science and clinicaltrials.gov databases was conducted for studies published between 2004 and 2024. Keywords included "bariatric surgery," "gut microbiota" and "obesity." Inclusion criteria focused on human studies with pre and post-surgical microbiota analysis. Non-human studies, pediatric populations and studies without microbiota assessment were excluded. Data extraction covered microbiota profiles, metabolic outcomes and clinical markers. RESULTS Total 27 articles and 28 clinical trials met the inclusion criteria. Pre-surgery, obese patients exhibited dysbiosis characterized by reduced microbial diversity and imbalances in key bacterial phyla. Post-surgery, especially after Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG), patients showed increased microbial diversity, reduced Firmicutes and elevated beneficial bacteria such as Akkermansia muciniphila and short-chain fatty acid (SCFA) producing bacteria. These microbiota changes were correlated with significant improvements in weight loss, insulin sensitivity and lipid profiles. However, some studies reported inconsistent or modest microbiota changes. CONCLUSION Bariatric surgery leads to significant gut microbiota alterations that are closely linked to metabolic improvements, including enhanced glucose control and lipid metabolism. However, the long-term sustainability of these microbial changes remains unclear. Longitudinal studies are essential to determine whether these alterations persist over time and how they continuously impact metabolic health. Further research should explore targeted microbiota interventions to maintain beneficial microbial profiles post-surgery.
Collapse
Affiliation(s)
- Nima Mohammadzadeh
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Shabnam Razavi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahla Shahriari
- Bacteriology Department, Faculty of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Gholamhossein Ebrahimipour
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
3
|
Gopalakrishnan V, Kumar C, Robertsen I, Morehouse C, Sparklin B, Khader S, Henry I, Johnson LK, Hertel JK, Christensen H, Sandbu R, Greasley PJ, Sellman BR, Åsberg A, Andersson S, Löfmark RJ, Hjelmesæth J, Karlsson C, Cohen TS. A multi-omics microbiome signature is associated with the benefits of gastric bypass surgery and is differentiated from diet induced weight loss through 2 years of follow-up. Mucosal Immunol 2025:S1933-0219(25)00040-6. [PMID: 40222615 DOI: 10.1016/j.mucimm.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/04/2025] [Accepted: 04/07/2025] [Indexed: 04/15/2025]
Abstract
Roux-en-Y gastric bypass (GBP) surgery is an effective treatment for reducing body weight and correcting metabolic dysfunction in individuals with severe obesity. Herein, we characterize the differences between very low energy diet (VLED) and GBP induced weight loss by multi-omic analyses of microbiome and host features in a non-randomized, controlled, single-center study. Eighty-eight participants with severe obesity were recruited into two arms - GBP versus VLED with matching weight loss for 6 weeks and 2-years of follow-up. A dramatic shift in the distribution of gut microbial taxa and their functional capacity was seen in the GBP group at Week 2 after surgery and was sustained through 2 years. Multi-omic analyses were performed after 6 weeks of matching weight loss between the GBP and VLED groups, which pointed to microbiome derived metabolites such as indoxyl sulphate as characterizing the GBP group. We also identified an inverse association between Streptococcus parasanguinis (an oral commensal) and plasma levels of tryptophan and tyrosine. These data have important implications, as they reveal a significant robust restructuring of the microbiome away from a baseline dysbiotic state in the GBP group. Furthermore, multi-omics modelling points to potentially novel mechanistic insights at the intersection of the microbiome and host.
Collapse
Affiliation(s)
| | - Chanchal Kumar
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Ida Robertsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, PO 1068 Blindern, 0316 Oslo, Norway
| | - Christopher Morehouse
- Discovery Microbiome, Early Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, USA
| | - Ben Sparklin
- Discovery Microbiome, Early Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, USA
| | - Shameer Khader
- Data Science and Artificial Intelligence, Biopharmaceuticals R&D, AstraZeneca, USA.
| | - Ian Henry
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Line Kristin Johnson
- Department of Endocrinology, Obesity and Nutrition, Vestfold Hospital Trust, P.O.Box 2168, 3103 Tønsberg, Norway
| | - Jens K Hertel
- Department of Endocrinology, Obesity and Nutrition, Vestfold Hospital Trust, P.O.Box 2168, 3103 Tønsberg, Norway
| | - Hege Christensen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, PO 1068 Blindern, 0316 Oslo, Norway
| | - Rune Sandbu
- Department of Endocrinology, Obesity and Nutrition, Vestfold Hospital Trust, P.O.Box 2168, 3103 Tønsberg, Norway
| | - Peter J Greasley
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Bret R Sellman
- Discovery Microbiome, Early Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, USA
| | - Anders Åsberg
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, PO 1068 Blindern, 0316 Oslo, Norway; Department of Transplantation Medicine, Oslo University Hospital, P.O.Box 4950 Nydalen 0424 Oslo, Norway
| | - Shalini Andersson
- Oligonucleotide Discovery, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Rasmus Jansson Löfmark
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jøran Hjelmesæth
- Department of Endocrinology, Obesity and Nutrition, Vestfold Hospital Trust, P.O.Box 2168, 3103 Tønsberg, Norway; Department of Endocrinology, Morbid Obesity and Preventive Medicine, Institute of Clinical Medicine, University of Oslo, P.O. Box 1171, 0318 Oslo, Norway
| | - Cecilia Karlsson
- Late-stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Taylor S Cohen
- Late Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, USA.
| |
Collapse
|
4
|
Cao Z, Mai W, Gan L, Huang L. A metabolomics and proteomics-based study on the metabolic effects of arecoline on the liver. Toxicon 2025; 260:108338. [PMID: 40216365 DOI: 10.1016/j.toxicon.2025.108338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/15/2025] [Accepted: 03/31/2025] [Indexed: 04/19/2025]
Abstract
Arecoline is one of the primary constituents of the areca nut. Its pharmacological effects include analgesia, anti-inflammation, and anti-allergy. Current researches on the toxicity of arecoline mainly focuse on oral carcinogenesis and immunotoxicity, so there are relatively little systematic study on its hepatotoxicity and underlying mechanisms. Therefore, this study aims to explore the mechanisms of hepatotoxicity induced by different doses of arecoline in mice by integrating metabolomics and proteomics. In our pathological results, we found that the medium and high dose groups of arecoline can cause fatty degeneration in the livers of mice. Additionally, the different doses of arecoline increased the levels of ALT and AST in the serum of mice. Proteomics research identified that exposure to different doses of arecoline primarily affected the PPARs signaling pathway, thereby influencing fatty acid metabolism, amino acid metabolism, and arachidonic acid metabolism pathways. Metabolomics research identified differential metabolites in each group after arecoline exposure. We observed that with increasing doses of arecoline, the metabolites of lipids and lipid-like molecules in mice gradually increased. The results suggested arecoline may induce fatty degeneration in the liver of mice through the PPARα/Acox-1 mediated pathways of oxidative stress, inflammatory response, energy, and lipid metabolism.
Collapse
Affiliation(s)
- Zhenjuan Cao
- School of Pharmacy, Hainan Medical University, Haikou, 571199, Hainan, China
| | - Wenzhi Mai
- School of Pharmacy, Hainan Medical University, Haikou, 571199, Hainan, China
| | - Lianfang Gan
- Research Center for Drug Safety Evaluation of Hainan Province, Haikou, China
| | - Ling Huang
- Center For Pharmacovigilance Of Hainan Province, Haikou, 570216, China.
| |
Collapse
|
5
|
Zhou H, Gizlenci M, Xiao Y, Martin F, Nakamori K, Zicari EM, Sato Y, Tullius SG. Obesity-associated Inflammation and Alloimmunity. Transplantation 2025; 109:588-596. [PMID: 39192462 PMCID: PMC11868468 DOI: 10.1097/tp.0000000000005183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Obesity is a worldwide health problem with a rapidly rising incidence. In organ transplantation, increasing numbers of patients with obesity accumulate on waiting lists and undergo surgery. Obesity is in general conceptualized as a chronic inflammatory disease, potentially impacting alloimmune response and graft function. Here, we summarize our current understanding of cellular and molecular mechanisms that control obesity-associated adipose tissue inflammation and provide insights into mechanisms affecting transplant outcomes, emphasizing on the beneficial effects of weight loss on alloimmune responses.
Collapse
Affiliation(s)
- Hao Zhou
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
| | - Merih Gizlenci
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Department of General, Visceral, Cancer and Transplant Surgery, University Hospital of Cologne, Cologne, Germany
| | - Yao Xiao
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
| | - Friederike Martin
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Department of Surgery, CVK/CCM, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Keita Nakamori
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Elizabeth M. Zicari
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Faculté de Pharmacie, Université Paris Cité, Paris, France
| | - Yuko Sato
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
| | - Stefan G. Tullius
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
| |
Collapse
|
6
|
Segrestin B, Cherifi BG. Obesity and its treatment: Toward new approaches. ANNALES D'ENDOCRINOLOGIE 2025; 86:101712. [PMID: 39961481 DOI: 10.1016/j.ando.2025.101712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/03/2025]
Abstract
Obesity has until now been defined in terms of body mass index, a parameter which lacks sensitivity. The definition rather needs to take account of the pathophysiology and impact of the mechanical, metabolic and psychological complications associated with excess adiposity, as proposed by the French Health Authority in 2022. Initial phenotyping of pathophysiological determinants and complications make it possible to precisely define treatment objectives and the means used to achieve them. For example, excess adiposity is associated with hypervolemia, which increases the risk of heart failure independently of classic cardiovascular risk factors, due in particular to myocardial remodeling and diastolic dysfunction. In this indication, SGLT2 (sodium/glucose cotransporter 2) inhibitors may be relevant. A global approach, involving assessment of eating behavior and sensations, energy expenditure and emotional eating, enables us to identify drug treatments suited to each profile. Obesity is a recurrent pathology requiring the development of new combined or sequential drug treatments. Currently under development, inhibitors of the myostatin/activin A pathway conserve lean body mass during weight loss, a crucial factor in limiting weight regain and maintaining functional capacity. Beyond medical or surgical approaches to obesity, lifelong lifestyle interventions are central to improving quality of life and limiting the risks associated with excess adiposity.
Collapse
Affiliation(s)
- Bérénice Segrestin
- F-CRIN - FORCE (French Obesity Research Centre of Excellence), CRNH, CARMEN lab INSERM U1060/University of Lyon/INRA U1235, Lyon1 University, Lyon, France; Endocrinology-Diabetes-Nutrition Department, hospices civils de Lyon, Lyon, France.
| | - Blandine Gatta Cherifi
- Service d'endocrinologie, diabétologie et maladies métaboliques, CRMR Pradort, CHU de Bordeaux, Bordeaux, France; Inserm U1215, "Physiopathologie de la balance enérgétique et obésité", Neurocentre Magendie, université de Bordeaux, Bordeaux, France; UFR des sciences médicales, université de Bordeaux, Bordeaux, France
| |
Collapse
|
7
|
Shi H, Li J. MAGs-based genomic comparison of gut significantly enriched microbes in obese individuals pre- and post-bariatric surgery across diverse locations. Front Cell Infect Microbiol 2025; 15:1485048. [PMID: 40171165 PMCID: PMC11958714 DOI: 10.3389/fcimb.2025.1485048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
Introduction Obesity, a pressing global health issue, is intricately associated with distinct gut microbiota profiles. Bariatric surgeries, such as Laparoscopic Sleeve Gastrectomy (LSG), Sleeve Gastrectomy (SG), and Roux-en-Y Gastric Bypass (RYGB), induce substantial weight loss and reshape gut microbiota composition and functionality, yet their comparative impacts remain underexplored. Methods This study integrated four published metagenomic datasets, encompassing 500 samples, and employed a unified bioinformatics workflow for analysis. We assessed gut microbiota α-diversity, identified species biomarkers using three differential analysis approaches, and constructed high-quality Metagenome-Assembled Genomes (MAGs). Comparative genomic, functional profiling and KEGG pathway analyses were performed, alongside estimation of microbial growth rates via Peak-to-Trough Ratios (PTRs). Results RYGB exhibited the most pronounced enhancement of gut microbiota α-diversity compared to LSG and SG. Cross-cohort analysis identified 39 species biomarkers: 27 enriched in the non-obesity group (NonOB_Enrich) and 12 in the obesity group (OB_Enrich). Among the MAGs, 177 were NonOB_Enrich and 14 were OB_Enrich. NonOB_Enrich MAGs displayed enriched carbohydrate degradation profiles (e.g., GH105, GH2, GH23, GH43, and GT0 families) and higher gene diversity in fatty acid biosynthesis and secondary metabolite pathways, alongside significant enrichment in amino acid metabolism (KEGG analysis). Post-surgery, Akkermansia muciniphila and Bacteroides uniformis showed elevated growth rates based on PTRs. Discussion These findings underscore RYGB's superior impact on gut microbiota diversity and highlight distinct microbial functional adaptations linked to weight loss, offering insights for targeted therapeutic strategies.
Collapse
Affiliation(s)
| | - Jia Li
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
8
|
Lazaro A, Tiago I, Mendes J, Ribeiro J, Bernardes A, Oliveira F, Regateiro F, Caramelo F, Silva H. Sleeve Gastrectomy and Gastric Bypass Impact in Patient's Metabolic, Gut Microbiome, and Immuno-inflammatory Profiles-A Comparative Study. Obes Surg 2025; 35:733-745. [PMID: 39870942 PMCID: PMC11906558 DOI: 10.1007/s11695-025-07708-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 08/20/2024] [Accepted: 01/18/2025] [Indexed: 01/29/2025]
Abstract
BACKGROUND Bariatric surgery is the most long-term effective treatment option for severe obesity. The role of gut microbiome (GM) in either the development of obesity or in response to obesity management strategies has been a matter of debate. This study aims to compare the impact of two of the most popular procedures, sleeve gastrectomy (SG) and Roux-en-Y gastric bypass (GB), on metabolic syndrome parameters and gut bacterial microbiome and in systemic immuno-inflammatory response. METHODS A prospective observational study enrolled 24 patients with severe obesity, 14 underwent SG and 10 GB. Evaluations before (0 M) and 6 months (6 M) after surgical procedures included clinical and biochemical parameters, expression of 17 immuno-inflammatory genes in peripheral blood leukocytes, and assessment of gut microbiome profile using 16 s rRNA next-generation sequencing approach. Statistical significance was set to a p value < 0.05 with an FDR < 0.1. RESULTS A significant and similar decrease in weight-associated parameters and for most metabolic markers was achieved with both surgeries. Considering the gut microbiome in the whole study population, there was an increase in alpha diversity at family-level taxa. Beta diversity between SG and GB at 6 M showed near significant differences (p = 0.042) at genus levels. Analysis of the relative abundance of individual taxonomic groups highlighted differences between pre- and post-surgical treatment and between both approaches, namely, a higher representation of family Enterobacteriaceae and genera Veillonella and Enterobacteriaceae_unclassified after GB. Increased expression of immune-inflammatory genes was observed mainly for SG patients. CONCLUSIONS We conclude that SG and GB have similar clinical and metabolic outcomes but different impacts in the gut bacterial microbiome. Results also suggest reactivation of immune response after bariatric surgery.
Collapse
Affiliation(s)
- Andre Lazaro
- General Surgery Unit, Centro Hospitalar E Universitário de Coimbra, ULS Coimbra, Coimbra, Portugal.
- Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| | - Igor Tiago
- Department of Life Sciences, Centre for Functional Ecology (CFE)-Science for People & the Planet, , University of Coimbra, Coimbra, Portugal
| | - Joao Mendes
- Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Laboratory of Sequencing and Functional Genomics of UC Genomics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Joana Ribeiro
- Laboratory of Sequencing and Functional Genomics of UC Genomics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Antonio Bernardes
- General Surgery Unit, Centro Hospitalar E Universitário de Coimbra, ULS Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Fernando Oliveira
- General Surgery Unit, Centro Hospitalar E Universitário de Coimbra, ULS Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Fernando Regateiro
- Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Laboratory of Sequencing and Functional Genomics of UC Genomics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Francisco Caramelo
- Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Laboratory of Biostatistics and Medical Informatics (LBIM), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Henriqueta Silva
- Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Laboratory of Sequencing and Functional Genomics of UC Genomics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
9
|
Ribeiro G, Schellekens H, Cuesta-Marti C, Maneschy I, Ismael S, Cuevas-Sierra A, Martínez JA, Silvestre MP, Marques C, Moreira-Rosário A, Faria A, Moreno LA, Calhau C. A menu for microbes: unraveling appetite regulation and weight dynamics through the microbiota-brain connection across the lifespan. Am J Physiol Gastrointest Liver Physiol 2025; 328:G206-G228. [PMID: 39811913 DOI: 10.1152/ajpgi.00227.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/14/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025]
Abstract
Appetite, as the internal drive for food intake, is often dysregulated in a broad spectrum of conditions associated with over- and under-nutrition across the lifespan. Appetite regulation is a complex, integrative process comprising psychological and behavioral events, peripheral and metabolic inputs, and central neurotransmitter and metabolic interactions. The microbiota-gut-brain axis has emerged as a critical mediator of multiple physiological processes, including energy metabolism, brain function, and behavior. Therefore, the role of the microbiota-gut-brain axis in appetite and obesity is receiving increased attention. Omics approaches such as genomics, epigenomics, transcriptomics, proteomics, and metabolomics in appetite and weight regulation offer new opportunities for featuring obesity phenotypes. Furthermore, gut-microbiota-targeted approaches such as pre-, pro-, post-, and synbiotic, personalized nutrition, and fecal microbiota transplantation are novel avenues for precision treatments. The aim of this narrative review is 1) to provide an overview of the role of the microbiota-gut-brain axis in appetite regulation across the lifespan and 2) to discuss the potential of omics and gut microbiota-targeted approaches to deepen understanding of appetite regulation and obesity.
Collapse
Affiliation(s)
- Gabriela Ribeiro
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CHRC - Center for Health Technology and Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Harriët Schellekens
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Cristina Cuesta-Marti
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Ivie Maneschy
- Growth, Exercise, Nutrition and Development Research Group, Instituto Agroalimentario de Aragón, University of Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, University of Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Shámila Ismael
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CHRC - Center for Health Technology and Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CINTESIS - Comprehensive Health Research Centre, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Amanda Cuevas-Sierra
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, Spanish National Research Council, Madrid, Spain
| | - J Alfredo Martínez
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, Spanish National Research Council, Madrid, Spain
| | - Marta P Silvestre
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CHRC - Center for Health Technology and Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Cláudia Marques
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CHRC - Center for Health Technology and Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - André Moreira-Rosário
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CINTESIS - Comprehensive Health Research Centre, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Ana Faria
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CHRC - Center for Health Technology and Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CINTESIS - Comprehensive Health Research Centre, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Luis A Moreno
- Growth, Exercise, Nutrition and Development Research Group, Instituto Agroalimentario de Aragón, University of Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, University of Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Conceição Calhau
- Metabolism and Nutrition Department, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
- CHRC - Center for Health Technology and Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
10
|
de Souza K, Defante MLR, Franco MDS, Mendes BX, Monteiro SON, Castro GC, Tavares JG, Tavares DG. Rheumatic Diseases Following Metabolic and Bariatric Surgery: A Systematic Review and Meta-Analysis. Obes Surg 2025; 35:624-634. [PMID: 39743656 DOI: 10.1007/s11695-024-07652-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/29/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025]
Abstract
Metabolic and bariatric surgery (MBS) has been associated with weight reduction and obesity complications improvement. However, there is no clear evidence of the extent and consistency of the effects of this procedure on rheumatic diseases. This study aims to conduct a meta-analysis to address the impact of MBS on rheumatic diseases. We searched PubMed, Cochrane, and Embase for studies reporting the prevalence of rheumatic diseases, the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), and the medication use after MBS. We conducted a random-effects meta-analysis using odds ratios (OR) and mean differences (MD) with 95% confidence intervals (CI). P-values < 0.05 were considered statistically significant. We included 28 studies comprising 43,421 patients, with 13,347 patients with rheumatic diseases. The prevalence of osteoarthritis (OA), rheumatoid arthritis, and psoriatic arthritis was significantly reduced after MBS (OR 0.20; 95% CI 0.12 to 0.33; P = 0.01). The WOMAC index for patients with OA had a statistically significant overall reduction after MBS at 6 months (MD - 20.60 points; 95% CI - 28.73 to - 12.47; P < 0.01) and at 12 months (MD - 15.88 points; 95% CI - 19.09 to - 12.66; P < 0.01). Medication use significantly decreased after MBS, both at the follow-up beyond 2 years (OR 0.49; 95% CI 0.35 to 0.69; P < 0.01) and up to 2 years (OR 0.32; 95% CI 0.15 to 0.69; P < 0.01). In this meta-analysis, we found a significant decrease in the prevalence of rheumatic diseases, improvements in the WOMAC index, and reduced medication use among patients undergoing MBS.
Collapse
|
11
|
Jankowski WM, Fichna J, Tarasiuk-Zawadzka A. A systematic review of the relationship between gut microbiota and prevalence of pancreatic diseases. Microb Pathog 2025; 199:107214. [PMID: 39653281 DOI: 10.1016/j.micpath.2024.107214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 11/24/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Acute pancreatitis (AP) represents one of the most common gastrointestinal (GI) diseases; it can manifest in varying degrees of severity, sometimes leading to a life-threatening condition for the patient. Pancreatic ductal adenocarcinoma (PDAC), due to its high malignancy and uncertain prognosis, is widely regarded as one of the most fatal diseases. The increasing prevalence of AP and PDAC represents a major burden on public health and the healthcare system worldwide. The aim of this systematic review was to discuss the current state of knowledge regarding the relationship between the gut microbiota and the incidence, prognosis, diagnosis and treatment of AP and PDAC. To identify studies that analyzed the relationship between the gut microbiota and the occurrence/development of pancreatic diseases or PDAC, the online databases PubMed, Scopus and Google Scholar were searched between November 2023 and January 2024. Finally, 14 publications met the inclusion criteria (1. were conducted exclusively in humans and/or animals; 2. original, published in English in peer-reviewed journals after 2019; 3. described the relationship between gut microbiota and the occurrence of AP or PDAC). The collected studies indicated significant changes in the gut microbiota of patients with AP and PDAC. Moreover, they highlighted the presence of a relationship between the gut microbiota and the occurrence, course, treatment efficiency and prognosis of the disease in question. Further research is needed to understand precisely the relationship between the gut microbiota and the occurrence of pancreatic diseases and whether it may be a starting point for the development of modern forms of therapy based on the use of prebiotics and/or diet to restore the normal composition of the intestinal bacteria.
Collapse
Affiliation(s)
- Wojciech Michał Jankowski
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland
| | - Aleksandra Tarasiuk-Zawadzka
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland.
| |
Collapse
|
12
|
Song K, Kong X, Xian Y, Yu Z, He M, Xiao D, Liang D, Zhang Z, Liu T, Huang Z, Liao X, Ren Y. Roux-en-Y gastric bypass improves liver and glucose homeostasis in Zucker diabetic fatty rats by upregulating hepatic trefoil factor family 3 and activating the phosphatidylinositol 3-kinase/protein kinase B pathway. Surg Obes Relat Dis 2025:S1550-7289(25)00007-3. [PMID: 39893149 DOI: 10.1016/j.soard.2024.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/25/2024] [Accepted: 12/22/2024] [Indexed: 02/04/2025]
Abstract
BACKGROUND Roux-en-Y gastric bypass (RYGB) surgery is effective in ameliorating type 2 diabetes mellitus (T2DM); but its mechanism remains incompletely understood. OBJECTIVES This study aimed to investigate whether RYGB improves glucose metabolism by upregulating hepatic trefoil factor family 3 (TFF3) and thereby activating the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway. SETTING Affiliated Hospital of North Sichuan Medical college in Nanchong city, China. METHODS Zucker diabetic fatty (ZDF) rats underwent RYGB or sham surgery (SHAM), and Zucker lean (ZL) rats served as controls (CON). TFF3 expression and PI3K/Akt pathway activity were compared between groups using western blot, immunofluorescence, and RT-qPCR. Adeno-associated virus (AAV) was used to specifically overexpress and interfere with hepatic TFF3. Liver fibrosis and steatosis were assessed using Masson trichrome and Oil Red O staining. HepG2 cells overexpressing or knocking out TFF3 were constructed using lentiviral transfection and CRISPR/Cas9 technology. After verifying the activity of the PI3K/Akt pathway by western blot, rescue experiments were performed on HepG2 cell overexpressing and knocking out TFF3 using LY294002 and 740Y-P, respectively. The activities of gluconeogenic enzymes and glucose uptake capacity in different HepG2 cells were evaluated using qPCR and flow cytometry. RESULTS Compared with the SHAM group, the blood glucose, body weight, insulin resistance, and lipid metabolism of ZDF rats in the RYGB group were significantly improved. The expression of TFF3 and PI3K/Akt phosphorylation in the liver of the RYGB group were higher than those of the rats that had undergone SHAM. In addition, compared with the SHAM group, the liver fibrosis and fatty degeneration of RYGB rats were milder, and the activity of gluconeogenic enzymes was lower. After tail vein injection of AAV that specifically overexpresses liver TTF3 in rats in the SHAM group, rats' insulin resistance, glucose tolerance, gluconeogenic enzymes, and other glucose metabolism indicators improved. After tail vein injection of AAV that interferes with liver TFF3 in rats in the RYGB group, rats' glucose metabolism indicators deteriorated. In in vitro experiments, the PI3K/Akt activity of TFF3-knocked-out HepG2 cells was lower than that of other groups. Lower glucose concentration were observed in TFF3-overexpressing cell lines. After rescue experiments, differences were found. The glucose metabolism level of the TFF3-expressing HepG2 cell line was positively correlated with the activity of the PI3K/Akt pathway. CONCLUSIONS RYGB regulates the expression of TFF3 in the liver of ZDF rats, thereby activating the PI3K/Akt pathway and improving T2DM.
Collapse
Affiliation(s)
- Ke Song
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China; Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Xiangxin Kong
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Yin Xian
- Department of Otolaryngology & Head and Neck Surgery, Nanchong Psychosomatic Hospital, Nanchong, China
| | - Zhenghang Yu
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China; Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Ming He
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Dingqi Xiao
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Dianyuan Liang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China; Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Zhongyang Zhang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China; Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Ting Liu
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Ziyan Huang
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Xinxin Liao
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Yixing Ren
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China; General Surgery, Chengdu XinHua Hospital Affiliated to North Sichuan Medical College, Chengdu, China.
| |
Collapse
|
13
|
Xie Y, Li Z, Fan Y, Liu X, Yi R, Gan Y, Yang Z, Liu S. Integrated gut microbiome and UHPLC-MS metabolomics to reveal the prevention mechanism of pidanjiangtang granules on IGT Rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156201. [PMID: 39531936 DOI: 10.1016/j.phymed.2024.156201] [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: 09/02/2024] [Revised: 10/16/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Pidanjiangtang (PDJT) is a traditional Chinese medicine formula empirically used to treat impaired glucose tolerance (IGT) based on the "Pidan" theory from the classic ancient book Nei Jing. However, the mechanism of PDJT intervention for IGT remains to be studied. OBJECTIVE This study aims to explore the mechanism of PDJT granules intervention in IGT by integrating gut microbiome and UHPLC-MS untargeted metabolomics. MATERIALS AND METHODS The IGT model was established in 6-week-old male Sprague-Dawley (SD) rats by feeding them a high-fat diet and using an STZ injection. The low, medium, and high doses of PDJT were used for six weeks. metformin (Glucophage) was used as the positive control drug. The efficacy of PDJT was evaluated using fasting blood glucose (FBG), blood glucose maximum (BGmax), blood lipid, and inflammatory factor levels. Finally, 16S rDNA gut microbiome sequencing with metabolomics analysis was used to explore the pharmacological mechanism of PDJT intervention in IGT. RESULTS PDJT could reverse the phenotype of IGT rats, reduce blood glucose levels, improve lipid metabolism disorder, and reduce inflammatory response. Gut microbiome analysis found that PDJT can improve gut microbiota composition and abundance of three phyla (Firmicutes, Bacteroidota, Desulfobacterota) and four genera (unclassified_f__Lachnospiraceae, Ruminococcus, Allobaculum, Desulfovibrio), which play an important role in the process of PDJT intervention on glucose metabolism and lipid metabolism in IGT rats. UHPLC-MS untargeted metabolomics showed that PDJT could regulate the levels of 258 metabolites in lipid metabolism pathways, inflammatory response pathways, fat and protein digestion, and absorption. The combined analysis of the two omics showed that improving the body's metabolism by gut microbes may be the possible mechanism of PDJT in treating IGT. Thus, this study provides a new method to integrate gut microbiome and UHPLC-MS untargeted metabolomics to evaluate the pharmacodynamics and mechanism of PDJT intervention in IGT, providing valuable ideas and insights for future research on the treatment of IGT with traditional Chinese medicine.
Collapse
Affiliation(s)
- Yu Xie
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zirong Li
- Beijing University of Chinese Medicine, Beijing, China
| | - Yue Fan
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyi Liu
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ran Yi
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yaoyao Gan
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zixuan Yang
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shangjian Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
14
|
Jakubowicz D, Matz Y, Landau Z, Rosenblum RC, Twito O, Wainstein J, Tsameret S. Interaction Between Early Meals (Big-Breakfast Diet), Clock Gene mRNA Expression, and Gut Microbiome to Regulate Weight Loss and Glucose Metabolism in Obesity and Type 2 Diabetes. Int J Mol Sci 2024; 25:12355. [PMID: 39596418 PMCID: PMC11594859 DOI: 10.3390/ijms252212355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/09/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
The circadian clock gene system plays a pivotal role in coordinating the daily rhythms of most metabolic processes. It is synchronized with the light-dark cycle and the eating-fasting schedule. Notably, the interaction between meal timing and circadian clock genes (CGs) allows for optimizing metabolic processes at specific times of the day. Breakfast has a powerful resetting effect on the CG network. A misaligned meal pattern, such as skipping breakfast, can lead to a discordance between meal timing and the endogenous CGs, and is associated with obesity and T2D. Conversely, concentrating most calories and carbohydrates (CH) in the early hours of the day upregulates metabolic CG expression, thus promoting improved weight loss and glycemic control. Recently, it was revealed that microorganisms in the gastrointestinal tract, known as the gut microbiome (GM), and its derived metabolites display daily oscillation, and play a critical role in energy and glucose metabolism. The timing of meal intake coordinates the oscillation of GM and GM-derived metabolites, which in turn influences CG expression, playing a crucial role in the metabolic response to food intake. An imbalance in the gut microbiota (dysbiosis) can also reciprocally disrupt CG rhythms. Evidence suggests that misaligned meal timing may cause such disruptions and can lead to obesity and hyperglycemia. This manuscript focuses on the reciprocal interaction between meal timing, GM oscillation, and circadian CG rhythms. It will also review studies demonstrating how aligning meal timing with the circadian clock can reset and synchronize CG rhythms and GM oscillations. This synchronization can facilitate weight loss and improve glycemic control in obesity and those with T2D.
Collapse
Affiliation(s)
- Daniela Jakubowicz
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| | - Yael Matz
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Zohar Landau
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| | - Rachel Chava Rosenblum
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| | - Orit Twito
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| | - Julio Wainstein
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| | - Shani Tsameret
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| |
Collapse
|
15
|
Custers E, van der Burgh YG, Vreeken D, Schuren F, van den Broek TJ, Verschuren L, de Blaauw I, Bouwens M, Kleemann R, Kiliaan AJ, Hazebroek EJ. Gastrointestinal complaints after Roux-en-Y gastric bypass surgery. Impact of microbiota and its metabolites. Heliyon 2024; 10:e39899. [PMID: 39559236 PMCID: PMC11570293 DOI: 10.1016/j.heliyon.2024.e39899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/20/2024] Open
Abstract
Unexplainable gastrointestinal complaints occasionally occur after Roux-en-Y Gastric Bypass (RYGB) surgery. We therefor investigated the impact of microbiota composition and metabolites on gastrointestinal complaints after RYGB. In the BARICO study (Bariatric surgery Rijnstate and Radboudumc neuroimaging and Cognition in Obesity), microbiota and metabolites were measured before surgery, and 6, and 24 months after surgery. Gastrointestinal complaints were assessed with the Irritable Bowel Syndrome Severity Scoring System (IBS-SSS) questionnaire 24 months after surgery. 65 participants (86.2 % female) with a mean age of 46.2 ± 6.0 years, and mean BMI of 41.2 ± 3.6 kg/m2 were included. According to the IBS-SSS questionnaire, 32.3 % had moderate/severe gastrointestinal complaints 24 months after surgery. Microbiota alpha diversity remained stable, while beta diversity significantly changed over time. Bile acids and short-chain fatty acids were significantly higher, and inflammatory markers significantly lower after surgery. Barnesiella sp., Escherichia/Shigella sp., and Faecalibacterium prausnitzii correlated positively, while Akkermansia sp correlated inversely with gastrointestinal complaints. Patients with mild and moderate/severe gastrointestinal complaints showed higher levels of GLC-3S. These findings suggest involvement of microbiota and metabolite changes in gastrointestinal complaints after surgery. However, it remains unclear whether bacteria influence gastrointestinal complaints directly or indirectly. Further exploration is required for development of interventions against gastrointestinal symptoms after surgery.
Collapse
Affiliation(s)
- Emma Custers
- Department of Bariatric Surgery, Vitalys, Rijnstate Hospital, Arnhem, the Netherlands
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Radboud Alzheimer Center, Donders Institute for Brain Cognition and Behaviour, Center for Medical Neuroscience, Nijmegen, the Netherlands
| | - Yonta G.R. van der Burgh
- Department of Bariatric Surgery, Vitalys, Rijnstate Hospital, Arnhem, the Netherlands
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, the Netherlands
| | - Debby Vreeken
- Department of Bariatric Surgery, Vitalys, Rijnstate Hospital, Arnhem, the Netherlands
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Radboud Alzheimer Center, Donders Institute for Brain Cognition and Behaviour, Center for Medical Neuroscience, Nijmegen, the Netherlands
| | - Frank Schuren
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Tim J. van den Broek
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Lars Verschuren
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Ivo de Blaauw
- Department of Surgery, Division of Paediatric Surgery, Radboudumc-Amalia Children's Hospital, Nijmegen, the Netherlands
| | - Mark Bouwens
- Dutch Digestive Foundation, Amersfoort, the Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Amanda J. Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Radboud Alzheimer Center, Donders Institute for Brain Cognition and Behaviour, Center for Medical Neuroscience, Nijmegen, the Netherlands
| | - Eric J. Hazebroek
- Department of Bariatric Surgery, Vitalys, Rijnstate Hospital, Arnhem, the Netherlands
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, the Netherlands
| |
Collapse
|
16
|
Mallet M, Silaghi CA, Sultanik P, Conti F, Rudler M, Ratziu V, Thabut D, Pais R. Current challenges and future perspectives in treating patients with NAFLD-related cirrhosis. Hepatology 2024; 80:1270-1290. [PMID: 37183906 DOI: 10.1097/hep.0000000000000456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/20/2023] [Indexed: 05/16/2023]
Abstract
Despite the slow, progressive nature of NAFLD, the number of patients with NAFLD-related cirrhosis has significantly increased. Although the management of patients with cirrhosis is constantly evolving, improving the prognosis of patients with NAFLD-related cirrhosis is a challenge because it is situated at the crossroads between the liver, the metabolic, and the cardiovascular diseases. Therefore, the therapeutic interventions should not only target the liver but also the associated cardiometabolic conditions and should be adapted accordingly. The objective of the current review is to critically discuss the particularities in the management of patients with NAFLD-related cirrhosis. We relied on the recommendations of scientific societies and discussed them in the specific context of NAFLD cirrhosis and the surrounding cardiometabolic milieu. Herein, we covered the following aspects: (1) the weight loss strategies through lifestyle interventions to avoid sarcopenia and improve portal hypertension; (2) the optimal control of metabolic comorbidities in particular type 2 diabetes aimed not only to improve cardiovascular morbidity/mortality but also to lower the incidence of cirrhosis-related complications (we discussed various aspects related to the safety of oral antidiabetic drugs in cirrhosis); (3) the challenges in performing bariatric surgery in patients with cirrhosis related to the portal hypertension and the risk of cirrhosis decompensation; (4) the particularities in the diagnosis and management of the portal hypertension and the difficulties in managing patients awaiting for liver transplantation; and (5) the difficulties in developing drugs and conducting clinical trials in patients with NAFLD-related cirrhosis. Moreover, we discussed the emerging options to overcome these obstacles.
Collapse
Affiliation(s)
- Maxime Mallet
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Service d'hepato-gastroentérologie, Hôpital Pitié-Salpêtrière, Paris, France
| | - Cristina Alina Silaghi
- Department of Endocrinology, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj-Napoca, Roumanie
| | - Philippe Sultanik
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Service d'hepato-gastroentérologie, Hôpital Pitié-Salpêtrière, Paris, France
- Brain Liver Pitié-Salpêtrière Study Group (BLIPS), Paris, France
| | - Filomena Conti
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Service d'hepato-gastroentérologie, Hôpital Pitié-Salpêtrière, Paris, France
- Centre de Recherche Saint Antoine, INSERM UMRS_938 Paris, France
| | - Marika Rudler
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Service d'hepato-gastroentérologie, Hôpital Pitié-Salpêtrière, Paris, France
- Brain Liver Pitié-Salpêtrière Study Group (BLIPS), Paris, France
- Centre de Recherche Saint Antoine, INSERM UMRS_938 Paris, France
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Vlad Ratziu
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Service d'hepato-gastroentérologie, Hôpital Pitié-Salpêtrière, Paris, France
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
- INSERM UMRS 1138 CRC, Paris, France
| | - Dominique Thabut
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Service d'hepato-gastroentérologie, Hôpital Pitié-Salpêtrière, Paris, France
- Brain Liver Pitié-Salpêtrière Study Group (BLIPS), Paris, France
- Centre de Recherche Saint Antoine, INSERM UMRS_938 Paris, France
| | - Raluca Pais
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Service d'hepato-gastroentérologie, Hôpital Pitié-Salpêtrière, Paris, France
- Centre de Recherche Saint Antoine, INSERM UMRS_938 Paris, France
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| |
Collapse
|
17
|
Zhao J, Alimu A, Li Y, Lin Z, Li J, Wang X, Wang Y, Lv G, Lin H, Lin Z. Potential Anti-Obesity Effect of Hazel Leaf Extract in Mice and Network Pharmacology of Selected Polyphenols. Pharmaceuticals (Basel) 2024; 17:1349. [PMID: 39458990 PMCID: PMC11510286 DOI: 10.3390/ph17101349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/28/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Obesity is gradually becoming a widespread health problem, and treatment using natural compounds has seen an increasing trend. As a by-product of hazelnut, hazel leaf is usually disposed of as waste, but it is widely used in traditional and folk medicines around the world. Aim of this study: Based on previous studies, the effects of the regulation of lipid metabolism and the mechanism of hazel leaf polyphenol extraction obesity were investigated. Methods: In this study, a high-fat diet-fed mouse model of obesity and 3T3-L1 preadipocytes were established. The ameliorative effects of the hazel leaf polyphenol extract on obesity and the regulating lipid metabolisms were explored based on network pharmacology, gut microbiota, and molecular docking. Results: Network pharmacology showed that hazel leaf polyphenols may play a role by targeting key targets, including PPARγ, and regulating the PPAR signaling pathway. They significantly improved body weight gain, the liver index, and adiposity and lipid levels; regulated the gut microbiota and short-chain fatty acid contents; down-regulated the expression of lipid synthesis proteins SREBP1c, PPARγ, and C/EBP-α; and up-regulated the expression of p-AMPK in obese mice. They inhibited the differentiation of 3T3-L1 cells, and the expression of related proteins is consistent with the results in vivo. The molecular docking results indicated that gallic acid, quercetin-3-O-beta-D-glucopyranoside, quercetin, myricetin, and luteolin-7-O-glucoside in the hazel leaf polyphenol extract had strong binding activities with PPARγ, C/EBP-α, and AMPK. Conclusions: The results demonstrate that the hazel leaf polyphenol extract can improve obesity by regulating lipid metabolism, which provides a valuable basis for developing health products made from hazel leaf polyphenols in the future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - He Lin
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (J.Z.); (A.A.); (Y.L.); (Z.L.); (J.L.); (X.W.); (Y.W.); (G.L.)
| | - Zhe Lin
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (J.Z.); (A.A.); (Y.L.); (Z.L.); (J.L.); (X.W.); (Y.W.); (G.L.)
| |
Collapse
|
18
|
Qian Y, Sorgen AA, Steffen KJ, Heinberg LJ, Reed K, Carroll IM. Intestinal Energy Harvest Mediates Gut Microbiota-Associated Weight Loss Following Bariatric Surgery. Obes Surg 2024; 34:3771-3780. [PMID: 39196507 DOI: 10.1007/s11695-024-07467-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024]
Abstract
PURPOSE Metabolic and bariatric surgery (MBS) is the most effective treatment for class III obesity. The capacity to efficiently extract intestinal energy is potentially a determinant of varying weight loss outcomes post-MBS. Prior research indicated that intestinal energy harvest is correlated with post-MBS weight loss. Studies have also demonstrated that the gut microbiota is associated with weight loss post-MBS. We aim to investigate whether gut microbiota-associated weight loss is mediated by intestinal energy harvest in patients post-MBS. MATERIALS AND METHODS We examined the relationship between specific gut microbiota, intestinal energy harvest, diet, and weight loss using fecal metagenomic sequence data, bomb calorimetry (fecal energy content as a proxy for calorie absorption), and a validated dietary questionnaire on 67 individuals before and after MBS. Mediation analysis and a machine learning algorithm were conducted. RESULTS Intestinal energy harvest was a mediator in the relationship between the intestinal microbiota (Bacteroides caccae) and weight loss outcomes in patients post-MBS at 18 months (M). The association between the abundance of B. caccae and post-MBS weight loss rate at 18 M was partly mediated by 1 M intestinal energy harvest (β = 0.001 ± 0.001, P = 0.020). This mediation represents 2.83% of the total effect (β = 0.050 ± 0.047; P = 0.028). Intestinal microbiota and energy harvest improved random forest model's accuracy in predicting weight loss results. CONCLUSION Energy harvest partly mediates the relationship between the intestinal microbiota and weight loss outcomes among patients post-MBS. This study elucidates a potential mechanism regarding how intestinal energy absorption facilitates the effect of intestinal microbiota on energy metabolism and weight loss outcomes.
Collapse
Affiliation(s)
- Yunzhi Qian
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alicia A Sorgen
- Department of Bioinformatics and Genomics, University of North Carolina, Charlotte, NC, 28262, USA
| | - Kristine J Steffen
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, 58103, USA
- Sanford Center for Biobehavioral Research, Fargo, ND, 58103, USA
| | - Leslie J Heinberg
- Department of Psychiatry and Psychology, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA
| | - Kylie Reed
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, 27708, USA
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ian M Carroll
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
19
|
Higuera-de-la-Tijera F, Servín-Caamaño A, Lajud-Barquín F, Tovar-Aguilar A. Cambios en el peso y modificaciones en el estilo de vida implementados durante el confinamiento por la pandemia de COVID-19 se asocian como desencadenantes del desarrollo de síntomas gastrointestinales. REVISTA DE GASTROENTEROLOGÍA DE MÉXICO 2024; 89:481-490. [DOI: 10.1016/j.rgmx.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
20
|
Lu M, Feng R, Li M, Liu L, Xiao Y, Liu Y, Yin C. Causal relationship between gut microbiota and childhood obesity: A Mendelian randomization study and case-control study. Clin Nutr ESPEN 2024; 63:197-206. [PMID: 38963766 DOI: 10.1016/j.clnesp.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/28/2024] [Accepted: 05/17/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Gut microbiota and obesity are deeply interconnected. However, the causality in the relationship between these factors remains unclear. Therefore, this study aimed to elucidate the genetic relationship between gut microbiota and childhood obesity. METHODS Genetic summary statistics for the gut microbiota were obtained from the MiBioGen consortium. Genome-wide association studies (GWAS) summary data for childhood obesity were obtained from North American, Australian, and European collaborative genome-wide meta-analyses. Mendelian randomization (MR) analyses were performed using the inverse variance weighting method. 16 children with obesity and 16 without obesity were included for clinical observation, and their weight, body mass index, blood lipid levels, and gut microbiology were assessed. Paired t-test was the primary method of data analysis, and statistical significance was set at P < 0.05. RESULTS MR identified 16 causal relationships between the gut microbiome and childhood obesity. In the case-control study, we found that five gut microorganisms differed between children with and without obesity, whereas three gut microorganisms changed after weight loss in children with obesity. CONCLUSION Our study provides new insights into the genetic mechanisms underlying gut microbiota and childhood obesity. TRIAL REGISTRATION NUMBER ChiCTR2300072179. NAME OF REGISTRY Change of intestinal flora and plasma metabolome in obese children and their weight loss intervention: a randomized controlled tria URL OF REGISTRY: https://www.chictr.org.cn/showproj.html. DATE OF REGISTRATION 2023-06-06. DATE OF ENROLMENT OF THE FIRST PARTICIPANT TO THE TRIAL 2023-06-07.
Collapse
Affiliation(s)
- Mengnan Lu
- Department of Pediatrics, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an, Shanxi, 710054, China
| | - Ruoyang Feng
- Department of Joint Surgery, Xi'an Jiaotong University HongHui Hospital, Xi'an, Shanxi, 710054, China
| | - Meng Li
- Department of Pediatrics, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an, Shanxi, 710054, China
| | - Lujie Liu
- Department of Pediatrics, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an, Shanxi, 710054, China
| | - Yanfeng Xiao
- Department of Pediatrics, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an, Shanxi, 710054, China.
| | - Yuesheng Liu
- Department of Pediatrics, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an, Shanxi, 710054, China.
| | - Chunyan Yin
- Department of Pediatrics, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an, Shanxi, 710054, China.
| |
Collapse
|
21
|
Higuera-de-la-Tijera F, Servín-Caamaño A, Lajud-Barquín F, Tovar-Aguilar A. Weight change and lifestyle modifications implemented during the COVID-19 pandemic lockdown are associated with the development of gastrointestinal symptoms. REVISTA DE GASTROENTEROLOGIA DE MEXICO (ENGLISH) 2024; 89:481-490. [PMID: 39332979 DOI: 10.1016/j.rgmxen.2024.09.001] [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: 08/15/2023] [Accepted: 02/20/2024] [Indexed: 09/29/2024]
Abstract
INTRODUCTION AND AIMS Pandemic lockdown measures are a cause of concern, regarding their negative impact on the mental health of individuals. The results of numerous studies have associated the appearance of gastrointestinal symptoms with different psychologic disorders, such as stress, depression, and anxiety, due to gut-brain axis interaction. The aim of the present study was to determine the prevalence of, and factors associated with, gastrointestinal symptom onset related to the COVID-19 pandemic lockdown and various lifestyle modifications. METHODS An analytic, observational, and cross-sectional study was conducted on an open population that agreed to participate within the time frame of January to May 2021. RESULTS A total of 298 subjects, 165 of whom were women (55.4%), agreed to participate and the mean patient age was 36.1 ± 12.6 years. There was a significant increase in the frequency of several gastrointestinal symptoms: epigastric burning, early satiety, heartburn, regurgitation, constipation, and diarrhea. Changes in weight and modifications in lifestyle were found to be associated variables. CONCLUSIONS The results of this study showed a significant increase in a wide variety of gastrointestinal symptoms related to lifestyle changes due to the pandemic lockdown. Weight change, supplement and multivitamin intake, and reduced physical activity were the main associated risk factors. Public healthcare systems should take a multidisciplinary approach into consideration for the care of affected individuals.
Collapse
Affiliation(s)
- F Higuera-de-la-Tijera
- Departamento de Gastroenterología y Hepatología, Hospital General de México Dr. Eduardo Liceaga, Mexico City, Mexico; Profesora de Medicina, Escuela de Medicina Saint Luke, Universidad Alliant, Mexico City, Mexico.
| | - A Servín-Caamaño
- Departamento de Medicina Interna, Hospital General de México Dr. Eduardo Liceaga, Mexico City, Mexico
| | - F Lajud-Barquín
- Departamento de Gastroenterología y Hepatología, Hospital General de México Dr. Eduardo Liceaga, Mexico City, Mexico
| | - A Tovar-Aguilar
- Departamento de Gastroenterología y Hepatología, Hospital General de México Dr. Eduardo Liceaga, Mexico City, Mexico
| |
Collapse
|
22
|
Petrinović M, Majetić D, Bakula M, Pećin I, Fabris-Vitković D, Deškin M, Tešanović Perković D, Bakula M, Gradišer M, Ćurčić IB, Canecki-Varžić S. Molecular Mechanisms Affecting Statin Pharmacokinetics after Bariatric Surgery. Int J Mol Sci 2024; 25:10375. [PMID: 39408705 PMCID: PMC11476770 DOI: 10.3390/ijms251910375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
According to recent data, one in eight people in the world struggle with obesity. Obesity management is increasingly dependent on bariatric surgical interventions, as the combination of lifestyle modifications and pharmacotherapy could have a modest long-term effect. Surgery is recommended only for individuals whose body mass index (BMI) ≥ 40 kg/m2 and ≥ 35 kg/m2 in the presence of weight-related comorbidities. The most commonly performed procedures are sleeve gastrectomy and roux-en-Y gastric bypass. Pharmacokinetic and pharmacodynamic alterations occur as a result of the anatomical and physiological changes caused by surgery, which further differ depending on physicochemical drug factors and factors related to the dosage form. The following modifications are distinguished based on the type of bariatric surgery performed. Most bariatric patients have accompanying comorbidities, including dyslipidemia treated with hydroxymethylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors or statins. Significant improvements in the lipid profile are observed early in the postoperative period. The data reported in this review on statin pharmacokinetic alterations have demonstrated substantial inter- and intravariability, making it difficult to adopt clear guidelines. Based on the current literature review, reducing the statin dose to the lowest effective with continuous monitoring is considered an optimal approach in clinical practice.
Collapse
Affiliation(s)
- Matea Petrinović
- Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (M.P.); (D.M.)
- Polyclinic Slavonija, 31000 Osijek, Croatia
| | - Domagoj Majetić
- Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (M.P.); (D.M.)
- The Clinic for Internal Diseases, Department for Gastroenterology and Hepatology, Clinical Hospital Centre Osijek, 31000 Osijek, Croatia
| | - Miro Bakula
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolic Diseases, Sveti Duh University Hospital, 10000 Zagreb, Croatia;
| | - Ivan Pećin
- Department of Internal Medicine, Unit for Metabolic Diseases, University Hospital Center Zagreb, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | | | - Marin Deškin
- Division of Endocrinology and Diabetes, General Hospital Pula, 52100 Pula, Croatia
| | | | - Maja Bakula
- Vuk Vrhovac University Clinic for Diabetes and Metabolism, Merkur University Hospital, 10000 Zagreb, Croatia
| | - Marina Gradišer
- Internal Medicine Department, County Hospital Čakovec, 40000 Čakovec, Croatia
- School of Medicine, University of Split, 21000 Split, Croatia
| | - Ines Bilić Ćurčić
- Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (M.P.); (D.M.)
- Clinic for Internal Diseases, Department for Endocrinology and Diabetes, Clinical Hospital Centre Osijek, 31000 Osijek, Croatia
| | - Silvija Canecki-Varžić
- Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (M.P.); (D.M.)
- Clinic for Internal Diseases, Department for Endocrinology and Diabetes, Clinical Hospital Centre Osijek, 31000 Osijek, Croatia
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| |
Collapse
|
23
|
Cheng T, Wen P, Yu R, Zhang F, Li H, Xu X, Zhao D, Liu F, Su W, Zheng Z, Yang H, Yao J, Jin L. Integrative microbiome and metabolome profiles reveal the impacts of periodontitis via oral-gut axis in first-trimester pregnant women. J Transl Med 2024; 22:819. [PMID: 39227984 PMCID: PMC11370083 DOI: 10.1186/s12967-024-05579-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/04/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Periodontitis results from host-microbe dysbiosis and the resultant dysregulated immunoinflammatory response. Importantly, it closely links to numerous systemic comorbidities, and perplexingly contributes to adverse pregnancy outcomes (APOs). Currently, there are limited studies on the distal consequences of periodontitis via oral-gut axis in pregnant women. This study investigated the integrative microbiome-metabolome profiles through multi-omics approaches in first-trimester pregnant women and explored the translational potentials. METHODS We collected samples of subgingival plaques, saliva, sera and stool from 54 Chinese pregnant women at the first trimester, including 31 maternal periodontitis (Perio) subjects and 23 Non-Perio controls. By integrating 16S rRNA sequencing, untargeted metabolomics and clinical traits, we explored the oral-gut microbial and metabolic connection resulting from periodontitis among early pregnant women. RESULTS We demonstrated a novel bacterial distinguisher Coprococcus from feces of periodontitis subjects in association with subgingival periodontopathogens, being different from other fecal genera in Lachnospiraceae family. The ratio of fecal Coprococcus to Lachnoclostridium could discriminate between Perio and Non-Perio groups as the ratio of subgingival Porphyromonas to Rothia did. Furthermore, there were differentially abundant fecal metabolic features pivotally enriched in periodontitis subjects like L-urobilin and kynurenic acid. We revealed a periodontitis-oriented integrative network cluster, which was centered with fecal Coprococcus and L-urobilin as well as serum triglyceride. CONCLUSIONS The current findings about the notable influence of periodontitis on fecal microbiota and metabolites in first-trimester pregnant women via oral-gut axis signify the importance and translational implications of preconceptional oral/periodontal healthcare for enhancing maternal wellbeing.
Collapse
Affiliation(s)
- Tianfan Cheng
- Division of Periodontology & Implant Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China.
| | - Ping Wen
- Institute of Maternal and Child Medicine & Shenzhen Key Laboratory of Maternal and Child Health and Diseases, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Rong Yu
- Institute of Maternal and Child Medicine & Shenzhen Key Laboratory of Maternal and Child Health and Diseases, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Feng Zhang
- Division of Stomatology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Huijun Li
- Division of Stomatology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Xiaoyi Xu
- Institute of Maternal and Child Medicine & Shenzhen Key Laboratory of Maternal and Child Health and Diseases, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
- Center for Disease Control and Prevention, Shenzhen, China
| | - Dan Zhao
- Division of Periodontology & Implant Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
- Department of Implant Dentistry, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Fang Liu
- Division of Obstetrics & Gynecology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Weilan Su
- Division of Obstetrics & Gynecology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Zheng Zheng
- Division of Obstetrics & Gynecology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Hong Yang
- Division of Obstetrics & Gynecology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Jilong Yao
- Division of Obstetrics & Gynecology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Lijian Jin
- Division of Periodontology & Implant Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
24
|
Shieh C, Thompson HJ, McLaughlin E, Chiang CW, Hussan H. Advancements in Understanding and Preventing Obesity-Related Colon Cancer. Cancer J 2024; 30:357-369. [PMID: 39312456 DOI: 10.1097/ppo.0000000000000744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
ABSTRACT Obesity and colorectal cancer are global public health issues, with the prevalence of both conditions increasing over the last 4 decades. In the United States alone, the prevalence of obesity is greater than 40%, and this percentage is projected to increase past 50% by 2030. This review focuses on understanding the association between obesity and the risk of colorectal cancer while also highlighting hypotheses about molecular mechanisms underlying the link between these disease processes. We also consider whether those linkages can be disrupted via weight loss therapies, including lifestyle modifications, pharmacotherapy, bariatric surgery, and endobariatrics.
Collapse
Affiliation(s)
- Christine Shieh
- From the Department of Gastroenterology, University of California, Davis, Sacramento, CA
| | - Henry J Thompson
- Cancer Prevention Laboratory, Colorado State University, Fort Collins, CO
| | | | - Chien-Wei Chiang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH
| | | |
Collapse
|
25
|
Lauwers E, Sabino J, Hoffman I, van Hoeve K. Faecal microbiota transplantation in children: A systematic review. Acta Paediatr 2024; 113:1991-2002. [PMID: 38391047 DOI: 10.1111/apa.17167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/25/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024]
Abstract
AIM Novel technologies offer insights into the potential role of the intestinal microbiota in human health and disease. Dysbiosis has been associated with several diseases, and it is thought to play a role in the pathogenesis of different gastrointestinal diseases. Faecal microbiota transplantation (FMT) is emerging as a method to modulate the gastrointestinal microbial ecosystem. While recurrent Clostridioides difficile infection is the recognised FMT indication, exploration of other therapeutic uses is ongoing. METHODS Following PRISMA guidelines, we conducted a systematic review, extracting 583 articles from Embase and PubMed (index date to October 2022). RESULTS The search yielded 58 studies for full review, with 50 included in the systematic review. Articles were categorised by FMT indication, study design, efficacy, adverse events, donor selection and administration route. FMT appears safe and effective for recurrent Clostridioides difficile infection, although severe adverse events are reported in children. However, there are currently insufficient data to support the use of FMT for other potential therapeutic indications (such as irritable or inflammatory bowel disease or obesity), beside the potential to decolonise multi-drug resistant organisms. CONCLUSION This underscores the need for randomised, controlled, prospective cohort studies in children to assess FMT effectiveness in diverse conditions and counteract publication bias.
Collapse
Affiliation(s)
- Ella Lauwers
- Department of Paediatric Gastroenterology & Hepatology & Nutrition, University Hospitals Leuven, Leuven, Belgium
| | - João Sabino
- TARGID, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
- Department of Gastroenterology & Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Ilse Hoffman
- Department of Paediatric Gastroenterology & Hepatology & Nutrition, University Hospitals Leuven, Leuven, Belgium
| | - Karen van Hoeve
- Department of Paediatric Gastroenterology & Hepatology & Nutrition, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
26
|
Angelini G, Russo S, Mingrone G. Incretin hormones, obesity and gut microbiota. Peptides 2024; 178:171216. [PMID: 38636809 DOI: 10.1016/j.peptides.2024.171216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/20/2024]
Abstract
Over the past 40 years, the prevalence of obesity has risen dramatically, reaching epidemic proportions. By 2030 the number of people affected by obesity will reach 1.12 billion worldwide. Gastrointestinal hormones, namely incretins, play a vital role in the pathogenesis of obesity and its comorbidities. GIP (glucose-dependent insulinotropic polypeptide) and GLP-1 (glucagon-like peptide-1), which are secreted from the intestine after nutrient intake and stimulate insulin secretion from pancreatic β cells, influence lipid metabolism, gastric empting, appetite and body weight. The gut microbiota plays an important role in various metabolic conditions, including obesity and type 2 diabetes and influences host metabolism through the interaction with enteroendocrine cells that modulate incretins secretion. Gut microbiota metabolites, such as short-chain fatty acids (SCFAs) and indole, directly stimulate the release of incretins from colonic enteroendocrine cells influencing host satiety and food intake. Moreover, bariatric surgery and incretin-based therapies are associated with increase gut bacterial richness and diversity. Understanding the role of incretins, gut microbiota, and their metabolites in regulating metabolic processes is crucial to develop effective strategies for the management of obesity and its associated comorbidities.
Collapse
Affiliation(s)
| | - Sara Russo
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Geltrude Mingrone
- Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Division of Diabetes & Nutritional Sciences, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, United Kingdom.
| |
Collapse
|
27
|
Wang R, Mijiti S, Xu Q, Liu Y, Deng C, Huang J, Yasheng A, Tian Y, Cao Y, Su Y. The Potential Mechanism of Remission in Type 2 Diabetes Mellitus After Vertical Sleeve Gastrectomy. Obes Surg 2024; 34:3071-3083. [PMID: 38951388 DOI: 10.1007/s11695-024-07378-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 07/03/2024]
Abstract
In recent years, there has been a gradual increase in the prevalence of obesity and type 2 diabetes mellitus (T2DM), with bariatric surgery remaining the most effective treatment strategy for these conditions. Vertical sleeve gastrectomy (VSG) has emerged as the most popular surgical procedure for bariatric/metabolic surgeries, effectively promoting weight loss and improving or curing T2DM. The alterations in the gastrointestinal tract following VSG may improve insulin secretion and resistance by increasing incretin secretion (especially GLP-1), modifying the gut microbiota composition, and through mechanisms dependent on weight loss. This review focuses on the potential mechanisms through which the enhanced action of incretin and metabolic changes in the digestive system after VSG may contribute to the remission of T2DM.
Collapse
Affiliation(s)
- Rongfei Wang
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No.57 Mei Hua East Road, Xiang Zhou District, Zhuhai, 519000, Guangdong, China
| | - Salamu Mijiti
- Department of General Surgery, The First People's Hospital of Kashi, Autonomous Region, Kashi, 844000, Xinjiang Uygur, China
| | - Qilin Xu
- Department of General Surgery, The First People's Hospital of Kashi, Autonomous Region, Kashi, 844000, Xinjiang Uygur, China
| | - Yile Liu
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No.57 Mei Hua East Road, Xiang Zhou District, Zhuhai, 519000, Guangdong, China
| | - Chaolun Deng
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No.57 Mei Hua East Road, Xiang Zhou District, Zhuhai, 519000, Guangdong, China
| | - Jiangtao Huang
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No.57 Mei Hua East Road, Xiang Zhou District, Zhuhai, 519000, Guangdong, China
| | - Abudoukeyimu Yasheng
- Department of General Surgery, The First People's Hospital of Kashi, Autonomous Region, Kashi, 844000, Xinjiang Uygur, China
| | - Yunping Tian
- Department of General Surgery, The First People's Hospital of Kashi, Autonomous Region, Kashi, 844000, Xinjiang Uygur, China.
| | - Yanlong Cao
- Department of General Surgery, The First People's Hospital of Kashi, Autonomous Region, Kashi, 844000, Xinjiang Uygur, China.
| | - Yonghui Su
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No.57 Mei Hua East Road, Xiang Zhou District, Zhuhai, 519000, Guangdong, China.
| |
Collapse
|
28
|
Gryaznova M, Smirnova Y, Burakova I, Syromyatnikov M, Chizhkov P, Popov E, Popov V. Changes in the Human Gut Microbiome Caused by the Short-Term Impact of Lactic Acid Bacteria Consumption in Healthy People. Probiotics Antimicrob Proteins 2024; 16:1240-1250. [PMID: 37365419 DOI: 10.1007/s12602-023-10111-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2023] [Indexed: 06/28/2023]
Abstract
The gut microbiome is one of the main factors affecting human health. It has been proven that probiotics can regulate the metabolism in the host body. A large number of people use probiotics not as medicines, but as a prophylactic supplement. The aim of our study was to evaluate the effect of lactic acid bacteria on the gut microbiome of healthy people using the V3 region of the 16S rRNA gene. Our study showed changes in the generic composition in the gut of healthy people when taking the supplement. There was an increase in the members responsible for the production of short-chain fatty acids in the gut of the host (Blautia, Fusicatenibacter, Eubacterium hallii group, Ruminococcus), as well as bacteria that improve intestinal homeostasis (Dorea and Barnesiella). There was also a decrease in the abundance of bacteria in the genera Catenibacterium, Hungatella, Escherichia-Shigella, and Pseudomonas, associated with an unhealthy profile of the human gut microbiome. An increase in members of the phylum Actinobacteriota was also observed, which has a positive effect on the host organism. Our results indicate that short-term prophylactic use of lactic acid bacteria-based supplements can be effective, as it contributes to a beneficial effect on the gut microbiome of healthy people.
Collapse
Affiliation(s)
- Mariya Gryaznova
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036, Voronezh, Russia
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018, Voronezh, Russia
| | - Yuliya Smirnova
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036, Voronezh, Russia
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018, Voronezh, Russia
| | - Inna Burakova
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036, Voronezh, Russia
| | - Mikhail Syromyatnikov
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036, Voronezh, Russia.
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018, Voronezh, Russia.
| | - Pavel Chizhkov
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018, Voronezh, Russia
| | - Evgeny Popov
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036, Voronezh, Russia
| | - Vasily Popov
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036, Voronezh, Russia
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018, Voronezh, Russia
| |
Collapse
|
29
|
Rubio Garcia E, Casadellà M, Parera M, Vila J, Paredes R, Noguera-Julian M. Gut resistome linked to sexual preference and HIV infection. BMC Microbiol 2024; 24:201. [PMID: 38851693 PMCID: PMC11162057 DOI: 10.1186/s12866-024-03335-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 05/16/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND People living with HIV (PLWH) are at increased risk of acquisition of multidrug resistant organisms due to higher rates of predisposing factors. The gut microbiome is the main reservoir of the collection of antimicrobial resistance determinants known as the gut resistome. In PLWH, changes in gut microbiome have been linked to immune activation and HIV-1 associated complications. Specifically, gut dysbiosis defined by low microbial gene richness has been linked to low Nadir CD4 + T-cell counts. Additionally, sexual preference has been shown to strongly influence gut microbiome composition in PLWH resulting in different Prevotella or Bacteroides enriched enterotypes, in MSM (men-who-have-sex-with-men) or no-MSM, respectively. To date, little is known about gut resistome composition in PLWH due to the scarcity of studies using shotgun metagenomics. The present study aimed to detect associations between different microbiome features linked to HIV-1 infection and gut resistome composition. RESULTS Using shotgun metagenomics we characterized the gut resistome composition of 129 HIV-1 infected subjects showing different HIV clinical profiles and 27 HIV-1 negative controls from a cross-sectional observational study conducted in Barcelona, Spain. Most no-MSM showed a Bacteroides-enriched enterotype and low microbial gene richness microbiomes. We did not identify differences in resistome diversity and composition according to HIV-1 infection or immune status. However, gut resistome was more diverse in MSM group, Prevotella-enriched enterotype and gut micorbiomes with high microbial gene richness compared to no-MSM group, Bacteroides-enriched enterotype and gut microbiomes with low microbial gene richness. Additionally, gut resistome beta-diversity was different according to the defined groups and we identified a set of differentially abundant antimicrobial resistance determinants based on the established categories. CONCLUSIONS Our findings reveal a significant correlation between gut resistome composition and various host variables commonly associated with gut microbiome, including microbiome enterotype, microbial gene richness, and sexual preference. These host variables have been previously linked to immune activation and lower Nadir CD4 + T-Cell counts, which are prognostic factors of HIV-related comorbidities. This study provides new insights into the relationship between antibiotic resistance and clinical characteristics of PLWH.
Collapse
Affiliation(s)
- Elisa Rubio Garcia
- Department of Microbiology, CDB, Hospital Clinic, University of Barcelona, Barcelona, Spain.
- Molecuar Core Facilty, Hospital Clínic de Barcelona, Barcelona, Spain.
- ISGlobal Barcelona Institute for Global Health, Barcelona, Spain.
| | | | | | - Jordi Vila
- Department of Microbiology, CDB, Hospital Clinic, University of Barcelona, Barcelona, Spain
- ISGlobal Barcelona Institute for Global Health, Barcelona, Spain
- Infectious Disease Networking Biomedical Research Center (CIBERINFEC), Carlos III Health Institute, Madrid, Spain
| | - Roger Paredes
- IrsiCaixa, Ctra de Canyet S/N, 08916, Badalona, Spain
- Universitat de Vic-Universitat Central de Catalunya, Vic, Spain
- Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Department of Infectious Diseasest &, Lluita Contra La SIDA Foundation, Hospital Universitari Germans Trias I Pujol, Badalona, Spain
- Infectious Disease Networking Biomedical Research Center (CIBERINFEC), Carlos III Health Institute, Madrid, Spain
| | - Marc Noguera-Julian
- IrsiCaixa, Ctra de Canyet S/N, 08916, Badalona, Spain
- Universitat de Vic-Universitat Central de Catalunya, Vic, Spain
- Infectious Disease Networking Biomedical Research Center (CIBERINFEC), Carlos III Health Institute, Madrid, Spain
| |
Collapse
|
30
|
Kühnen P, Argente J, Clément K, Dollfus H, Dubern B, Farooqi S, de Groot C, Grüters A, Holm JC, Hopkins M, Kleinendorst L, Körner A, Meeker D, Rydén M, von Schnurbein J, Tschöp M, Yeo GSH, Zorn S, Wabitsch M. IMPROVE 2022 International Meeting on Pathway-Related Obesity: Vision of Excellence. Clin Obes 2024; 14:e12659. [PMID: 38602039 DOI: 10.1111/cob.12659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 03/08/2024] [Indexed: 04/12/2024]
Abstract
Nearly 90 clinicians and researchers from around the world attended the first IMPROVE 2022 International Meeting on Pathway-Related Obesity. Delegates attended in person or online from across Europe, Argentina and Israel to hear the latest scientific and clinical developments in hyperphagia and severe, early-onset obesity, and set out a vision of excellence for the future for improving the diagnosis, treatment, and care of patients with melanocortin-4 receptor (MC4R) pathway-related obesity. The meeting co-chair Peter Kühnen, Charité Universitätsmedizin Berlin, Germany, indicated that change was needed with the rapidly increasing prevalence of obesity and the associated complications to improve the understanding of the underlying mechanisms and acknowledge that monogenic forms of obesity can play an important role, providing insights that can be applied to a wider group of patients with obesity. World-leading experts presented the latest research and led discussions on the underlying science of obesity, diagnosis (including clinical and genetic approaches such as the role of defective MC4R signalling), and emerging clinical data and research with targeted pharmacological approaches. The aim of the meeting was to agree on the questions that needed to be addressed in future research and to ensure that optimised diagnostic work-up was used with new genetic testing tools becoming available. This should aid the planning of new evidence-based treatment strategies for the future, as explained by co-chair Martin Wabitsch, Ulm University Medical Center, Germany.
Collapse
Affiliation(s)
- Peter Kühnen
- Department of Pediatric Endocrinology and Diabetology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jesús Argente
- Departments of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Karine Clément
- Assistance Publique-Hôpitaux de Paris, Nutrition Department, Pitié-Salpêtrière Hospital, Paris, France
- INSERM, Nutrition and Obesity: Systemic Approaches, NutriOmics, Research Unit, Sorbonne Université, Paris, France
| | - Hélène Dollfus
- CARGO and Department of Medical Genetics, University of Strasbourg, Strasbourg, France
| | - Béatrice Dubern
- INSERM, Nutrition and Obesity: Systemic Approaches, NutriOmics, Research Unit, Sorbonne Université, Paris, France
- Sorbonne Université, Trousseau Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Sadaf Farooqi
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Corjan de Groot
- Sophia Children's Hospital, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Annette Grüters
- Department of Pediatric Endocrinology and Diabetes, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jens-Christian Holm
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Pediatrics, Copenhagen University Hospital Holbæk, Copenhagen, Denmark
| | - Mark Hopkins
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Lotte Kleinendorst
- Department of Clinical Genetics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Antje Körner
- Center for Pediatric Research, Department of Pediatrics, LIFE Research Center for Civilization Diseases, University Hospital Leipzig, Leipzig, Germany
| | - David Meeker
- Rhythm Pharmaceuticals, Boston, Massachusetts, USA
| | - Mikael Rydén
- Department of Medicine H7, Karolinska Institute, Stockholm, Sweden
- Department of Endocrinology and Metabolism, Karolinska University Hospital, Stockholm, Sweden
| | - Julia von Schnurbein
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Matthias Tschöp
- Institute for Diabetes and Obesity, Helmholtz Zentrum, Munich, Germany
| | - Giles S H Yeo
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Stefanie Zorn
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Martin Wabitsch
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
31
|
Deehan EC, Mocanu V, Madsen KL. Effects of dietary fibre on metabolic health and obesity. Nat Rev Gastroenterol Hepatol 2024; 21:301-318. [PMID: 38326443 DOI: 10.1038/s41575-023-00891-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/18/2023] [Indexed: 02/09/2024]
Abstract
Obesity and metabolic syndrome represent a growing epidemic worldwide. Body weight is regulated through complex interactions between hormonal, neural and metabolic pathways and is influenced by numerous environmental factors. Imbalances between energy intake and expenditure can occur due to several factors, including alterations in eating behaviours, abnormal satiation and satiety, and low energy expenditure. The gut microbiota profoundly affects all aspects of energy homeostasis through diverse mechanisms involving effects on mucosal and systemic immune, hormonal and neural systems. The benefits of dietary fibre on metabolism and obesity have been demonstrated through mechanistic studies and clinical trials, but many questions remain as to how different fibres are best utilized in managing obesity. In this Review, we discuss the physiochemical properties of different fibres, current findings on how fibre and the gut microbiota interact to regulate body weight homeostasis, and knowledge gaps related to using dietary fibres as a complementary strategy. Precision medicine approaches that utilize baseline microbiota and clinical characteristics to predict individual responses to fibre supplementation represent a new paradigm with great potential to enhance weight management efficacy, but many challenges remain before these approaches can be fully implemented.
Collapse
Affiliation(s)
- Edward C Deehan
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, USA
- Nebraska Food for Health Center, Lincoln, NE, USA
| | - Valentin Mocanu
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Karen L Madsen
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
32
|
Garcia MM, Romero AS, Merkley SD, Meyer-Hagen JL, Forbes C, Hayek EE, Sciezka DP, Templeton R, Gonzalez-Estrella J, Jin Y, Gu H, Benavidez A, Hunter RP, Lucas S, Herbert G, Kim KJ, Cui JY, Gullapalli RR, In JG, Campen MJ, Castillo EF. In Vivo Tissue Distribution of Polystyrene or Mixed Polymer Microspheres and Metabolomic Analysis after Oral Exposure in Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:47005. [PMID: 38598326 PMCID: PMC11005960 DOI: 10.1289/ehp13435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 01/05/2024] [Accepted: 02/23/2024] [Indexed: 04/12/2024]
Abstract
BACKGROUND Global plastic use has consistently increased over the past century with several different types of plastics now being produced. Much of these plastics end up in oceans or landfills leading to a substantial accumulation of plastics in the environment. Plastic debris slowly degrades into microplastics (MPs) that can ultimately be inhaled or ingested by both animals and humans. A growing body of evidence indicates that MPs can cross the gut barrier and enter into the lymphatic and systemic circulation leading to accumulation in tissues such as the lungs, liver, kidney, and brain. The impacts of mixed MPs exposure on tissue function through metabolism remains largely unexplored. OBJECTIVES This study aims to investigate the impacts of polymer microspheres on tissue metabolism in mice by assessing the microspheres ability to translocate across the gut barrier and enter into systemic circulation. Specifically, we wanted to examine microsphere accumulation in different organ systems, identify concentration-dependent metabolic changes, and evaluate the effects of mixed microsphere exposures on health outcomes. METHODS To investigate the impact of ingested microspheres on target metabolic pathways, mice were exposed to either polystyrene (5 μ m ) microspheres or a mixture of polymer microspheres consisting of polystyrene (5 μ m ), polyethylene (1 - 4 μ m ), and the biodegradability and biocompatible plastic, poly-(lactic-co-glycolic acid) (5 μ m ). Exposures were performed twice a week for 4 weeks at a concentration of either 0, 2, or 4 mg / week via oral gastric gavage. Tissues were collected to examine microsphere ingress and changes in metabolites. RESULTS In mice that ingested microspheres, we detected polystyrene microspheres in distant tissues including the brain, liver, and kidney. Additionally, we report on the metabolic differences that occurred in the colon, liver, and brain, which showed differential responses that were dependent on concentration and type of microsphere exposure. DISCUSSION This study uses a mouse model to provide critical insight into the potential health implications of the pervasive issue of plastic pollution. These findings demonstrate that orally consumed polystyrene or mixed polymer microspheres can accumulate in tissues such as the brain, liver, and kidney. Furthermore, this study highlights concentration-dependent and polymer type-specific metabolic changes in the colon, liver, and brain after plastic microsphere exposure. These results underline the mobility within and between biological tissues of MPs after exposure and emphasize the importance of understanding their metabolic impact. https://doi.org/10.1289/EHP13435.
Collapse
Affiliation(s)
- Marcus M. Garcia
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences, Albuquerque, New Mexico, USA
| | - Aaron S. Romero
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Seth D. Merkley
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Jewel L. Meyer-Hagen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Charles Forbes
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Eliane El Hayek
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences, Albuquerque, New Mexico, USA
| | - David P. Sciezka
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences, Albuquerque, New Mexico, USA
| | - Rachel Templeton
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences, Albuquerque, New Mexico, USA
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jorge Gonzalez-Estrella
- School of Civil & Environmental Engineering, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Yan Jin
- Center for Translational Science, Florida International University, Port St. Lucie, Florida, USA
| | - Haiwei Gu
- Center for Translational Science, Florida International University, Port St. Lucie, Florida, USA
| | - Angelica Benavidez
- Center for Micro-Engineered Materials, University of New Mexico, Albuquerque, New Mexico, USA
| | - Russell P. Hunter
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences, Albuquerque, New Mexico, USA
| | - Selita Lucas
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences, Albuquerque, New Mexico, USA
| | - Guy Herbert
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences, Albuquerque, New Mexico, USA
| | - Kyle Joohyung Kim
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, Washington, USA
| | - Julia Yue Cui
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, Washington, USA
| | - Rama R. Gullapalli
- Department of Pathology, University of New Mexico Health Sciences, Albuquerque, New Mexico, USA
| | - Julie G. In
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Matthew J. Campen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences, Albuquerque, New Mexico, USA
| | - Eliseo F. Castillo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| |
Collapse
|
33
|
Deng Y, Hu M, Huang S, Fu N. Molecular mechanism and therapeutic significance of essential amino acids in metabolically associated fatty liver disease. J Nutr Biochem 2024; 126:109581. [PMID: 38219809 DOI: 10.1016/j.jnutbio.2024.109581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/01/2024] [Accepted: 01/06/2024] [Indexed: 01/16/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD), also known as metabolically associated fatty liver disease (MAFLD), is a systemic metabolic disease characterized by lipid accumulation in the liver, lipid toxicity, insulin resistance, intestinal dysbiosis, and inflammation that can progress from simple steatosis to nonalcoholic steatohepatitis (NASH) and even cirrhosis or cancer. It is the most prevalent illness threatening world health. Currently, there are almost no approved drug interventions for MAFLD, mainly dietary changes and exercise to control weight and regulate metabolic disorders. Meanwhile, the metabolic pathway involved in amino acid metabolism also influences the onset and development of MAFLD in the body, and most amino acid metabolism takes place in the liver. Essential amino acids are those amino acids that must be supplemented from outside the diet and that cannot be synthesized in the body or cannot be synthesized at a rate sufficient to meet the body's needs, including leucine, isoleucine, valine (collectively known as branched-chain amino acids), tryptophan, phenylalanine (which are aromatic amino acids), histidine, methionine, threonine and lysine. The metabolic balance of the body is closely linked to these essential amino acids, and essential amino acids are closely linked to the pathophysiological process of MAFLD. In this paper, we will focus on the metabolism of essential amino acids in the body and further explore the therapeutic strategies for MAFLD based on the studies conducted in recent years.
Collapse
Affiliation(s)
- Yuting Deng
- The Affiliated Nanhua Hospital, Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Mengsi Hu
- The Affiliated Nanhua Hospital, Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Shufang Huang
- The Affiliated Nanhua Hospital, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China.
| | - Nian Fu
- The Affiliated Nanhua Hospital, Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China; The Affiliated Nanhua Hospital, Institute of Clinical Research, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China.
| |
Collapse
|
34
|
Roy G, Prifti E, Belda E, Zucker JD. Deep learning methods in metagenomics: a review. Microb Genom 2024; 10:001231. [PMID: 38630611 PMCID: PMC11092122 DOI: 10.1099/mgen.0.001231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/27/2024] [Indexed: 04/19/2024] Open
Abstract
The ever-decreasing cost of sequencing and the growing potential applications of metagenomics have led to an unprecedented surge in data generation. One of the most prevalent applications of metagenomics is the study of microbial environments, such as the human gut. The gut microbiome plays a crucial role in human health, providing vital information for patient diagnosis and prognosis. However, analysing metagenomic data remains challenging due to several factors, including reference catalogues, sparsity and compositionality. Deep learning (DL) enables novel and promising approaches that complement state-of-the-art microbiome pipelines. DL-based methods can address almost all aspects of microbiome analysis, including novel pathogen detection, sequence classification, patient stratification and disease prediction. Beyond generating predictive models, a key aspect of these methods is also their interpretability. This article reviews DL approaches in metagenomics, including convolutional networks, autoencoders and attention-based models. These methods aggregate contextualized data and pave the way for improved patient care and a better understanding of the microbiome's key role in our health.
Collapse
Affiliation(s)
- Gaspar Roy
- IRD, Sorbonne University, UMMISCO, 32 avenue Henry Varagnat, Bondy Cedex, France
| | - Edi Prifti
- IRD, Sorbonne University, UMMISCO, 32 avenue Henry Varagnat, Bondy Cedex, France
- Sorbonne University, INSERM, Nutriomics, 91 bvd de l’hopital, 75013 Paris, France
| | - Eugeni Belda
- IRD, Sorbonne University, UMMISCO, 32 avenue Henry Varagnat, Bondy Cedex, France
- Sorbonne University, INSERM, Nutriomics, 91 bvd de l’hopital, 75013 Paris, France
| | - Jean-Daniel Zucker
- IRD, Sorbonne University, UMMISCO, 32 avenue Henry Varagnat, Bondy Cedex, France
- Sorbonne University, INSERM, Nutriomics, 91 bvd de l’hopital, 75013 Paris, France
| |
Collapse
|
35
|
Zhang L, Wang P, Huang J, Xing Y, Wong FS, Suo J, Wen L. Gut microbiota and therapy for obesity and type 2 diabetes. Front Endocrinol (Lausanne) 2024; 15:1333778. [PMID: 38596222 PMCID: PMC11002083 DOI: 10.3389/fendo.2024.1333778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/06/2024] [Indexed: 04/11/2024] Open
Abstract
There has been a major increase in Type 2 diabetes and obesity in many countries, and this will lead to a global public health crisis, which not only impacts on the quality of life of individuals well but also places a substantial burden on healthcare systems and economies. Obesity is linked to not only to type 2 diabetes but also cardiovascular diseases, musculoskeletal disorders, and certain cancers, also resulting in increased medical costs and diminished quality of life. A number of studies have linked changes in gut in obesity development. Dysbiosis, a deleterious change in gut microbiota composition, leads to altered intestinal permeability, associated with obesity and Type 2 diabetes. Many factors affect the homeostasis of gut microbiota, including diet, genetics, circadian rhythms, medication, probiotics, and antibiotics. In addition, bariatric surgery induces changes in gut microbiota that contributes to the metabolic benefits observed post-surgery. Current obesity management strategies encompass dietary interventions, exercise, pharmacotherapy, and bariatric surgery, with emerging treatments including microbiota-altering approaches showing promising efficacy. While pharmacotherapy has demonstrated significant advancements in recent years, bariatric surgery remains one of the most effective treatments for sustainable weight loss. However, access to this is generally limited to those living with severe obesity. This underscores the need for non-surgical interventions, particularly for adolescents and mildly obese patients. In this comprehensive review, we assess longitudinal alterations in gut microbiota composition and functionality resulting from the two currently most effective anti-obesity treatments: pharmacotherapy and bariatric surgery. Additionally, we highlight the functions of gut microbiota, focusing on specific bacteria, their metabolites, and strategies for modulating gut microbiota to prevent and treat obesity. This review aims to provide insights into the evolving landscape of obesity management and the potential of microbiota-based approaches in addressing this pressing global health challenge.
Collapse
Affiliation(s)
- Luyao Zhang
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - Pai Wang
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - Juan Huang
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha, Hunan, China
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanpeng Xing
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - F Susan Wong
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Jian Suo
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Li Wen
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
36
|
d'Humières C, Delavy M, Alla L, Ichou F, Gauliard E, Ghozlane A, Levenez F, Galleron N, Quinquis B, Pons N, Mullaert J, Bridier-Nahmias A, Condamine B, Touchon M, Rainteau D, Lamazière A, Lesnik P, Ponnaiah M, Lhomme M, Sertour N, Devente S, Docquier JD, Bougnoux ME, Tenaillon O, Magnan M, Ruppé E, Grall N, Duval X, Ehrlich D, Mentré F, Denamur E, Rocha EPC, Le Chatelier E, Burdet C. Perturbation and resilience of the gut microbiome up to 3 months after β-lactams exposure in healthy volunteers suggest an important role of microbial β-lactamases. MICROBIOME 2024; 12:50. [PMID: 38468305 DOI: 10.1186/s40168-023-01746-0] [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: 03/14/2023] [Accepted: 12/20/2023] [Indexed: 03/13/2024]
Abstract
BACKGROUND Antibiotics notoriously perturb the gut microbiota. We treated healthy volunteers either with cefotaxime or ceftriaxone for 3 days, and collected in each subject 12 faecal samples up to day 90. Using untargeted and targeted phenotypic and genotypic approaches, we studied the changes in the bacterial, phage and fungal components of the microbiota as well as the metabolome and the β-lactamase activity of the stools. This allowed assessing their degrees of perturbation and resilience. RESULTS While only two subjects had detectable concentrations of antibiotics in their faeces, suggesting important antibiotic degradation in the gut, the intravenous treatment perturbed very significantly the bacterial and phage microbiota, as well as the composition of the metabolome. In contrast, treatment impact was relatively low on the fungal microbiota. At the end of the surveillance period, we found evidence of resilience across the gut system since most components returned to a state like the initial one, even if the structure of the bacterial microbiota changed and the dynamics of the different components over time were rarely correlated. The observed richness of the antibiotic resistance genes repertoire was significantly reduced up to day 30, while a significant increase in the relative abundance of β-lactamase encoding genes was observed up to day 10, consistent with a concomitant increase in the β-lactamase activity of the microbiota. The level of β-lactamase activity at baseline was positively associated with the resilience of the metabolome content of the stools. CONCLUSIONS In healthy adults, antibiotics perturb many components of the microbiota, which return close to the baseline state within 30 days. These data suggest an important role of endogenous β-lactamase-producing anaerobes in protecting the functions of the microbiota by de-activating the antibiotics reaching the colon. Video Abstract.
Collapse
Affiliation(s)
- Camille d'Humières
- Université Paris Cité, IAME, INSERM, Paris, F-75018, France
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Microbial Evolutionary Genomics, Paris, 75015, France
| | - Margot Delavy
- Institut Pasteur, Université Paris Cité, INRAE, USC2019, Unité Biologie Et Pathogénicité Fongiques, Paris, F-75015, France
| | - Laurie Alla
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, F-78350, France
| | - Farid Ichou
- ICANomics, Foundation of Innovation in Cardiometabolism and Nutrition (IHU ICAN), Paris, F-75013, France
| | - Emilie Gauliard
- Sorbonne Université, INSERM U938, Centre de Recherche Saint-Antoine, Paris, F-75012, France
| | - Amine Ghozlane
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, F-75015, France
| | - Florence Levenez
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, F-78350, France
| | - Nathalie Galleron
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, F-78350, France
| | - Benoit Quinquis
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, F-78350, France
| | - Nicolas Pons
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, F-78350, France
| | - Jimmy Mullaert
- Université Paris Cité, IAME, INSERM, Paris, F-75018, France
- AP-HP, Département d'Epidemiologie, Biostatistique and Recherche Clinique, Hôpital Bichat, Paris, F-75018, France
| | | | | | - Marie Touchon
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Microbial Evolutionary Genomics, Paris, 75015, France
| | - Dominique Rainteau
- Sorbonne Université, INSERM U938, Centre de Recherche Saint-Antoine, Paris, F-75012, France
| | - Antonin Lamazière
- Sorbonne Université, INSERM U938, Centre de Recherche Saint-Antoine, Paris, F-75012, France
| | - Philippe Lesnik
- INSERM UMR-S 1166, Institute of Cardiometabolism and Nutrition, Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, F-75013, France
- ICANomics, Foundation of Innovation in Cardiometabolism and Nutrition (IHU ICAN), Paris, F-75013, France
| | - Maharajah Ponnaiah
- ICANomics, Foundation of Innovation in Cardiometabolism and Nutrition (IHU ICAN), Paris, F-75013, France
| | - Marie Lhomme
- ICANomics, Foundation of Innovation in Cardiometabolism and Nutrition (IHU ICAN), Paris, F-75013, France
| | - Natacha Sertour
- Institut Pasteur, Université Paris Cité, INRAE, USC2019, Unité Biologie Et Pathogénicité Fongiques, Paris, F-75015, France
| | - Savannah Devente
- Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, I-53100, Italy
| | - Jean-Denis Docquier
- Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, I-53100, Italy
| | - Marie-Elisabeth Bougnoux
- Institut Pasteur, Université Paris Cité, INRAE, USC2019, Unité Biologie Et Pathogénicité Fongiques, Paris, F-75015, France
- AP-HP, Unité de Parasitologie-Mycologie, Service de Microbiologie Clinique, Hôpital Necker-Enfants-Malades, Paris, F-75015, France
| | | | - Mélanie Magnan
- Université Paris Cité, IAME, INSERM, Paris, F-75018, France
| | - Etienne Ruppé
- Université Paris Cité, IAME, INSERM, Paris, F-75018, France
- AP-HP, Laboratoire de Bactériologie, Hôpital Bichat, Paris, F-75018, France
| | - Nathalie Grall
- Université Paris Cité, IAME, INSERM, Paris, F-75018, France
- AP-HP, Laboratoire de Bactériologie, Hôpital Bichat, Paris, F-75018, France
| | - Xavier Duval
- Université Paris Cité, IAME, INSERM, Paris, F-75018, France
- AP-HP, Centre d'Investigation Clinique, INSERM CIC 1425, Hôpital Bichat, Paris, F-75018, France
| | - Dusko Ehrlich
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, F-78350, France
- University College London, Institute for Neurology, London, UK
| | - France Mentré
- Université Paris Cité, IAME, INSERM, Paris, F-75018, France
- AP-HP, Département d'Epidemiologie, Biostatistique and Recherche Clinique, Hôpital Bichat, Paris, F-75018, France
| | - Erick Denamur
- Université Paris Cité, IAME, INSERM, Paris, F-75018, France
- AP-HP, Laboratoire de Génétique Moléculaire, Hôpital Bichat, Paris, F-75018, France
| | - Eduardo P C Rocha
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Microbial Evolutionary Genomics, Paris, 75015, France
| | | | - Charles Burdet
- Université Paris Cité, IAME, INSERM, Paris, F-75018, France.
- AP-HP, Département d'Epidemiologie, Biostatistique and Recherche Clinique, Hôpital Bichat, Paris, F-75018, France.
| |
Collapse
|
37
|
Lee HK, Kim NE, Shin CM, Oh TJ, Yoon H, Park YS, Kim N, Won S, Lee DH. Gut microbiome signature of metabolically healthy obese individuals according to anthropometric, metabolic and inflammatory parameters. Sci Rep 2024; 14:3449. [PMID: 38342934 PMCID: PMC10859373 DOI: 10.1038/s41598-024-53837-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 02/06/2024] [Indexed: 02/13/2024] Open
Abstract
In this study, we investigated the characteristics of gut microbiome in the metabolically healthy obese (MHO) patients, and how they correlate with metabolic and inflammatory profiles. A total of 120 obese people without metabolic comorbidities were recruited, and their clinical phenotypes, metabolic and inflammatory parameters were analysed. The faecal microbial markers originating from bacterial cell and extracellular vesicle (EV) were profiled using 16S rDNA sequencing. The total study population could be classified into two distinct enterotypes (enterotype I: Prevotellaceae-predominant, enterotype II: Akkermansia/Bacteroides-predominant), based on their stool EV-derived microbiome profile. When comparing the metabolic and inflammatory profiles, subjects in enterotype I had higher levels of serum IL-1β [false discovery rate (FDR) q = 0.050] and had a lower level of microbial diversity than enterotype II (Wilcoxon rank-sum test p < 0.01). Subjects in enterotype I had relatively higher abundance of Bacteroidetes, Prevotellaceae and Prevotella-derived EVs, and lower abundance of Actinobacteria, Firmicutes, Proteobacteria, Akkermansia and Bacteroides-derived EVs (FDR q < 0.05). In conclusion, HMO patients can be categorised into two distinct enterotypes by the faecal EV-derived microbiome profile. The enterotyping may be associated with different metabolic and inflammatory profiles. Further studies are warranted to elucidate the long-term prognostic impact of EV-derived microbiome in the obese population.
Collapse
Affiliation(s)
- Ho-Kyoung Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
| | - Nam-Eun Kim
- Institute of Health and Environment, Seoul National University, Seoul, South Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea.
| | - Tae Jung Oh
- Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
| | - Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
| | - Young Soo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
| | - Sungho Won
- Department of Public Health Sciences, Seoul National University, Seoul, South Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea.
| |
Collapse
|
38
|
Liu C, Xu Q, Dong S, Ding H, Li B, Zhang D, Liang Y, Li L, Liu Q, Cheng Y, Wu J, Zhu J, Zhong M, Cao Y, Zhang G. New mechanistic insights of anti-obesity by sleeve gastrectomy-altered gut microbiota and lipid metabolism. Front Endocrinol (Lausanne) 2024; 15:1338147. [PMID: 38375198 PMCID: PMC10875461 DOI: 10.3389/fendo.2024.1338147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/09/2024] [Indexed: 02/21/2024] Open
Abstract
Background The obesity epidemic has been on the rise due to changes in living standards and lifestyles. To combat this issue, sleeve gastrectomy (SG) has emerged as a prominent bariatric surgery technique, offering substantial weight reduction. Nevertheless, the mechanisms that underlie SG-related bodyweight loss are not fully understood. Methods In this study, we conducted a collection of preoperative and 3-month postoperative serum and fecal samples from patients who underwent laparoscopic SG at the First Affiliated Hospital of Shandong First Medical University (Jinan, China). Here, we took an unbiased approach of multi-omics to investigate the role of SG-altered gut microbiota in anti-obesity of these patients. Non-target metabolome sequencing was performed using the fecal and serum samples. Results Our data show that SG markedly increased microbiota diversity and Rikenellaceae, Alistipes, Parabacteroides, Bactreoidales, and Enterobacteraies robustly increased. These compositional changes were positively correlated with lipid metabolites, including sphingolipids, glycerophospholipids, and unsaturated fatty acids. Increases of Rikenellaceae, Alistipes, and Parabacteroide were reversely correlated with body mass index (BMI). Conclusion In conclusion, our findings provide evidence that SG induces significant alterations in the abundances of Rikenellaceae, Alistipes, Parabacteroides, and Bacteroidales, as well as changes in lipid metabolism-related metabolites. Importantly, these changes were found to be closely linked to the alleviation of obesity. On the basis of these findings, we have identified a number of microbiotas that could be potential targets for treatment of obesity.
Collapse
Affiliation(s)
- Chuxuan Liu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Qian Xu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Shuohui Dong
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Huanxin Ding
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Bingjun Li
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Dexu Zhang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Yongjuan Liang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Linchuan Li
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Qiaoran Liu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Yugang Cheng
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Jing Wu
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Jiankang Zhu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Mingwei Zhong
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Guangyong Zhang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
39
|
Nogacka AM, Saturio S, Alvarado-Jasso GM, Salazar N, de los Reyes Gavilán CG, Martínez-Faedo C, Suarez A, Wang R, Miyazawa K, Harata G, Endo A, Arboleya S, Gueimonde M. Probiotic-Induced Modulation of Microbiota Composition and Antibiotic Resistance Genes Load, an In Vitro Assessment. Int J Mol Sci 2024; 25:1003. [PMID: 38256076 PMCID: PMC10816173 DOI: 10.3390/ijms25021003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
The imbalance of the gut microbiota (GM) is known as dysbiosis and is associated with disorders such as obesity. The increasing prevalence of microorganisms harboring antibiotic resistance genes (ARG) in the GM has been reported as a potential risk for spreading multi-drug-resistant pathogens. The objective of this work was the evaluation, in a fecal culture model, of different probiotics for their ability to modulate GM composition and ARG levels on two population groups, extremely obese (OB) and normal-weight (NW) subjects. Clear differences in the basal microbiota composition were observed between NW and OB donors. The microbial profile assessed by metataxonomics revealed the broader impact of probiotics on the OB microbiota composition. Also, supplementation with probiotics promoted significant reductions in the absolute levels of tetM and tetO genes. Regarding the blaTEM gene, a minor but significant decrease in both donor groups was detected after probiotic addition. A negative association between the abundance of Bifidobacteriaceae and the tetM gene was observed. Our results show the ability of some of the tested strains to modulate GM. Moreover, the results suggest the potential application of probiotics for reducing the levels of ARG, which constitutes an interesting target for the future development of probiotics.
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), 33300 Villaviciosa, Spain; (A.M.N.); (S.S.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.G.); (S.A.)
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (DIMISA-ISPA), 33011 Oviedo, Spain;
| | - Silvia Saturio
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain; (A.M.N.); (S.S.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.G.); (S.A.)
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (DIMISA-ISPA), 33011 Oviedo, Spain;
| | - Guadalupe Monserrat Alvarado-Jasso
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain; (A.M.N.); (S.S.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.G.); (S.A.)
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain; (A.M.N.); (S.S.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.G.); (S.A.)
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (DIMISA-ISPA), 33011 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), 33300 Villaviciosa, Spain; (A.M.N.); (S.S.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.G.); (S.A.)
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (DIMISA-ISPA), 33011 Oviedo, Spain;
| | - Ceferino Martínez-Faedo
- Endocrinology and Nutrition Service, Central University Hospital of Asturias (HUCA), 33011 Oviedo, Spain;
- Endocrinology, Nutrition, Diabetes and Obesity Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Adolfo Suarez
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (DIMISA-ISPA), 33011 Oviedo, Spain;
- Digestive Service, Central University Hospital of Asturias (HUCA), 33011 Oviedo, Spain
| | - Ruipeng Wang
- Technical Research Laboratory, Takanashi Milk Products Co., Ltd., Yokohama 241-0021, Japan; (R.W.); (K.M.); (G.H.)
| | - Kenji Miyazawa
- Technical Research Laboratory, Takanashi Milk Products Co., Ltd., Yokohama 241-0021, Japan; (R.W.); (K.M.); (G.H.)
| | - Gaku Harata
- Technical Research Laboratory, Takanashi Milk Products Co., Ltd., Yokohama 241-0021, Japan; (R.W.); (K.M.); (G.H.)
| | - Akihito Endo
- Department of Food, Aroma and Cosmetic Chemistry, Tokyo University of Agriculture, Abashiri, Hokkaido 099-2493, Japan;
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain; (A.M.N.); (S.S.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.G.); (S.A.)
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (DIMISA-ISPA), 33011 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), 33300 Villaviciosa, Spain; (A.M.N.); (S.S.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.G.); (S.A.)
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (DIMISA-ISPA), 33011 Oviedo, Spain;
| |
Collapse
|
40
|
Saberi-Karimian M, Safarian-Bana H, Fazeli M, Tabatabaeizadeh SA, Ferns GA, Ghayour-Mobarhan M. Gut microbiota and metabolic syndrome: What's new? METABOLIC SYNDROME 2024:527-541. [DOI: 10.1016/b978-0-323-85732-1.00050-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
41
|
Steinbach E, Belda E, Alili R, Adriouch S, Dauriat CJG, Donatelli G, Dumont JL, Pacini F, Tuszynski T, Pelloux V, Jacques F, Creusot L, Coles E, Taillandier P, Vazquez Gomez M, Masi D, Mateo V, André S, Kordahi M, Rouault C, Zucker JD, Sokol H, Genser L, Chassaing B, Le Roy T, Clément K. Comparative analysis of the duodenojejunal microbiome with the oral and fecal microbiomes reveals its stronger association with obesity and nutrition. Gut Microbes 2024; 16:2405547. [PMID: 39679619 DOI: 10.1080/19490976.2024.2405547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/24/2024] [Accepted: 09/10/2024] [Indexed: 12/17/2024] Open
Abstract
The intestinal microbiota is increasingly recognized as a crucial player in the development and maintenance of various chronic conditions, including obesity and associated metabolic diseases. While most research focuses on the fecal microbiota due to its easier accessibility, the small intestine, as a major site for nutrient sensing and absorption, warrants further investigation to determine its microbiota composition and functions. Here, we conducted a clinical research project in 30 age- and sex-matched participants with (n = 15) and without (n = 15) obesity. Duodenojejunal fluid was obtained by aspiration during endoscopy. Phenotyping included clinical variables related to metabolic status, lifestyle, and psychosocial factors using validated questionnaires. We performed metagenomic analyses of the oral, duodenojejunal, and fecal microbiome, alongside metabolomic data from duodenojejunal fluid and feces, integrating these data with clinical and lifestyle information. Our results highlight significant associations between duodenojejunal microbiota composition and usual dietary intake, as well as clinical phenotypes, with larger effect sizes than the associations between these variables and fecal microbiota. Notably, we found that the duodenojejunal microbiota of patients with obesity exhibited higher diversity and showed distinct differences in the abundance of several duodenojejunal microbiota species compared with individuals without obesity. Our findings support the relevance of studying the role of the small intestinal microbiota in the pathogenesis of nutrition-related diseases.
Collapse
Affiliation(s)
- Emilie Steinbach
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Eugeni Belda
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
- Unité de Modélisation Mathématique et Informatique des Systèmes Complexes, UMMISCO, Sorbonne Université, Institut de Recherche pour le Développement (IRD), Paris, France
| | - Rohia Alili
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Solia Adriouch
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Charlène J G Dauriat
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM, Paris, France
- Mucosal Microbiota in Chronic Inflammatory Diseases, INSERM, CNRS UMR8104, Université de Paris, Paris, France
| | - Gianfranco Donatelli
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
- Endoscopy Department, Peupliers Hospital, Ramsay-Santé, Paris, France
| | - Jean-Loup Dumont
- Endoscopy Department, Peupliers Hospital, Ramsay-Santé, Paris, France
| | - Filippo Pacini
- Endoscopy Department, Peupliers Hospital, Ramsay-Santé, Paris, France
| | - Thierry Tuszynski
- Endoscopy Department, Peupliers Hospital, Ramsay-Santé, Paris, France
| | - Véronique Pelloux
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Flavien Jacques
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Laura Creusot
- Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Paris, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, France
| | - Emavieve Coles
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Paul Taillandier
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Marta Vazquez Gomez
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Davide Masi
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, Sapienza University of Rome, Rome, Italy
| | - Véronique Mateo
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Sébastien André
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Melissa Kordahi
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM, Paris, France
- Mucosal Microbiota in Chronic Inflammatory Diseases, INSERM, CNRS UMR8104, Université de Paris, Paris, France
| | - Christine Rouault
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Jean-Daniel Zucker
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
- Unité de Modélisation Mathématique et Informatique des Systèmes Complexes, UMMISCO, Sorbonne Université, Institut de Recherche pour le Développement (IRD), Paris, France
| | - Harry Sokol
- Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Paris, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, France
- INRAE, UMR1319 Micalis & AgroParisTech, Jouy en Josas, France
| | - Laurent Genser
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
- Sorbonne Université, Department of Hepato-Biliary and Pancreatic Surgery, Assistance Publique-Hôpitaux de Paris, AP-HP, Pitié-Salpêtrière University Hospital, Paris, France
| | - Benoit Chassaing
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM, Paris, France
- Mucosal Microbiota in Chronic Inflammatory Diseases, INSERM, CNRS UMR8104, Université de Paris, Paris, France
| | - Tiphaine Le Roy
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Karine Clément
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
- Assistance Publique Hôpitaux de Paris, Nutrition Department, Pitié-Salpêtrière Hospital, Paris, France
| |
Collapse
|
42
|
Zhang Z, Mocanu V, Deehan EC, Hotte N, Zhu Y, Wei S, Kao DH, Karmali S, Birch DW, Walter J, Madsen KL. Recipient microbiome-related features predicting metabolic improvement following fecal microbiota transplantation in adults with severe obesity and metabolic syndrome: a secondary analysis of a phase 2 clinical trial. Gut Microbes 2024; 16:2345134. [PMID: 38685731 PMCID: PMC11062372 DOI: 10.1080/19490976.2024.2345134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 04/16/2024] [Indexed: 05/02/2024] Open
Abstract
Microbial-based therapeutics in clinical practice are of considerable interest, and a recent study demonstrated fecal microbial transplantation (FMT) followed by dietary fiber supplements improved glucose homeostasis. Previous evidence suggests that donor and recipient compatibility and FMT protocol are key determinants, but little is known about the involvement of specific recipient factors. Using data from our recent randomized placebo-control phase 2 clinical trial in adults with obesity and metabolic syndrome, we grouped participants that received FMT from one of 4 donors with either fiber supplement into HOMA-IR responders (n = 21) and HOMA-IR non-responders (n = 8). We further assessed plasma bile acids using targeted metabolomics and performed subgroup analyzes to evaluate the effects of recipient parameters and gastrointestinal factors on microbiota engraftment and homeostatic model assessment of insulin resistance (HOMA2-IR) response. The baseline fecal microbiota composition at genus level of recipients could predict the improvements in HOMA2-IR at week 6 (ROC-AUC = 0.70). Prevotella was identified as an important predictor, with responders having significantly lower relative abundance than non-responders (p = .02). In addition, recipients displayed a highly individualized degree of microbial engraftment from donors. Compared to the non-responders, the responders had significantly increased bacterial richness (Chao1) after FMT and a more consistent engraftment of donor-specific bacteria ASVs (amplicon sequence variants) such as Faecalibacillus intestinalis (ASV44), Roseburia spp. (ASV103), and Christensenellaceae spp. (ASV140) (p < .05). Microbiota engraftment was strongly associated with recipients' factors at baseline including initial gut microbial diversity, fiber and nutrient intakes, inflammatory markers, and bile acid derivative levels. This study identified that responders to FMT therapy had a higher engraftment rate in the transplantation of specific donor-specific microbes, which were strongly correlated with insulin sensitivity improvements. Further, the recipient baseline gut microbiota and related factors were identified as the determinants for responsiveness to FMT and fiber supplementation. The findings provide a basis for the development of precision microbial therapeutics for the treatment of metabolic syndrome.
Collapse
Affiliation(s)
- Zhengxiao Zhang
- College of Ocean Food and Biological Engineering, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian, China
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Valentin Mocanu
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Edward C. Deehan
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, USA
| | - Naomi Hotte
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Yuanyuan Zhu
- College of Ocean Food and Biological Engineering, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian, China
| | - Shanshan Wei
- College of Ocean Food and Biological Engineering, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian, China
| | - Dina H. Kao
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Shahzeer Karmali
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Daniel W. Birch
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Jens Walter
- APC Microbiome Ireland, School of Microbiology and Department of Medicine, University College Cork, Cork, Ireland
| | - Karen L. Madsen
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
43
|
Cabanillas-Lazo M, Quispe-Vicuña C, Pascual-Guevara M, Guerrero ME, Barja-Ore J, Espinoza-Carhuancho F, Mayta-Tovalino F. Effect of Bariatric Surgery on Gut Microbiota: A Scientometric Analysis. SAUDI JOURNAL OF MEDICINE & MEDICAL SCIENCES 2024; 12:65-70. [PMID: 38362093 PMCID: PMC10866388 DOI: 10.4103/sjmms.sjmms_146_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/21/2023] [Accepted: 10/08/2023] [Indexed: 02/17/2024]
Abstract
Objective To perform a bibliometric analysis of the scientific production related to intestinal microbiota and bariatric surgery between January 2016 and December 2022. Materials and Methods A bibliographic search was performed in the Scopus database to identify published papers. Free and controlled terms (MeSH and Emtree) were used. The information collected was analyzed with SciVal. Results A total of 518 published papers were included in the analysis. Carel Le Roux was the author with the highest scientific production; however, Edi Prifti had the highest impact. French National Institute of Health and Medical Research (Institut national de la santé et de la recherche médicale) was the institution with the highest number of published articles. Six of the 10 institutions with the highest production were in France, yet the United States had the highest volume of scientific production in this research topic. Most papers were published in first quartile journals. Articles with international collaboration had the highest impact. There is a sustained increase in the number of publications since 2019. Conclusions The study found that the vast majority of research on gut microbiota changes following bariatric surgery are conducted in the United States and European countries. In addition, the sustained increase in production coupled with the articles being published in high-quality journals and having good citation impact are indictors of the current interest in this research field.
Collapse
Affiliation(s)
- Miguel Cabanillas-Lazo
- Grupo Peruano de Investigación Epidemiológica, Unidad Para la Generación y Síntesis de Evidencias en Salud, Universidad San Ignacio de Loyola, Lima, Peru
- Sociedad Científica de San Fernando, Lima, Peru
| | - Carlos Quispe-Vicuña
- Grupo Peruano de Investigación Epidemiológica, Unidad Para la Generación y Síntesis de Evidencias en Salud, Universidad San Ignacio de Loyola, Lima, Peru
- Sociedad Científica de San Fernando, Lima, Peru
| | - Milagros Pascual-Guevara
- Sociedad Científica de San Fernando, Lima, Peru
- Facultad de Medicina, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | | | - John Barja-Ore
- Academic Department, Universidad Privada del Norte, Lima, Peru
| | - Fran Espinoza-Carhuancho
- Grupo de Bibliometría, Evaluación de evidencia y Revisiones Sistemáticas (BEERS), Human Medicine Career, Universidad Cientifica del Sur, Lima, Peru
| | - Frank Mayta-Tovalino
- Grupo de Bibliometría, Evaluación de evidencia y Revisiones Sistemáticas (BEERS), Human Medicine Career, Universidad Cientifica del Sur, Lima, Peru
| |
Collapse
|
44
|
Steinbach E, Masi D, Ribeiro A, Serradas P, Le Roy T, Clément K. Upper small intestine microbiome in obesity and related metabolic disorders: A new field of investigation. Metabolism 2024; 150:155712. [PMID: 37884078 DOI: 10.1016/j.metabol.2023.155712] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 10/08/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023]
Abstract
The study of the gut microbiome holds great promise for understanding and treating metabolic diseases, as its functions and derived metabolites can influence the metabolic status of the host. While research on the fecal microbiome has provided valuable insights, it tells us only part of the story. This limitation arises from the substantial variations in microorganism distribution throughout the gastrointestinal tract due to changes in physicochemical conditions. Thus, relying solely on the fecal microbiome may not be sufficient to draw comprehensive conclusions about metabolic diseases. The proximal part of the small intestine, particularly the jejunum, indeed, serves as the crucial site for digestion and absorption of nutrients, suggesting a potential role of its microbiome in metabolic regulation. Unfortunately, it remains relatively underexplored due to limited accessibility. This review presents current evidence regarding the relationships between the microbiome in the upper small intestine and various phenotypes, focusing on obesity and type 2 diabetes, in both humans and rodents. Research on humans is still limited with variability in the population and methods used. Accordingly, to better understand the role of the whole gut microbiome in metabolic diseases, studies exploring the human microbiome in different niches are needed.
Collapse
Affiliation(s)
- Emilie Steinbach
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches (NutriOmics) Research Unit, 75013, Paris, France
| | - Davide Masi
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches (NutriOmics) Research Unit, 75013, Paris, France; Sapienza University of Rome, Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, 00161 Rome, Italy
| | - Agnès Ribeiro
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches (NutriOmics) Research Unit, 75013, Paris, France
| | - Patricia Serradas
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches (NutriOmics) Research Unit, 75013, Paris, France
| | - Tiphaine Le Roy
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches (NutriOmics) Research Unit, 75013, Paris, France
| | - Karine Clément
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches (NutriOmics) Research Unit, 75013, Paris, France; Assistance Publique Hôpitaux de Paris, Nutrition Department, Pitié-Salpêtrière Hospital, 75013 Paris, France.
| |
Collapse
|
45
|
Effenberger M, Grander C, Grabherr F, Tilg H. Nonalcoholic Fatty Liver Disease and the Intestinal Microbiome: An Inseparable Link. J Clin Transl Hepatol 2023; 11:1498-1507. [PMID: 38161503 PMCID: PMC10752805 DOI: 10.14218/jcth.2023.00069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/21/2023] [Accepted: 07/18/2023] [Indexed: 01/03/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) particularly affects patients with type 2 diabetes and obesity. The incidence of NAFLD has increased significantly over the last decades and is now pandemically across the globe. It is a complex systemic disease comprising hepatic lipid accumulation, inflammation, lipotoxicity, gut dysbiosis, and insulin resistance as main features and with the potential to progress to cirrhosis and hepatocellular carcinoma (HCC). In numerous animal and human studies the gut microbiota plays a key role in the pathogenesis of NAFLD, NAFLD-cirrhosis and NAFLD-associated HCC. Lipotoxicity is the driver of inflammation, insulin resistance, and liver injury. Likewise, western diet, obesity, and metabolic disorders may alter the gut microbiota, which activates innate and adaptive immune responses and fuels hereby hepatic and systemic inflammation. Indigestible carbohydrates are fermented by the gut microbiota to produce important metabolites, such as short-chain fatty acids and succinate. Numerous animal and human studies suggested a pivotal role of these metabolites in the progression of NAFLD and its comorbidities. Though, modification of the gut microbiota and/or the metabolites could even be beneficial in patients with NAFLD, NAFLD-cirrhosis, and NAFLD-associated HCC. In this review we collect the evidence that exogenous and endogenous hits drive liver injury in NAFLD and propel liver fibrosis and the progressing to advanced disease stages. NAFLD can be seen as the product of a complex interplay between gut microbiota, the immune response and metabolism. Thus, the challenge will be to understand its pathogenesis and to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Maria Effenberger
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph Grander
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
46
|
Pontes-Silva A, Lopes AL, Maciel EDS, Quaresma FRP, Dibai-Filho AV. Human metabolism and body composition: prospects for novel studies. Nutr Rev 2023; 82:5-8. [PMID: 38073333 DOI: 10.1093/nutrit/nuad040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
CONTEXT Most articles on gut microbiota argue the importance of body composition assessment in patients; however, body composition assessments are fragile (ie, with methodological limitations) in the most recent studies. OBJECTIVE To present two suggestions for further research using the human body composition assessment. METHODS The methods used in this study are based on a Pinto et al article published in Nutrition Reviews. DATA EXTRACTION On the basis of data. obtained from the PubMed, SCOPUS, LILACS, and Web of Science databases, Pinto et al provided a current survey of intermittent fasting protocols and an understanding of the outcomes to date in terms of the profile of the intestinal microbiota in obese organisms. DATA ANALYSIS Of the 82 original articles identified from the databases, 35 were eliminated because of duplication and 32 were excluded for not meeting the inclusion criteria. Two additional articles found in a new search were added, yielding a total of 17 studies to be included in this review. Among the protocols, alternate-day fasting and time-restricted feeding were the most common, and they were shown to have different mechanisms of metabolic signaling. Time-restricted feeding influences body mass control and biochemical parameters by regulating the circadian system and improving satiety control systems by acting on leptin secretion. In contrast, alternate-day fasting leads to a reduction of ±75% of all energy consumption regardless of dietary composition, in addition to promoting hormonal adjustments that promote body mass control. Furthermore, both protocols could remodel the intestinal microbiota by changing the Firmicutes to Bacteroidetes ratio and increasing the abundance of strains such as Lactobacillus spp. and Akkermansia that have a protective effect on metabolism against the effects of body mass gain. CONCLUSION Changes in adipose tissue (eg, body mass loss, control, gain) should be interpreted via the sum of skinfolds in absolute values, waist perimeter, and patients' body proportionality, because fat is just a fraction of the adipocyte (lipid).
Collapse
Affiliation(s)
- André Pontes-Silva
- is with the Physical Therapy Post-Graduate Program, Physical Therapy Department, Federal University of São Carlos, São Carlos, São Paulo, Brazil
| | - André Luiz Lopes
- is with the Human Movement Sciences Post-Graduate Program, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Erika da Silva Maciel
- are with the Sciences and Health Teaching Post-Graduate Program, Federal University of Tocantins, Palmas, Tocantins, Brazil
| | | | - Almir Vieira Dibai-Filho
- is with the Physical Education Post-Graduate Program, Physical Education Department, Federal University of Maranhão, São Luís, Maranhão, Brazil
| |
Collapse
|
47
|
Hussan H, Clinton SK, Grainger EM, Webb M, Wang C, Webb A, Needleman B, Noria S, Zhu J, Choueiry F, Pietrzak M, Bailey MT. Distinctive patterns of sulfide- and butyrate-metabolizing bacteria after bariatric surgery: potential implications for colorectal cancer risk. Gut Microbes 2023; 15:2255345. [PMID: 37702461 PMCID: PMC10501170 DOI: 10.1080/19490976.2023.2255345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/09/2023] [Accepted: 08/31/2023] [Indexed: 09/14/2023] Open
Abstract
Despite improved cardiometabolic outcomes following bariatric surgery, its long-term impact on colorectal cancer (CRC) risk remains uncertain. In parallel, the influence of bariatric surgery on the host microbiome and relationships with disease outcomes is beginning to be appreciated. Therefore, we investigated the impact of Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG) on the patterns of sulfide-reducing and butyrate-producing bacteria, which are hypothesized to modulate CRC risk after bariatric surgery. In this single-center, cross-sectional study, we included 15 pre-surgery subjects with severe obesity and patients who are at a median (range) of 25.6 (9.9-46.5) months after RYGB (n = 16) or VSG (n = 10). The DNA abundance of fecal bacteria and enzymes involved in butyrate and sulfide metabolism were identified using metagenomic sequencing. Differences between pre-surgery and post-RYGB or post-VSG cohorts were quantified using the linear discriminant analysis (LDA) effect size (LEfSe) method. Our sample was predominantly female (87%) with a median (range) age of 46 (23-71) years. Post-RYGB and post-VSG patients had a higher DNA abundance of fecal sulfide-reducing bacteria than pre-surgery controls (LDA = 1.3-4.4, p < .05). The most significant enrichments were for fecal E. coli, Acidaminococcus and A. finegoldii after RYGB, and for A. finegoldii, S. vestibularis, V. parvula after VSG. As for butyrate-producing bacteria, R. faecis was more abundant, whereas B. dentium and A. hardus were lower post-RYGB vs. pre-surgery. B. dentium was also lower in post-VSG vs. pre-surgery. Consistent with these findings, our analysis showed a greater enrichment of sulfide-reducing enzymes after bariatric surgery, especially RYGB, vs. pre-surgery. The DNA abundance of butyrate-producing enzymes was lower post-RYGB. In conclusion, the two most used bariatric surgeries, RYGB and VSG, are associated with microbiome patterns that are potentially implicated in CRC risk. Future studies are needed to validate and understand the impact of these microbiome changes on CRC risk after bariatric surgery.
Collapse
Affiliation(s)
- Hisham Hussan
- Division of Gastroenterology, Department of Internal Medicine, University of California, Davis; Sacramento, CA, USA
- The UC Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Steven K. Clinton
- Division of Medical Oncology; Department of Internal Medicine, The Ohio StateUniversity, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Elizabeth M. Grainger
- Division of Medical Oncology; Department of Internal Medicine, The Ohio StateUniversity, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Maxine Webb
- Division of Medical Oncology; Department of Internal Medicine, The Ohio StateUniversity, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Cankun Wang
- Division of Biomedical Informatics, Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Amy Webb
- Division of Biomedical Informatics, Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Bradley Needleman
- Center for Minimally Invasive Surgery; Department of General Surgery, The Ohio State University, Columbus, OH, USA
| | - Sabrena Noria
- Center for Minimally Invasive Surgery; Department of General Surgery, The Ohio State University, Columbus, OH, USA
| | - Jiangjiang Zhu
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- The Department of Human Sciences, The Ohio State University, Columbus, OH, USA
| | - Fouad Choueiry
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- The Department of Human Sciences, The Ohio State University, Columbus, OH, USA
| | - Maciej Pietrzak
- Division of Biomedical Informatics, Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Michael T. Bailey
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital and Department of Pediatrics, Columbus, OH, USA
- The Oral and Gastrointestinal Microbiology Research Affinity Group, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| |
Collapse
|
48
|
Kheloufi L, Weiss N, Thabut D, Pais R. Neurocognitive impairment in obesity and liver disease-An unsolved mystery. Hepatol Commun 2023; 7:e0305. [PMID: 37902509 PMCID: PMC10617873 DOI: 10.1097/hc9.0000000000000305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 08/17/2023] [Indexed: 10/31/2023] Open
Affiliation(s)
- Lyes Kheloufi
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
- Brain Liver Pitié-Salpêtrière Study Group (BLIPS)
- Centre de Recherche Saint Antoine, INSERM, Paris, France
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Nicolas Weiss
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
- Brain Liver Pitié-Salpêtrière Study Group (BLIPS)
- Centre de Recherche Saint Antoine, INSERM, Paris, France
| | - Dominique Thabut
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
- Brain Liver Pitié-Salpêtrière Study Group (BLIPS)
- Centre de Recherche Saint Antoine, INSERM, Paris, France
| | - Raluca Pais
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
- Brain Liver Pitié-Salpêtrière Study Group (BLIPS)
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| |
Collapse
|
49
|
Lalloyer F, Mogilenko DA, Verrijken A, Haas JT, Lamazière A, Kouach M, Descat A, Caron S, Vallez E, Derudas B, Gheeraert C, Baugé E, Despres G, Dirinck E, Tailleux A, Dombrowicz D, Van Gaal L, Eeckhoute J, Lefebvre P, Goossens JF, Francque S, Staels B. Roux-en-Y gastric bypass induces hepatic transcriptomic signatures and plasma metabolite changes indicative of improved cholesterol homeostasis. J Hepatol 2023; 79:898-909. [PMID: 37230231 DOI: 10.1016/j.jhep.2023.05.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 04/18/2023] [Accepted: 05/08/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND & AIMS Roux-en-Y gastric bypass (RYGB), the most effective surgical procedure for weight loss, decreases obesity and ameliorates comorbidities, such as non-alcoholic fatty liver (NAFLD) and cardiovascular (CVD) diseases. Cholesterol is a major CVD risk factor and modulator of NAFLD development, and the liver tightly controls its metabolism. How RYGB surgery modulates systemic and hepatic cholesterol metabolism is still unclear. METHODS We studied the hepatic transcriptome of 26 patients with obesity but not diabetes before and 1 year after undergoing RYGB. In parallel, we measured quantitative changes in plasma cholesterol metabolites and bile acids (BAs). RESULTS RYGB surgery improved systemic cholesterol metabolism and increased plasma total and primary BA levels. Transcriptomic analysis revealed specific alterations in the liver after RYGB, with the downregulation of a module of genes implicated in inflammation and the upregulation of three modules, one associated with BA metabolism. A dedicated analysis of hepatic genes related to cholesterol homeostasis pointed towards increased biliary cholesterol elimination after RYGB, associated with enhancement of the alternate, but not the classical, BA synthesis pathway. In parallel, alterations in the expression of genes involved in cholesterol uptake and intracellular trafficking indicate improved hepatic free cholesterol handling. Finally, RYGB decreased plasma markers of cholesterol synthesis, which correlated with an improvement in liver disease status after surgery. CONCLUSIONS Our results identify specific regulatory effects of RYGB on inflammation and cholesterol metabolism. RYGB alters the hepatic transcriptome signature, likely improving liver cholesterol homeostasis. These gene regulatory effects are reflected by systemic post-surgery changes of cholesterol-related metabolites, corroborating the beneficial effects of RYGB on both hepatic and systemic cholesterol homeostasis. IMPACT AND IMPLICATIONS Roux-en-Y gastric bypass (RYGB) is a widely used bariatric surgery procedure with proven efficacy in body weight management, combatting cardiovascular disease (CVD) and non-alcoholic fatty liver disease (NAFLD). RYGB exerts many beneficial metabolic effects, by lowering plasma cholesterol and improving atherogenic dyslipidemia. Using a cohort of patients undergoing RYGB, studied before and 1 year after surgery, we analyzed how RYGB modulates hepatic and systemic cholesterol and bile acid metabolism. The results of our study provide important insights on the regulation of cholesterol homeostasis after RYGB and open avenues that could guide future monitoring and treatment strategies targeting CVD and NAFLD in obesity.
Collapse
Affiliation(s)
- Fanny Lalloyer
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Denis A Mogilenko
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France; Department of Medicine, Department of Pathology, Microbiology and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Ann Verrijken
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, 2610, Wilrijk, Antwerp, Belgium; Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, 2650, Edegem, Antwerp, Belgium
| | - Joel T Haas
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Antonin Lamazière
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Clinical Metabolomic Department, Sorbonne Université, Inserm, F-75012, Paris, France
| | - Mostafa Kouach
- University of Lille, CHU Lille, EA 7365-GRITA-Groupe de Recherche sur les formes Injectables et les Technologies Associées, F-59000, Lille, France
| | - Amandine Descat
- University of Lille, CHU Lille, EA 7365-GRITA-Groupe de Recherche sur les formes Injectables et les Technologies Associées, F-59000, Lille, France
| | - Sandrine Caron
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Emmanuelle Vallez
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Bruno Derudas
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Céline Gheeraert
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Eric Baugé
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Gaëtan Despres
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Clinical Metabolomic Department, Sorbonne Université, Inserm, F-75012, Paris, France
| | - Eveline Dirinck
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, 2610, Wilrijk, Antwerp, Belgium; Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, 2650, Edegem, Antwerp, Belgium
| | - Anne Tailleux
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - David Dombrowicz
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Luc Van Gaal
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, 2610, Wilrijk, Antwerp, Belgium; Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, 2650, Edegem, Antwerp, Belgium
| | - Jerôme Eeckhoute
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Philippe Lefebvre
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Jean-François Goossens
- University of Lille, CHU Lille, EA 7365-GRITA-Groupe de Recherche sur les formes Injectables et les Technologies Associées, F-59000, Lille, France
| | - Sven Francque
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, 2610, Wilrijk, Antwerp, Belgium; Department of Gastroenterology and Hepatology, Antwerp University Hospital, ERN RARE-LIVER, 2650, Edegem, Antwerp, Belgium
| | - Bart Staels
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France.
| |
Collapse
|
50
|
Camp KK, Coleman MF, McFarlane TL, Doerstling SS, Khatib SA, Rezeli ET, Lewis AG, Pfeil AJ, Smith LA, Bowers LW, Fouladi F, Gong W, Glenny EM, Parker JS, Milne GL, Carroll IM, Fodor AA, Seeley RJ, Hursting SD. Calorie restriction outperforms bariatric surgery in a murine model of obesity and triple-negative breast cancer. JCI Insight 2023; 8:e172868. [PMID: 37698918 PMCID: PMC10629811 DOI: 10.1172/jci.insight.172868] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023] Open
Abstract
Obesity promotes triple-negative breast cancer (TNBC), and effective interventions are urgently needed to break the obesity-TNBC link. Epidemiologic studies indicate that bariatric surgery reduces TNBC risk, while evidence is limited or conflicted for weight loss via low-fat diet (LFD) or calorie restriction (CR). Using a murine model of obesity-driven TNBC, we compared the antitumor effects of vertical sleeve gastrectomy (VSG) with LFD, chronic CR, and intermittent CR. Each intervention generated weight and fat loss and suppressed tumor growth relative to obese mice (greatest suppression with CR). VSG and CR regimens exerted both similar and unique effects, as assessed using multiomics approaches, in reversing obesity-associated transcript, epigenetics, secretome, and microbiota changes and restoring antitumor immunity. Thus, in a murine model of TNBC, bariatric surgery and CR each reverse obesity-driven tumor growth via shared and distinct antitumor mechanisms, and CR is superior to VSG in reversing obesity's procancer effects.
Collapse
Affiliation(s)
- Kristina K. Camp
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michael F. Coleman
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Tori L. McFarlane
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Steven S. Doerstling
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Subreen A. Khatib
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Erika T. Rezeli
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Alfor G. Lewis
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Alexander J. Pfeil
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Laura A. Smith
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Laura W. Bowers
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Farnaz Fouladi
- College of Computing and Informatics, University of North Carolina at Charlotte, Charlotte, North Carolina, USA
| | - Weida Gong
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Elaine M. Glenny
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joel S. Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ginger L. Milne
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Ian M. Carroll
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Anthony A. Fodor
- College of Computing and Informatics, University of North Carolina at Charlotte, Charlotte, North Carolina, USA
| | - Randy J. Seeley
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Stephen D. Hursting
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina, USA
| |
Collapse
|