1
|
Ren M, Xia Y, Pan H, Zhou X, Yu M, Ji F. Duodenal-jejunal bypass ameliorates MASLD in rats by regulating gut microbiota and bile acid metabolism through FXR pathways. Hepatol Commun 2025; 9:e0615. [PMID: 39813598 PMCID: PMC11737483 DOI: 10.1097/hc9.0000000000000615] [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: 07/09/2024] [Accepted: 10/16/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Although bariatric and metabolic surgical methods, including duodenal-jejunal bypass (DJB), were shown to improve metabolic dysfunction-associated steatotic liver disease (MASLD) in clinical trials and experimental rodent models, their underlying mechanisms remain unclear. The present study therefore evaluated the therapeutic effects and mechanisms of action of DJB in rats with MASLD. METHODS Rats with MASLD were randomly assigned to undergo DJB or sham surgery. Rats were orally administered a broad-spectrum antibiotic cocktail (Abx) or underwent fecal microbiota transplantation to assess the role of gut microbiota in DJB-induced improvement of MASLD. Gut microbiota were profiled by 16S rRNA gene sequencing and metagenomic sequencing, and bile acids (BAs) were analyzed by BA-targeted metabolomics. RESULTS DJB alleviated hepatic steatosis and insulin resistance in rats with diet-induced MASLD. Abx depletion of bacteria abrogated the ameliorating effects of DJB on MASLD. Fecal microbiota transplantation from rats that underwent DJB improved MASLD in high-fat diet-fed recipients by reshaping the gut microbiota, especially by significantly reducing the abundance of Clostridium. This, in turn, suppressed secondary BA biosynthesis and activated the hepatic BA receptor, farnesoid X receptor. Inhibition of farnesoid X receptor attenuated the ameliorative effects of post-DJB microbiota on MASLD. CONCLUSIONS DJB ameliorates MASLD by regulating gut microbiota and BA metabolism through hepatic farnesoid X receptor pathways.
Collapse
Affiliation(s)
- Mengting Ren
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Gastroenterology, Cancer Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yi Xia
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hanghai Pan
- Department of Gastroenterology, Cancer Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xinxin Zhou
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Mosang Yu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Feng Ji
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
Chen Y, Chen S, Xu C, Yu L, Chu S, Bao J, Wang J, Wang J. Identification of Diagnostic Biomarkers for Compensatory Liver Cirrhosis Based on Gut Microbiota and Urine Metabolomics Analyses. Mol Biotechnol 2024; 66:3164-3181. [PMID: 37875653 PMCID: PMC11549169 DOI: 10.1007/s12033-023-00922-9] [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: 07/06/2023] [Accepted: 09/26/2023] [Indexed: 10/26/2023]
Abstract
Liver cirrhosis is one of the most prevalent chronic liver disorders with high mortality. We aimed to explore changed gut microbiome and urine metabolome in compensatory liver cirrhosis (CLC) patients, thus providing novel diagnostic biomarkers for CLC. Forty fecal samples from healthy volunteers (control: 19) and CLC patients (patient: 21) were undertaken 16S rDNA sequencing. Chromatography-mass spectrometry was performed on 40 urine samples (20 controls and 20 patients). Microbiome and metabolome data were separately analyzed using corresponding bioinformatics approaches. The diagnostic model was constructed using the least absolute shrinkage and selection operator regression. The optimal diagnostic model was determined by five-fold cross-validation. Pearson correlation analysis was applied to clarify the relations among the diagnostic markers. 16S rDNA sequencing analyses showed changed overall alpha diversity and beta diversity in patient samples compared with those of controls. Similarly, we identified 841 changed metabolites. Pathway analysis revealed that the differential metabolites were mainly associated with pathways, such as tryptophan metabolism, purine metabolism, and steroid hormone biosynthesis. A 9-maker diagnostic model for CLC was determined, including 7 microorganisms and 2 metabolites. In this model, there were multiple correlations between microorganisms and metabolites. Subdoligranulum, Agathobacter, norank_f_Eubacterium_coprostanoligenes_group, Butyricicoccus, Lachnospiraceae_UCG_004, and L-2,3-Dihydrodipicolinate were elevated in CLC patients, whereas Blautia, Monoglobus, and 5-Acetamidovalerate were reduced. A novel diagnostic model for CLC was constructed and verified to be reliable, which provides new strategies for the diagnosis and treatment of CLC.
Collapse
Affiliation(s)
- Yingjun Chen
- Department of Infectious Diseases, Tiantai People's Hospital of Zhejiang Province, Taizhou, 317200, People's Republic of China
| | - Shaoxian Chen
- Department of Infectious Diseases, Tiantai People's Hospital of Zhejiang Province, Taizhou, 317200, People's Republic of China
| | - Chandi Xu
- Department of Infectious Diseases, Tiantai People's Hospital of Zhejiang Province, Taizhou, 317200, People's Republic of China
| | - Li Yu
- Department of Infectious Diseases, Tiantai People's Hospital of Zhejiang Province, Taizhou, 317200, People's Republic of China
| | - Shanshan Chu
- Department of Infectious Diseases, Tiantai People's Hospital of Zhejiang Province, Taizhou, 317200, People's Republic of China
| | - Jianzhi Bao
- Department of Infectious Diseases, Tiantai People's Hospital of Zhejiang Province, Taizhou, 317200, People's Republic of China
| | - Jinwei Wang
- Department of General Medicine, Tiantai People's Hospital of Zhejiang Province, Taizhou, 317200, People's Republic of China
| | - Junwei Wang
- Department of Infectious Diseases, Tiantai People's Hospital of Zhejiang Province, Taizhou, 317200, People's Republic of China.
| |
Collapse
|
3
|
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
|
4
|
Román-Sagüillo S, Quiñones Castro R, Juárez-Fernández M, Soluyanova P, Stephens C, Robles-Díaz M, Jorquera Plaza F, González-Gallego J, Martínez-Flórez S, García-Mediavilla MV, Nistal E, Jover R, Sánchez-Campos S. Idiosyncratic Drug-Induced Liver Injury and Amoxicillin-Clavulanate: Spotlight on Gut Microbiota, Fecal Metabolome and Bile Acid Profile in Patients. Int J Mol Sci 2024; 25:6863. [PMID: 38999973 PMCID: PMC11241776 DOI: 10.3390/ijms25136863] [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: 05/20/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Several hepatic disorders are influenced by gut microbiota, but its role in idiosyncratic drug-induced liver injury (iDILI), whose main causative agent is amoxicillin-clavulanate, remains unknown. This pioneering study aims to unravel particular patterns of gut microbiota composition and associated metabolites in iDILI and iDILI patients by amoxicillin-clavulanate (iDILI-AC). Thus, serum and fecal samples from 46 patients were divided into three study groups: healthy controls (n = 10), non-iDILI acute hepatitis (n = 12) and iDILI patients (n = 24). To evaluate the amoxicillin-clavulanate effect, iDILI patients were separated into two subgroups: iDILI non-caused by amoxicillin-clavulanate (iDILI-nonAC) (n = 18) and iDILI-AC patients (n = 6). Gut microbiota composition and fecal metabolome plus serum and fecal bile acid (BA) analyses were performed, along with correlation analyses. iDILI patients presented a particular microbiome profile associated with reduced fecal secondary BAs and fecal metabolites linked to lower inflammation, such as dodecanedioic acid and pyridoxamine. Moreover, certain taxa like Barnesiella, Clostridia UCG-014 and Eubacterium spp. correlated with significant metabolites and BAs. Additionally, comparisons between iDILI-nonAC and iDILI-AC groups unraveled unique features associated with iDILI when caused by amoxicillin-clavulanate. In conclusion, specific gut microbiota profiles in iDILI and iDILI-AC patients were associated with particular metabolic and BA status, which could affect disease onset and progression.
Collapse
Affiliation(s)
- Sara Román-Sagüillo
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain
| | - Raisa Quiñones Castro
- Servicio de Aparato Digestivo, Complejo Asistencial Universitario de León, 24008 León, Spain
| | - María Juárez-Fernández
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Polina Soluyanova
- Unidad Mixta de Investigación en Hepatología Experimental, IIS Hospital La Fe, 46026 Valencia, Spain
- Departamento de Bioquímica y Biología Molecular, Universidad de Valencia, 46010 Valencia, Spain
| | - Camilla Stephens
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unidad de Gestión Clínica de Aparato Digestivo y Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma BIONAND, Hospital Universitario Virgen de la Victoria, Facultad de Medicina, Universidad de Málaga, 29010 Málaga, Spain
| | - Mercedes Robles-Díaz
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unidad de Gestión Clínica de Aparato Digestivo y Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma BIONAND, Hospital Universitario Virgen de la Victoria, Facultad de Medicina, Universidad de Málaga, 29010 Málaga, Spain
| | - Francisco Jorquera Plaza
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain
- Servicio de Aparato Digestivo, Complejo Asistencial Universitario de León, 24008 León, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier González-Gallego
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Susana Martínez-Flórez
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain
| | - María Victoria García-Mediavilla
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Esther Nistal
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ramiro Jover
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unidad Mixta de Investigación en Hepatología Experimental, IIS Hospital La Fe, 46026 Valencia, Spain
- Departamento de Bioquímica y Biología Molecular, Universidad de Valencia, 46010 Valencia, Spain
| | - Sonia Sánchez-Campos
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
5
|
Sun L, Wen L, Li Q, Chen R, Wen S, Lai X, Lai Z, Cao J, Zhang Z, Hao M, Cao F, Sun S. Microbial Fermentation Enhances the Effect of Black Tea on Hyperlipidemia by Mediating Bile Acid Metabolism and Remodeling Intestinal Microbes. Nutrients 2024; 16:998. [PMID: 38613030 PMCID: PMC11013065 DOI: 10.3390/nu16070998] [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: 03/01/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Black tea (BT), the most consumed tea worldwide, can alleviate hyperlipidemia which is a serious threat to human health. However, the quality of summer BT is poor. It was improved by microbial fermentation in a previous study, but whether it affects hypolipidemic activity is unknown. Therefore, we compared the hypolipidemic activity of BT and microbially fermented black tea (EFT). The results demonstrated that BT inhibited weight gain and improved lipid and total bile acid (TBA) levels, and microbial fermentation reinforced this activity. Mechanistically, both BT and EFT mediate bile acid circulation to relieve hyperlipidemia. In addition, BT and EFT improve dyslipidemia by modifying the gut microbiota. Specifically, the increase in Lactobacillus johnsonii by BT, and the increase in Mucispirillum and Colidextribacter by EFT may also be potential causes for alleviation of hyperlipidemia. In summary, we demonstrated that microbial fermentation strengthened the hypolipidemic activity of BT and increased the added value of BT.
Collapse
Affiliation(s)
- Lingli Sun
- Tea Research Institute, Guangdong Provincial Key Laboratory of Tea Plant Resources Innovation and Utilization, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (L.S.); (Q.L.); (R.C.); (S.W.); (X.L.); (Z.L.); (J.C.); (Z.Z.); (M.H.)
| | - Lianghua Wen
- College of Horticulture, South China Agricultural University, Guangzhou 510000, China; (L.W.); (F.C.)
| | - Qiuhua Li
- Tea Research Institute, Guangdong Provincial Key Laboratory of Tea Plant Resources Innovation and Utilization, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (L.S.); (Q.L.); (R.C.); (S.W.); (X.L.); (Z.L.); (J.C.); (Z.Z.); (M.H.)
| | - Ruohong Chen
- Tea Research Institute, Guangdong Provincial Key Laboratory of Tea Plant Resources Innovation and Utilization, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (L.S.); (Q.L.); (R.C.); (S.W.); (X.L.); (Z.L.); (J.C.); (Z.Z.); (M.H.)
| | - Shuai Wen
- Tea Research Institute, Guangdong Provincial Key Laboratory of Tea Plant Resources Innovation and Utilization, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (L.S.); (Q.L.); (R.C.); (S.W.); (X.L.); (Z.L.); (J.C.); (Z.Z.); (M.H.)
| | - Xingfei Lai
- Tea Research Institute, Guangdong Provincial Key Laboratory of Tea Plant Resources Innovation and Utilization, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (L.S.); (Q.L.); (R.C.); (S.W.); (X.L.); (Z.L.); (J.C.); (Z.Z.); (M.H.)
| | - Zhaoxiang Lai
- Tea Research Institute, Guangdong Provincial Key Laboratory of Tea Plant Resources Innovation and Utilization, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (L.S.); (Q.L.); (R.C.); (S.W.); (X.L.); (Z.L.); (J.C.); (Z.Z.); (M.H.)
| | - Junxi Cao
- Tea Research Institute, Guangdong Provincial Key Laboratory of Tea Plant Resources Innovation and Utilization, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (L.S.); (Q.L.); (R.C.); (S.W.); (X.L.); (Z.L.); (J.C.); (Z.Z.); (M.H.)
| | - Zhenbiao Zhang
- Tea Research Institute, Guangdong Provincial Key Laboratory of Tea Plant Resources Innovation and Utilization, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (L.S.); (Q.L.); (R.C.); (S.W.); (X.L.); (Z.L.); (J.C.); (Z.Z.); (M.H.)
| | - Mengjiao Hao
- Tea Research Institute, Guangdong Provincial Key Laboratory of Tea Plant Resources Innovation and Utilization, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (L.S.); (Q.L.); (R.C.); (S.W.); (X.L.); (Z.L.); (J.C.); (Z.Z.); (M.H.)
| | - Fanrong Cao
- College of Horticulture, South China Agricultural University, Guangzhou 510000, China; (L.W.); (F.C.)
| | - Shili Sun
- Tea Research Institute, Guangdong Provincial Key Laboratory of Tea Plant Resources Innovation and Utilization, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (L.S.); (Q.L.); (R.C.); (S.W.); (X.L.); (Z.L.); (J.C.); (Z.Z.); (M.H.)
| |
Collapse
|
6
|
Amin U, Huang D, Dhir A, Shindler AE, Franks AE, Thomas CJ. Effects of gastric bypass bariatric surgery on gut microbiota in patients with morbid obesity. Gut Microbes 2024; 16:2427312. [PMID: 39551972 PMCID: PMC11581163 DOI: 10.1080/19490976.2024.2427312] [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: 07/09/2024] [Revised: 10/11/2024] [Accepted: 11/04/2024] [Indexed: 11/19/2024] Open
Abstract
The Western diet is associated with gastrointestinal dysbiosis, an active contributor to the pathophysiology of obesity and its comorbidities. Gastrointestinal dysbiosis is strongly linked to increased adiposity, low-grade inflammation, dyslipidaemia, and insulin resistance in individuals with morbid obesity. Bariatric bypass surgery remains the most effective treatment for achieving significant weight loss and alleviating obesity-related comorbidities. A growing body of evidence indicates that traditional Roux-en-Y Gastric Bypass (RYGB) improves the disrupted gut microbiota linked with obesity, potentially contributing to sustained weight loss and reduction of comorbidities. One Anastomosis Gastric Bypass (OAGB), a relatively new and technically simpler bariatric procedure, has shown both safety and efficacy in promoting weight loss and improving comorbidities. Few studies have investigated the impact of OAGB on gut microbiota. This review provides insights into the pathogenesis of obesity, current treatment strategies and our current understanding of the gut microbiota in health and disease, including modulating the gut microbiota as a promising and novel way to alleviate the burden of obesity and cardiometabolic conditions. By exploring the impact of gastric bypass surgery on gut microbiota-host interactions, we aim to shed light on this evolving field of research and uncover potential therapeutic targets for elevating outcomes in bariatric surgery.
Collapse
Affiliation(s)
- Urja Amin
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, Victoria, Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Bundoora, Victoria, Australia
| | - Dora Huang
- Department of Surgery, Austin Health, Heidelberg, Victoria, Australia
- Body Genesis Institute, Bundoora, Victoria, Australia
| | - Arun Dhir
- Department of Surgery, Austin Health, Heidelberg, Victoria, Australia
- Body Genesis Institute, Bundoora, Victoria, Australia
| | - Anya E Shindler
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, Victoria, Australia
| | - Ashley E Franks
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, Victoria, Australia
| | - Colleen J Thomas
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, Victoria, Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Bundoora, Victoria, Australia
- Pre-Clinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
7
|
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
|
8
|
Yin M, Wang Y, Han M, Liang R, Li S, Wang G, Gang X. Mechanisms of bariatric surgery for weight loss and diabetes remission. J Diabetes 2023; 15:736-752. [PMID: 37442561 PMCID: PMC10509523 DOI: 10.1111/1753-0407.13443] [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: 02/22/2023] [Revised: 06/12/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Obesity and type 2 diabetes(T2D) lead to defects in intestinal hormones secretion, abnormalities in the composition of bile acids (BAs), increased systemic and adipose tissue inflammation, defects of branched-chain amino acids (BCAAs) catabolism, and dysbiosis of gut microbiota. Bariatric surgery (BS) has been shown to be highly effective in the treatment of obesity and T2D, which allows us to view BS not simply as weight-loss surgery but as a means of alleviating obesity and its comorbidities, especially T2D. In recent years, accumulating studies have focused on the mechanisms of BS to find out which metabolic parameters are affected by BS through which pathways, such as which hormones and inflammatory processes are altered. The literatures are saturated with the role of intestinal hormones and the gut-brain axis formed by their interaction with neural networks in the remission of obesity and T2D following BS. In addition, BAs, gut microbiota and other factors are also involved in these benefits after BS. The interaction of these factors makes the mechanisms of metabolic improvement induced by BS more complicated. To date, we do not fully understand the exact mechanisms of the metabolic alterations induced by BS and its impact on the disease process of T2D itself. This review summarizes the changes of intestinal hormones, BAs, BCAAs, gut microbiota, signaling proteins, growth differentiation factor 15, exosomes, adipose tissue, brain function, and food preferences after BS, so as to fully understand the actual working mechanisms of BS and provide nonsurgical therapeutic strategies for obesity and T2D.
Collapse
Affiliation(s)
- Mengsha Yin
- Department of Endocrinology and MetabolismThe First Hospital of Jilin UniversityChangchunChina
| | - Yao Wang
- Department of OrthopedicsThe Second Hospital Jilin UniversityChangchunChina
| | - Mingyue Han
- Department of Endocrinology and MetabolismThe First Hospital of Jilin UniversityChangchunChina
| | - Ruishuang Liang
- Department of Endocrinology and MetabolismThe First Hospital of Jilin UniversityChangchunChina
| | - Shanshan Li
- Department of Endocrinology and MetabolismThe First Hospital of Jilin UniversityChangchunChina
| | - Guixia Wang
- Department of Endocrinology and MetabolismThe First Hospital of Jilin UniversityChangchunChina
| | - Xiaokun Gang
- Department of Endocrinology and MetabolismThe First Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
9
|
Hankir MK, Kovatcheva-Datchary P, Springer R, Hoffmann A, Vogel J, Seyfried F, Arora T. Gut Microbiota Contribution to Weight-Independent Glycemic Improvements after Gastric Bypass Surgery. Microbiol Spectr 2023; 11:e0510922. [PMID: 37022171 PMCID: PMC10269853 DOI: 10.1128/spectrum.05109-22] [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: 12/13/2022] [Accepted: 03/02/2023] [Indexed: 04/07/2023] Open
Abstract
Roux-en-Y gastric bypass surgery (RYGB) leads to improved glycemic control in individuals with severe obesity beyond the effects of weight loss alone. Here, We addressed the potential contribution of gut microbiota in mediating this favourable surgical outcome by using an established preclinical model of RYGB. 16S rRNA sequencing revealed that RYGB-treated Zucker fatty rats had altered fecal composition of various bacteria at the phylum and species levels, including lower fecal abundance of an unidentified Erysipelotrichaceae species, compared with both sham-operated (Sham) and body weight-matched to RYGB-treated (BWM) rats. Correlation analysis further revealed that fecal abundance of this unidentified Erysipelotrichaceae species linked with multiple indices of glycemic control uniquely in RYGB-treated rats. Sequence alignment of this Erysipelotrichaceae species identified Longibaculum muris to be the most closely related species, and its fecal abundance positively correlated with oral glucose intolerance in RYGB-treated rats. In fecal microbiota transplant experiments, the improved oral glucose tolerance of RYGB-treated compared with BWM rats could partially be transferred to recipient germfree mice, independently of body weight. Unexpectedly, providing L. muris as a supplement to RYGB recipient mice further improved oral glucose tolerance, while administering L. muris alone to chow-fed or Western style diet-challenged conventionally raised mice had minimal metabolic impact. Taken together, our findings provide evidence that the gut microbiota contributes to weight loss-independent improvements in glycemic control after RYGB and demonstrate how correlation of a specific gut microbiota species with a host metabolic trait does not imply causation. IMPORTANCE Metabolic surgery remains the most effective treatment modality for severe obesity and its comorbidities, including type 2 diabetes. Roux-en-Y gastric bypass (RYGB) is a commonly performed type of metabolic surgery that reconfigures gastrointestinal anatomy and profoundly remodels the gut microbiota. While it is clear that RYGB is superior to dieting when it comes to improving glycemic control, the extent to which the gut microbiota contributes to this effect remains untested. In the present study, we uniquely linked fecal Erysipelotrichaceae species, including Longibaculum muris, with indices of glycemic control after RYGB in genetically obese and glucose-intolerant rats. We further show that the weight loss-independent improvements in glycemic control in RYGB-treated rats can be transmitted via their gut microbiota to germfree mice. Our findings provide rare causal evidence that the gut microbiota contributes to the health benefits of metabolic surgery and have implications for the development of gut microbiota-based treatments for type 2 diabetes.
Collapse
Affiliation(s)
- Mohammed K. Hankir
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital Wurzburg, Wurzburg, Germany
| | - Petia Kovatcheva-Datchary
- Institute for Molecular Infection Biology, University of Wurzburg, Wurzburg, Germany
- Department of Pediatrics, University Hospital Wurzburg, Wurzburg, Germany
| | - Rebecca Springer
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital Wurzburg, Wurzburg, Germany
| | - Annett Hoffmann
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital Wurzburg, Wurzburg, Germany
| | - Jörg Vogel
- Institute for Molecular Infection Biology, University of Wurzburg, Wurzburg, Germany
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Wurzburg, Germany
| | - Florian Seyfried
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital Wurzburg, Wurzburg, Germany
| | - Tulika Arora
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
10
|
Yan Q, Hu W, Tian Y, Li X, Yu Y, Li X, Feng B. Probiotics intervention in preventing conversion of impaired glucose tolerance to diabetes: The PPDP follow-on study. Front Endocrinol (Lausanne) 2023; 14:1113611. [PMID: 36875472 PMCID: PMC9982119 DOI: 10.3389/fendo.2023.1113611] [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: 12/01/2022] [Accepted: 02/07/2023] [Indexed: 02/19/2023] Open
Abstract
OBJECTIVES The purpose of this study was to assess the incidence of type 2 diabetes mellitus (T2DM) after 6 years in patients with IGT who received early probiotic intervention in the Probiotics Prevention Diabetes Program (PPDP) trial. METHODS 77 patients with IGT in the PPDP trial were randomized to either probiotic or placebo. After the completion of the trial, 39 non-T2DM patients were invited to follow up glucose metabolism after the next 4 years. The incidence of T2DM in each group was assessed using Kaplan-Meier analysis. The 16S rDNA sequencing technology was used to analyze gut microbiota's structural composition and abundance changes between the groups. RESULTS The cumulative incidence of T2DM was 59.1% with probiotic treatment versus 54.5% with placebo within 6 years, there was no significant difference in the risk of developing T2DM between the two groups (P=0.674). CONCLUSIONS Supplemental probiotic therapy does not reduce the risk of IGT conversion to T2DM. CLINICAL TRIAL REGISTRATION https://www.chictr.org.cn/showproj.aspx?proj=5543, identifier ChiCTR-TRC-13004024.
Collapse
Affiliation(s)
- Qun Yan
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Weiting Hu
- The Second Clinical Medical College, Shanxi Medical University, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yan Tian
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xu Li
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuan Yu
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xing Li
- The Second Clinical Medical College, Shanxi Medical University, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Bo Feng
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Bo Feng,
| |
Collapse
|
11
|
Huang YL, Xiang Q, Zou JJ, Wu Y, Yu R. Zuogui Jiangtang Shuxin formula Ameliorates diabetic cardiomyopathy mice via modulating gut-heart axis. Front Endocrinol (Lausanne) 2023; 14:1106812. [PMID: 36843604 PMCID: PMC9948445 DOI: 10.3389/fendo.2023.1106812] [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: 11/24/2022] [Accepted: 01/16/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND There is growing evidence demonstrating that the gut microbiota plays a crucial role in multiple endocrine disorders, including diabetic cardiomyopathy (DCM). Research shows that the Chinese herb reduces disease occurrence by regulating gut microbiota. Zuogui Jiangtang Shuxin formula (ZGJTSXF), a Chinese medicinal formula, has been clinically used for treatment of DCM for many years. However, there is still no clear understanding of how ZGJTSXF treatment contributes to the prevention and treatment of DCM through its interaction with gut microbiota and metabolism. METHODS In this study, mice models of DCM were established, and ZGJTSXF's therapeutic effects were assessed. Specifically, serum glycolipid, echocardiography, histological staining, myocardial apoptosis rate were assessed. Using 16s rRNA sequencing and high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS), we determined the impact of ZGJTSXF on the structure of gut microbiota and content of its metabolite TMAO. The mechanism of ZGJTSXF action on DCM was analyzed using quantitative real-time PCR and western blots. RESULTS We found that ZGJTSXF significantly ameliorated DCM mice by modulating gut-heart axis: ZGJTSXF administration improved glycolipid levels, heart function, cardiac morphological changes, inhibited cardiomyocytes apoptosis, and regulate the gut microbiota in DCM mice. Specifically, ZGJTSXF treatment reverse the significant changes in the abundance of certain genera closely related to DCM phenotype, including Lactobacillus, Alloprevotella and Alistipes. Furthermore, ZGJTSXF alleviated DCM in mice by blunting TMAO/PERK/FoxO1 signaling pathway genes and proteins. CONCLUSION ZGJTSXF administration could ameliorate DCM mice by remodeling gut microbiota structure, reducing serum TMAO generation and suppressing TMAO/PERK/FoxO1 signaling pathway.
Collapse
Affiliation(s)
- Ya-lan Huang
- The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Qin Xiang
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Jun-ju Zou
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yongjun Wu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
- *Correspondence: Rong Yu, ; Yongjun Wu,
| | - Rong Yu
- The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- *Correspondence: Rong Yu, ; Yongjun Wu,
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Yang M, Liu S, Zhang C. The Related Metabolic Diseases and Treatments of Obesity. Healthcare (Basel) 2022; 10:1616. [PMID: 36141228 PMCID: PMC9498506 DOI: 10.3390/healthcare10091616] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Obesity is a chronic disease characterized by the abnormal or excessive accumulation of body fat, affecting more than 1 billion people worldwide. Obesity is commonly associated with other metabolic disorders, such as type 2 diabetes, non-alcoholic fatty liver disease, cardiovascular diseases, chronic kidney disease, and cancers. Factors such as a sedentary lifestyle, overnutrition, socioeconomic status, and other environmental and genetic conditions can cause obesity. Many molecules and signaling pathways are involved in the pathogenesis of obesity, such as nuclear factor (NF)-κB, Toll-like receptors (TLRs), adhesion molecules, G protein-coupled receptors (GPCRs), programmed cell death 1 (PD-1)/programmed death-ligand 1 (PD-L1), and sirtuin 1 (SIRT1). Commonly used strategies of obesity management and treatment include exercise and dietary change or restriction for the early stage of obesity, bariatric surgery for server obesity, and Food and Drug Administration (FDA)-approved medicines such as semaglutide and liraglutide that can be used as monotherapy or as a synergistic treatment. In addition, psychological management, especially for patients with obesity and distress, is a good option. Gut microbiota plays an important role in obesity and its comorbidities, and gut microbial reprogramming by fecal microbiota transplantation (FMT), probiotics, prebiotics, or synbiotics shows promising potential in obesity and metabolic syndrome. Many clinical trials are ongoing to evaluate the therapeutic effects of different treatments. Currently, prevention and early treatment of obesity are the best options to prevent its progression to many comorbidities.
Collapse
Affiliation(s)
- Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
| | - Shuai Liu
- The First Affiliated Hospital, Zhejiang University, Hangzhou 310006, China
| | - Chunye Zhang
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
15
|
The Effects of Time-Restricted Eating on Metabolism and Gut Microbiota: A Real-Life Study. Nutrients 2022; 14:nu14132569. [PMID: 35807750 PMCID: PMC9267969 DOI: 10.3390/nu14132569] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/13/2022] [Accepted: 06/17/2022] [Indexed: 12/26/2022] Open
Abstract
The metabolic benefits of time-restricted eating (TRE) in humans are statistically significant but not clinically relevant. Few data are available about the effects of TRE on the gut microbiota. We compared the effects of a TRE regimen (<12 h feeding; n = 25) with a time-unrestricted (TUE) regimen (>12 h feeding; n = 24), on the clinical and dietary variables and gut-microbiota composition in patients with obesity, who were subjected for 12 weeks to the same caloric restriction. Median weight loss was 4.0 kg and 2.2 kg in the TRE and TUE groups, respectively, with a between-group borderline difference (p = 0.049). No significant between-group difference was found in other dietary, anthropometric, or laboratory variables. There were no substantial between-group differences in alpha and beta diversity or gut-microbiota composition. The TRE group showed a significant increase in the frequency of Lachnospiraceae, Parasutterella, and Romboutsia at the study’s end. A TRE regimen induced small changes both in metabolic/dietary variables and in the gut-microbiota composition, with respect to the TUE. The microbial changes we have found were of uncertain clinical significance.
Collapse
|
16
|
Li R, Yi X, Yang J, Zhu Z, Wang Y, Liu X, Huang X, Wan Y, Fu X, Shu W, Zhang W, Wang Z. Gut Microbiome Signatures in the Progression of Hepatitis B Virus-Induced Liver Disease. Front Microbiol 2022; 13:916061. [PMID: 35733959 PMCID: PMC9208012 DOI: 10.3389/fmicb.2022.916061] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 04/27/2022] [Indexed: 12/12/2022] Open
Abstract
The gut microbiome is associated with hepatitis B virus (HBV)-induced liver disease, which progresses from chronic hepatitis B, to liver cirrhosis, and eventually to hepatocellular carcinoma. Studies have analyzed the gut microbiome at each stage of HBV-induced liver diseases, but a consensus has not been reached on the microbial signatures across these stages. Here, we conducted by a systematic meta-analysis of 486 fecal samples from publicly available 16S rRNA gene datasets across all disease stages, and validated the results by a gut microbiome characterization on an independent cohort of 15 controls, 23 chronic hepatitis B, 20 liver cirrhosis, and 22 hepatocellular carcinoma patients. The integrative analyses revealed 13 genera consistently altered at each of the disease stages both in public and validation datasets, suggesting highly robust microbiome signatures. Specifically, Colidextribacter and Monoglobus were enriched in healthy controls. An unclassified Lachnospiraceae genus was specifically elevated in chronic hepatitis B, whereas Bilophia was depleted. Prevotella and Oscillibacter were depleted in liver cirrhosis. And Coprococcus and Faecalibacterium were depleted in hepatocellular carcinoma. Classifiers established using these 13 genera showed diagnostic power across all disease stages in a cross-validation between public and validation datasets (AUC = 0.65–0.832). The identified microbial taxonomy serves as non-invasive biomarkers for monitoring the progression of HBV-induced liver disease, and may contribute to microbiome-based therapies.
Collapse
Affiliation(s)
- Ranxi Li
- South China Normal University-Panyu Central Hospital Joint Laboratory of Basic and Translational Medical Research, Guangzhou Panyu Central Hospital, Guangzhou, China
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xinzhu Yi
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Junhao Yang
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Zhou Zhu
- South China Normal University-Panyu Central Hospital Joint Laboratory of Basic and Translational Medical Research, Guangzhou Panyu Central Hospital, Guangzhou, China
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Yifei Wang
- South China Normal University-Panyu Central Hospital Joint Laboratory of Basic and Translational Medical Research, Guangzhou Panyu Central Hospital, Guangzhou, China
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xiaomin Liu
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xili Huang
- South China Normal University-Panyu Central Hospital Joint Laboratory of Basic and Translational Medical Research, Guangzhou Panyu Central Hospital, Guangzhou, China
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Yu Wan
- Department of Gastroenterology, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Xihua Fu
- Department of Infectious Diseases, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Wensheng Shu
- School of Life Sciences, South China Normal University, Guangzhou, China
- *Correspondence: Wensheng Shu
| | - Wenjie Zhang
- Department of Science and Education, Guangzhou Panyu Central Hospital, Guangzhou, China
- Wenjie Zhang
| | - Zhang Wang
- South China Normal University-Panyu Central Hospital Joint Laboratory of Basic and Translational Medical Research, Guangzhou Panyu Central Hospital, Guangzhou, China
- School of Life Sciences, South China Normal University, Guangzhou, China
- Zhang Wang
| |
Collapse
|
17
|
Chen JH, Yu ZH, Liu QLF, Meng QG, Chen X. Research Progress of Duodenal-Jejunal Bypass Liner in the Treatment of Obesity and Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2022; 15:3319-3327. [PMID: 36329806 PMCID: PMC9624146 DOI: 10.2147/dmso.s382324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
With the development of economy and improvement of people's living standards, the incidence of obesity and type 2 diabetes mellitus (T2DM) has increased significantly and obesity has also become one of the most important risk factors of T2DM. In light of these trends, there have been many ways to take effect in losing weight. However, they also have corresponding deficiencies including inapparent curative effect, complex and incomplete reversible procedures and severe complications. Duodenal-Jejunal Bypass Liner (DJBL), which mimics Roux-en-Y gastric bypass (RYGB), is proved to play a key role in weight loss and control of T2DM. DJBL is reversible, less invasive and is more suitable for the treatment of obesity and T2DM, which is associated with multiple mechanisms, including incretin effect, gastric emptying mechanism, bile acid regulation, intestinal microbiota, inflammatory reaction mechanism and neural mechanism. In our review, we aimed to elaborate DJBL's clinical efficacy, safety and mechanisms in detail.
Collapse
Affiliation(s)
- Ji-Hua Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Zi-Han Yu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Qin-Ling Fei Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Qing-Guo Meng
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Correspondence: Xin Chen, Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China, Email
| |
Collapse
|