1
|
Yao J, Ning B, Ding J. The gut microbiota: an emerging modulator of drug resistance in hepatocellular carcinoma. Gut Microbes 2025; 17:2473504. [PMID: 40042184 PMCID: PMC11901387 DOI: 10.1080/19490976.2025.2473504] [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: 06/12/2024] [Revised: 11/08/2024] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Liver cancer is usually diagnosed at an advanced stage and is the third most common cause of cancer-related death worldwide. In addition to the lack of effective treatment options, resistance to therapeutic drugs is a major clinical challenge. The gut microbiota has recently been recognized as one of the key factors regulating host health. The microbiota and its metabolites can directly or indirectly regulate gene expression in the liver, leading to gut-liver axis dysregulation, which is closely related to liver cancer occurrence and the treatment response. Gut microbiota disturbance may participate in tumor progression and drug resistance through metabolite production, gene transfer, immune regulation, and other mechanisms. However, systematic reviews on the role of the gut microbiota in drug resistance in liver cancer are lacking. Herein, we review the relationships between the gut microbiota and the occurrence and drug resistance of hepatocellular carcinoma, summarize the emerging mechanisms underlying gut microbiota-mediated drug resistance, and propose new personalized treatment options to overcome this resistance.
Collapse
Affiliation(s)
- Jiali Yao
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, China
| | - Beifang Ning
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jin Ding
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, China
| |
Collapse
|
2
|
Du W, Zou ZP, Ye BC, Zhou Y. Gut microbiota and associated metabolites: key players in high-fat diet-induced chronic diseases. Gut Microbes 2025; 17:2494703. [PMID: 40260760 DOI: 10.1080/19490976.2025.2494703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/26/2025] [Accepted: 04/11/2025] [Indexed: 04/24/2025] Open
Abstract
Excessive intake of dietary fats is strongly associated with an increased risk of various chronic diseases, such as obesity, diabetes, hepatic metabolic disorders, cardiovascular disease, chronic intestinal inflammation, and certain cancers. A significant portion of the adverse effects of high-fat diet on disease risk is mediated through modifications in the gut microbiota. Specifically, high-fat diets are linked to reduced microbial diversity, an overgrowth of gram-negative bacteria, an elevated Firmicutes-to-Bacteroidetes ratio, and alterations at various taxonomic levels. These microbial alterations influence the intestinal metabolism of small molecules, which subsequently increases intestinal permeability, exacerbates inflammatory responses, disrupts metabolic functions, and impairs signal transduction pathways in the host. Consequently, diet-induced changes in the gut microbiota play a crucial role in the initiation and progression of chronic diseases. This review explores the relationship between high-fat diets and gut microbiota, highlighting their roles and underlying mechanisms in the development of chronic metabolic diseases. Additionally, we propose probiotic interventions may serve as a promising adjunctive therapy to counteract the negative effects of high-fat diet-induced alterations in gut microbiota composition.
Collapse
Affiliation(s)
- Wei Du
- Laboratory of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Zhen-Ping Zou
- Laboratory of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Bang-Ce Ye
- Laboratory of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Ying Zhou
- Laboratory of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
3
|
Qin LN, Yu YF, Ma L, Yu R. Intestinal bacteria-derived extracellular vesicles in metabolic dysfunction-associated steatotic liver disease: from mechanisms to therapeutics. Mol Cells 2025:100216. [PMID: 40239896 DOI: 10.1016/j.mocell.2025.100216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/06/2025] [Accepted: 04/06/2025] [Indexed: 04/18/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a progressive disease that affects the health of approximately one-third of the world's population. It is the primary cause of end-stage liver disease, liver malignancy, and liver transplantation, resulting in a great medical burden. No medications have yet been approved by the US Food and Drug Administration (FDA) for treating MASLD without liver inflammation or scarring. Therefore, the development of specific drugs to treat MASLD remains a key task in the ongoing research objective. Extracellular vesicles (EVs) play an important role in the communication between organs, tissues, and cells. Recent studies have found that intestinal microbiota are closely related to the pathogenesis and progression of MASLD. EVs produced by bacteria (BEVs) play an indispensable role in this process. Thus, this study provides a new direction for MASLD treatment. However, the mechanism by which BEVs affect MASLD remains unclear. Therefore, this study investigated the influence and function of intestinal microbiota in MASLD. Additionally, we focus on the research progress of BEVs in recent years, and explain the relationship between BEVs and MASLD from the perspectives of glucose and lipid metabolism, immune responses, and intestinal homeostasis. Finally, we summarized the potential therapeutic value of BEVs and EVs from other sources, such as adipocytes, immunocytes, stem cells, and plants.
Collapse
Affiliation(s)
- Li-Na Qin
- Department of endocrinology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yun-Feng Yu
- Department of endocrinology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lie Ma
- The Third Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Rong Yu
- Department of endocrinology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China; Hunan University of Chinese Medicine, Changsha, Hunan, China.
| |
Collapse
|
4
|
Li C, Cai C, Wang C, Chen X, Zhang B, Huang Z. Gut microbiota-mediated gut-liver axis: a breakthrough point for understanding and treating liver cancer. Clin Mol Hepatol 2025; 31:350-381. [PMID: 39659059 PMCID: PMC12016628 DOI: 10.3350/cmh.2024.0857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/22/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024] Open
Abstract
The trillions of commensal microorganisms living in the gut lumen profoundly influence the physiology and pathophysiology of the liver through a unique gut-liver axis. Disruptions in the gut microbial communities, arising from environmental and genetic factors, can lead to altered microbial metabolism, impaired intestinal barrier and translocation of microbial components to the liver. These alterations collaboratively contribute to the pathogenesis of liver disease, and their continuous impact throughout the disease course plays a critical role in hepatocarcinogenesis. Persistent inflammatory responses, metabolic rearrangements and suppressed immunosurveillance induced by microbial products underlie the pro-carcinogenic mechanisms of gut microbiota. Meanwhile, intrahepatic microbiota derived from the gut also emerges as a novel player in the development and progression of liver cancer. In this review, we first discuss the causes of gut dysbiosis in liver disease, and then specify the pivotal role of gut microbiota in the malignant progression from chronic liver diseases to hepatobiliary cancers. We also delve into the cellular and molecular interactions between microbes and liver cancer microenvironment, aiming to decipher the underlying mechanism for the malignant transition processes. At last, we summarize the current progress in the clinical implications of gut microbiota for liver cancer, shedding light on microbiota-based strategies for liver cancer prevention, diagnosis and therapy.
Collapse
Affiliation(s)
- Chenyang Li
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chujun Cai
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chendong Wang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences; NHC Key Laboratory of Organ Transplantation, Wuhan, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences; NHC Key Laboratory of Organ Transplantation, Wuhan, China
| | - Zhao Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Zhang Q, Du M, Wei S, Zhu L, Yan R, Jin M, Wang Y. Variation of Meat Quality and Relationship to Gut Microbiota Among Different Pig Breeds. Microb Biotechnol 2025; 18:e70139. [PMID: 40181253 PMCID: PMC11968327 DOI: 10.1111/1751-7915.70139] [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: 12/30/2024] [Revised: 02/23/2025] [Accepted: 03/18/2025] [Indexed: 04/05/2025] Open
Abstract
Meat production is of great importance to the world's food supply and economic development, and meat quality determines the purchasing desire of consumers. Recent studies show intestinal microorganisms are involved in several physiological functions of the host and therefore are likely to regulate meat quality. This study aimed to compare the carcass performance, meat quality traits and serum parameters of three different pig breeds, Jinhua (JH) pigs (n = 8), Duroc × Berkshire × Jiaxinghei (DBJ) pigs (n = 8), Duroc × Landrace × Yorkshire (DLY) pigs (n = 8) and to investigate a possible relationship between gut microbiota composition and these traits. Meat quality results showed that compared with DLY pigs, JH pigs had lower water loss and shear force in the longissimus dorsi muscle, and higher intramuscular fat content and inosine monophosphate content were observed in JH pigs. Serum biochemical indicators showed the content of nonesterified fatty acid in JH pigs was lowest. Furthermore, the gut microbiota analysis indicated that JH pigs harboured more abundant Lachnospiraceae, Prevotellaceae NK3B31 and Marvinbryantia. Spearman correlation analysis showed that unidentified genus of family Lachnospiraceae, genus Prevotella and genus Alloprevotella were positively correlated with IMF content and marbling score in the longissimus dorsi muscle of pigs. In conclusion, our results indicated the quality of JH pork was superior to DBJ and DLY pigs, and the difference in meat qualities was related to the abundance of fibre-degrading bacteria. Our study provides insight into further understanding of the relationship between microbiota and meat quality, nutrient metabolism and fat deposition, which is critical to the pork industry and swine intestinal health.
Collapse
Affiliation(s)
- Qifan Zhang
- Key Laboratory of Molecular Animal Nutrition, Ministry of EducationZhejiang UniversityZhejiangHangzhouChina
- National Engineering Research Center of Green Feeds and Healthy Livestock IndustryZhejiang UniversityZhejiangHangzhouChina
- Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural AffairsZhejiang UniversityZhejiangHangzhouChina
- Zhejiang Key Laboratory of Nutrition and Breeding for High‐Quality Animal ProductsZhejiang UniversityZhejiangHangzhouChina
- College of Animal Sciences, Institute of Feed ScienceZhejiang UniversityZhejiangHangzhouChina
| | - Man Du
- Key Laboratory of Molecular Animal Nutrition, Ministry of EducationZhejiang UniversityZhejiangHangzhouChina
- National Engineering Research Center of Green Feeds and Healthy Livestock IndustryZhejiang UniversityZhejiangHangzhouChina
- Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural AffairsZhejiang UniversityZhejiangHangzhouChina
- Zhejiang Key Laboratory of Nutrition and Breeding for High‐Quality Animal ProductsZhejiang UniversityZhejiangHangzhouChina
- College of Animal Sciences, Institute of Feed ScienceZhejiang UniversityZhejiangHangzhouChina
| | - Siyu Wei
- Key Laboratory of Molecular Animal Nutrition, Ministry of EducationZhejiang UniversityZhejiangHangzhouChina
- National Engineering Research Center of Green Feeds and Healthy Livestock IndustryZhejiang UniversityZhejiangHangzhouChina
- Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural AffairsZhejiang UniversityZhejiangHangzhouChina
- Zhejiang Key Laboratory of Nutrition and Breeding for High‐Quality Animal ProductsZhejiang UniversityZhejiangHangzhouChina
- College of Animal Sciences, Institute of Feed ScienceZhejiang UniversityZhejiangHangzhouChina
| | - Luoyi Zhu
- Key Laboratory of Molecular Animal Nutrition, Ministry of EducationZhejiang UniversityZhejiangHangzhouChina
- National Engineering Research Center of Green Feeds and Healthy Livestock IndustryZhejiang UniversityZhejiangHangzhouChina
- Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural AffairsZhejiang UniversityZhejiangHangzhouChina
- Zhejiang Key Laboratory of Nutrition and Breeding for High‐Quality Animal ProductsZhejiang UniversityZhejiangHangzhouChina
- College of Animal Sciences, Institute of Feed ScienceZhejiang UniversityZhejiangHangzhouChina
| | - Rong Yan
- Key Laboratory of Molecular Animal Nutrition, Ministry of EducationZhejiang UniversityZhejiangHangzhouChina
- National Engineering Research Center of Green Feeds and Healthy Livestock IndustryZhejiang UniversityZhejiangHangzhouChina
- Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural AffairsZhejiang UniversityZhejiangHangzhouChina
- Zhejiang Key Laboratory of Nutrition and Breeding for High‐Quality Animal ProductsZhejiang UniversityZhejiangHangzhouChina
- College of Animal Sciences, Institute of Feed ScienceZhejiang UniversityZhejiangHangzhouChina
| | - Mingliang Jin
- Key Laboratory of Molecular Animal Nutrition, Ministry of EducationZhejiang UniversityZhejiangHangzhouChina
- National Engineering Research Center of Green Feeds and Healthy Livestock IndustryZhejiang UniversityZhejiangHangzhouChina
- Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural AffairsZhejiang UniversityZhejiangHangzhouChina
- Zhejiang Key Laboratory of Nutrition and Breeding for High‐Quality Animal ProductsZhejiang UniversityZhejiangHangzhouChina
- College of Animal Sciences, Institute of Feed ScienceZhejiang UniversityZhejiangHangzhouChina
| | - Yizhen Wang
- Key Laboratory of Molecular Animal Nutrition, Ministry of EducationZhejiang UniversityZhejiangHangzhouChina
- National Engineering Research Center of Green Feeds and Healthy Livestock IndustryZhejiang UniversityZhejiangHangzhouChina
- Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural AffairsZhejiang UniversityZhejiangHangzhouChina
- Zhejiang Key Laboratory of Nutrition and Breeding for High‐Quality Animal ProductsZhejiang UniversityZhejiangHangzhouChina
- College of Animal Sciences, Institute of Feed ScienceZhejiang UniversityZhejiangHangzhouChina
| |
Collapse
|
6
|
Zhang QR, Dong Y, Fan JG. Early-life exposure to gestational diabetes mellitus predisposes offspring to pediatric nonalcoholic fatty liver disease. Hepatobiliary Pancreat Dis Int 2025; 24:128-137. [PMID: 38195352 DOI: 10.1016/j.hbpd.2023.12.007] [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: 08/05/2023] [Accepted: 12/28/2023] [Indexed: 01/11/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as the prevailing chronic liver disease in the pediatric population due to the global obesity pandemic. Evidence shows that prenatal and postnatal exposure to maternal abnormalities leads to a higher risk of pediatric NAFLD through persistent alterations in developmental programming. Gestational diabetes mellitus (GDM) is a hyperglycemic syndrome which has become the most prevalent complication in pregnant women. An increasing number of both epidemiologic investigations and animal model studies have validated adverse and long-term outcomes in offspring following GDM exposure in utero. Similarly, GDM is considered a crucial risk factor for pediatric NAFLD. This review aimed to summarize currently published studies concerning the inductive roles of GDM in offspring NAFLD development during childhood and adolescence. Dysregulations in hepatic lipid metabolism and gut microbiota in offspring, as well as dysfunctions in the placenta are potential factors in the pathogenesis of GDM-associated pediatric NAFLD. In addition, potentially effective interventions for GDM-associated offspring NAFLD are also discussed in this review. However, most of these therapeutic approaches still require further clinical research for validation.
Collapse
Affiliation(s)
- Qian-Ren Zhang
- Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yan Dong
- Department of Endocrinology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jian-Gao Fan
- Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China.
| |
Collapse
|
7
|
Chen H, Cao T, Lin C, Jiao S, He Y, Zhu Z, Guo Q, Wu R, Cai H, Zhang B. Akkermansia muciniphila ameliorates olanzapine-induced metabolic dysfunction-associated steatotic liver disease via PGRMC1/SIRT1/FOXO1 signaling pathway. Front Pharmacol 2025; 16:1550015. [PMID: 40176900 PMCID: PMC11961884 DOI: 10.3389/fphar.2025.1550015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Akkermansia muciniphila (AKK), classified as "lean bacteria," has emerged as a promising candidate for ameliorating metabolic disorders, including obesity, diabetes, and liver disease. In this study, we investigated the therapeutic potential of AKK to counteract metabolic dysfunctions induced by Olanzapine (OLZ), a first-class antipsychotic known for its high therapeutic efficacy but also its association with metabolic disturbances, particularly Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Previous studies have implicated progesterone receptor membrane component 1 (PGRMC1) as a key player in antipsychotic-induced metabolic side effects. Using male C57BL/6J mice fed a high-fat diet, we assessed the effects of AKK supplementation on OLZ-induced metabolic disturbances. Key parameters such as body weight, hepatic injury markers, glucose tolerance, insulin resistance, and lipid metabolism were analyzed. The study revealed that AKK supplementation reduced hepatic lipid accumulation, oxidative stress, and insulin resistance, while normalizing lipid and glucose metabolism. These effects are likely mediated through the restoration of PGRMC1/SIRT1/FOXO1 signaling pathway by AKK. Additionally, changes in gut microbiota composition, including a reduction in pathogenic bacteria such as Lactococcus and enrichment of beneficial bacteria, were observed. Overall, the study suggests that AKK has therapeutic potential to counteract OLZ-induced MASLD by modulating gut microbiota and key metabolic pathways, making it a promising strategy for managing metabolic side effects in patients receiving antipsychotic treatment.
Collapse
Affiliation(s)
- Hui Chen
- Department of Pharmacy, Changsha Stomatological Hospital, Changsha, Hunan, China
- Department of pharmacy, Institute of Clinical Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, Hunan, China
| | - Ting Cao
- Department of pharmacy, Institute of Clinical Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, Hunan, China
| | - ChenQuan Lin
- Department of pharmacy, Institute of Clinical Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, Hunan, China
| | - ShiMeng Jiao
- Department of pharmacy, Institute of Clinical Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, Hunan, China
| | - YiFang He
- Department of pharmacy, Institute of Clinical Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, Hunan, China
| | - ZhenYu Zhu
- Department of pharmacy, Institute of Clinical Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, Hunan, China
| | - QiuJin Guo
- Department of pharmacy, Institute of Clinical Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, Hunan, China
| | - RenRong Wu
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - HuaLin Cai
- Department of pharmacy, Institute of Clinical Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, Hunan, China
| | - BiKui Zhang
- Department of pharmacy, Institute of Clinical Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, Hunan, China
| |
Collapse
|
8
|
Ye L, Yao Z, Xuan Q, Liu Q, Bo T. The impact of sleeve gastrectomy on MASH development by regulating the composition of gut microbiota and metabolic homeostasis. Biochem Biophys Res Commun 2025; 752:151466. [PMID: 39938449 DOI: 10.1016/j.bbrc.2025.151466] [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: 11/22/2024] [Revised: 01/21/2025] [Accepted: 02/07/2025] [Indexed: 02/14/2025]
Abstract
The prevalence of metabolic dysfunction-associated steatohepatitis (MASH) is increasing annually, which is a global public health issue. Although clinical trials are lacking, observational studies indicate that bariatric surgery can alleviate the progression of MASH. Here, we performed sleeve gastrectomy (SG) and Sham surgery on 8-week-old mice, and then fed a AMLN diet for 24 weeks to construct a diet-inducted MASH mice model after 4-week post-surgery recovery. Applying a multi-omics approach combining metagenomics, metabolomics, and transcriptomics, we found that SG prevents the development of hepatic steatosis, inflammation, and fibrosis in MASH mice not only by significantly altering the structure of gut microbiota including s_Akkermansia muciniphila, s_Alistiples dispar, g_Helicobacter and s_uc_Oscillospiraceae, but also by modulating the levels of serum metabolites including l-arginine and taurocholic acid (TCA). These results suggest that SG and the alteration of gut microbiota and its related serum metabolites can be served as the effective therapeutic strategies for MASH.
Collapse
Affiliation(s)
- Lingxi Ye
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Zhenyu Yao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Qiuhui Xuan
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Qiaoran Liu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250021, China; Key Laboratory of Metabolism and Gastrointestinal Tumor, the First Affiliated Hospital of Shandong First Medical University, China; Key Laboratory of Laparoscopic Technology, the First Affiliated Hospital of Shandong First Medical University, China.
| | - Tao Bo
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
| |
Collapse
|
9
|
Xu J, Chen N, Li Z, Liu Y. Gut microbiome and liver diseases. FUNDAMENTAL RESEARCH 2025; 5:890-901. [PMID: 40242515 PMCID: PMC11997574 DOI: 10.1016/j.fmre.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 04/18/2025] Open
Abstract
Symbiotic microbiota plays a crucial role in the education, development, and maintenance of the host immune system, significantly contributing to overall health. Through the gut-liver axis, the gut microbiota and liver have a bidirectional relationship that is becoming increasingly evident as more research highlights the translocation of the gut microbiota and its metabolites. The focus of this narrative review is to examine and discuss the importance of the gut-liver axis and the enterohepatic barrier in maintaining overall health. Additionally, we emphasize the crucial role of the gut microbiome in liver diseases and explore potential therapeutic strategies for liver diseases by manipulating the microbiota.
Collapse
Affiliation(s)
- Jun Xu
- Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing 100044, China
| | - Ning Chen
- Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing 100044, China
| | - Zhou Li
- Beijing Key Laboratory of Micro-nano Energy and Sensor, Beijing Institute of Nanoenergy and Nanosystems Chinese Academy of Sciences, Beijing 101400, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yulan Liu
- Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
10
|
Caesar R. The impact of novel probiotics isolated from the human gut on the gut microbiota and health. Diabetes Obes Metab 2025; 27 Suppl 1:3-14. [PMID: 39726216 PMCID: PMC11894790 DOI: 10.1111/dom.16129] [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: 10/25/2024] [Revised: 12/02/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024]
Abstract
The gut microbiota plays a pivotal role in influencing the metabolism and immune responses of the body. A balanced microbial composition promotes metabolic health through various mechanisms, including the production of beneficial metabolites, which help regulate inflammation and support immune functions. In contrast, imbalance in the gut microbiota, known as dysbiosis, can disrupt metabolic processes and increase the risk of developing diseases, such as obesity, type 2 diabetes, and inflammatory disorders. The composition of the gut microbiota is dynamic and can be influenced by environmental factors such as diet, medication, and the consumption of live bacteria. Since the early 1900s, bacteria isolated from food and have been used as probiotics. However, the human gut also offers an enormous reservoir of bacterial strains, and recent advances in microbiota research have led to the discovery of strains with probiotic potentials. These strains, derived from a broad spectrum of microbial taxa, differ in their ecological properties and how they interact with their hosts. For most probiotics bacterial structural components and metabolites, such as short-chain fatty acids, contribute to the maintenance of metabolic and immunological homeostasis by regulating inflammation and reinforcing gut barrier integrity. Metabolites produced by probiotic strains can also be used for bacterial cross-feeding to promote a balanced microbiota. Despite the challenges related to safety, stability, and strain-specific properties, several newly identified strains offer great potential for personalized probiotic interventions, allowing for targeted health strategies.
Collapse
Affiliation(s)
- Robert Caesar
- The Wallenberg Laboratory, Department of Molecular and Clinical MedicineUniversity of GothenburgGothenburgSweden
| |
Collapse
|
11
|
Mahmoud M, Nwankwo E, Zhang Z, Matiwala N, Tripathi R, Mohamed I, Barrios C, Syn WK, Hachem C. Low prevalence of peptic ulcer disease in hospitalized patients with cystic fibrosis: A national database study. Am J Med Sci 2025; 369:321-325. [PMID: 39245183 DOI: 10.1016/j.amjms.2024.09.001] [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: 04/09/2024] [Revised: 08/31/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Although cystic fibrosis (CF) is widely considered a lung disease, the prevalence of CF-specific gastrointestinal symptoms and diseases has continued to rise. Peptic ulcer disease (PUD) has not been well-studied among people with CF (PwCF) and may be a common cause of abdominal symptoms. In PwCF, impaired bicarbonate secretion and unbuffered gastric acid production have been attributed to the development of ulcers, although ulcers remain uncommon. The objective of this study was to evaluate the prevalence of PUD in PwCF and assess for possible contributing factors. METHODS This study utilized the National Inpatient Sample (NIS) database. All patients 18 years or older with CF were identified from 2014 to 2019. Relevant patient characteristics and procedures were identified using ICD-9 and ICD-10 codes. Linear trend, bivariate analyses, and multiple regression analysis were performed. The outcomes of interest were peptic ulcer disease, pancreatic insufficiency, and nonalcoholic steatohepatitis or NASH. All analyses accounted for complex sampling scheme of the NIS. RESULTS The total prevalence of PwCF in the National Inpatient Sample (NIS) database was 0.08 %, and the number was stable year to year from 2014 to 2019. Hispanic patients were more likely to be diagnosed with PUD than other white (aOR 1.802 [1.311,2.476]). Multiple regression analysis indicated that PUD in PwCF was strongly associated with a diagnosis of NASH (aOR 2.421[1.197, 4.898]). PUD patients were less likely to have pancreatic insufficiency compared to the non-PUD group (aOR 0.583 [0.455, 0.745]). CONCLUSION Although cystic fibrosis has been historically known as a disease of childhood, advancements in therapy have led to prolonged life expectancy and higher prevalence for cystic fibrosis-related digestive diseases. This study revealed a low prevalence of PUD in PwCF. Hispanics and those with NASH are more likely to develop peptic ulcers.
Collapse
Affiliation(s)
- Maya Mahmoud
- Department of Internal Medicine, Saint Louis University, St Louis Missouri, United States.
| | - Eugene Nwankwo
- Department of Internal Medicine, Saint Louis University, St Louis Missouri, United States
| | - Zidong Zhang
- Advanced Health Data (AHEAD) Institute, Saint Louis University School of Medicine, United States
| | - Neel Matiwala
- Saint Louis University School of medicine, St Louis Missouri, United States
| | - Rohan Tripathi
- Saint Louis University School of medicine, St Louis Missouri, United States
| | - Islam Mohamed
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas city Missouri, United States
| | - Christopher Barrios
- Division of Pulmonary, Department of Internal Medicine, Saint Louis University, St Louis Missouri, United States
| | - Wing-Kin Syn
- Division of Gastroenterology & Hepatology, Department of Internal Medicine, Saint Louis University, St Louis Missouri, United States; Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, Vizcaya, Spain
| | - Christine Hachem
- Division of Gastroenterology & Hepatology, Department of Internal Medicine, Saint Louis University, St Louis Missouri, United States
| |
Collapse
|
12
|
Jiang Y, Jin H, Liang Q, Zhu X. Causal relationship between gut microbiota and dental caries: A Mendelian randomization analysis. Medicine (Baltimore) 2025; 104:e41555. [PMID: 39993094 PMCID: PMC11856918 DOI: 10.1097/md.0000000000041555] [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: 09/27/2024] [Revised: 12/02/2024] [Accepted: 01/29/2025] [Indexed: 02/26/2025] Open
Abstract
The onset of dental caries is associated with multiple factors, including oral microbiota, dietary sugars, the defensive mechanisms of saliva and teeth, oral hygiene practices, and socioeconomic factors. However, its relationship with the gut microbiota remains to be further explored. It remains crucial to establish a definitive causal link between the gut microbiota and the development of dental caries. This study aimed to investigate the causal relationship between gut microbiota and the risk of dental caries, focusing on identifying specific microbial communities potentially implicated in its pathogenesis. Gut microbiota data from genome-wide association studies (GWAS) conducted by the MiBioGen consortium were utilized as the exposure variable, with dental caries as the outcome variable. A Mendelian randomization (MR) approach was employed, leveraging comprehensive, publicly available GWAS summary data from European populations. The primary analytical method was the inverse variance weighted method, supplemented by additional techniques such as the weighted median model, MR-Egger, simple mode, and weighted mode, to ensure the robustness of the results. Heterogeneity was evaluated using Cochran Q test, and potential pleiotropy was assessed through MR-Egger regression. Sensitivity analyses were performed using the leave-one-out method to further validate the findings. The results revealed that a higher relative abundance of Christensenellaceae, FamilyXIII, Ruminococcaceae, and Senegalimassilia was associated with a reduced risk of dental caries. In contrast, a higher relative abundance of Erysipelotrichia, Erysipelotrichales, Pasteurellales, Erysipelotrichaceae, Pasteurellaceae, Methanobrevibacter, Roseburia, and Terrisporobacter was linked to an elevated risk of dental caries. This study provides compelling evidence for a causal relationship between gut microbiota and the development of dental caries, offering novel insights into the potential role of specific gut microbial communities in the pathogenesis of dental caries.
Collapse
Affiliation(s)
- Yongyuan Jiang
- Department of Stomatology, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, P.R. China
| | - Huan Jin
- Department of Emergency, Qingdao Municipal Hospital, Qingdao, Shandong, P.R. China
| | - Qian Liang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xuan Zhu
- Department of Stomatology, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, P.R. China
| |
Collapse
|
13
|
Rondanelli M, Borromeo S, Cavioni A, Gasparri C, Gattone I, Genovese E, Lazzarotti A, Minonne L, Moroni A, Patelli Z, Razza C, Sivieri C, Valentini EM, Barrile GC. Therapeutic Strategies to Modulate Gut Microbial Health: Approaches for Chronic Metabolic Disorder Management. Metabolites 2025; 15:127. [PMID: 39997751 PMCID: PMC11857149 DOI: 10.3390/metabo15020127] [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: 12/02/2024] [Revised: 01/17/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Numerous recent studies have suggested that the composition of the intestinal microbiota can trigger metabolic disorders, such as diabetes, prediabetes, obesity, metabolic syndrome, sarcopenia, dyslipidemia, hyperhomocysteinemia, and non-alcoholic fatty liver disease. Since then, considerable effort has been made to understand the link between the composition of intestinal microbiota and metabolic disorders, as well as the role of probiotics in the modulation of the intestinal microbiota. The aim of this review was to summarize the reviews and individual articles on the state of the art regarding ideal therapy with probiotics and prebiotics in order to obtain the reversion of dysbiosis (alteration in microbiota) to eubiosis during metabolic diseases, such as diabetes, prediabetes, obesity, hyperhomocysteinemia, dyslipidemia, sarcopenia, and non-alcoholic fatty liver diseases. This review includes 245 eligible studies. In conclusion, a condition of dysbiosis, or in general, alteration of the intestinal microbiota, could be implicated in the development of metabolic disorders through different mechanisms, mainly linked to the release of pro-inflammatory factors. Several studies have already demonstrated the potential of using probiotics and prebiotics in the treatment of this condition, detecting significant improvements in the specific symptoms of metabolic diseases. These findings reinforce the hypothesis that a condition of dysbiosis can lead to a generalized inflammatory picture with negative consequences on different organs and systems. Moreover, this review confirms that the beneficial effects of probiotics on metabolic diseases are promising, but more research is needed to determine the optimal probiotic strains, doses, and administration forms for specific metabolic conditions.
Collapse
Affiliation(s)
- Mariangela Rondanelli
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy;
| | - Sara Borromeo
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Alessandro Cavioni
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Clara Gasparri
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Ilaria Gattone
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Elisa Genovese
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Alessandro Lazzarotti
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Leonardo Minonne
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Alessia Moroni
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Zaira Patelli
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Claudia Razza
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Claudia Sivieri
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Eugenio Marzio Valentini
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Gaetan Claude Barrile
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| |
Collapse
|
14
|
Hu W, Gong W, Yang F, Cheng R, Zhang G, Gan L, Zhu Y, Qin W, Gao Y, Li X, Liu J. Dual GIP and GLP-1 receptor agonist tirzepatide alleviates hepatic steatosis and modulates gut microbiota and bile acid metabolism in diabetic mice. Int Immunopharmacol 2025; 147:113937. [PMID: 39752752 DOI: 10.1016/j.intimp.2024.113937] [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: 10/29/2024] [Revised: 12/14/2024] [Accepted: 12/21/2024] [Indexed: 01/29/2025]
Abstract
Tirzepatide is a dual agonist of glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) receptors and is a promising therapeutic option for type 2 diabetes mellitus (T2DM). Nevertheless, its effect and underlying mechanism on hepatic steatosis remain ambiguous. Herein, we explored the impact of tirzepatide on improving hepatic steatosis in diabetic mice, with a particular focus on the gut microbiota and bile acids (BAs) using animal models. The tirzepatide effectively reduced body weight, improved insulin resistance, decreased serum and hepatic lipid levels, and mitigated liver injury. Compared to semaglutide, tirzepatide exhibited superior efficacy in reducing hepatic lipid accumulation. 16S rRNA gene sequencing and targeted metabolomics of BAs revealed that tirzepatide ameliorated gut microbiota dysbiosis and BAs metabolism in diabetic mice. Notably, tirzepatide observably increased the abundance of beneficial genera such as Akkermansia, elevated the ratio of farnesoid X receptor (FXR) antagonists (glycoursodeoxycholic acid: GUDCA, β-muricholic acid: β-MCA, hyodeoxycholic acid: HDCA, ursodeoxycholic acid: UDCA) to natural agonists (cholic acid: CA, lithocholic acid: LCA, chenodeoxycholic acid: CDCA, glycocholic acid: GCA, taurodeoxycholic acid: TDCA), and reduced FXR expression in intestinal tissues. In conclusion, tirzepatide attenuated hepatic steatosis in diabetic mice and regulated the gut microbiota and BAs metabolism, which may help to provide a novel therapeutic approach and therapeutic target for metabolic dysfunction-associated steatotic liver disease (MASLD).
Collapse
Affiliation(s)
- Weiting Hu
- Department of Clinical Medicine, The Second Clinical Medical College, Shanxi Medical University, Taiyuan 030000, China; Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Wenyu Gong
- Department of Clinical Medicine, The Second Clinical Medical College, Shanxi Medical University, Taiyuan 030000, China
| | - Fan Yang
- The First Clinical Medical College, Shanxi Medical University, Taiyuan 030000, China
| | - Rui Cheng
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Gerong Zhang
- Department of Clinical Medicine, The Second Clinical Medical College, Shanxi Medical University, Taiyuan 030000, China
| | - Lu Gan
- Department of Emergency Medicine and National Clinical Research Center for Geriatrics, Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Yikun Zhu
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Weiwei Qin
- Department of Cardiology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Ying Gao
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Xing Li
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China.
| | - Jing Liu
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China.
| |
Collapse
|
15
|
Gan C, Yuan Y, Shen H, Gao J, Kong X, Che Z, Guo Y, Wang H, Dong E, Xiao J. Liver diseases: epidemiology, causes, trends and predictions. Signal Transduct Target Ther 2025; 10:33. [PMID: 39904973 PMCID: PMC11794951 DOI: 10.1038/s41392-024-02072-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/06/2024] [Accepted: 11/12/2024] [Indexed: 02/06/2025] Open
Abstract
As a highly complex organ with digestive, endocrine, and immune-regulatory functions, the liver is pivotal in maintaining physiological homeostasis through its roles in metabolism, detoxification, and immune response. Various factors including viruses, alcohol, metabolites, toxins, and other pathogenic agents can compromise liver function, leading to acute or chronic injury that may progress to end-stage liver diseases. While sharing common features, liver diseases exhibit distinct pathophysiological, clinical, and therapeutic profiles. Currently, liver diseases contribute to approximately 2 million deaths globally each year, imposing significant economic and social burdens worldwide. However, there is no cure for many kinds of liver diseases, partly due to a lack of thorough understanding of the development of these liver diseases. Therefore, this review provides a comprehensive examination of the epidemiology and characteristics of liver diseases, covering a spectrum from acute and chronic conditions to end-stage manifestations. We also highlight the multifaceted mechanisms underlying the initiation and progression of liver diseases, spanning molecular and cellular levels to organ networks. Additionally, this review offers updates on innovative diagnostic techniques, current treatments, and potential therapeutic targets presently under clinical evaluation. Recent advances in understanding the pathogenesis of liver diseases hold critical implications and translational value for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Can Gan
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan Yuan
- Aier Institute of Ophthalmology, Central South University, Changsha, China
| | - Haiyuan Shen
- Department of Oncology, the First Affiliated Hospital; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jinhang Gao
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangxin Kong
- Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Zhaodi Che
- Clinical Medicine Research Institute and Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yangkun Guo
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China.
| | - Erdan Dong
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| | - Jia Xiao
- Clinical Medicine Research Institute and Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China.
- Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China.
| |
Collapse
|
16
|
Liang S, Yao Z, Chen J, Qian J, Dai Y, Li H. Structural characterization of a α-d-glucan from Ginkgo biloba seeds and its protective effects on non-alcoholic fatty liver disease in mice. Carbohydr Polym 2025; 349:123022. [PMID: 39638527 DOI: 10.1016/j.carbpol.2024.123022] [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: 06/05/2024] [Revised: 10/03/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) poses a great global challenge to public health, yet it holds promise for amelioration through plant-derived polysaccharide. Ginkgo biloba seeds have long been used as medicine and food, which has potential benefits for various chronic diseases. However, the protective role of Ginkgo biloba seed polysaccharide against NAFLD remains unclear. In this study, we isolated and purified polysaccharide (GBSP-2) from Ginkgo biloba seeds. GBSP-2 is composed of α-d-glucopyranose residues, which are interconnected with α-d-glucopyranose units linked by (1→4) bonds, (1→4,6) bonds and (1→3,4) bonds, the ratio distribution is 15:1:1. By studying a mouse model, we investigated the effect of GBSP-2 (100 or 200 mg/kg) on high-fat-diet-induced NAFLD. We demonstrated that GBSP-2 significantly alleviated NAFLD, as evidenced by reduced hepatic steatosis, decreased inflammation, improved oxidative stress and ameliorative glucolipid metabolic disorders. Furthermore, GBSP-2 mitigated gut microbiota disturbance of NAFLD mice and markedly increased the abundance of Akkermansia, Romboutsia, Lactobacillus and Bacteroides. Mechanistically, GBSP-2 could activate AMPK/ACC signaling pathway to inhibit lipid synthesis by generating 3,4-dihydroxyphenylpropionic acid (DHPPA). Overall, these findings suggest that GBSP-2 plays a multi-channel and multi-target role in improving NAFLD through the gut-liver axis.
Collapse
Affiliation(s)
- Shuxiao Liang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhijie Yao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jinxiang Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jin Qian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yufeng Dai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Haitao Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
17
|
Saeed H, Díaz LA, Gil-Gómez A, Burton J, Bajaj JS, Romero-Gomez M, Arrese M, Arab JP, Khan MQ. Microbiome-centered therapies for the management of metabolic dysfunction-associated steatotic liver disease. Clin Mol Hepatol 2025; 31:S94-S111. [PMID: 39604327 PMCID: PMC11925441 DOI: 10.3350/cmh.2024.0811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a significant global health issue, affecting over 30% of the population worldwide due to the rising prevalence of metabolic risk factors such as obesity and type 2 diabetes mellitus. This spectrum of liver disease ranges from isolated steatosis to more severe forms such as steatohepatitis, fibrosis, and cirrhosis. Recent studies highlight the role of gut microbiota in MASLD pathogenesis, showing that dysbiosis significantly impacts metabolic health and the progression of liver disease. This review critically evaluates current microbiome-centered therapies in MASLD management, including prebiotics, probiotics, synbiotics, fecal microbiota transplantation, and emerging therapies such as engineered bacteria and bacteriophage therapy. We explore the scientific rationale, clinical evidence, and potential mechanisms by which these interventions influence MASLD. The gut-liver axis is crucial in MASLD, with notable changes in microbiome composition linked to disease progression. For instance, specific microbial profiles and reduced alpha diversity are associated with MASLD severity. Therapeutic strategies targeting the microbiome could modulate disease progression by improving gut permeability, reducing endotoxin-producing bacteria, and altering bile acid metabolism. Although promising, these therapies require further research to fully understand their mechanisms and optimize their efficacy. This review integrates findings from clinical trials and experimental studies, providing a comprehensive overview of microbiome-centered therapies' potential in managing MASLD. Future research should focus on personalized strategies, utilizing microbiome features, blood metabolites, and customized dietary interventions to enhance the effectiveness of these therapies.
Collapse
Affiliation(s)
- Huma Saeed
- Division of Infectious Diseases, Department of Medicine, University of Western Ontario, London, ON, Canada
| | - Luis Antonio Díaz
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, CA, USA
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Antonio Gil-Gómez
- SeLiver Group, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Jeremy Burton
- Department of Microbiology & Immunology, Western University, London, ON, Canada
| | - Jasmohan S Bajaj
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Manuel Romero-Gomez
- SeLiver Group, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- UCM Digestive diseases, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Marco Arrese
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Pablo Arab
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Mohammad Qasim Khan
- Division of Gastroenterology, Department of Medicine, University of Western Ontario, London, ON, Canada
- Department of Epidemiology and Biostatistics, University of Western Ontario, London, ON, Canada
| |
Collapse
|
18
|
Redruello-Requejo M, del Mar Blaya M, González-Reguero D, Robas-Mora M, Arranz-Herrero J, Partearroyo T, Varela-Moreiras G, Penalba-Iglesias D, Jiménez-Gómez P, Reche-Sainz P. Cross-Sectional Comparative Analysis of Gut Microbiota in Spanish Adolescents with Mediterranean and Western Diets. Nutrients 2025; 17:388. [PMID: 39940246 PMCID: PMC11820480 DOI: 10.3390/nu17030388] [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: 12/23/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/14/2025] Open
Abstract
Dietary patterns, such as the Mediterranean diet (MD) and the Western diet (WD), influence gut microbiota composition and functionality, which play important roles in energy metabolism and nutrient absorption. OBJECTIVES A descriptive cross-sectional study was designed to evaluate the gut microbiota of 19 Spanish adolescents and to investigate the association of MD and ultra-processed food (UPF) intake with microbial diversity and community structure. METHODS Functional diversity of gut microbiota was evaluated using Biolog EcoPlates, taxonomic composition was assessed with 16S rRNA sequencing via MinION, and phenotypic responses to antibiotics were analyzed using the cenoantibiogram technique under aerobic and anaerobic conditions. RESULTS Adolescents with higher adherence to the MD exhibited greater functional diversity, as per the Shannon-Weaver index. In addition, this group showed higher abundance of bacterial genera previously described as beneficial, such as Paraclostridium, Anaerobutyricum, Romboutsia, and Butyricicoccus. In contrast, adolescents reporting greater UPF intakes had a microbiota composition similar to those with low adherence to the MD, characterized by decreased abundance of beneficial genera. Regarding antibiotic resistance, significant differences were only observed under anaerobic conditions, with individuals with low adherence to the MD showing more sensitivity for most antibiotics tested. CONCLUSIONS These results suggest that the MD promotes a healthier and more balanced gut environment, potentially improving metabolic functions in adolescents. Despite the lack of differences in α-diversity, comparisons of microbial community structure between adolescents following the MD and those with high UPF (characteristic of the WD) showed clear differences in terms of β-diversity. These findings suggest that dietary patterns influence the composition of the gut microbiota in a more complex manner, beyond just taxonomic richness. The outcomes of this exploratory study highlight opportunities for future research to deepen understanding of the long-term health implications of these dietary patterns, as well as the mechanisms regulating the composition, functionality, and phenotypic responses to antibiotics of gut microbial communities.
Collapse
Affiliation(s)
- Marina Redruello-Requejo
- Grupo USP-CEU de Excelencia “Nutrición para la vida (Nutrition for Life)”, Ref: E02/0720, Department of Pharmaceutical and Health Sciences, Faculty of Pharmacy, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (T.P.); (G.V.-M.)
- Instituto Universitario CEU Alimentación y Sociedad, Faculty of Pharmacy, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain
| | - María del Mar Blaya
- Department of Pharmaceutical and Health Sciences, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (M.d.M.B.); (D.G.-R.); (M.R.-M.); (J.A.-H.); (D.P.-I.); (P.R.-S.)
| | - Daniel González-Reguero
- Department of Pharmaceutical and Health Sciences, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (M.d.M.B.); (D.G.-R.); (M.R.-M.); (J.A.-H.); (D.P.-I.); (P.R.-S.)
| | - Marina Robas-Mora
- Department of Pharmaceutical and Health Sciences, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (M.d.M.B.); (D.G.-R.); (M.R.-M.); (J.A.-H.); (D.P.-I.); (P.R.-S.)
| | - Javier Arranz-Herrero
- Department of Pharmaceutical and Health Sciences, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (M.d.M.B.); (D.G.-R.); (M.R.-M.); (J.A.-H.); (D.P.-I.); (P.R.-S.)
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA) Nemesio Díez, Medicine Faculty, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain
| | - Teresa Partearroyo
- Grupo USP-CEU de Excelencia “Nutrición para la vida (Nutrition for Life)”, Ref: E02/0720, Department of Pharmaceutical and Health Sciences, Faculty of Pharmacy, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (T.P.); (G.V.-M.)
- Instituto Universitario CEU Alimentación y Sociedad, Faculty of Pharmacy, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain
| | - Gregorio Varela-Moreiras
- Grupo USP-CEU de Excelencia “Nutrición para la vida (Nutrition for Life)”, Ref: E02/0720, Department of Pharmaceutical and Health Sciences, Faculty of Pharmacy, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (T.P.); (G.V.-M.)
- Instituto Universitario CEU Alimentación y Sociedad, Faculty of Pharmacy, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain
| | - Diana Penalba-Iglesias
- Department of Pharmaceutical and Health Sciences, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (M.d.M.B.); (D.G.-R.); (M.R.-M.); (J.A.-H.); (D.P.-I.); (P.R.-S.)
| | - Pedro Jiménez-Gómez
- Department of Pharmaceutical and Health Sciences, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (M.d.M.B.); (D.G.-R.); (M.R.-M.); (J.A.-H.); (D.P.-I.); (P.R.-S.)
| | - Paloma Reche-Sainz
- Department of Pharmaceutical and Health Sciences, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (M.d.M.B.); (D.G.-R.); (M.R.-M.); (J.A.-H.); (D.P.-I.); (P.R.-S.)
| |
Collapse
|
19
|
Wang LJ, Sun JG, Chen SC, Sun YL, Zheng Y, Feng JC. The role of intestinal flora in metabolic dysfunction-associated steatotic liver disease and treatment strategies. Front Med (Lausanne) 2025; 11:1490929. [PMID: 39839647 PMCID: PMC11746088 DOI: 10.3389/fmed.2024.1490929] [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: 09/04/2024] [Accepted: 11/28/2024] [Indexed: 01/23/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a common multi-factorial liver disease, and its incidence is gradually increasing worldwide. Many reports have revealed that intestinal flora plays a crucial role for the occurrence and development of MASLD, through mechanisms such as flora translocation, endogenous ethanol production, dysregulation of choline metabolism and bile acid, and endotoxemia. Here, we review the relationship between intestinal flora and MASLD, as well as interventions for MASLD, such as prebiotics, probiotics, synbiotics, and intestinal flora transplantation. Intervention strategies targeting the intestinal flora along with its metabolites may be new targets for preventing and treating MASLD.
Collapse
Affiliation(s)
- Li Jun Wang
- Department of Traditional Chinese Medicine, Binzhou Medical University, Yantai, China
| | - Jian Guang Sun
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shu Cheng Chen
- School of Nursing, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Yu Li Sun
- Department of Hepatology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yang Zheng
- Department of Acupuncture and Moxibustion, Zibo Hospital, Zibo, China
| | - Jian Chao Feng
- Department of Acupuncture and Moxibustion, Zibo Hospital, Zibo, China
| |
Collapse
|
20
|
Lin Z, Li T, Huang F, Wu M, Zhu L, Zhou Y, Ming Y, Lu Z, Peng W, Gao F, Zhu Y. Comparison of diet and exercise on cardiometabolic factors in young adults with overweight/obesity: multiomics analysis and gut microbiota prediction, a randomized controlled trial. MedComm (Beijing) 2025; 6:e70044. [PMID: 39802638 PMCID: PMC11725046 DOI: 10.1002/mco2.70044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/13/2024] [Accepted: 11/22/2024] [Indexed: 01/16/2025] Open
Abstract
The optimal strategy for improving cardiometabolic factors (CMFs) in young obese individuals through diet and exercise remains unclear, as do the potential mechanisms. We conducted an 8-week randomized controlled trial to compare the effects of different interventions in youth with overweight/obesity. Gut microbes and serum metabolites were examined to identify regulating mechanisms. A total of 129 undergraduates were randomly assigned to fiber-rich (FR) diet, rope-skipping (RS), combined FR-RS and control groups. The results showed that single interventions were as effective as combined interventions in improving weight, waist circumference, body fat, and lipid profile compared with control group. Notably, the FR group further reduced low-density lipoprotein (LDL-C) and uric acid (UA) (all p < 0.05). Mediation analysis revealed four gut microbiota-metabolite-host axes in improving CMFs. Additionally, we used machine learning algorithms to further predict individual responses based on baseline gut microbiota composition, with specific microbial genera guiding targeted intervention selection. In conclusion, FR diet and/or RS were effective in improving CMFs, with the FR diet particular effectiveness in reducing LDL-C and UA levels. These benefits may drive by gut microbiome-metabolite-host interactions. Moreover, the predictability of gut microbiota composition supports making targeted decisions in selecting interventions. Trial Registration: NCT04834687.
Collapse
Affiliation(s)
- Zongyu Lin
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Tianze Li
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Fenglian Huang
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Miao Wu
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Lewei Zhu
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Yueqin Zhou
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Ying‐An Ming
- Department of Physical EducationSun Yat‐sen UniversityGuangzhouChina
| | - Zhijun Lu
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Wei Peng
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Fei Gao
- Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
| | - Yanna Zhu
- Department of Maternal and Child HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of FoodNutrition and HealthGuangzhouChina
| |
Collapse
|
21
|
Saha S, Schnabl B. Modulating the microbiome in chronic liver diseases - current evidence on the role of fecal microbiota transplantation. Expert Rev Gastroenterol Hepatol 2025; 19:53-64. [PMID: 39760535 PMCID: PMC11882407 DOI: 10.1080/17474124.2025.2450707] [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: 10/29/2024] [Revised: 12/23/2024] [Accepted: 01/04/2025] [Indexed: 01/07/2025]
Abstract
INTRODUCTION The gut microbiota has a complex relationship with the human host and is key to maintaining health. Disruption of the healthy diverse gut microbial milieu plays an important role in the pathogenesis of several diseases including Clostridioides difficile infection (CDI), inflammatory bowel disease, irritable bowel syndrome, alcohol-related liver disease and metabolic-dysfunction associated steatotic liver disease (MASLD). Fecal microbiota transplantation (FMT) is highly effective in treating CDI, though its utility in other diseases is still being explored. AREAS COVERED In this narrative review, we explore the role of gut microbiota in liver diseases, focusing on key changes in the microbial composition and function. We summarize current evidence on the role of FMT, identifying gaps in current research and outlining future directions for investigation. We comprehensively searched PubMed through 15 October 2024 to identify relevant studies. EXPERT OPINION While data from available studies shows promise, more research is necessary before we can use FMT for liver diseases. Key areas that require further study are - determining the optimal FMT regimen for each disease, establishing efficacy and safety with larger clinical trials, ensuring safe and equitable access to the FMT product and mechanistic insights into the reasons for success or failure of FMT.
Collapse
Affiliation(s)
- Srishti Saha
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of California San Diego, San Diego, CA
| | - Bernd Schnabl
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of California San Diego, San Diego, CA
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
22
|
Chen T, Wang N, Hao Y, Fu L. Fecal microbiota transplantation from postmenopausal osteoporosis human donors accelerated bone mass loss in mice. Front Cell Infect Microbiol 2024; 14:1488017. [PMID: 39703374 PMCID: PMC11655470 DOI: 10.3389/fcimb.2024.1488017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/13/2024] [Indexed: 12/21/2024] Open
Abstract
Objectives To investigate the effect of gut microbiota from postmenopausal osteoporosis patients on bone mass in mice. Methods Fecal samples were collected from postmenopausal women with normal bone mass (Con, n=5) and postmenopausal women with osteoporosis (Op, n=5). Microbial composition was identified by shallow shotgun sequencing. Then fecal samples were transplanted into pseudo-sterile mice previously treated with antibiotics for 4 weeks. These mice were categorized into two groups: the Vehicle group (n=7) received fecal samples from individuals with normal bone mass, and the FMT group (n=7) received fecal samples from individuals with osteoporosis. After 8 weeks, bone mass, intestinal microbial composition, intestinal permeability and inflammation were assessed, followed by a correlation analysis. Results The bone mass was significantly reduced in the FMT group. Microbiota sequencing showed that Shannon index (p < 0.05) and Simpson index (p < 0.05) were significantly increased in Op groups, and β diversity showed significant differences. the recipient mice were similar. linear discriminant analysis effect size (LEfSe) analysis of mice showed that Halobiforma, Enterorhabdus, Alistipes, and Butyricimonas were significantly enriched in the FMT group. Lachnospiraceae and Oscillibacter were significantly enriched in the Vehicle group. H&E staining of intestinal tissues showed obvious intestinal mucosal injury in mice. Intestinal immunohistochemistry showed that the expression of Claudin and ZO-1 in the intestinal tissue of the FMT group mice was decreased. The FITC-Dextran (FD-4) absorption rate and serum soluble CD14 (sCD14) content were increased in FMT mice. Correlation analysis showed that these dominant genera were significantly associated with bone metabolism and intestinal permeability, and were associated with the enrichment of specific enzymes. Serum and bone tissue inflammatory cytokines detection showed that the expression of TNF-α and IL-17A in the FMT group were significantly increased. Conclusion Overall, our findings suggested gut microbiota from postmenopausal osteoporosis patients accelerate bone mass loss in mice. Aberrant gut microbiota might play a causal role in the process of bone mass loss mediated by inflammation after the destruction of the intestinal barrier.
Collapse
Affiliation(s)
- Tinglong Chen
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Innovative Orthopaedic Instruments and Personalized Medicine, Clinical and Translational Research Center for 3D Printing Technology, Shanghai, China
| | - Ning Wang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Innovative Orthopaedic Instruments and Personalized Medicine, Clinical and Translational Research Center for 3D Printing Technology, Shanghai, China
| | - Yongqiang Hao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Innovative Orthopaedic Instruments and Personalized Medicine, Clinical and Translational Research Center for 3D Printing Technology, Shanghai, China
| | - Lingjie Fu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Innovative Orthopaedic Instruments and Personalized Medicine, Clinical and Translational Research Center for 3D Printing Technology, Shanghai, China
| |
Collapse
|
23
|
Miao S, Li J, Chen Y, Zhao W, Xu M, Liu F, Zou X, Dong X. Targeting gut microbiota and metabolism profiles with coated sodium butyrate to ameliorate high-energy and low-protein diet-induced intestinal barrier dysfunction in laying hens. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 19:104-116. [PMID: 39635416 PMCID: PMC11615920 DOI: 10.1016/j.aninu.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 12/07/2024]
Abstract
High energy diets are a risk factor for intestinal barrier damage. Butyrate, a major energy source for intestinal epithelial cells, has been shown to improve barrier dysfunction and modulate the gut microbiota. In this trial, we examined the preventative effect of coated sodium butyrate (CSB) on high-energy and low-protein (HELP)-induced intestinal barrier injury in laying hens, and also worked to determine the underlying mechanisms by an integrative analysis of gut microbiota and the metabolome. A total of 216 healthy 28-week-old Huafeng laying hens were randomly assigned to 3 groups with 6 replicates each: the CON group (normal diet), HELP group (HELP diet) and CH500 group (500 mg/kg CSB added to HELP diet). The duration of the trial encompassed a period of 10 weeks. The results revealed that CSB treatment improved the laying rate and mitigated the detrimental effects on intestinal barrier function and the inflammatory response induced by the HELP diet in laying hens (P < 0.05). Microbial profiling analysis revealed that the CSB treatment reshaped the HELP-perturbed gut microbiota and promoted the growth of beneficial bacteria (P < 0.05). Untargeted metabolomics analysis revealed that CSB reduced the metabolites associated with intestinal inflammation (P < 0.05). In conclusion, CSB did not merely modulate alterations in the gut microbiota composition and microbial metabolites but also yielded increased egg production, while mitigating intestinal barrier dysfunction and inflammatory responses induced by HELP in laying hens.
Collapse
Affiliation(s)
- Sasa Miao
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiankui Li
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ying Chen
- Hangzhou Zhejiang University Animal Hospital Co., Ltd., Hangzhou 310058, China
| | - Wenyan Zhao
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mengru Xu
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fang Liu
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaoting Zou
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinyang Dong
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
24
|
Zha A, Qi M, Deng Y, Li H, Wang N, Wang C, Liao S, Wan D, Xiong X, Liao P, Wang J, Yin Y, Tan B. Gut Bifidobacterium pseudocatenulatum protects against fat deposition by enhancing secondary bile acid biosynthesis. IMETA 2024; 3:e261. [PMID: 39742294 PMCID: PMC11683477 DOI: 10.1002/imt2.261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025]
Abstract
Gut microbiome is crucial for lipid metabolism in humans and animals. However, how specific gut microbiota and their associated metabolites impact fat deposition remains unclear. In this study, we demonstrated that the colonic microbiome of lean and obese pigs differentially contributes to fat deposition, as evidenced by colonic microbiota transplantation experiments. Notably, the higher abundance of Bifidobacterium pseudocatenulatum was significantly associated with lower backfat thickness in lean pigs. Microbial-derived lithocholic acid (LCA) species were also significantly enriched in lean pigs and positively correlated with the abundance of B. pseudocatenulatum. In a high-fat diet (HFD)-fed mice model, administration of live B. pseudocatenulatum decreased fat deposition and enhances colonic secondary bile acid biosynthesis. Importantly, pharmacological inhibition of the bile salt hydrolase (BSH), which mediates secondary bile acid biosynthesis, impaired the anti-fat deposition effect of B. pseudocatenulatum in antibiotic-pretreated, HFD-fed mice. Furthermore, dietary LCA also decreased fat deposition in HFD-fed rats and obese pig models. These findings provide mechanistic insights into the anti-fat deposition role of B. pseudocatenulatum and identify BSH as a potential target for preventing excessive fat deposition in humans and animals.
Collapse
Affiliation(s)
- Andong Zha
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
- School of Basic Medical Science, Central South UniversityChangshaChina
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Ming Qi
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Yuankun Deng
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| | - Hao Li
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| | - Nan Wang
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| | - Chengming Wang
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| | - Simeng Liao
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| | - Dan Wan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Xia Xiong
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Peng Liao
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Jing Wang
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| | - Yulong Yin
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Bi'e Tan
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| |
Collapse
|
25
|
Zhou L, Zhang Y, Wu S, Kuang Y, Jiang P, Zhu X, Yin K. Type III Secretion System in Intestinal Pathogens and Metabolic Diseases. J Diabetes Res 2024; 2024:4864639. [PMID: 39544522 PMCID: PMC11561183 DOI: 10.1155/2024/4864639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 10/08/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
Modern lifestyle changes, especially the consumption of a diet high in salt, sugar, and fat, have contributed to the increasing incidence and prevalence of chronic metabolic diseases such as diabetes, obesity, and gout. Changing lifestyles continuously shape the gut microbiota which is closely related to the occurrence and development of metabolic diseases due to its specificity of composition and structural diversity. A large number of pathogenic bacteria such as Yersinia, Salmonella, Shigella, and pathogenic E. coli in the gut utilize the type III secretion system (T3SS) to help them resist host defenses and cause disease. Although the T3SS is critical for the virulence of many important human pathogens, its relationship with metabolic diseases remains unknown. This article reviews the structure and function of the T3SS, the disruption of intestinal barrier integrity by the T3SS, the changes in intestinal flora containing the T3SS in metabolic diseases, the possible mechanisms of the T3SS affecting metabolic diseases, and the application of the T3SS in the treatment of metabolic diseases. The aim is to provide insights into metabolic diseases targeting the T3SS, thereby serving as a valuable reference for future research on disease diagnosis, prevention, and treatment.
Collapse
Affiliation(s)
- Le Zhou
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Yaoyuan Zhang
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou 510900, China
| | - Shiqi Wu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Yiyu Kuang
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Pengfei Jiang
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Kai Yin
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou 510900, China
| |
Collapse
|
26
|
López-Tenorio II, Aguilar-Villegas ÓR, Espinoza-Palacios Y, Segura-Real L, Peña-Aparicio B, Amedei A, Aguirre-García MM. Primary Prevention Strategy for Non-Communicable Diseases (NCDs) and Their Risk Factors: The Role of Intestinal Microbiota. Biomedicines 2024; 12:2529. [PMID: 39595097 PMCID: PMC11591598 DOI: 10.3390/biomedicines12112529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/22/2024] [Accepted: 11/01/2024] [Indexed: 11/28/2024] Open
Abstract
Non-communicable diseases (NCDs) are the leading cause of morbidity and mortality worldwide. These conditions have numerous health consequences and significantly impact patients' lifestyles. Effective long-term treatment is essential since NCDs are irreversible. Therefore, primary healthcare must be both exclusive and of the highest quality, ensuring comprehensive care. The primary goal should be to improve quality of life with a focus on patients, families, and communities, as most of these diseases can be prevented and controlled, although not cured. Several factors have been linked to individual health, including social, cultural, and economic aspects, lifestyle, and certain environmental factors, including work, that can have positive or negative effects. More of these variables may contribute to the onset of NCDs, which are defined by their chronic nature, propensity for prolongation, and generally slow rate of progression. Examples of NCDs include hypertension, type 2 diabetes (T2D), dyslipidemia, and fatty liver disease linked to metabolic dysfunction. The onset of these diseases has been associated with an imbalance in certain microbial niches, such as the gut, which hosts billions of microorganisms performing multiple metabolic functions, such as the production of metabolites like bile acids (BAs), short-chain fatty acids (SCFAs), and trimethylamine N-oxide (TMAO). Therefore, lifestyle changes and personal habits can significantly impact the gut microbiota (GM), potentially preventing chronic diseases associated with metabolism. NCDs are highly prevalent worldwide, prompting increased attention to strategies for modifying the intestinal microbiota (IM). Approaches such as probiotics, prebiotics, synbiotics, and fecal transplantation (FMT) have demonstrated improvements in the quality of life for individuals with these conditions. Additionally, lifestyle changes and the adoption of healthy habits can significantly impact IM and may help prevent chronic diseases related to metabolism. Therefore, the main aim of this review is to analyze and understand the importance of microbiota intervention in the prevention of non-communicable diseases. R3:A1.
Collapse
Affiliation(s)
- Itzel Ivonn López-Tenorio
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina UNAM, Instituto Nacional de Cardiología Ignacio Cháve, Mexico City 14080, Mexico; (I.I.L.-T.); (Ó.R.A.-V.); (Y.E.-P.); (L.S.-R.)
| | - Óscar Rodrigo Aguilar-Villegas
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina UNAM, Instituto Nacional de Cardiología Ignacio Cháve, Mexico City 14080, Mexico; (I.I.L.-T.); (Ó.R.A.-V.); (Y.E.-P.); (L.S.-R.)
| | - Yoshua Espinoza-Palacios
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina UNAM, Instituto Nacional de Cardiología Ignacio Cháve, Mexico City 14080, Mexico; (I.I.L.-T.); (Ó.R.A.-V.); (Y.E.-P.); (L.S.-R.)
| | - Lorena Segura-Real
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina UNAM, Instituto Nacional de Cardiología Ignacio Cháve, Mexico City 14080, Mexico; (I.I.L.-T.); (Ó.R.A.-V.); (Y.E.-P.); (L.S.-R.)
| | - Berenice Peña-Aparicio
- Consulta Externa Endocrinología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy;
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 50134 Florence, Italy
| | - María Magdalena Aguirre-García
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina UNAM, Instituto Nacional de Cardiología Ignacio Cháve, Mexico City 14080, Mexico; (I.I.L.-T.); (Ó.R.A.-V.); (Y.E.-P.); (L.S.-R.)
| |
Collapse
|
27
|
Maher S, Rajapakse J, El-Omar E, Zekry A. Role of the Gut Microbiome in Metabolic Dysfunction-Associated Steatotic Liver Disease. Semin Liver Dis 2024; 44:457-473. [PMID: 39389571 DOI: 10.1055/a-2438-4383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD)-previously described as nonalcoholic fatty liver disease-continues to rise globally. Despite this, therapeutic measures for MASLD remain limited. Recently, there has been a growing interest in the gut microbiome's role in the pathogenesis of MASLD. Understanding this relationship may allow for the administration of therapeutics that target the gut microbiome and/or its metabolic function to alleviate MASLD development or progression. This review will discuss the interplay between the gut microbiome's structure and function in relation to the development of MASLD, assess the diagnostic yield of gut microbiome-based signatures as a noninvasive tool to identify MASLD severity, and examine current and emerging therapies targeting the gut microbiome-liver axis.
Collapse
Affiliation(s)
- Salim Maher
- Department of Gastroenterology and Hepatology, St George Hospital, Sydney, Australia
- School of Clinical Medicine, UNSW Medicine & Health, St George & Sutherland Clinical Campuses
| | - Jayashi Rajapakse
- School of Clinical Medicine, UNSW Medicine & Health, St George & Sutherland Clinical Campuses
| | - Emad El-Omar
- Department of Gastroenterology and Hepatology, St George Hospital, Sydney, Australia
- School of Clinical Medicine, UNSW Medicine & Health, St George & Sutherland Clinical Campuses
| | - Amany Zekry
- Department of Gastroenterology and Hepatology, St George Hospital, Sydney, Australia
- School of Clinical Medicine, UNSW Medicine & Health, St George & Sutherland Clinical Campuses
| |
Collapse
|
28
|
Hermanson JB, Tolba SA, Chrisler EA, Leone VA. Gut microbes, diet, and genetics as drivers of metabolic liver disease: a narrative review outlining implications for precision medicine. J Nutr Biochem 2024; 133:109704. [PMID: 39029595 PMCID: PMC11480923 DOI: 10.1016/j.jnutbio.2024.109704] [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: 04/05/2024] [Revised: 07/01/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is rapidly increasing in prevalence, impacting over a third of the global population. The advanced form of MASLD, Metabolic dysfunction-associated steatohepatitis (MASH), is on track to become the number one indication for liver transplant. FDA-approved pharmacological agents are limited for MASH, despite over 400 ongoing clinical trials, with only a single drug (resmetirom) currently on the market. This is likely due to the heterogeneous nature of disease pathophysiology, which involves interactions between highly individualized genetic and environmental factors. To apply precision medicine approaches that overcome interpersonal variability, in-depth insights into interactions between genetics, nutrition, and the gut microbiome are needed, given that each have emerged as dynamic contributors to MASLD and MASH pathogenesis. Here, we discuss the associations and molecular underpinnings of several of these factors individually and outline their interactions in the context of both patient-based studies and preclinical animal model systems. Finally, we highlight gaps in knowledge that will require further investigation to aid in successfully implementing precision medicine to prevent and alleviate MASLD and MASH.
Collapse
Affiliation(s)
- Jake B Hermanson
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Samar A Tolba
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Evan A Chrisler
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Vanessa A Leone
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|
29
|
Yaghmaei H, Bahanesteh A, Soltanipur M, Takaloo S, Rezaei M, Siadat SD. The Role of Gut Microbiota Modification in Nonalcoholic Fatty Liver Disease Treatment Strategies. Int J Hepatol 2024; 2024:4183880. [PMID: 39444759 PMCID: PMC11498984 DOI: 10.1155/2024/4183880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 08/25/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024] Open
Abstract
One of the most common chronic liver diseases is nonalcoholic fatty liver disease (NAFLD), which affects many people around the world. Gut microbiota (GM) dysbiosis seems to be an influential factor in the pathophysiology of NAFLD because changes in GM lead to fundamental changes in host metabolism. Therefore, the study of the effect of dysbiosis on the pathogenicity of NAFLD is important. European clinical guidelines state that the best advice for people with NAFLD is to lose weight and improve their lifestyle, but only 40% of people can achieve this goal. Accordingly, it is necessary to provide new treatment approaches for prevention and treatment. In addition to dietary interventions and lifestyle modifications, GM modification-based therapies are of interest. These therapies include probiotics, synbiotics, fecal microbiota transplantation (FMT), and next-generation probiotics. All of these treatments have had promising results in animal studies, and it can be imagined that acceptable results will be obtained in human studies as well. However, further investigations are required to generalize the outcomes of animal studies to humans.
Collapse
Affiliation(s)
- Hessam Yaghmaei
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | | | - Masood Soltanipur
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sobhan Takaloo
- Biomedical Engineering Department, Hamedan University of Technology, Hamedan, Iran
| | - Mahdi Rezaei
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
30
|
Kumar AR, Nair B, Kamath AJ, Nath LR, Calina D, Sharifi-Rad J. Impact of gut microbiota on metabolic dysfunction-associated steatohepatitis and hepatocellular carcinoma: pathways, diagnostic opportunities and therapeutic advances. Eur J Med Res 2024; 29:485. [PMID: 39367507 PMCID: PMC11453073 DOI: 10.1186/s40001-024-02072-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/22/2024] [Indexed: 10/06/2024] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) and progression to hepatocellular carcinoma (HCC) exhibits distinct molecular and immune characteristics. These traits are influenced by multiple factors, including the gut microbiome, which interacts with the liver through the "gut-liver axis". This bidirectional relationship between the gut and its microbiota and the liver plays a key role in driving various liver diseases, with microbial metabolites and immune responses being central to these processes. Our review consolidates the latest research on how gut microbiota contributes to MASH development and its progression to HCC, emphasizing new diagnostic and therapeutic possibilities. We performed a comprehensive literature review across PubMed/MedLine, Scopus, and Web of Science from January 2000 to August 2024, focusing on both preclinical and clinical studies that investigate the gut microbiota's roles in MASH and HCC. This includes research on pathogenesis, as well as diagnostic and therapeutic advancements related to the gut microbiota. This evidence emphasizes the critical role of the gut microbiome in the pathogenesis of MASH and HCC, highlighting the need for further clinical studies and trials. This is to refine diagnostic techniques and develop targeted therapies that exploit the microbiome's capabilities, aiming to enhance patient care in liver diseases.
Collapse
Affiliation(s)
- Ayana R Kumar
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - Bhagyalakshmi Nair
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - Adithya Jayaprakash Kamath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health. Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - Lekshmi R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, 092301, Ecuador.
- Centro de Estudios Tecnológicos y Universitarios del Golfo, Veracruz, Mexico.
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
31
|
Alisi A, McCaughan G, Grønbæk H. Role of gut microbiota and immune cells in metabolic-associated fatty liver disease: clinical impact. Hepatol Int 2024; 18:861-872. [PMID: 38995341 DOI: 10.1007/s12072-024-10674-6] [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: 01/24/2024] [Accepted: 03/18/2024] [Indexed: 07/13/2024]
Abstract
In 2020, a revised definition of fatty liver disease associated with metabolic dysfunction (MAFLD) was proposed to replace non-alcoholic fatty liver (NAFLD). Liver steatosis and at least one of the three metabolic risk factors, including type 2 diabetes, obesity, or signs of metabolic dysregulation, are used to diagnose MAFLD. MAFLD, similarly to NAFLD, is characterized by a spectrum of disease ranging from simple steatosis to advanced metabolic steatohepatitis with or without fibrosis, and may progress to cirrhosis and liver cancer, including increased risk of other critical extrahepatic diseases. Even though the pathophysiology of MAFLD and potential therapeutic targets have been explored in great detail, there is yet no Food and Drug Administration approved treatment. Recently, gut microbiome-derived products (e.g., endotoxins and metabolites) involved in intestinal barrier disruption, systemic inflammation, and modification of intrahepatic immunity have been associated with MAFLD development and progression. Therefore, different strategies could be adopted to modify the gut microbiome to improve outcomes in early and progressive MAFLD. Here, we provide an overview of mechanisms that may link the gut microbiome and immune response during the onset of liver steatosis and progression to steatohepatitis and fibrosis in patients with MAFLD. Finally, gut microbiota-based approaches are discussed as potential personalized treatments against MAFLD.
Collapse
Affiliation(s)
- Anna Alisi
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesu' Children Hospital, IRCCS, Rome, Italy.
| | - Geoffrey McCaughan
- A.W Morrow Gastroenterology and Liver Center, Royal Prince Alfred Hospital, Sydney, Australia
- Centenary Institute, University of Sydney, Sydney, Australia
| | - Henning Grønbæk
- Department of Hepatology & Gastroenterology, Aarhus University Hospital and Clinical Institute, Aarhus University, Aarhus, Denmark
| |
Collapse
|
32
|
Kwan SY, Sabotta CM, Cruz LR, Wong MC, Ajami NJ, McCormick JB, Fisher-Hoch SP, Beretta L. Gut phageome in Mexican Americans: a population at high risk for metabolic dysfunction-associated steatotic liver disease and diabetes. mSystems 2024; 9:e0043424. [PMID: 39166873 PMCID: PMC11406975 DOI: 10.1128/msystems.00434-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024] Open
Abstract
Mexican Americans are disproportionally affected by metabolic dysfunction-associated steatotic liver disease (MASLD), which often co-occurs with diabetes. Despite extensive evidence on the causative role of the gut microbiome in MASLD, studies determining the involvement of the gut phageome are scarce. In this cross-sectional study, we characterized the gut phageome in Mexican Americans of South Texas by stool shotgun metagenomic sequencing of 340 subjects, concurrently screened for liver steatosis by transient elastography. Inter-individual variations in the phageome were associated with gender, country of birth, diabetes, and liver steatosis. The phage signatures for diabetes and liver steatosis were subsequently determined. Enrichment of Inoviridae was associated with both diabetes and liver steatosis. Diabetes was further associated with the enrichment of predominantly temperate Escherichia phages, some of which possessed virulence factors. Liver steatosis was associated with the depletion of Lactococcus phages r1t and BK5-T, and enrichment of the globally prevalent Crassvirales phages, including members of genus cluster IX (Burzaovirus coli, Burzaovirus faecalis) and VI (Kahnovirus oralis). The Lactococcus phages showed strong correlations and co-occurrence with Lactococcus lactis, while the Crassvirales phages, B. coli, B. faecalis, and UAG-readthrough crAss clade correlated and co-occurred with Prevotella copri. In conclusion, we identified the gut phageome signatures for two closely linked metabolic diseases with significant global burden. These phage signatures may have utility in risk modeling and disease prevention in this high-risk population, and identification of potential bacterial targets for phage therapy.IMPORTANCEPhages influence human health and disease by shaping the gut bacterial community. Using stool samples from a high-risk Mexican American population, we provide insights into the gut phageome changes associated with diabetes and liver steatosis, two closely linked metabolic diseases with significant global burden. Common to both diseases was an enrichment of Inoviridae, a group of phages that infect bacterial hosts chronically without lysis, allowing them to significantly influence bacterial growth, virulence, motility, biofilm formation, and horizontal gene transfer. Diabetes was additionally associated with the enrichment of Escherichia coli-infecting phages, some of which contained virulence factors. Liver steatosis was additionally associated with the depletion of Lactococcus lactis-infecting phages, and enrichment of Crassvirales phages, a group of virulent phages with high global prevalence and persistence across generations. These phageome signatures may have utility in risk modeling, as well as identify potential bacterial targets for phage therapy.
Collapse
Affiliation(s)
- Suet-Ying Kwan
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Caroline M. Sabotta
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lorenzo R. Cruz
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Matthew C. Wong
- The Platform for Innovative Microbiome and Translational Research (PRIME-TR), Moon Shots Program, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nadim J. Ajami
- The Platform for Innovative Microbiome and Translational Research (PRIME-TR), Moon Shots Program, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Joseph B. McCormick
- School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, Texas, USA
| | - Susan P. Fisher-Hoch
- School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, Texas, USA
| | - Laura Beretta
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
33
|
Fu Y, Hua Y, Alam N, Liu E. Progress in the Study of Animal Models of Metabolic Dysfunction-Associated Steatotic Liver Disease. Nutrients 2024; 16:3120. [PMID: 39339720 PMCID: PMC11435380 DOI: 10.3390/nu16183120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has recently been proposed as an alternative term to NAFLD. MASLD is a globally recognized chronic liver disease that poses significant health concerns and is frequently associated with obesity, insulin resistance, and hyperlipidemia. To better understand its pathogenesis and to develop effective treatments, it is essential to establish suitable animal models. Therefore, attempts have been made to establish modelling approaches that are highly similar to human diet, physiology, and pathology to better replicate disease progression. Here, we reviewed the pathogenesis of MASLD disease and summarised the used animal models of MASLD in the last 7 years through the PubMed database. In addition, we have summarised the commonly used animal models of MASLD and describe the advantages and disadvantages of various models of MASLD induction, including genetic models, diet, and chemically induced models, to provide directions for research on the pathogenesis and treatment of MASLD.
Collapse
Affiliation(s)
- Yu Fu
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China; (Y.F.); (Y.H.)
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, 76 Yanta West Road, Xi’an 710061, China;
| | - Yuxin Hua
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China; (Y.F.); (Y.H.)
| | - Naqash Alam
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, 76 Yanta West Road, Xi’an 710061, China;
| | - Enqi Liu
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, 76 Yanta West Road, Xi’an 710061, China;
| |
Collapse
|
34
|
Singh S, Kriti M, Catanzaro R, Marotta F, Malvi M, Jain A, Verma V, Nagpal R, Tiwari R, Kumar M. Deciphering the Gut–Liver Axis: A Comprehensive Scientific Review of Non-Alcoholic Fatty Liver Disease. LIVERS 2024; 4:435-454. [DOI: 10.3390/livers4030032] [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] [Indexed: 12/11/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged as a significant global health issue. The condition is closely linked to metabolic dysfunctions such as obesity and type 2 diabetes. The gut–liver axis, a bidirectional communication pathway between the liver and the gut, plays a crucial role in the pathogenesis of NAFLD. This review delves into the mechanisms underlying the gut–liver axis, exploring the influence of gut microbiota, intestinal permeability, and inflammatory pathways. This review also explores the potential therapeutic strategies centered on modulating gut microbiota such as fecal microbiota transplantation; phage therapy; and the use of specific probiotics, prebiotics, and postbiotics in managing NAFLD. By understanding these interactions, we can better comprehend the development and advancement of NAFLD and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Samradhi Singh
- ICMR-National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal 462030, India
| | - Mona Kriti
- ICMR-National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal 462030, India
| | - Roberto Catanzaro
- Internal Medicine Unit, Department of Clinical and Experimental Medicine, Gastroenterology and Hepatology Service, University Hospital Policlinico “G. Rodolico”, University of Catania, 95123 Catania, Italy
| | | | - Mustafa Malvi
- Choithram Hospital and Research Centre Indore, Indore 452014, India
| | - Ajay Jain
- Choithram Hospital and Research Centre Indore, Indore 452014, India
| | - Vinod Verma
- Stem Cell Research Centre, Department of Hematology, Sanjay Gandhi Post-Graduate Institute of Medical Sciences, Lucknow 226014, India
| | - Ravinder Nagpal
- Department of Nutrition & Integrative Physiology, College of Health & Human Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Rajnarayan Tiwari
- ICMR-National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal 462030, India
| | - Manoj Kumar
- ICMR-National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal 462030, India
| |
Collapse
|
35
|
Zhang Y, Qiao Y, Li Z, Liu D, Jin Q, Guo J, Li X, Chen L, Liu L, Peng L. Intestinal NSD2 Aggravates Nonalcoholic Steatohepatitis Through Histone Modifications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402551. [PMID: 38923875 PMCID: PMC11434126 DOI: 10.1002/advs.202402551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/09/2024] [Indexed: 06/28/2024]
Abstract
Mounting clinical evidence suggests that a comprised intestinal barrier contributes to the progression of nonalcoholic steatohepatitis (NASH); nevertheless, the precise mechanism remains elusive. This study unveils a significant upregulation of nuclear receptor-binding SET domain protein 2 (NSD2) in the intestines of obese humans and mice subjected to a high-fat cholesterol diet (HFCD). Intestine-specific NSD2 knockout attenuated the progression of intestinal barrier impairment and NASH, whereas NSD2 overexpression exacerbated this progression. Mechanistically, NSD2 directly regulates the transcriptional activation of Ern1 by demethylating histone H3 at lysine 36 (H3K36me2), thus activating the ERN1-JNK axis to intensify intestinal barrier impairment and subsequently foster NASH progression. These findings elucidate the crucial role of NSD2-mediated H3K36me2 in intestinal barrier impairment, suggesting that targeting intestinal NSD2 can represent a novel therapeutic approach for NASH.
Collapse
Affiliation(s)
- Yijia Zhang
- Beijing Key Laboratory of BioprocessCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
- Beijing Key Laboratory for Immune‐Mediated Inflammatory DiseasesInstitute of Clinical Medical SciencesChina‐Japan Friendship HospitalBeijing100029P. R. China
| | - Yuan Qiao
- Beijing Key Laboratory for Immune‐Mediated Inflammatory DiseasesInstitute of Clinical Medical SciencesChina‐Japan Friendship HospitalBeijing100029P. R. China
| | - Zecheng Li
- Beijing Key Laboratory for Immune‐Mediated Inflammatory DiseasesInstitute of Clinical Medical SciencesChina‐Japan Friendship HospitalBeijing100029P. R. China
| | - Donghai Liu
- Beijing Key Laboratory for Immune‐Mediated Inflammatory DiseasesInstitute of Clinical Medical SciencesChina‐Japan Friendship HospitalBeijing100029P. R. China
| | - Qi Jin
- Beijing Key Laboratory for Immune‐Mediated Inflammatory DiseasesInstitute of Clinical Medical SciencesChina‐Japan Friendship HospitalBeijing100029P. R. China
| | - Jing Guo
- Beijing Key Laboratory for Immune‐Mediated Inflammatory DiseasesInstitute of Clinical Medical SciencesChina‐Japan Friendship HospitalBeijing100029P. R. China
| | - Xin Li
- Beijing Key Laboratory for Immune‐Mediated Inflammatory DiseasesInstitute of Clinical Medical SciencesChina‐Japan Friendship HospitalBeijing100029P. R. China
| | - Long Chen
- Beijing Key Laboratory of BioprocessCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Lihong Liu
- Beijing Key Laboratory for Immune‐Mediated Inflammatory DiseasesInstitute of Clinical Medical SciencesChina‐Japan Friendship HospitalBeijing100029P. R. China
| | - Liang Peng
- Beijing Key Laboratory for Immune‐Mediated Inflammatory DiseasesInstitute of Clinical Medical SciencesChina‐Japan Friendship HospitalBeijing100029P. R. China
| |
Collapse
|
36
|
Wu Q, Zhu F, Yao Y, Chen L, Ding Y, Su Y, Ge C. Sini san regulates intestinal flora and short-chain fatty acids to ameliorate hepatocyte apoptosis and relieve CCl 4-induced liver fibrosis in mice. Front Pharmacol 2024; 15:1408459. [PMID: 39281277 PMCID: PMC11392872 DOI: 10.3389/fphar.2024.1408459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/12/2024] [Indexed: 09/18/2024] Open
Abstract
Introduction Si-Ni-San (SNS), a traditional Chinese medicine, is effective in treating liver fibrosis with an unclear mechanism. Although disturbance of intestinal flora and the subsequent secretion of short-chain fatty acids (SCFAs) is suggested to be involved in the progression of liver fibrosis, whether SNS produces the anti-fibrosis effect through the regulation of intestinal flora and SCFAs remains unclear. Methods In the current study, carbon tetrachloride (CCl4)-treated mice were dosed with SNS to examine the anti-fibrotic effects and the involved mechanism. Biochemical parameters, histological staining, and analyses of fibrotic gene expression were used to evaluate the anti-fibrotic effect of SNS, while intestinal flora and SCFA content were determined by 16S rRNA and LC-MS to evaluate the mechanism. Results In vivo results showed that SNS improved liver function, reduced hepatocyte apoptosis and FFAR2/3 expression, and restored intestinal dysbiosis and reduced PA, BA, and IsA levels. In vitro experiments showed that PA, BA, and IsA exacerbated TNF-α-induced HepG2 apoptosis. Notably, the protective effects of SNS were compromised in pseudo-sterile mice. Discussion In conclusion, our experimental results suggest that the disturbance in intestinal flora results in elevated SCFA levels, which further exacerbates hepatocyte apoptosis in liver fibrosis, while SNS suppresses CCl4-induced liver fibrosis at least partially by reinstating intestinal flora homeostasis and reducing SCFA levels.
Collapse
Affiliation(s)
- Qiong Wu
- School of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Fangsi Zhu
- School of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Yu Yao
- School of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
- Department of Pharmacy, Anhui No. 2 Provincial People’s Hospital, Hefei, Anhui, China
| | - Luyun Chen
- School of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Yijie Ding
- School of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Yong Su
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Chaoliang Ge
- School of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
37
|
Gao XF, Ji BY, Zhang JJ, Wang Z, Jiang S, Hu JN, Gong XJ, Zhang JT, Tsopmejio ISN, Li W. Ginsenoside Rg2 Attenuates Aging-Induced Liver Injury via Inhibiting Caspase 8-Mediated Pyroptosis, Apoptosis and Modulating Gut Microbiota. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1427-1449. [PMID: 39192676 DOI: 10.1142/s0192415x24500563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Aging is an irresistible natural law of the progressive decline of body molecules, organs, and overall function with the passage of time, resulting in eventual death. World Health Organization data show that aging is correlated with a wide range of common chronic diseases in the elderly, and is an essential driver of many diseases. Panax Ginseng C.A Meyer is an ancient herbal medicine, which has an effect of "long service, light weight, and longevity" recorded in the ancient Chinese medicine book "Compendium of Materia Medica." Ginsenoside Rg2, the main active ingredient of ginseng, also exerts a marked effect on the treatment of liver injury. However, it remains unclear whether Rg2 has the potential to ameliorate aging-induced liver injury. Hence, exploring the hepatoprotective properties of Rg2 and its possible molecular mechanism by Senescence Accelerate Mouse Prone 8 (SAMP8) and gut microbiota. Our study demonstrated that Rg2 can inhibit pyroptosis and apoptosis through caspase 8, and regulate the gut-liver axis to alleviate liver inflammation by changing the composition of gut microbiota, thus improving aging-induced liver injury. These findings provide theoretical support for the pharmacological effects of ginsenosides in delaying aging-induced liver injury.
Collapse
Affiliation(s)
- Xu-Fei Gao
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Bao-Yu Ji
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, P. R. China
- Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Jun-Jie Zhang
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Zi Wang
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Shuang Jiang
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Jun-Nan Hu
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Xiao-Jie Gong
- Department of Biological Engineering, College of Life Science, Dalian Minzu University, Dalian 116600, P. R. China
| | - Jing-Tian Zhang
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Ivan Stève Nguepi Tsopmejio
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
- College of Life Sciences, Jilin Agricultural University, Changchun 130118, P. R. China
| |
Collapse
|
38
|
Zhang Y, Wang X, Lin J, Liu J, Wang K, Nie Q, Ye C, Sun L, Ma Y, Qu R, Mao Y, Zhang X, Lu H, Xia P, Zhao D, Wang G, Zhang Z, Fu W, Jiang C, Pang Y. A microbial metabolite inhibits the HIF-2α-ceramide pathway to mediate the beneficial effects of time-restricted feeding on MASH. Cell Metab 2024; 36:1823-1838.e6. [PMID: 39079531 DOI: 10.1016/j.cmet.2024.07.004] [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: 05/05/2023] [Revised: 01/24/2024] [Accepted: 07/05/2024] [Indexed: 08/10/2024]
Abstract
Time-restricted feeding (TRF) is a potent dietary intervention for improving metabolic diseases, including metabolic dysfunction-associated steatotic liver disease/metabolic dysfunction-associated steatohepatitis (MASLD/MASH). However, the mechanism of this efficacy has remained elusive. Here, we show that TRF improves MASLD, which is associated with a significant enrichment of Ruminococcus torques (R. torques). Mechanistically, R. torques suppresses the intestinal HIF-2α-ceramide pathway via the production of 2-hydroxy-4-methylpentanoic acid (HMP). We identify rtMor as a 4-methyl-2-oxopentanoate reductase that synthesizes HMP in R. torques. Finally, we show that either the colonization of R. torques or oral HMP supplementation can ameliorate inflammation and fibrosis in a MASH mouse model. These findings identify R. torques and HMP as potential TRF mimetics for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Yi Zhang
- Department of General Surgery, Cancer Center, Peking University Third Hospital, Beijing, China; Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China
| | - Xuemei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jun Lin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China; Department of Endocrinology, Capital Medical University Chaoyang Hospital, Beijing, China
| | - Jia Liu
- Department of Endocrinology, Capital Medical University Chaoyang Hospital, Beijing, China
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Qixing Nie
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chuan Ye
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Lulu Sun
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China; State Key Laboratory of Female Fertility Preservation, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Yanpeng Ma
- Department of General Surgery, Cancer Center, Peking University Third Hospital, Beijing, China; Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China
| | - Ruize Qu
- Department of General Surgery, Cancer Center, Peking University Third Hospital, Beijing, China; Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China
| | - Yuejian Mao
- Mengniu Institute of Nutrition Science, Shanghai, China
| | - Xuguang Zhang
- Mengniu Institute of Nutrition Science, Shanghai, China; Shanghai Institute of Nutrition and Health, The Chinese Academy of Sciences, Shanghai, China
| | - Hua Lu
- National Laboratory for Molecular Sciences, Center for Soft Matter Science and Engineering, Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Pengyan Xia
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Dongyu Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China; Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Guang Wang
- Department of Endocrinology, Capital Medical University Chaoyang Hospital, Beijing, China.
| | - Zhipeng Zhang
- Department of General Surgery, Cancer Center, Peking University Third Hospital, Beijing, China; Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China.
| | - Wei Fu
- Department of General Surgery, Cancer Center, Peking University Third Hospital, Beijing, China; Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China.
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China; Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China; Department of Immunology, School of Basic Medical Sciences, State Key Laboratory of Female Fertility Promotion, Peking University, Beijing, China; NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China.
| | - Yanli Pang
- State Key Laboratory of Female Fertility Preservation, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
39
|
Adolph TE, Tilg H. Western diets and chronic diseases. Nat Med 2024; 30:2133-2147. [PMID: 39085420 DOI: 10.1038/s41591-024-03165-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024]
Abstract
'Westernization', which incorporates industrial, cultural and dietary trends, has paralleled the rise of noncommunicable diseases across the globe. Today, the Western-style diet emerges as a key stimulus for gut microbial vulnerability, chronic inflammation and chronic diseases, affecting mainly the cardiovascular system, systemic metabolism and the gut. Here we review the diet of modern times and evaluate the threat it poses for human health by summarizing recent epidemiological, translational and clinical studies. We discuss the links between diet and disease in the context of obesity and type 2 diabetes, cardiovascular diseases, gut and liver diseases and solid malignancies. We collectively interpret the evidence and its limitations and discuss future challenges and strategies to overcome these. We argue that healthcare professionals and societies must react today to the detrimental effects of the Western diet to bring about sustainable change and improved outcomes in the future.
Collapse
Affiliation(s)
- Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
40
|
Duan Y, Yang Y, Zhao S, Bai Y, Yao W, Gao X, Yin J. Crosstalk in extrahepatic and hepatic system in NAFLD/NASH. Liver Int 2024; 44:1856-1871. [PMID: 38717072 DOI: 10.1111/liv.15967] [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: 01/09/2024] [Revised: 02/28/2024] [Accepted: 04/26/2024] [Indexed: 07/17/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged as the most prevalent chronic liver disease globally. Non-alcoholic steatohepatitis (NASH) represents an extremely progressive form of NAFLD, which, without timely intervention, may progress to cirrhosis or hepatocellular carcinoma. Presently, a definitive comprehension of the pathogenesis of NAFLD/NASH eludes us, and pharmacological interventions targeting NASH specifically remain constrained. The aetiology of NAFLD encompasses a myriad of external factors including environmental influences, dietary habits and gender disparities. More significantly, inter-organ and cellular interactions within the human body play a role in the development or regression of the disease. In this review, we categorize the influences affecting NAFLD both intra- and extrahepatically, elaborating meticulously on the mechanisms governing the onset and progression of NAFLD/NASH. This exploration delves into progress in aetiology and promising therapeutic targets. As a metabolic disorder, the development of NAFLD involves complexities related to nutrient metabolism, liver-gut axis interactions and insulin resistance, among other regulatory functions of extraneous organs. It further encompasses intra-hepatic interactions among hepatic cells, Kupffer cells (KCs) and hepatic stellate cells (HSCs). A comprehensive understanding of the pathogenesis of NAFLD/NASH from a macroscopic standpoint is instrumental in the formulation of future therapies for NASH.
Collapse
Affiliation(s)
- Yiliang Duan
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yan Yang
- The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Shuqiang Zhao
- Jiangsu Institute for Food and Drug Control, NMPA Key Laboratory for Impurity Profile of Chemical Drugs, Nanjing, Jiangsu, China
| | - Yuesong Bai
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wenbing Yao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xiangdong Gao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jun Yin
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
41
|
Ezenabor EH, Adeyemi AA, Adeyemi OS. Gut Microbiota and Metabolic Syndrome: Relationships and Opportunities for New Therapeutic Strategies. SCIENTIFICA 2024; 2024:4222083. [PMID: 39041052 PMCID: PMC11262881 DOI: 10.1155/2024/4222083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/10/2024] [Accepted: 07/04/2024] [Indexed: 07/24/2024]
Abstract
Since its discovery, numerous studies have shown the role of the microbiota in well-being and disease. The gut microbiota represents an essential factor that plays a multidirectional role that affects not just the gut but also other parts of the body, including the brain, endocrine system, humoral system, immune system, and metabolic pathways, as well as host-microbiome interactions. Through a comprehensive analysis of existing literature using the desktop research methodology, this review elucidates the mechanisms by which gut microbiota dysbiosis contributes to metabolic dysfunction, including obesity, dyslipidaemia, hypertension, atherosclerosis, hyperuricemia, and hyperglycaemia. Furthermore, it examines the bidirectional communication pathways between gut microbiota and host metabolism, highlighting the role of microbial-derived metabolites, immune modulation, and gut barrier integrity in shaping metabolic homeostasis. Importantly, the review identifies promising therapeutic strategies targeting the gut microbiota as potential interventions for metabolic syndrome, including probiotics, prebiotics, symbiotics, dietary modifications, and faecal microbiota transplantation. By delineating the bidirectional interactions between gut microbiota and metabolic syndrome, the review not only advances our understanding of disease pathophysiology but also underscores the potential for innovative microbiota-based interventions to mitigate the global burden of metabolic syndrome and its associated complications.
Collapse
Affiliation(s)
- Emmanuel Henry Ezenabor
- Department of BiochemistryMedicinal Biochemistry, Nanomedicine & Toxicology LaboratoryBowen University, Iwo 232102, Osun State, Nigeria
| | - Aishat Abimbola Adeyemi
- Department of BiochemistryMedicinal Biochemistry, Nanomedicine & Toxicology LaboratoryBowen University, Iwo 232102, Osun State, Nigeria
| | - Oluyomi Stephen Adeyemi
- Department of BiochemistryMedicinal Biochemistry, Nanomedicine & Toxicology LaboratoryBowen University, Iwo 232102, Osun State, Nigeria
| |
Collapse
|
42
|
Xu H, Luo Y, Li Q, Zhu H. Acupuncture influences multiple diseases by regulating gut microbiota. Front Cell Infect Microbiol 2024; 14:1371543. [PMID: 39040602 PMCID: PMC11260648 DOI: 10.3389/fcimb.2024.1371543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 06/13/2024] [Indexed: 07/24/2024] Open
Abstract
Acupuncture, an important green and side effect-free therapy in traditional Chinese medicine, is widely use both domestically and internationally. Acupuncture can interact with the gut microbiota and influence various diseases, including metabolic diseases, gastrointestinal diseases, mental disorders, nervous system diseases, and other diseases. This review presents a thorough analysis of these interactions and their impacts and examines the alterations in the gut microbiota and the potential clinical outcomes following acupuncture intervention to establish a basis for the future utilization of acupuncture in clinical treatments.
Collapse
Affiliation(s)
- Huimin Xu
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yingzhe Luo
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qiaoqi Li
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Zhu
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
43
|
Zyoud SH, Alalalmeh SO, Hegazi OE, Shakhshir M, Abushamma F, Al-Jabi SW. An examination of global research trends for exploring the associations between the gut microbiota and nonalcoholic fatty liver disease through bibliometric and visualization analysis. Gut Pathog 2024; 16:31. [PMID: 38961453 PMCID: PMC11223324 DOI: 10.1186/s13099-024-00624-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 06/28/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is increasingly recognized as a significant health issue. Emerging research has focused on the role of the gut microbiota in NAFLD, emphasizing the gut-liver axis. This study aimed to identify key research trends and guide future investigations in this evolving area. METHODS This bibliometric study utilized Scopus to analyze global research on the link between the gut microbiota and NAFLD. The method involved a search strategy focusing on relevant keywords in article titles, refined by including only peer-reviewed journal articles. The data analysis included bibliometric indicators such as publication counts and trends, which were visualized using VOSviewer software version 1.6.20 for network and co-occurrence analysis, highlighting key research clusters and emerging topics. RESULTS Among the 479 publications on the gut microbiota and NAFLD, the majority were original articles (n = 338; 70.56%), followed by reviews (n = 119; 24.84%). The annual publication count increased from 1 in 2010 to 118 in 2022, with a significant growth phase starting in 2017 (R2 = 0.9025, p < 0.001). The research was globally distributed and dominated by China (n = 231; 48.23%) and the United States (n = 90; 18.79%). The University of California, San Diego, led institutional contributions (n = 18; 3.76%). Funding was prominent, with 62.8% of the articles supported, especially by the National Natural Science Foundation of China (n = 118; 24.63%). The average citation count was 43.23, with an h-index of 70 and a citation range of 0 to 1058 per article. Research hotspots shifted their focus post-2020 toward the impact of high-fat diets on NAFLD incidence. CONCLUSIONS This study has effectively mapped the growing body of research on the gut microbiota-NAFLD relationship, revealing a significant increase in publications since 2017. There is significant interest in gut microbiota and NAFLD research, mainly led by China and the United States, with diverse areas of focus. Recently, the field has moved toward exploring the interconnections among diet, lifestyle, and the gut-liver axis. We hypothesize that with advanced technologies, new opportunities for personalized medicine and a holistic understanding of NAFLD will emerge.
Collapse
Affiliation(s)
- Sa'ed H Zyoud
- Poison Control and Drug Information Center (PCDIC), College of Medicine and Health Sciences, An-Najah National University, Nablus, 44839, Palestine.
- Department of Clinical and Community Pharmacy, College of Medicine and Health Sciences, An-Najah National University, Nablus, 44839, Palestine.
- Clinical Research Centre, An-Najah National University Hospital, Nablus, 44839, Palestine.
| | - Samer O Alalalmeh
- College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
| | - Omar E Hegazi
- College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
| | - Muna Shakhshir
- Department of Nutrition, An-Najah National University Hospital, Nablus, 44839, Palestine
| | - Faris Abushamma
- Department of Medicine, College of Medicine and Health Sciences, An-Najah National University, Nablus, 44839, Palestine
- Department of Urology, An-Najah National University Hospital, Nablus, 44839, Palestine
| | - Samah W Al-Jabi
- Department of Clinical and Community Pharmacy, College of Medicine and Health Sciences, An-Najah National University, Nablus, 44839, Palestine.
| |
Collapse
|
44
|
Long J, Xu Y, Zhang X, Wu B, Wang C. Role of FXR in the development of NAFLD and intervention strategies of small molecules. Arch Biochem Biophys 2024; 757:110024. [PMID: 38703803 DOI: 10.1016/j.abb.2024.110024] [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: 12/26/2023] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) remains a prevailing etiological agent behind hepatocyte diseases like chronic liver disease. The spectrum of processes involved in NAFLD stages includes hepatic steatosis, non-alcoholic fatty liver, and non-alcoholic steatohepatitis (NASH). Without intervention, the progression of NASH can further deteriorate into cirrhosis and ultimately, hepatocellular carcinoma. The cardinal features that characterize NAFLD are insulin resistance, lipogenesis, oxidative stress and inflammation, extracellular matrix deposition and fibrosis. Due to its complex pathogenesis, existing pharmaceutical agents fail to take a curative or ameliorative effect on NAFLD. Consequently, it is imperative to identify novel therapeutic targets and strategies for NAFLD, ideally to improve the aforementioned key features in patients. As an enterohepatic regulator of bile acid homeostasis, lipid metabolism, and inflammation, FarnesoidX receptor (FXR) is an important pharmacological target for the treatment of NAFLD. Manipulating FXR to regulate lipid metabolic signaling pathways is a potential mechanism to mitigate NAFLD. Therefore, elucidating the modulatory character of FXR in regulating lipid metabolism in NAFLD has the potential to yield groundbreaking perspectives for drug design. This review details recent advances in the regulation of lipid depletion in hepatocytes and investigates the pivotal function of FXR in the progress of NAFLD.
Collapse
Affiliation(s)
- Jiachan Long
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yuanhang Xu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xuerong Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Bingxing Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Caiyan Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
45
|
Ronen D, Rokach Y, Abedat S, Qadan A, Daana S, Amir O, Asleh R. Human Gut Microbiota in Cardiovascular Disease. Compr Physiol 2024; 14:5449-5490. [PMID: 39109979 DOI: 10.1002/cphy.c230012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The gut ecosystem, termed microbiota, is composed of bacteria, archaea, viruses, protozoa, and fungi and is estimated to outnumber human cells. Microbiota can affect the host by multiple mechanisms, including the synthesis of metabolites and toxins, modulating inflammation and interaction with other organisms. Advances in understanding commensal organisms' effect on human conditions have also elucidated the importance of this community for cardiovascular disease (CVD). This effect is driven by both direct CV effects and conditions known to increase CV risk, such as obesity, diabetes mellitus (DM), hypertension, and renal and liver diseases. Cardioactive metabolites, such as trimethylamine N -oxide (TMAO), short-chain fatty acids (SCFA), lipopolysaccharides, bile acids, and uremic toxins, can affect atherosclerosis, platelet activation, and inflammation, resulting in increased CV incidence. Interestingly, this interaction is bidirectional with microbiota affected by multiple host conditions including diet, bile acid secretion, and multiple diseases affecting the gut barrier. This interdependence makes manipulating microbiota an attractive option to reduce CV risk. Indeed, evolving data suggest that the benefits observed from low red meat and Mediterranean diet consumption can be explained, at least partially, by the changes that these diets may have on the gut microbiota. In this article, we depict the current epidemiological and mechanistic understanding of the role of microbiota and CVD. Finally, we discuss the potential therapeutic approaches aimed at manipulating gut microbiota to improve CV outcomes. © 2024 American Physiological Society. Compr Physiol 14:5449-5490, 2024.
Collapse
Affiliation(s)
- Daniel Ronen
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yair Rokach
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Suzan Abedat
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Abed Qadan
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Samar Daana
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Offer Amir
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rabea Asleh
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
46
|
Parthasarathy G, Malhi H, Bajaj JS. Therapeutic manipulation of the microbiome in liver disease. Hepatology 2024:01515467-990000000-00932. [PMID: 38922826 DOI: 10.1097/hep.0000000000000987] [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: 05/24/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024]
Abstract
Myriad associations between the microbiome and various facets of liver physiology and pathology have been described in the literature. Building on descriptive and correlative sequencing studies, metagenomic studies are expanding our collective understanding of the functional and mechanistic role of the microbiome as mediators of the gut-liver axis. Based on these mechanisms, the functional activity of the microbiome represents an attractive, tractable, and precision medicine therapeutic target in several liver diseases. Indeed, several therapeutics have been used in liver disease even before their description as a microbiome-dependent approach. To bring successful microbiome-targeted and microbiome-inspired therapies to the clinic, a comprehensive appreciation of the different approaches to influence, collaborate with, or engineer the gut microbiome to coopt a disease-relevant function of interest in the right patient is key. Herein, we describe the various levels at which the microbiome can be targeted-from prebiotics, probiotics, synbiotics, and antibiotics to microbiome reconstitution and precision microbiome engineering. Assimilating data from preclinical animal models, human studies as well as clinical trials, we describe the potential for and rationale behind studying such therapies across several liver diseases, including metabolic dysfunction-associated steatotic liver disease, alcohol-associated liver disease, cirrhosis, HE as well as liver cancer. Lastly, we discuss lessons learned from previous attempts at developing such therapies, the regulatory framework that needs to be navigated, and the challenges that remain.
Collapse
Affiliation(s)
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jasmohan S Bajaj
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University and Central Virginia Veterans Healthcare System, Richmond, Virginia, USA
| |
Collapse
|
47
|
Lara-Romero C, Romero-Gómez M. Treatment Options and Continuity of Care in Metabolic-associated Fatty Liver Disease: A Multidisciplinary Approach. Eur Cardiol 2024; 19:e06. [PMID: 38983581 PMCID: PMC11231815 DOI: 10.15420/ecr.2023.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 02/14/2024] [Indexed: 07/11/2024] Open
Abstract
The terms non-alcoholic fatty liver disease and non-alcoholic steatohepatitis have some limitations as they use exclusionary confounder terms and the use of potentially stigmatising language. Recently, a study with content experts and patients has been set to change this nomenclature. The term chosen to replace non-alcoholic fatty liver disease was metabolic dysfunction-associated steatotic liver disease (MASLD), which avoids stigmatising and helps improve awareness and patient identification. MASLD is the most common cause of chronic liver disease with an increasing prevalence, accounting for 25% of the global population. It is considered the hepatic manifestation of the metabolic syndrome with lifestyle playing a fundamental role in its physiopathology. Diet change and physical activity are the cornerstones of treatment, encompassing weight loss and healthier behaviours and a holistic approach. In Europe, there is no approved drug for MASLD to date and there is a substantial unmet medical need for effective treatments for patients with MASLD. This review not only provides an update on advances in evidence for nutrition and physical activity interventions but also explores the different therapeutic options that are being investigated and whose development focuses on the restitution of metabolic derangements and halting inflammatory and fibrogenic pathways.
Collapse
Affiliation(s)
- Carmen Lara-Romero
- Gastroenterology and Hepatology Department, Virgen del Rocío University Hospital Seville, Spain
- Clinical and Translational Research in Digestive Diseases, Institute of Biomedicine of Seville, University of Seville Seville, Spain
| | - Manuel Romero-Gómez
- Gastroenterology and Hepatology Department, Virgen del Rocío University Hospital Seville, Spain
- Clinical and Translational Research in Digestive Diseases, Institute of Biomedicine of Seville, University of Seville Seville, Spain
| |
Collapse
|
48
|
Wang P, Sun J, Zhao W, Wang D, Ma Y, Zhao Y, Wang Y, Zhao X. Tomato Pectin Ameliorated Hepatic Steatosis in High-Fat-Diet Mice by Modulating Gut Microbiota and Bile Acid Metabolism. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38856079 DOI: 10.1021/acs.jafc.4c01598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a worldwide public health issue. Changes in the gut microbiota structure and composition are closely related to host pathophysiology processes. Pectin is associated with several beneficial health effects. In the present study, we aimed at investigating the effect of tomato pectin (TP) on hepatic steatosis and exploring the underlying mechanisms by focusing on the regulation of the gut microbiota-bile acid axis. Our results showed that TP attenuated high-fat diet (HFD)-induced liver steatosis and inflammation. TP administration increased the diversity of gut microbiota, enhancing the abundance of beneficial bacteria and suppressing the abundance of harmful or conditional pathogenic bacteria. Further antibiotic-caused microbiome depletion confirmed that the anti-NAFLD activities of TP were dependent on the regulation of gut microbiota. Besides, TP intervention affected feces bile acid metabolism and caused significant changes in functional conjugated bile acids, which in turn inhibited the ileum FXR/FGF15 signaling, leading to stimulation of the hepatic bile acid (BA) production. Furthermore, TP treatment accelerated BA excretion, promoted BA transportation, inhibited BA reabsorption, and facilitated cholesterol efflux to relieve HFD-induced hyperlipidemia. These findings provide a potential dietary intervention strategy for TP against NAFLD via modulation of cross-talk between BAs and gut bacteria.
Collapse
Affiliation(s)
- Pan Wang
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing 100097, China
| | - Jing Sun
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing 100097, China
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China
| | - Wenting Zhao
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing 100097, China
| | - Dan Wang
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing 100097, China
| | - Yue Ma
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing 100097, China
| | - Yuanyuan Zhao
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing 100097, China
| | - Yubin Wang
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing 100097, China
| | - Xiaoyan Zhao
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing 100097, China
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China
| |
Collapse
|
49
|
Ghani I, An Y, Qiao Q, He S, Li Z. Polyphenols from Foxtail Millet Improve Non-Alcoholic Fatty Liver Disease by Regulating Intestinal Microbiome in Mice. Foods 2024; 13:1683. [PMID: 38890912 PMCID: PMC11172357 DOI: 10.3390/foods13111683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/18/2024] [Accepted: 05/19/2024] [Indexed: 06/20/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic hepatic manifestation of metabolic dysfunction for which effective interventions are lacking. The burden of NAFLD is increasing at an alarming rate. NAFLD is frequently associated with morbidities such as dyslipidemia, type 2 diabetes mellitus and obesity, etc. The current study explored the potential role of bound polyphenols from foxtail millet (BPIS) in treating mice with NAFLD induced by a high-fat diet (HFD). The results indicated the critical role of BPIS in treating NAFLD by effectively restoring the gut microbiota in C57BL/6 mice that received a high-fat diet (HFD) for 12 weeks. At the same time, 16S rRNA analysis demonstrated that BPIS remodeled the overall structure of the gut microbiota from fatty liver diseases towards that of normal counterparts, including ten phylum and twenty genus levels. Further study found that the expression of tight junction proteins was upregulated in the BPIS-treated group. This study provides new insights into the potential NAFLD protective effects induced by polyphenols of foxtail millet.
Collapse
Affiliation(s)
| | | | | | | | - Zhuoyu Li
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China; (I.G.); (Y.A.); (Q.Q.); (S.H.)
| |
Collapse
|
50
|
Zhang P, Li J, Miao Y, Zhao X, Zhu L, Yao J, Wan M, Tang W. Sheng-Jiang powder ameliorates NAFLD via regulating intestinal microbiota in mice. Front Microbiol 2024; 15:1387401. [PMID: 38860223 PMCID: PMC11163104 DOI: 10.3389/fmicb.2024.1387401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 05/13/2024] [Indexed: 06/12/2024] Open
Abstract
Background Intestinal microbiota have been demonstrated to be involved in the development of NAFLD, while the relationship between the severity of NAFLD and intestinal microbiota is still not fully elucidated. Sheng-Jiang Powder (SJP) showed exact efficacy in treating SFL and great potential in regulating intestinal microbiota, but the effects need to be further addressed in NASH and liver fibrosis. Objectives To investigate the differences in intestinal microbiota of NAFLD with different severity and the effect of SJP on liver damage and intestinal microbiota. Design NAFLD mice models with different severity were induced by high-fat diet (HFD) or choline-deficient, L-amino acid-defined high-fat diet (CDAHFD) feeding and then treated with SJP/normal saline. Methods Biochemical blood tests, H&E/Masson/Oil Red O/IHC staining, Western blot, and 16SrDNA sequencing were performed to explore intestinal microbiota alteration in different NAFLD models and the effect of SJP on liver damage and intestinal microbiota. Results Intestinal microbiota alteration was detected in all NAFLD mice. SJP induced increased expression of Pparγ and alleviated liver lipid deposition in all NAFLD mice. Microbiome analysis revealed obvious changes in intestinal microbiota composition, while SJP significantly elevated the relative abundance of Roseburia and Akkermansia, which were demonstrated to be beneficial for improving inflammation and intestinal barrier function. Conclusion Our results demonstrated that SJP was effective in improving lipid metabolism in NAFLD mice, especially in mice with SFL. The potential mechanism may be associated with the regulation of intestinal microbiota.
Collapse
Affiliation(s)
- Pengcheng Zhang
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, China
| | - Juan Li
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yifan Miao
- Department of Emergency Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xianlin Zhao
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lv Zhu
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaqi Yao
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Meihua Wan
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Wenfu Tang
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, China
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|