1
|
Hamamah S, Hajnal A, Covasa M. Influence of Bariatric Surgery on Gut Microbiota Composition and Its Implication on Brain and Peripheral Targets. Nutrients 2024; 16:1071. [PMID: 38613104 PMCID: PMC11013759 DOI: 10.3390/nu16071071] [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: 03/14/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Obesity remains a significant global health challenge, with bariatric surgery remaining as one of the most effective treatments for severe obesity and its related comorbidities. This review highlights the multifaceted impact of bariatric surgery beyond mere physical restriction or nutrient malabsorption, underscoring the importance of the gut microbiome and neurohormonal signals in mediating the profound effects on weight loss and behavior modification. The various bariatric surgery procedures, such as Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG), act through distinct mechanisms to alter the gut microbiome, subsequently impacting metabolic health, energy balance, and food reward behaviors. Emerging evidence has shown that bariatric surgery induces profound changes in the composition of the gut microbiome, notably altering the Firmicutes/Bacteroidetes ratio and enhancing populations of beneficial bacteria such as Akkermansia. These microbiota shifts have far-reaching effects beyond gut health, influencing dopamine-mediated reward pathways in the brain and modulating the secretion and action of key gut hormones including ghrelin, leptin, GLP-1, PYY, and CCK. The resultant changes in dopamine signaling and hormone levels contribute to reduced hedonic eating, enhanced satiety, and improved metabolic outcomes. Further, post-bariatric surgical effects on satiation targets are in part mediated by metabolic byproducts of gut microbiota like short-chain fatty acids (SCFAs) and bile acids, which play a pivotal role in modulating metabolism and energy expenditure and reducing obesity-associated inflammation, as well as influencing food reward pathways, potentially contributing to the regulation of body weight and reduction in hedonic eating behaviors. Overall, a better understanding of these mechanisms opens the door to developing non-surgical interventions that replicate the beneficial effects of bariatric surgery on the gut microbiome, dopamine signaling, and gut hormone regulation, offering new avenues for obesity treatment.
Collapse
Affiliation(s)
- Sevag Hamamah
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 9176, USA;
| | - Andras Hajnal
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA;
| | - Mihai Covasa
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 9176, USA;
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 7200229 Suceava, Romania
| |
Collapse
|
2
|
Igwe JK, Surapaneni PK, Cruz E, Cole C, Njoku K, Kim J, Alaribe U, Weze K, Mohammed B. Bariatric Surgery and Inflammatory Bowel Disease: National Trends and Outcomes Associated with Procedural Sleeve Gastrectomy vs Historical Bariatric Surgery Among US Hospitalized Patients 2009-2020. Obes Surg 2023; 33:3472-3486. [PMID: 37804470 PMCID: PMC10603008 DOI: 10.1007/s11695-023-06833-7] [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: 04/18/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 10/09/2023]
Abstract
PURPOSE The association between bariatric surgery and IBD-related inpatient outcomes is not well characterized. We report, analyze, and compare inpatient trends and outcomes among encounters with a history of bariatric surgery (Hx-MBS) compared to those receiving bariatric surgery during index admission (PR-MBS) admitted from 2009 to 2020. METHODS Retrospective cohort design: the 2009-2020 National Inpatient Sample (NIS) databases were used to identify hospital encounters with patients aged ≥ 18 years with a history of MBS (Hx-MBS) or with procedure coding indicating MBS procedure (PR-MBS) according to International Classification of Diseases, Ninth (ICD-9-CM/ ICD-9-PCS) or Tenth Revision (ICD-10-CM/ICD-10-PCS) Clinical Modification/Procedure Coding System during index admission (ICD-9-CM: V4586; ICD-10-CM: Z9884; ICD-9-PR: 4382, 4389; ICD-10-PR: 0DB64Z3, 0DB63ZZ). Pearson χ2 analysis, analysis of variance, multivariable regression analyses, and propensity matching on independent variables were conducted to analyze significant associations between variables and for primary outcome inflammatory bowel disease-related admission, and secondary outcomes: diagnosis of nonalcoholic steatohepatitis, nonalcoholic fatty liver disease, or chronic mesenteric ischemia during admission. RESULTS We identified 3,365,784 (76.20%) Hx-MBS hospitalizations and 1,050,900 hospitalizations with PR-MBS (23.80%). Propensity score matching analysis demonstrated significantly higher odds of inflammatory bowel disease, and chronic mesenteric ischemia for Hx-MBS compared to PR-MBS, and significantly lower odds of nonalcoholic steatohepatitis and nonalcoholic fatty liver disease for Hx-MBS compared to PR-MBS. CONCLUSION In our study, Hx-MBS was associated with significantly increased odds of inflammatory bowel disease and other GI pathologies compared to matched controls. The mechanism by which this occurs is unclear. Additional studies are needed to examine these findings.
Collapse
Affiliation(s)
- Joseph-Kevin Igwe
- School of Medicine, Department of Medicine, Stanford University, 291 Campus Drive, Stanford, CA, 94305, USA.
- Department of Medicine, Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA, 30313, USA.
- American Heart Association Strategically Focused Research Network on the Science of Diversity in Clinical Trials Research Fellowship, 5001 S Miami Blvd #300, Durham, NC, 27703, USA.
| | | | - Erin Cruz
- School of Medicine, Department of Medicine, Stanford University, 291 Campus Drive, Stanford, CA, 94305, USA
| | - Cedric Cole
- Department of Medicine, Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA, 30313, USA
| | - Kingsley Njoku
- Department of Medicine, Emory University School of Medicine, Atlanta, USA
| | - Jisoo Kim
- Department of Surgery, Texas Tech University Health Sciences Center at El Paso, El Paso, USA
| | - Ugo Alaribe
- School of Medicine, Caribbean Medical University, Willemstad, USA
| | - Kelechi Weze
- Department of Medicine, Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA, 30313, USA
| | - Bilal Mohammed
- Department of Medicine, Ascension Saint Vincent, Indianapolis, USA
| |
Collapse
|
3
|
Frick LD, Hankir MK, Borner T, Malagola E, File B, Gero D. Novel Insights into the Physiology of Nutrient Sensing and Gut-Brain Communication in Surgical and Experimental Obesity Therapy. Obes Surg 2023; 33:2906-2916. [PMID: 37474864 PMCID: PMC10435392 DOI: 10.1007/s11695-023-06739-4] [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: 05/10/2023] [Revised: 07/05/2023] [Accepted: 07/14/2023] [Indexed: 07/22/2023]
Abstract
Despite standardized surgical technique and peri-operative care, metabolic outcomes of bariatric surgery are not uniform. Adaptive changes in brain function may play a crucial role in achieving optimal postbariatric weight loss. This review follows the anatomic-physiologic structure of the postbariatric nutrient-gut-brain communication chain through its key stations and provides a concise summary of recent findings in bariatric physiology, with a special focus on the composition of the intestinal milieu, intestinal nutrient sensing, vagal nerve-mediated gastrointestinal satiation signals, circulating hormones and nutrients, as well as descending neural signals from the forebrain. The results of interventional studies using brain or vagal nerve stimulation to induce weight loss are also summarized. Ultimately, suggestions are made for future diagnostic and therapeutic research for the treatment of obesity.
Collapse
Affiliation(s)
- Lukas D Frick
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Mohammed K Hankir
- Department of Experimental Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Tito Borner
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ermanno Malagola
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY, 10032, USA
| | - Bálint File
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary, Institute of Cognitive Neuroscience and Psychology, Research Centre for Natural Sciences, Budapest, Hungary
- Wigner Research Centre for Physics, Budapest, Hungary
| | - Daniel Gero
- Department of Surgery and Transplantation, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091, Zürich, Switzerland.
| |
Collapse
|
4
|
Abstract
Overweight, obesity, undernutrition and their respective sequelae have devastating tolls on personal and public health worldwide. Traditional approaches for treating these conditions with diet, exercise, drugs and/or surgery have shown varying degrees of success, creating an urgent need for new solutions with long-term efficacy. Owing to transformative advances in sequencing, bioinformatics and gnotobiotic experimentation, we now understand that the gut microbiome profoundly impacts energy balance through diverse mechanisms affecting both sides of the energy balance equation. Our growing knowledge of microbial contributions to energy metabolism highlights new opportunities for weight management, including the microbiome-aware improvement of existing tools and novel microbiome-targeted therapies. In this Review, we synthesize current knowledge concerning the bidirectional influences between the gut microbiome and existing weight management strategies, including behaviour-based and clinical approaches, and incorporate a subject-level meta-analysis contrasting the effects of weight management strategies on microbiota composition. We consider how emerging understanding of the gut microbiome alters our prospects for weight management and the challenges that must be overcome for microbiome-focused solutions to achieve success.
Collapse
Affiliation(s)
- Rachel N Carmody
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA.
| | - Jordan E Bisanz
- Department of Biochemistry and Molecular Biology, Penn State Microbiome Center, Huck Institutes of the Life Sciences, The Pennsylvania State University, State College, PA, USA.
| |
Collapse
|
5
|
Stroud AM, Coleman MF. Bariatric surgery in the prevention of obesity-associated cancers: mechanistic implications. Surg Obes Relat Dis 2023; 19:772-780. [PMID: 37120355 DOI: 10.1016/j.soard.2023.02.016] [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: 01/30/2023] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 03/04/2023]
Abstract
Obesity is associated with an increased risk of at least 13 different cancers, as well as worse cancer outcomes and increased cancer mortality. As rates continue to rise both in the United States and worldwide, obesity is poised to become the leading lifestyle-related risk factor for cancer. Currently, the most effective treatment for patients with severe obesity is bariatric surgery. Multiple cohort studies have demonstrated a consistent >30% decreased risk of cancer incidence in women, but not men, following bariatric surgery. However, the physiologic mechanisms driving obesity-associated cancer and the cancer-protective effect of bariatric surgery are not clearly defined. In this review, we highlight emerging concepts in the mechanistic understanding of obesity-associated cancer. Evidence from both human studies and preclinical animal models suggest that obesity drives carcinogenesis through dysregulation of systemic metabolism, immune dysfunction, and an altered gut microbiome. Additionally, we present related findings to suggest that bariatric surgery may disrupt and even reverse many of these mechanisms. Finally, we discuss the use of preclinical bariatric surgery animal models in the study of cancer biology. The prevention of cancer is emerging as an important indication for bariatric surgery. Elucidating the mechanisms through which bariatric surgery limits carcinogenesis is critical to developing a variety of interventions that intercept obesity-driven cancer.
Collapse
Affiliation(s)
- Andrea M Stroud
- Division of Bariatric Surgery, Oregon Health & Science University, Portland, Oregon.
| | - Michael F Coleman
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
6
|
Gawey BJ, Khanna S. Clostridioides difficile Infection: Landscape and Microbiome Therapeutics. Gastroenterol Hepatol (N Y) 2023; 19:319-328. [PMID: 37706187 PMCID: PMC10496268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Clostridioides difficile infection (CDI) is the leading cause of hospital-acquired diarrhea and is common in the community. Both younger individuals who may be healthy otherwise and older individuals with comorbid conditions are at risk for developing CDI, with the predominant risk factor being antibiotic use. Unlike other gastrointestinal infections, CDI is not self-limited, requires antimicrobial therapy, and tends to recur at high rates even without additional risk factor exposure. The goals of CDI management include controlling active symptoms and using a recurrence prevention strategy such as a narrow-spectrum antibiotic, tapered and pulsed regimens, antibody- based therapies (directed against toxin B), or microbiome restoration. In recent years, fecal microbiota transplantation (FMT) has been the most used modality to prevent recurrent CDI with high cure rates. Heterogeneity, lack of scalability, and serious adverse events from FMT have led to development of standardized microbiota restoration therapies (MRTs). The US Food and Drug Administration has approved 2 stool-derived MRTs for prevention of recurrent CDI: fecal microbiota, live-jslm, an enema-based therapy; and fecal microbiota spores, live-brpk, an oral therapy. A phase 3 trial for a synthetic oral MRT is underway. This article outlines the pathophysiology and treatment of CDI, focusing primarily on the gut microbiome and standardized MRTs.
Collapse
Affiliation(s)
- Brent J. Gawey
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Sahil Khanna
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
7
|
Liao J, Liu Y, Pei Z, Wang H, Zhu J, Zhao J, Lu W, Chen W. Clostridium butyricum Reduces Obesity in a Butyrate-Independent Way. Microorganisms 2023; 11:1292. [PMID: 37317266 DOI: 10.3390/microorganisms11051292] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 06/16/2023] Open
Abstract
Accumulating evidence from recent studies links the gut microbiota to obesity, and microbiome therapy has been examined as a treatment. Clostridium butyricum (C. butyricum), an intestinal symbiont, protects the host from a range of diseases. Studies have shown a negative correlation between the relative abundance of C. butyricum and a predisposition for obesity. However, the physiological function and material basis of C. butyricum for obesity are unclear. Here, five C. butyricum isolates were administered to mice on a high-fat diet (HFD) to determine their anti-obesity effects. All isolates suppressed the formation and inflammation of subcutaneous fat, and the two effective strains considerably reduced weight gain and ameliorated dyslipidemia, hepatic steatosis, and inflammation. These positive effects were not achieved by increasing the concentration of intestinal butyrate, and the effective strains could not be replaced by sodium butyrate (NaB). We also discovered that oral supplementation with the two most effective strains changed the metabolism of tryptophan and purine and altered the composition of the gut microbiota. In summary, C. butyricum improved the metabolic phenotypes under the HFD by controlling the composition of the gut microbiota and modulating intestinal metabolites, thereby demonstrating its ability to fight obesity and providing a theoretical foundation for microbial preparations production.
Collapse
Affiliation(s)
- Jingyi Liao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yaoliang Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhangming Pei
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Hongchao Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jinlin Zhu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
8
|
Mukorako P, St-Pierre DH, Flamand N, Biertho L, Lebel S, Lemoine N, Plamondon J, Roy MC, Tchernof A, Varin TV, Marette A, Silvestri C, Di Marzo V, Richard D. Hypoabsorptive surgeries cause limb-dependent changes in the gut endocannabinoidome and microbiome in association with beneficial metabolic effects. Int J Obes (Lond) 2023:10.1038/s41366-023-01307-3. [PMID: 37142736 DOI: 10.1038/s41366-023-01307-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 03/27/2023] [Accepted: 04/03/2023] [Indexed: 05/06/2023]
Abstract
OBJECTIVE To determine whether the metabolic benefits of hypoabsorptive surgeries are associated with changes in the gut endocannabinoidome (eCBome) and microbiome. METHODS Biliopancreatic diversion with duodenal switch (BPD-DS) and single anastomosis duodeno-ileal bypass with sleeve gastrectomy (SADI-S) were performed in diet-induced obese (DIO) male Wistar rats. Control groups fed a high-fat diet (HF) included sham-operated (SHAM HF) and SHAM HF-pair-weighed to BPD-DS (SHAM HF-PW). Body weight, fat mass gain, fecal energy loss, HOMA-IR, and gut-secreted hormone levels were measured. The levels of eCBome lipid mediators and prostaglandins were quantified in different intestinal segments by LC-MS/MS, while expression levels of genes encoding eCBome metabolic enzymes and receptors were determined by RT-qPCR. Metataxonomic (16S rRNA) analysis was performed on residual distal jejunum, proximal jejunum, and ileum contents. RESULTS BPD-DS and SADI-S reduced fat gain and HOMA-IR, while increasing glucagon-like peptide-1 (GLP-1) and peptide tyrosine tyrosine (PYY) levels in HF-fed rats. Both surgeries induced potent limb-dependent alterations in eCBome mediators and in gut microbial ecology. In response to BPD-DS and SADI-S, changes in gut microbiota were significantly correlated with those of eCBome mediators. Principal component analyses revealed connections between PYY, N-oleoylethanolamine (OEA), N-linoleoylethanolamine (LEA), Clostridium, and Enterobacteriaceae_g_2 in the proximal and distal jejunum and in the ileum. CONCLUSIONS BPD-DS and SADI-S caused limb-dependent changes in the gut eCBome and microbiome. The present results indicate that these variables could significantly influence the beneficial metabolic outcome of hypoabsorptive bariatric surgeries.
Collapse
Affiliation(s)
- Paulette Mukorako
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
- Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada
| | - David H St-Pierre
- Institute of Nutrition and Functional Foods, Centre NUTRISS, Québec, QC, Canada
- Department of Exercise Sciences, Université du Québec à Montréal (UQAM), Québec, QC, Canada
- School of Nutrition, Faculty of Agriculture and Food Sciences, Québec, QC, Canada
| | - Nicolas Flamand
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
- Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Québec, QC, Canada
| | - Laurent Biertho
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
- Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada
| | - Stéfane Lebel
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
- Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada
| | - Natacha Lemoine
- Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada
| | - Julie Plamondon
- Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada
| | - Marie-Claude Roy
- Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada
| | - André Tchernof
- Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada
- School of Nutrition, Faculty of Agriculture and Food Sciences, Québec, QC, Canada
| | - Thibault V Varin
- Institute of Nutrition and Functional Foods, Centre NUTRISS, Québec, QC, Canada
| | - André Marette
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
- Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada
- Institute of Nutrition and Functional Foods, Centre NUTRISS, Québec, QC, Canada
| | - Cristoforo Silvestri
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada.
- Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada.
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Québec, QC, Canada.
| | - Vincenzo Di Marzo
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada.
- Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada.
- Institute of Nutrition and Functional Foods, Centre NUTRISS, Québec, QC, Canada.
- School of Nutrition, Faculty of Agriculture and Food Sciences, Québec, QC, Canada.
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Québec, QC, Canada.
| | - Denis Richard
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada.
- Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada.
| |
Collapse
|
9
|
Zhang Y, Chen R, Zhang D, Qi S, Liu Y. Metabolite interactions between host and microbiota during health and disease: Which feeds the other? Biomed Pharmacother 2023; 160:114295. [PMID: 36709600 DOI: 10.1016/j.biopha.2023.114295] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/20/2023] [Accepted: 01/20/2023] [Indexed: 01/30/2023] Open
Abstract
Metabolites produced by the host and microbiota play a crucial role in how human bodies develop and remain healthy. Most of these metabolites are produced by microbiota and hosts in the digestive tract. Metabolites in the gut have important roles in energy metabolism, cellular communication, and host immunity, among other physiological activities. Although numerous host metabolites, such as free fatty acids, amino acids, and vitamins, are found in the intestine, metabolites generated by gut microbiota are equally vital for intestinal homeostasis. Furthermore, microbiota in the gut is the sole source of some metabolites, including short-chain fatty acids (SCFAs). Metabolites produced by microbiota, such as neurotransmitters and hormones, may modulate and significantly affect host metabolism. The gut microbiota is becoming recognized as a second endocrine system. A variety of chronic inflammatory disorders have been linked to aberrant host-microbiota interplays, but the precise mechanisms underpinning these disturbances and how they might lead to diseases remain to be fully elucidated. Microbiome-modulated metabolites are promising targets for new drug discovery due to their endocrine function in various complex disorders. In humans, metabolotherapy for the prevention or treatment of various disorders will be possible if we better understand the metabolic preferences of bacteria and the host in specific tissues and organs. Better disease treatments may be possible with the help of novel complementary therapies that target host or bacterial metabolism. The metabolites, their physiological consequences, and functional mechanisms of the host-microbiota interplays will be highlighted, summarized, and discussed in this overview.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Anethesiology, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China.
| | - Rui Chen
- Department of Pediatrics, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China.
| | - DuoDuo Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin Province 130021, People's Republic of China.
| | - Shuang Qi
- Department of Anethesiology, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China.
| | - Yan Liu
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China.
| |
Collapse
|
10
|
Zheng Z, Hu Y, Tang J, Xu W, Zhu W, Zhang W. The implication of gut microbiota in recovery from gastrointestinal surgery. Front Cell Infect Microbiol 2023; 13:1110787. [PMID: 36926517 PMCID: PMC10011459 DOI: 10.3389/fcimb.2023.1110787] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
Recovery from gastrointestinal (GI) surgery is often interrupted by the unpredictable occurrence of postoperative complications, including infections, anastomotic leak, GI dysmotility, malabsorption, cancer development, and cancer recurrence, in which the implication of gut microbiota is beginning to emerge. Gut microbiota can be imbalanced before surgery due to the underlying disease and its treatment. The immediate preparations for GI surgery, including fasting, mechanical bowel cleaning, and antibiotic intervention, disrupt gut microbiota. Surgical removal of GI segments also perturbs gut microbiota due to GI tract reconstruction and epithelial barrier destruction. In return, the altered gut microbiota contributes to the occurrence of postoperative complications. Therefore, understanding how to balance the gut microbiota during the perioperative period is important for surgeons. We aim to overview the current knowledge to investigate the role of gut microbiota in recovery from GI surgery, focusing on the crosstalk between gut microbiota and host in the pathogenesis of postoperative complications. A comprehensive understanding of the postoperative response of the GI tract to the altered gut microbiota provides valuable cues for surgeons to preserve the beneficial functions and suppress the adverse effects of gut microbiota, which will help to enhance recovery from GI surgery.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei Zhang
- Department of General Surgery, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
11
|
Zhang XL, Chen L, Yang J, Zhao SS, Jin S, Ao N, Yang J, Liu HX, Du J. Vitamin D alleviates non-alcoholic fatty liver disease via restoring gut microbiota and metabolism. Front Microbiol 2023; 14:1117644. [PMID: 36819064 PMCID: PMC9932697 DOI: 10.3389/fmicb.2023.1117644] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/10/2023] [Indexed: 02/05/2023] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) represents a severe public health problem. Dysbiosis of gut microbiome has been identified as one of the key environmental factors contributing to NAFLD. As an essential nutrition, Vitamin D (VD) plays an important role in regulating gut microbiota based on its receptor (Vitamin D Receptor, VDR) which is highly expressed in the gastrointestinal tract. Methods Rats were fed with HFD (high-fat diet) for 12 weeks. And the rats were treated with VD two times a week by intraperitoneal injection for 12 weeks. H&E staining combined with plasma biochemical index was performed to characterize pathological changes and function of the liver. Fecal microbiota 16S rRNA gene sequencing and metabolomics were taken to reveal the altered gut microbiota and metabolites. Result The VD alleviates the HFD-induced lipid accumulation in the liver as well as decreases the levels of amlodipine besylate (ALT) and amlodipine aspartate (AST). VD supplement decreased the ratio of phylum Firmicutes/Bacteroidetes (F/B) but increased alpha diversity. In addition, the VD treatment improved the HFD-induced gut microbiota by increasing the Prevotella and Porphyromonadaceae and decreasing Mucispirillum, Acetatifactor, Desulfovibrio, and Oscillospira abundance. Furthermore, the capability of tyrosine metabolism, tryptophan metabolism, arginine biosynthesis, and sphingolipid metabolism was enhanced after VD treatment. Consistently, Prevotella positively correlated with tryptophan metabolism and sphingolipid metabolism. Importantly, the Prevotella abundance was positively associated with serotonin, melatonin, tryptamine, L-arginine, and 3-dehydrosphinganine which synthesize from tryptophan, tyrosine, arginosuccinate, and serine, respectively. Conclusion VD treatment inhibited HFD-induced NAFLD accompany by dysbiosis gut microbiota and metabolites, suggesting that VD supplement could be a potential intervention used for NAFLD treatment by targeting the specific microbiota.
Collapse
Affiliation(s)
- Xiao-Lei Zhang
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Lei Chen
- Institute of Health Sciences, China Medical University, Shenyang, Liaoning, China,Institute of Life Sciences, China Medical University, Shenyang, Liaoning, China,Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, Liaoning, China
| | - Jiang Yang
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shan-Shan Zhao
- Institute of Health Sciences, China Medical University, Shenyang, Liaoning, China,Institute of Life Sciences, China Medical University, Shenyang, Liaoning, China,Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, Liaoning, China
| | - Shi Jin
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Na Ao
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jing Yang
- Institute of Health Sciences, China Medical University, Shenyang, Liaoning, China,Institute of Life Sciences, China Medical University, Shenyang, Liaoning, China,Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, Liaoning, China
| | - Hui-Xin Liu
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China,Institute of Health Sciences, China Medical University, Shenyang, Liaoning, China,Institute of Life Sciences, China Medical University, Shenyang, Liaoning, China,Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, Liaoning, China,*Correspondence: Hui-Xin Liu,
| | - Jian Du
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China,Jian Du,
| |
Collapse
|
12
|
Fernando DG, Saravia FL, Atkinson SN, Barron M, Kirby JR, Kindel TL. A single, peri-operative antibiotic can persistently alter the post-operative gut microbiome after Roux-en-Y gastric bypass. Surg Endosc 2023; 37:1476-1486. [PMID: 35768736 DOI: 10.1007/s00464-022-09387-4] [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: 03/19/2022] [Accepted: 06/06/2022] [Indexed: 10/17/2022]
Abstract
INTRODUCTION Roux-en-Y gastric bypass (RYGB) significantly alters the gut microbiome and may be a mechanism for post-operative cardiovascular disease improvement. We have previously found an association between the class of peri-operative, intravenous antibiotic administered at the time of RYGB and the resolution rate of hypertension suggesting the gut microbiome as a mechanism. In this study, we performed a prospective study of RYGB to determine if a single intravenous antibiotic could alter the gastrointestinal microbial composition. METHODS Patients undergoing RYGB were randomized to a single, peri-operative antibiotic of intravenous cefazolin (n = 8) or clindamycin (n = 8). Stool samples were collected from four-time points: 2 weeks pre-op (- 2w), 2 days pre-op (- 2d), 2 weeks post-op (+ 2w) and 3 months post-op (+ 3m). Stool samples were processed for genomic DNA followed by Illumina 16S rRNA gene sequencing and shotgun metagenomic sequencing (MGS). RESULTS A total of 60 stool samples (- 2w, n = 16; - 2d, n = 15; + 2w, n = 16; + 3m, n = 13) from 16 patients were analyzed. 87.5% of patients were female with an average age of 48.6 ± 12.2 years and pre-operative BMI of 50.9 ± 23.3 kg/m2. RYGB induced statistically significant differences in alpha and beta diversity. There were statistically significant differences in alpha diversity at + 2w and beta diversity at + 3m due to antibiotic treatment. MGS revealed significantly distinct gut microbiota with 11 discriminatory metagenomic assembled genomes driven by antibiotic treatment at 3 months post-op, including increased Bifidobacterium spp. with clindamycin. CONCLUSION RYGB induces significant changes in the gut microbiome at 2 weeks that are maintained 3 months after surgery. However, the single peri-operative dose of antibiotic administered at the time of RYGB induces unique and persisting changes to the gut microbiome that are antibiotic-specific. Increased Bifidobacterium spp. with clindamycin administration may improve the metabolic efficacy of RYGB when considering gut-microbiome driven mechanisms for blood pressure resolution.
Collapse
Affiliation(s)
- Deemantha G Fernando
- Department of Surgery, Medical College of Wisconsin, 8900 W. Doyne Avenue, Milwaukee, WI, 53226, USA
| | - Fatima L Saravia
- Department of Microbiology & Immunology, Medical College of Wisconsin, 8900 W. Doyne Avenue, Milwaukee, WI, 53226, USA
| | - Samantha N Atkinson
- Department of Microbiology & Immunology, Medical College of Wisconsin, 8900 W. Doyne Avenue, Milwaukee, WI, 53226, USA.,Center for Microbiome Research, Medical College of Wisconsin, 8900 W. Doyne Avenue, Milwaukee, WI, 53226, USA
| | - Matthew Barron
- Department of Surgery, Medical College of Wisconsin, 8900 W. Doyne Avenue, Milwaukee, WI, 53226, USA
| | - John R Kirby
- Department of Microbiology & Immunology, Medical College of Wisconsin, 8900 W. Doyne Avenue, Milwaukee, WI, 53226, USA
| | - Tammy L Kindel
- Department of Surgery, Medical College of Wisconsin, 8900 W. Doyne Avenue, Milwaukee, WI, 53226, USA.
| |
Collapse
|
13
|
Dischinger U, Kötzner L, Kovatcheva-Datchary P, Kleinschmidt H, Haas C, Perez J, Presek C, Koschker AC, Miras AD, Hankir MK, Vogel J, Germer CT, Fassnacht M, Herrmann MJ, Seyfried F. Hypothalamic integrity is necessary for sustained weight loss after bariatric surgery: A prospective, cross-sectional study. Metabolism 2023; 138:155341. [PMID: 36341838 DOI: 10.1016/j.metabol.2022.155341] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The hypothalamus is the main integrator of peripheral and central signals in the control of energy homeostasis. Its functional relevance for the effectivity of bariatric surgery is not entirely elucidated. Studying the effects of bariatric surgery in patients with hypothalamic damage might provide insight. SUMMARY BACKGROUND DATA Prospective study to analyze the effects of bariatric surgery in patients with hypothalamic obesity (HO) vs. matched patients with common obesity (CO) with and without bariatric surgery. METHODS 65 participants were included (HO-surgery: n = 8, HO-control: n = 10, CO-surgery: n = 12, CO-control: n = 12, Lean-control: n = 23). Body weight, levels of anorexic hormones, gut microbiota, as well as subjective well-being/health status, eating behavior, and brain activity (via functional MRI) were evaluated. RESULTS Patients with HO lost significantly less weight after bariatric surgery than CO-participants (total body weight loss %: 5.5 % vs. 26.2 %, p = 0.0004). After a mixed meal, satiety and abdominal fullness tended to be lowest in HO-surgery and did not correlate with levels of GLP-1 or PYY. Levels of PYY (11,151 ± 1667 pmol/l/h vs. 8099 ± 1235 pmol/l/h, p = 0.028) and GLP-1 (20,975 ± 2893 pmol/l/h vs. 13,060 ± 2357 pmol/l/h, p = 0.009) were significantly higher in the HO-surgery vs. CO-surgery group. Abundance of Enterobacteriaceae and Streptococcus was increased in feces of HO and CO after bariatric surgery. Comparing HO patients with lean-controls revealed an increased activation in insula and cerebellum to viewing high-caloric foods in left insula and cerebellum in fMRI. CONCLUSIONS Hypothalamic integrity is necessary for the effectiveness of bariatric surgery in humans. Peripheral changes after bariatric surgery are not sufficient to induce satiety and long-term weight loss in patients with hypothalamic damage.
Collapse
Affiliation(s)
- Ulrich Dischinger
- Department of Internal Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Germany.
| | - Laura Kötzner
- Department of Internal Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Germany
| | | | - Helena Kleinschmidt
- Department of Internal Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Germany
| | - Christina Haas
- Department of Internal Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Germany
| | - Jose Perez
- Department of Neurosurgery, University Hospital, University of Würzburg, Germany
| | - Cornelius Presek
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital, University of Würzburg, Germany
| | - Ann-Cathrin Koschker
- Department of Internal Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Germany
| | - Alexander D Miras
- Department of Metabolism, Digestion and Reproduction, Imperial College London, United Kingdom
| | - Mohammed K Hankir
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital, University of Würzburg, Germany
| | - Jörg Vogel
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Germany; Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg, Germany
| | - Christoph-Thomas Germer
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital, University of Würzburg, Germany
| | - Martin Fassnacht
- Department of Internal Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Germany
| | - Martin J Herrmann
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital, University of Würzburg, Germany
| | - Florian Seyfried
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital, University of Würzburg, Germany
| |
Collapse
|
14
|
Canyelles M, Pérez A, Junza A, Miñambres I, Yanes O, Sardà H, Rotllan N, Julve J, Sánchez-Quesada JL, Tondo M, Escolà-Gil JC, Blanco-Vaca F. Divergent Effects of Glycemic Control and Bariatric Surgery on Circulating Concentrations of TMAO in Newly Diagnosed T2D Patients and Morbidly Obese. Diagnostics (Basel) 2022; 12:diagnostics12112783. [PMID: 36428843 PMCID: PMC9689652 DOI: 10.3390/diagnostics12112783] [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: 10/12/2022] [Revised: 11/07/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
High circulating concentrations of the gut microbiota-derived metabolite trimethylamine N-oxide (TMAO) are significantly associated with the risk of obesity and type 2 diabetes (T2D). We aimed at evaluating the impact of glycemic control and bariatric surgery on circulating concentrations of TMAO and its microbiota-dependent intermediate, γ-butyrobetaine (γBB), in newly diagnosed T2D patients and morbidly obese subjects following a within-subject design. Based on HbA1c concentrations, T2D patients achieved glycemic control. However, the plasma TMAO and γBB concentrations were significantly increased, without changes in estimated glomerular filtration rate. Bariatric surgery was very effective in reducing weight in obese subjects. Nevertheless, the surgery reduced plasma γBB concentrations without affecting TMAO concentrations and the estimated glomerular filtration rate. Considering these results, an additional experiment was carried out in male C57BL/6J mice fed a Western-type diet for twelve weeks. Neither diet-induced obesity nor insulin resistance were associated with circulating TMAO and γBB concentrations in these genetically defined mice strains. Our findings do not support that glycemic control or bariatric surgery improve the circulating concentrations of TMAO in newly diagnosed T2D and morbidly obese patients.
Collapse
Affiliation(s)
- Marina Canyelles
- Institut de Recerca de l’Hospital Santa Creu i Sant Pau, Institut d’Investigacions Biomèdiques IIB Sant Pau, 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Antonio Pérez
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Department of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, 08041 Barcelona, Spain
| | - Alexandra Junza
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Metabolomics Platform, Department of Electronic Engineering, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Inka Miñambres
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Department of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, 08041 Barcelona, Spain
| | - Oscar Yanes
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Metabolomics Platform, Department of Electronic Engineering, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Helena Sardà
- Department of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, 08041 Barcelona, Spain
| | - Noemí Rotllan
- Institut de Recerca de l’Hospital Santa Creu i Sant Pau, Institut d’Investigacions Biomèdiques IIB Sant Pau, 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Josep Julve
- Institut de Recerca de l’Hospital Santa Creu i Sant Pau, Institut d’Investigacions Biomèdiques IIB Sant Pau, 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - José Luis Sánchez-Quesada
- Institut de Recerca de l’Hospital Santa Creu i Sant Pau, Institut d’Investigacions Biomèdiques IIB Sant Pau, 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Mireia Tondo
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Department of Clinical Biochemistry, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, 08041 Barcelona, Spain
- Correspondence: (M.T.); (F.B.-V.)
| | - Joan Carles Escolà-Gil
- Institut de Recerca de l’Hospital Santa Creu i Sant Pau, Institut d’Investigacions Biomèdiques IIB Sant Pau, 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Francisco Blanco-Vaca
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Department of Clinical Biochemistry, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, 08041 Barcelona, Spain
- Department de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08041 Barcelona, Spain
- Correspondence: (M.T.); (F.B.-V.)
| |
Collapse
|
15
|
Martin WP, Nair M, Chuah YH, Malmodin D, Pedersen A, Abrahamsson S, Hutter M, Abdelaal M, Elliott JA, Fearon N, Eckhardt H, Godson C, Brennan EP, Fändriks L, le Roux CW, Docherty NG. Dietary restriction and medical therapy drives PPARα-regulated improvements in early diabetic kidney disease in male rats. Clin Sci (Lond) 2022; 136:1485-1511. [PMID: 36259366 PMCID: PMC7613831 DOI: 10.1042/cs20220205] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 11/17/2022]
Abstract
The attenuation of diabetic kidney disease (DKD) by metabolic surgery is enhanced by pharmacotherapy promoting renal fatty acid oxidation (FAO). Using the Zucker Diabetic Fatty and Zucker Diabetic Sprague Dawley rat models of DKD, we conducted studies to determine if these effects could be replicated with a non-invasive bariatric mimetic intervention. Metabolic control and renal injury were compared in rats undergoing a dietary restriction plus medical therapy protocol (DMT; fenofibrate, liraglutide, metformin, ramipril, and rosuvastatin) and ad libitum-fed controls. The global renal cortical transcriptome and urinary 1H-NMR metabolomic profiles were also compared. Kidney cell type-specific and medication-specific transcriptomic responses were explored through in silico deconvolution. Transcriptomic and metabolomic correlates of improvements in kidney structure were defined using a molecular morphometric approach. The DMT protocol led to ∼20% weight loss, normalized metabolic parameters and was associated with reductions in indices of glomerular and proximal tubular injury. The transcriptomic response to DMT was dominated by changes in fenofibrate- and peroxisome proliferator-activated receptor-α (PPARα)-governed peroxisomal and mitochondrial FAO transcripts localizing to the proximal tubule. DMT induced urinary excretion of PPARα-regulated metabolites involved in nicotinamide metabolism and reversed DKD-associated changes in the urinary excretion of tricarboxylic acid (TCA) cycle intermediates. FAO transcripts and urinary nicotinamide and TCA cycle metabolites were moderately to strongly correlated with improvements in glomerular and proximal tubular injury. Weight loss plus pharmacological PPARα agonism is a promising means of attenuating DKD.
Collapse
Affiliation(s)
- William P. Martin
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Meera Nair
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Yeong H.D. Chuah
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Daniel Malmodin
- Swedish NMR Centre, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Anders Pedersen
- Swedish NMR Centre, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Sanna Abrahamsson
- Bioinformatics Core Facility, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Michaela Hutter
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Mahmoud Abdelaal
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Jessie A. Elliott
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Naomi Fearon
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Hans Eckhardt
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Catherine Godson
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Eoin P. Brennan
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Lars Fändriks
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Carel W. le Roux
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
- Diabetes Research Group, Ulster University, Coleraine BT52 1SA, UK
| | - Neil G. Docherty
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| |
Collapse
|
16
|
Yang J, Chen L, Shang XY, Chen YL, Zhao SS, Jin S, Yang J, Liu HX, Du J. Roux-en-Y gastric bypass-induced perturbative changes in microbial communities and metabolic pathways in rats. Front Microbiol 2022; 13:1034839. [DOI: 10.3389/fmicb.2022.1034839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/07/2022] [Indexed: 11/11/2022] Open
Abstract
BackgroundObesity has become a global health and socioeconomic problem because of an inadequate balance between energy intake and energy expenditure. Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG) are the two most commonly used strategies for weight loss, which have been proven to benefit from gut microbiota restoration.MethodsRats received SG, RYGB, and sham operations for 10 weeks. At the end of the experiment, the fecal microbiota was analyzed using 16s rRNA gene sequencing. In addition, the shift in the plasma metabolism of rats that underwent RYGB surgery was analyzed using untargeted metabolomics. The crosstalk between microbiome and metabolites was revealed using metabolic pathway enrichment and integrated analysis.ResultThe SG surgery induced a modest shift in the gut microbiota relative to the RYGB. RYGB significantly decreased the alpha diversity and Firmicutes/Bacteroides (F/B) ratio and increased the proportion of Escherichia, Bacteroides, and Akkermansia genera compared to sham and SG operations. The predicted function of gut microbiota revealed that the RYGB surgery uniquely enhanced the capability of linoleic acid and sphingolipid metabolism. Furthermore, the circulating serine, phosphatidylcholine (PC) 20:5/22:5, riboflavin, L–carnitine, and linoleic acid were evaluated after RYGB surgery. In addition, the metabolic pathway enrichment and integrated analysis suggest that the RYGB induced Escherichia, Bacteroides, and Akkermansia might inhibit the sphingonine and phytosphingosine metabolisms from serine and promote the PC (20:5/22:5) metabolism to produce linoleic acid.ConclusionThis comprehensive analysis not only revealed the difference in the gut microbiota shifts after SG and RYGB but also discovered the perturbative changes in microbial communities and metabolic pathways after RYGB surgery, which provided clues for improving the beneficial effect of RYGB in metabolic disease intervention via regulating bacterial-metabolite crosstalk.
Collapse
|
17
|
Xia Y, Ren M, Yang J, Cai C, Cheng W, Zhou X, Lu D, Ji F. Gut microbiome and microbial metabolites in NAFLD and after bariatric surgery: Correlation and causality. Front Microbiol 2022; 13:1003755. [PMID: 36204626 PMCID: PMC9531827 DOI: 10.3389/fmicb.2022.1003755] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is currently related to a heavy socioeconomic burden and increased incidence. Since obesity is the most prevalent risk factor for NAFLD, weight loss is an effective therapeutic solution. Bariatric surgery (BS), which can achieve long-term weight loss, improves the overall health of patients with NAFLD. The two most common surgeries are the Roux-en-Y gastric bypass and sleeve gastrectomy. The gut-liver axis is the complex network of cross-talking between the gut, its microbiome, and the liver. The gut microbiome, involved in the homeostasis of the gut-liver axis, is believed to play a significant role in the pathogenesis of NAFLD and the metabolic improvement after BS. Alterations in the gut microbiome in NAFLD have been confirmed compared to that in healthy individuals. The mechanisms linking the gut microbiome to NAFLD have been proposed, including increased intestinal permeability, higher energy intake, and other pathophysiological alterations. Interestingly, several correlation studies suggested that the gut microbial signatures after BS become more similar to those of lean, healthy controls than that of patients with NAFLD. The resolution of NAFLD after BS is related to changes in the gut microbiome and its metabolites. However, confirming a causal link remains challenging. This review summarizes characteristics of the gut microbiome in patients with NAFLD before and after BS and accumulates existing evidence about the underlying mechanisms of the gut microbiome.
Collapse
Affiliation(s)
- Yi Xia
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengting Ren
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinpu Yang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Changzhou Cai
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weixin Cheng
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinxin Zhou
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan Lu
- Department of Endoscopy Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Ji
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Feng Ji,
| |
Collapse
|
18
|
Kim Y, Son D, Kim BK, Kim KH, Seo KW, Jung K, Park SJ, Lim S, Kim JH. Association between the Blautia/Bacteroides Ratio and Altered Body Mass Index after Bariatric Surgery. Endocrinol Metab (Seoul) 2022; 37:475-486. [PMID: 35798551 PMCID: PMC9262679 DOI: 10.3803/enm.2022.1481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 05/30/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGRUOUND Current evidence support that the gut microbiota plays a potential role in obesity. Bariatric surgery can reduce excess weight and decrease the risk of life-threatening weight-related health problems and may also influence gut microbiota. In this study, we aimed to investigate the changes in gut microbiota before and after bariatric surgery and evaluate the association of the gut microbial shift and altered body mass index (BMI) after bariatric surgery. METHODS Between January 2019 and July 2020, stools from 58 patients scheduled for bariatric surgery were collected. Six months after bariatric surgery, stools from 22 of these patients were re-collected, and the changes in gut microbiota before and after bariatric surgery were evaluated. In addition, the differences in gut microbiota between patients with severe obesity (BMI >35 kg/m2, n=42) and healthy volunteers with normal BMI (18.8 to 22.8 kg/m2, n=41) were investigated. RESULTS The gut microbiota of patients who underwent bariatric surgery showed increased α-diversity and differed β-diversity compared with those before surgery. Interestingly, Blautia was decreased and Bacteriodes was increased at the genus level after bariatric surgery. Further, the Blautia/Bacteroides ratio showed a positive correlation with BMI. To validate these results, we compared the gut microbiota from severely obese patients with high BMI with those from healthy volunteers and demonstrated that the Blautia/Bacteroides ratio correlated positively with BMI. CONCLUSION In the gut microbial analysis of patients who underwent bariatric surgery, we presented that the Blautia/Bacteroides ratio had changed after bariatric surgery and showed a positive correlation with BMI.
Collapse
Affiliation(s)
- Yoonhong Kim
- Department of Surgery, Kosin University College of Medicine, Busan, Korea
| | | | - Bu Kyung Kim
- Department of Internal Medicine, Kosin University College of Medicine, Busan, Korea
| | - Ki Hyun Kim
- Department of Surgery, Kosin University College of Medicine, Busan, Korea
| | - Kyung Won Seo
- Department of Surgery, Kosin University College of Medicine, Busan, Korea
| | - Kyoungwon Jung
- Department of Internal Medicine, Kosin University College of Medicine, Busan, Korea
| | - Seun Ja Park
- Department of Internal Medicine, Kosin University College of Medicine, Busan, Korea
| | - Sanghyun Lim
- Cell Biotech Co. Ltd., Gimpo, Korea
- Sanghyun Lim Cell Biotech Co. Ltd., 50 Aegibong-ro 409beon-gil, Wolgot-myeon, Gimpo 10003, Korea Tel: +82-31-987-6205, Fax: +82-31-987-6209, E-mail:
| | - Jae Hyun Kim
- Department of Internal Medicine, Kosin University College of Medicine, Busan, Korea
- Corresponding authors: Jae Hyun Kim Department of Internal Medicine, Kosin University College of Medicine, 262 Gamcheon-ro, Seo-gu, Busan 49267, Korea Tel: +82-51-990-5061, Fax: +82-51-990-5055, E-mail:
| |
Collapse
|
19
|
Lin Z, Ma X. Dietary nutrients mediate crosstalk between bile acids and gut microbes in animal host metabolism. Crit Rev Food Sci Nutr 2022; 63:9315-9329. [PMID: 35507502 DOI: 10.1080/10408398.2022.2067118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Bile acids (BAs) are synthesized by liver, then gut microbes embellish primary BAs into secondary BAs with diverse and biological functions. Over the past few decades, amounts of evidences demonstrated the importance of gut microbes in BA metabolism. There is also significant evidence that BAs are regarded as cell signals in gut-liver, gut-brain, and gut-testis axis. Moreover, the interaction between BAs and gut microbes plays a key role not only in the absorption and metabolism of nutrients, but the regulation of immune function. Herein, we collected the major information of the BA metabolism-related bacteria, nutrients, and cell signals, focused on the possible molecular mechanisms by "Microbes-Bile acids" crosstalk, highlighted the gut-liver, gut-brain, and gut-testis axis, and discussed the possibility and application of the regulation of BA metabolism by nutrients.
Collapse
Affiliation(s)
- Zishen Lin
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
20
|
Loo RL, Chan Q, Nicholson JK, Holmes E. Balancing the Equation: A Natural History of Trimethylamine and Trimethylamine- N-oxide. J Proteome Res 2022; 21:560-589. [PMID: 35142516 DOI: 10.1021/acs.jproteome.1c00851] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Trimethylamine (TMA) and its N-oxide (TMAO) are ubiquitous in prokaryote and eukaryote organisms as well as in the environment, reflecting their fundamental importance in evolutionary biology, and their diverse biochemical functions. Both metabolites have multiple biological roles including cell-signaling. Much attention has focused on the significance of serum and urinary TMAO in cardiovascular disease risk, yet this is only one of the many facets of a deeper TMA-TMAO partnership that reflects the significance of these metabolites in multiple biological processes spanning animals, plants, bacteria, and fungi. We report on analytical methods for measuring TMA and TMAO and attempt to critically synthesize and map the global functions of TMA and TMAO in a systems biology framework.
Collapse
Affiliation(s)
- Ruey Leng Loo
- Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, Western Australia 6150, Australia.,The Australian National Phenome Centre, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, Western Australia 6150, Australia
| | - Queenie Chan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, United Kingdom.,MRC Centre for Environment and Health, School of Public Health, Imperial College London, London W2 1PG, United Kingdom
| | - Jeremy K Nicholson
- Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, Western Australia 6150, Australia.,The Australian National Phenome Centre, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, Western Australia 6150, Australia.,Institute of Global Health Innovation, Imperial College London, Level 1, Faculty Building, South Kensington Campus, London SW7 2NA, United Kingdom
| | - Elaine Holmes
- Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, Western Australia 6150, Australia.,The Australian National Phenome Centre, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, Western Australia 6150, Australia.,Nutrition Research, Department of Metabolism, Nutrition and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, London SW7 2AZ, United Kingdom
| |
Collapse
|
21
|
Ramaboli M, Nesengani L, Katsidzira L, Haller D, Kinross J, Ocvirk S, O'Keefe SJD. Interactions between the environmental and human microbiota in the preservation of health and genesis of disease: symposium report. Curr Opin Gastroenterol 2022; 38:146-155. [PMID: 35098936 DOI: 10.1097/mog.0000000000000817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
PURPOSE OF REVIEW The purpose of this symposium was to bring thought leaders in the microbiome from the west to Africa to share their unique experiences with African investigators in order to build the foundations for scientifically rigorous explorations into the African human and environmental microbiome that may explain why disease patterns are different in Africa where the chief killers are infectious diseases, whereas noncommunicable diseases (NCDs) are the major threat to healthcare resources in the developed world. RECENT FINDINGS The application of new high throughput technologies to the investigation of the microbiome and its metabolome has revealed mechanisms whereby a traditional African high fiber diet can suppress NCDs which include colon cancer, inflammatory bowel diseases, obesity, type 2 diabetes and atherosclosis. There is concern that with migration and westernization, NCDs are becoming more common in Africa and that food security is becoming impaired by unbalanced obesogenic foods rather than inadequate food intake. SUMMARY There is an urgent need for the formation of combined African-Western research programs to identify what is good and bad in the African diet-microbiome axis to develop strategies to prevent the incidence of NCDs rising to western levels in Africa, at the same time offering novel prevention strategies against the #1 healthcare threat in the developed world.
Collapse
Affiliation(s)
- Matsepo Ramaboli
- African Microbiome Institute, Department of Internal Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Lucky Nesengani
- African Microbiome Institute, Department of Internal Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Leolin Katsidzira
- Department of Medicine, College of Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Dirk Haller
- ZIEL Institute for Food and Health, Technical University of Munich, Freising-Weihenstephan, Germany
| | - James Kinross
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Soeren Ocvirk
- Intestinal Microbiology Research Group, Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Stephen J D O'Keefe
- African Microbiome Institute, Department of Internal Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.,Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
22
|
Martin WP, Malmodin D, Pedersen A, Wallace M, Fändriks L, Aboud CM, Petry TBZ, Cunha da Silveira LP, da Costa Silva ACC, Cohen RV, le Roux CW, Docherty NG. Urinary Metabolomic Changes Accompanying Albuminuria Remission following Gastric Bypass Surgery for Type 2 Diabetic Kidney Disease. Metabolites 2022; 12:139. [PMID: 35186675 PMCID: PMC7612403 DOI: 10.3390/metabo12020139] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
In the Microvascular Outcomes after Metabolic Surgery randomised clinical trial (MOMS RCT, NCT01821508), combined metabolic surgery (gastric bypass) plus medical therapy (CSM) was superior to medical therapy alone (MTA) as a means of achieving albuminuria remission at 2-year follow-up in patients with obesity and early diabetic kidney disease (DKD). In the present study, we assessed the urinary 1H-NMR metabolome in a subgroup of patients from both arms of the MOMS RCT at baseline and 6-month follow-up. Whilst CSM and MTA both reduced the urinary excretion of sugars, CSM generated a distinctive urinary metabolomic profile characterised by increases in host–microbial co-metabolites (N-phenylacetylglycine, trimethylamine N-oxide, and 4-aminobutyrate (GABA)) and amino acids (arginine and glutamine). Furthermore, reductions in aromatic amino acids (phenylalanine and tyrosine), as well as branched-chain amino acids (BCAAs) and related catabolites (valine, leucine, 3-hydroxyisobutyrate, 3-hydroxyisovalerate, and 3-methyl-2-oxovalerate), were observed following CSM but not MTA. Improvements in BMI did not correlate with improvements in metabolic and renal indices following CSM. Conversely, urinary metabolites changed by CSM at 6 months were moderately to strongly correlated with improvements in blood pressure, glycaemia, triglycerides, and albuminuria up to 24 months following treatment initiation, highlighting the potential involvement of these shifts in the urinary metabolomic profile in the metabolic and renoprotective effects of CSM.
Collapse
Affiliation(s)
- William P. Martin
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland; (W.P.M.); (C.W.l.R.)
| | - Daniel Malmodin
- Swedish NMR Centre, University of Gothenburg, 40530 Gothenburg, Sweden; (D.M.); (A.P.)
| | - Anders Pedersen
- Swedish NMR Centre, University of Gothenburg, 40530 Gothenburg, Sweden; (D.M.); (A.P.)
| | - Martina Wallace
- Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland;
| | - Lars Fändriks
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden;
| | - Cristina M. Aboud
- The Centre for Obesity and Diabetes, Oswaldo Cruz German Hospital, São Paulo 01333-010, Brazil; (C.M.A.); (T.B.Z.P.); (L.P.C.d.S.); (A.C.C.d.C.S.); (R.V.C.)
| | - Tarissa B. Zanata Petry
- The Centre for Obesity and Diabetes, Oswaldo Cruz German Hospital, São Paulo 01333-010, Brazil; (C.M.A.); (T.B.Z.P.); (L.P.C.d.S.); (A.C.C.d.C.S.); (R.V.C.)
| | - Lívia P. Cunha da Silveira
- The Centre for Obesity and Diabetes, Oswaldo Cruz German Hospital, São Paulo 01333-010, Brazil; (C.M.A.); (T.B.Z.P.); (L.P.C.d.S.); (A.C.C.d.C.S.); (R.V.C.)
| | - Ana C. Calmon da Costa Silva
- The Centre for Obesity and Diabetes, Oswaldo Cruz German Hospital, São Paulo 01333-010, Brazil; (C.M.A.); (T.B.Z.P.); (L.P.C.d.S.); (A.C.C.d.C.S.); (R.V.C.)
| | - Ricardo V. Cohen
- The Centre for Obesity and Diabetes, Oswaldo Cruz German Hospital, São Paulo 01333-010, Brazil; (C.M.A.); (T.B.Z.P.); (L.P.C.d.S.); (A.C.C.d.C.S.); (R.V.C.)
| | - Carel W. le Roux
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland; (W.P.M.); (C.W.l.R.)
- Diabetes Research Group, Ulster University, Coleraine BT52 1SA, UK
| | - Neil G. Docherty
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland; (W.P.M.); (C.W.l.R.)
- Correspondence:
| |
Collapse
|
23
|
Yang Y, Huang Y, Lin W, Liu J, Chen X, Chen C, Yu X, Teng L. Host miRNAs-microbiota interactions in gastric cancer. J Transl Med 2022; 20:52. [PMID: 35093110 PMCID: PMC8800214 DOI: 10.1186/s12967-022-03264-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/17/2022] [Indexed: 12/24/2022] Open
Abstract
It is widely acknowledged that gastric cancer seriously affects the quality of life and survival of patients. The correlation between the microbiota and gastric cancer has attracted extensive attention in recent years, nonetheless the specific mechanism of its impact on gastric cancer remain largely unclear. Recent studies have shown that in addition to its role in the host’s inflammatory and immune response, the microbiota can also affect the occurrence and development of gastric cancer by affecting the expression of miRNAs. This paper brings together all currently available data on miRNAs, microbiota and gastric cancer, and preliminarily describes the relationship among them.
Collapse
|
24
|
Jones B, Sands C, Alexiadou K, Minnion J, Tharakan G, Behary P, Ahmed AR, Purkayastha S, Lewis MR, Bloom S, Li JV, Tan TM. The Metabolomic Effects of Tripeptide Gut Hormone Infusion Compared to Roux-en-Y Gastric Bypass and Caloric Restriction. J Clin Endocrinol Metab 2022; 107:e767-e782. [PMID: 34460933 PMCID: PMC8764224 DOI: 10.1210/clinem/dgab608] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Indexed: 12/23/2022]
Abstract
CONTEXT The gut-derived peptide hormones glucagon-like peptide-1 (GLP-1), oxyntomodulin (OXM), and peptide YY (PYY) are regulators of energy intake and glucose homeostasis and are thought to contribute to the glucose-lowering effects of bariatric surgery. OBJECTIVE To establish the metabolomic effects of a combined infusion of GLP-1, OXM, and PYY (tripeptide GOP) in comparison to a placebo infusion, Roux-en-Y gastric bypass (RYGB) surgery, and a very low-calorie diet (VLCD). DESIGN AND SETTING Subanalysis of a single-blind, randomized, placebo-controlled study of GOP infusion (ClinicalTrials.gov NCT01945840), including VLCD and RYGB comparator groups. PATIENTS AND INTERVENTIONS Twenty-five obese patients with type 2 diabetes or prediabetes were randomly allocated to receive a 4-week subcutaneous infusion of GOP (n = 14) or 0.9% saline control (n = 11). An additional 22 patients followed a VLCD, and 21 underwent RYGB surgery. MAIN OUTCOME MEASURES Plasma and urine samples collected at baseline and 4 weeks into each intervention were subjected to cross-platform metabolomic analysis, followed by unsupervised and supervised modeling approaches to identify similarities and differences between the effects of each intervention. RESULTS Aside from glucose, very few metabolites were affected by GOP, contrasting with major metabolomic changes seen with VLCD and RYGB. CONCLUSIONS Treatment with GOP provides a powerful glucose-lowering effect but does not replicate the broader metabolomic changes seen with VLCD and RYGB. The contribution of these metabolomic changes to the clinical benefits of RYGB remains to be elucidated.
Collapse
MESH Headings
- Adult
- Aged
- Blood Glucose/analysis
- Caloric Restriction/methods
- Caloric Restriction/statistics & numerical data
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/therapy
- Diabetes Mellitus, Type 2/urine
- Drug Therapy, Combination/methods
- Female
- Gastric Bypass/methods
- Gastric Bypass/statistics & numerical data
- Gastrointestinal Hormones/administration & dosage
- Glucagon-Like Peptide 1/administration & dosage
- Humans
- Infusions, Subcutaneous
- Male
- Metabolomics/statistics & numerical data
- Middle Aged
- Obesity, Morbid/blood
- Obesity, Morbid/metabolism
- Obesity, Morbid/therapy
- Obesity, Morbid/urine
- Oxyntomodulin/administration & dosage
- Peptide YY/administration & dosage
- Single-Blind Method
- Treatment Outcome
- Weight Loss
- Young Adult
Collapse
Affiliation(s)
- Ben Jones
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Caroline Sands
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Kleopatra Alexiadou
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - James Minnion
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - George Tharakan
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Preeshila Behary
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Ahmed R Ahmed
- Department of Surgery and Cancer, Imperial College Healthcare NHS Trust, London, UK
| | - Sanjay Purkayastha
- Department of Surgery and Cancer, Imperial College Healthcare NHS Trust, London, UK
| | - Matthew R Lewis
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Stephen Bloom
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Jia V Li
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Tricia M Tan
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
- Correspondence: Tricia M. Tan, MB, ChB, BSc, PhD, FRCP, FRCPath, 6th Floor, Commonwealth Building, Hammersmith Campus, Imperial College London, London W12 0HS, UK.
| |
Collapse
|
25
|
Yeung KTD, Penney N, Whiley L, Ashrafian H, Lewis MR, Purkayastha S, Darzi A, Holmes E. The impact of bariatric surgery on serum tryptophan-kynurenine pathway metabolites. Sci Rep 2022; 12:294. [PMID: 34996930 PMCID: PMC8741964 DOI: 10.1038/s41598-021-03833-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 12/03/2021] [Indexed: 11/09/2022] Open
Abstract
This study aims to explore the immediate effects of bariatric surgery on serum tryptophan–kynurenine pathway metabolites in individuals with type 2 diabetes and BMI > 30. With the goal of providing insight into the link between tryptophan pathway metabolites, type 2 diabetes, and chronic obesity-induced inflammation. This longitudinal study included 20 participants. Half were diagnosed with type 2 diabetes. 11 and 9 underwent RYGB and SG respectively. Blood samples were obtained at pre-operative and 3 months post-operative timepoints. Tryptophan and downstream metabolites of the kynurenine pathway were quantified with an ultrahigh-performance liquid chromatography tandem mass spectrometry with electrospray ionisation method. At 3 months post-operation, RYGB led to significant reductions in tryptophan, kynurenic acid and xanthurenic acid levels when compared to baseline. Significant reductions of the same metabolites after surgery were also observed in individuals with T2D irrespective of surgical procedure. These metabolites were significantly correlated with serum HbA1c levels and BMI. Bariatric surgery, in particular RYGB reduces serum levels of tryptophan and its downstream kynurenine metabolites. These metabolites are associated with T2D and thought to be potentially mechanistic in the systemic processes of obesity induced inflammation leading to insulin resistance. Its reduction after surgery is associated with an improvement in glycaemic control (HbA1c).
Collapse
Affiliation(s)
- Kai Tai Derek Yeung
- Department of Surgery & Cancer, Imperial College London, South Kensington, London, UK
| | - Nicholas Penney
- Department of Surgery & Cancer, Imperial College London, South Kensington, London, UK
| | - Luke Whiley
- Department of Surgery & Cancer, Imperial College London, South Kensington, London, UK.,Australian National Phenome Centre & Centre for Computational & Systems Medicine, Health Futures Institute, Murdoch University, Perth, WA, Australia.,Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington, London, UK
| | - Hutan Ashrafian
- Department of Surgery & Cancer, Imperial College London, South Kensington, London, UK
| | - Matthew R Lewis
- National Phenome Centre, Imperial College London, South Kensington, London, UK.,Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington, London, UK
| | - Sanjay Purkayastha
- Department of Surgery & Cancer, Imperial College London, South Kensington, London, UK
| | - Ara Darzi
- Department of Surgery & Cancer, Imperial College London, South Kensington, London, UK
| | - Elaine Holmes
- Department of Surgery & Cancer, Imperial College London, South Kensington, London, UK. .,Australian National Phenome Centre & Centre for Computational & Systems Medicine, Health Futures Institute, Murdoch University, Perth, WA, Australia.
| |
Collapse
|
26
|
Terburgh K, Lindeque JZ, van der Westhuizen FH, Louw R. Cross-comparison of systemic and tissue-specific metabolomes in a mouse model of Leigh syndrome. Metabolomics 2021; 17:101. [PMID: 34792662 DOI: 10.1007/s11306-021-01854-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/03/2021] [Indexed: 01/03/2023]
Abstract
INTRODUCTION The value of metabolomics in multi-systemic mitochondrial disease research has been increasingly recognized, with the ability to investigate a variety of biofluids and tissues considered a particular advantage. Although minimally invasive biofluids are the generally favored sample type, it remains unknown whether systemic metabolomes provide a clear reflection of tissue-specific metabolic alterations. OBJECTIVES Here we cross-compare urine and tissue-specific metabolomes in the Ndufs4 knockout mouse model of Leigh syndrome-a complex neurometabolic MD defined by progressive focal lesions in specific brain regions-to identify and evaluate the extent of common and unique metabolic alterations on a systemic and brain regional level. METHODS Untargeted and semi-targeted multi-platform metabolomics were performed on urine, four brain regions, and two muscle types of Ndufs4 KO (n≥19) vs wildtype (n≥20) mice. RESULTS Widespread alterations were evident in alanine, aspartate, glutamate, and arginine metabolism in Ndufs4 KO mice; while brain-region specific metabolic signatures include the accumulation of branched-chain amino acids, proline, and glycolytic intermediates. Furthermore, we describe a systemic dysregulation in one-carbon metabolism and the tricarboxylic acid cycle, which was not clearly reflected in the Ndufs4 KO brain. CONCLUSION Our results confirm the value of urinary metabolomics when evaluating MD-associated metabolites, while cautioning against mechanistic studies relying solely on systemic biofluids.
Collapse
Affiliation(s)
- Karin Terburgh
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), Private Bag X6001, Potchefstroom, South Africa
| | - Jeremie Z Lindeque
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), Private Bag X6001, Potchefstroom, South Africa
| | - Francois H van der Westhuizen
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), Private Bag X6001, Potchefstroom, South Africa
| | - Roan Louw
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), Private Bag X6001, Potchefstroom, South Africa.
| |
Collapse
|
27
|
Zhang C, Yang M. Current Options and Future Directions for NAFLD and NASH Treatment. Int J Mol Sci 2021; 22:ijms22147571. [PMID: 34299189 PMCID: PMC8306701 DOI: 10.3390/ijms22147571] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide, with a broad spectrum ranging from simple steatosis to advanced stage of nonalcoholic steatohepatitis (NASH). Although there are many undergoing clinical trials for NAFLD treatment, there is no currently approved treatment. NAFLD accounts as a major causing factor for the development of hepatocellular carcinoma (HCC), and its incidence rises accompanying the prevalence of obesity and diabetes. Reprogramming of antidiabetic and anti-obesity medicine is a major treatment option for NAFLD and NASH. Liver inflammation and cellular death, with or without fibrosis account for the progression of NAFLD to NASH. Therefore, molecules and signaling pathways involved in hepatic inflammation, fibrosis, and cell death are critically important targets for the therapy of NAFLD and NASH. In addition, the avoidance of aberrant infiltration of inflammatory cytokines by treating with CCR antagonists also provides a therapeutic option. Currently, there is an increasing number of pre-clinical and clinical trials undergoing to evaluate the effects of antidiabetic and anti-obesity drugs, antibiotics, pan-caspase inhibitors, CCR2/5 antagonists, and others on NAFLD, NASH, and liver fibrosis. Non-invasive serum diagnostic markers are developed for fulfilling the need of diagnostic testing in a large amount of NAFLD cases. Overall, a better understanding of the underlying mechanism of the pathogenesis of NAFLD is helpful to choose an optimized treatment.
Collapse
Affiliation(s)
- Chunye Zhang
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA;
| | - Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65211, USA
- Correspondence:
| |
Collapse
|