1
|
Wang Y, Li Y, Lin Y, Cao C, Chen D, Huang X, Li C, Xu H, Lai H, Chen H, Zhou Y. Roles of the gut microbiota in hepatocellular carcinoma: from the gut dysbiosis to the intratumoral microbiota. Cell Death Discov 2025; 11:140. [PMID: 40185720 PMCID: PMC11971373 DOI: 10.1038/s41420-025-02413-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/23/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is closely linked to alterations in the gut microbiota. This dysbiosis is characterized by significant changes in the microbial population, which correlate with the progression of HCC. Gut dysbiosis ultimately promotes HCC development in several ways: it damages the integrity of the gut-vascular barrier (GVB), alters the tumor microenvironment (TME), and even affects the intratumoral microbiota. Subsequently, intratumoral microbiota present a characteristic profile and play an essential role in HCC progression mainly by causing DNA damage, mediating tumor-related signaling pathways, altering the TME, promoting HCC metastasis, or through other mechanisms. Both gut microbiota and intratumoral microbiota have dual effects on HCC progression; a comprehensive understanding of their complex biological roles will provide a theoretical foundation for potential clinical applications in HCC treatment.
Collapse
Affiliation(s)
- Yiqin Wang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yongqiang Li
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yong Lin
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Chuangyu Cao
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Dongcheng Chen
- Department of Gastroenterology and Hepatology, Baiyun Hospital of Guangzhou First People's Hospital (The Second People's Hospital of Baiyun District), Guangzhou, China
| | - Xianguang Huang
- Department of Gastroenterology and Hepatology, Baiyun Hospital of Guangzhou First People's Hospital (The Second People's Hospital of Baiyun District), Guangzhou, China
| | - Canhua Li
- Department of Gastroenterology and Hepatology, Baiyun Hospital of Guangzhou First People's Hospital (The Second People's Hospital of Baiyun District), Guangzhou, China
| | - Haoming Xu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Huasheng Lai
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Huiting Chen
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
| | - Yongjian Zhou
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
| |
Collapse
|
2
|
Meadows V, Antonio JM, Ferraris RP, Gao N. Ruminococcus gnavus in the gut: driver, contributor, or innocent bystander in steatotic liver disease? FEBS J 2025; 292:1252-1264. [PMID: 39589934 PMCID: PMC11927045 DOI: 10.1111/febs.17327] [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: 03/07/2024] [Revised: 07/29/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024]
Abstract
The human gut microbiome plays a crucial role in regulating intestinal and systemic health, impacting host immune response and metabolic function. Dysbiosis of the gut microbiome is linked to various diseases, including steatotic liver diseases. Metabolic dysfunction-associated steatotic liver disease (MASLD), a chronic liver disease characterized by excess hepatic lipid content and impaired metabolism, is the leading cause of liver disease worldwide. Among the gut microbes, Ruminococcus gnavus (R. gnavus) has garnered attention for its association with inflammatory and metabolic diseases. While R. gnavus abundance correlates to liver fat accumulation, further research is needed to identify a causal role or therapeutic intervention in steatotic liver disease. This review surveys our current understanding of R. gnavus in the development and progression of steatotic liver diseases, highlighting its potential mechanisms through metabolite secretion, and emphasizes the need for comprehensive microbiome analyses and longitudinal studies to better understand R. gnavus' impact on liver health. This knowledge could pave the way for targeted interventions aimed at modulating gut microbiota to treat and prevent MASLD and its comorbidities.
Collapse
Affiliation(s)
- Vik Meadows
- Department of Biological Sciences, School of Arts & SciencesRutgers UniversityNewarkNJUSA
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical SchoolRutgers UniversityNewarkNJUSA
| | - Jayson M. Antonio
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical SchoolRutgers UniversityNewarkNJUSA
| | - Ronaldo P. Ferraris
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical SchoolRutgers UniversityNewarkNJUSA
| | - Nan Gao
- Department of Biological Sciences, School of Arts & SciencesRutgers UniversityNewarkNJUSA
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical SchoolRutgers UniversityNewarkNJUSA
| |
Collapse
|
3
|
Shen X, Zhang X, Li K, Huang G, Li X, Hou Y, Ge X. Combined bacterial translocation and cholestasis aggravates liver injury by activation pyroptosis in obstructive jaundice. Heliyon 2024; 10:e35793. [PMID: 39220957 PMCID: PMC11363856 DOI: 10.1016/j.heliyon.2024.e35793] [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: 02/02/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
This study explores the mechanism by which obstructive jaundice (OJ) induces liver damage through pyroptosis. We induced OJ in rats via bile duct ligation and assessed liver damage using serum biochemical markers and histological analysis of liver tissue. Pyroptosis was investigated through immunofluorescence, ELISA, Western blot, and quantitative RT-PCR techniques. Additionally, we examined intestinal function and fecal microbiota alterations in the rats using 16S rDNA sequencing. In vitro experiments involved co-culturing Kupffer cells and hepatocytes, which were then exposed to bile and lipopolysaccharide (LPS). Our findings indicated that OJ modified the gut microbiota, increasing LPS levels, which, in conjunction with bile, initiated a cycle of inflammation, fibrosis, and cell death in the liver. Mechanistically, OJ elevated necrotic markers such as ATP, which in turn activated pyroptotic pathways. Increased levels of pyroptosis-related molecules, including NLRP3, caspase-1, gasdermin D, and IL-18, were confirmed. In our co-cultured cell model, bile exposure resulted in cell death and ATP release, leading to the activation of the NLRP3 inflammasome and its downstream effectors, caspase-1 and IL-18. The combination of bile and LPS significantly intensified pyroptotic responses. This study is the first to demonstrate that LPS and bile synergistically exacerbate liver injury by promoting necrosis and pyroptosis, unveiling a novel mechanism of OJ-associated hepatic damage and suggesting avenues for potential preventive or therapeutic interventions.
Collapse
Affiliation(s)
- Xin Shen
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xin Zhang
- Luoyang Orthopedic-Traumatological Hospital of Henan Province (Henan Provincial Orthopedic Hospital), Luoyang, 471002, Henan, China
| | - Kaiyu Li
- Department of General Surgery, Heilongjiang Provincial Hospital, 82 Zhongshan Road, Harbin, 150036, Heilongjiang, China
| | - Guangming Huang
- Department of General Surgery, Heilongjiang Provincial Hospital, 82 Zhongshan Road, Harbin, 150036, Heilongjiang, China
| | - Xinyu Li
- Department of General Surgery, Heilongjiang Provincial Hospital, 82 Zhongshan Road, Harbin, 150036, Heilongjiang, China
| | - Yunlong Hou
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine, Shijiazhuang, 050000, Hebei, China
| | - Xin Ge
- Department of General Surgery, Heilongjiang Provincial Hospital, 82 Zhongshan Road, Harbin, 150036, Heilongjiang, China
| |
Collapse
|
4
|
Zhang Y, Wang W. Probiotics in reducing obesity by reconfiguring the gut microbiota. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2024; 49:1042-1051. [PMID: 39788492 PMCID: PMC11495983 DOI: 10.11817/j.issn.1672-7347.2024.240361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Indexed: 01/05/2025]
Abstract
Obesity, as a global health crisis, is increasingly linked to intestinal microecology. Probiotics colonise the body, effectively regulating the balance of intestinal flora, while strengthening the intestinal barrier, activating the immune response, releasing beneficial substances, and maintaining micro-ecological balance. This process not only enhances the defence against pathogens, but also reduces the production of inflammatory factors and lowers the level of chronic inflammation. However, the specific process and mechanism by which probiotics influence the intestinal microecology through the immune response, improve metabolic disorders caused by obesity, and participate in weight management are not clear. Through multiple neural pathways including the 'gut-brain axis' and their direct interaction with the intestine, probiotics increase the number of beneficial bacteria in the intestine and inhibit the growth of harmful bacteria, thus effectively restructuring the balance of the intestinal flora. This restructuring of the balance can optimise the intestinal environment and enhance the efficiency of food digestion and nutrient absorption. Probiotics show positive effects on obesity management by regulating the metabolic process and reducing fat accumulation, providing individuals with a new way to control body weight and prevent obesity. Therefore, the application of probiotics is of great significance in promoting gut health and weight management.
Collapse
Affiliation(s)
- Yunuo Zhang
- Department of Endocrinology, First Affiliated Hospital of Baotou Medical College, Baotou Inner Mongolia Autonomous Region 014010, China.
| | - Wei Wang
- Department of Endocrinology, First Affiliated Hospital of Baotou Medical College, Baotou Inner Mongolia Autonomous Region 014010, China.
| |
Collapse
|
5
|
Yang X, Yao S, Jiang Q, Chen H, Liu S, Shen G, Xiang X, Chen L. Exploring the Regulatory Effect of Tegillarca granosa Polysaccharide on High-Fat Diet-Induced Non-Alcoholic Fatty Liver Disease in Mice Based on Intestinal Flora. Mol Nutr Food Res 2024; 68:e2300453. [PMID: 38389187 DOI: 10.1002/mnfr.202300453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/21/2023] [Indexed: 02/24/2024]
Abstract
To explore the potential mechanism of action of Tegillarca granosa polysaccharide (TGP) in treating nonalcoholic fatty liver disease (NAFLD), the study conducts in vivo experiments using male C57BL/6 mice fed a high-fat diet while administering TGP for 16 weeks. The study measures body weight, liver weight, serum biochemical markers, pathological histology, liver lipid accumulation, oxidative stress and inflammation-related factors, lipid synthesis and metabolism-related gene and protein expression, and the composition and abundance of intestinal flora. Additionally, short-chain fatty acid (SCFAs) content and the correlation between intestinal flora and environmental factors are measured. The results show that TGP effectively reduces excessive hepatic lipid accumulation, dyslipidemia, abnormal liver function, and steatosis in the mice with NAFLD. Moreover, TGP effectively regulates intestinal flora disorder, increases the diversity of intestinal flora, and affects the relative abundance of specific bacteria while also increasing the content of SCFAs. These findings provide a basis for exploring the regulatory effect of T. granosa polysaccharide on NAFLD based on intestinal flora and highlight its potential as a natural liver nutraceutical.
Collapse
Affiliation(s)
- Xingwen Yang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Hangzhou, 310014, China
- National R&D Branch Center for Pelagic Aquatic Products Processing (Hangzhou), Hangzhou, 310014, China
| | - Shiwei Yao
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Hangzhou, 310014, China
- National R&D Branch Center for Pelagic Aquatic Products Processing (Hangzhou), Hangzhou, 310014, China
| | - Qihong Jiang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Hangzhou, 310014, China
- National R&D Branch Center for Pelagic Aquatic Products Processing (Hangzhou), Hangzhou, 310014, China
| | - Hui Chen
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Hangzhou, 310014, China
- National R&D Branch Center for Pelagic Aquatic Products Processing (Hangzhou), Hangzhou, 310014, China
| | - Shulai Liu
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Hangzhou, 310014, China
- National R&D Branch Center for Pelagic Aquatic Products Processing (Hangzhou), Hangzhou, 310014, China
| | - Guoxin Shen
- Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, 310021, China
| | - Xingwei Xiang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Hangzhou, 310014, China
- National R&D Branch Center for Pelagic Aquatic Products Processing (Hangzhou), Hangzhou, 310014, China
| | - Lin Chen
- Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, 310021, China
| |
Collapse
|
6
|
Satthawiwat N, Jinato T, Sutheeworapong S, Tanpowpong N, Chuaypen N, Tangkijvanich P. Distinct Gut Microbial Signature and Host Genetic Variants in Association with Liver Fibrosis Severity in Patients with MASLD. Nutrients 2024; 16:1800. [PMID: 38931155 PMCID: PMC11206871 DOI: 10.3390/nu16121800] [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: 03/20/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Gut microbiota might affect the severity and progression of metabolic dysfunction-associated steatotic liver disease (MASLD). We aimed to characterize gut dysbiosis and clinical parameters regarding fibrosis stages assessed by magnetic resonance elastography. This study included 156 patients with MASLD, stratified into no/mild fibrosis (F0-F1) and moderate/severe fibrosis (F2-F4). Fecal specimens were sequenced targeting the V4 region of the 16S rRNA gene and analyzed using bioinformatics. The genotyping of PNPLA3, TM6SF2, and HSD17B13 was assessed by allelic discrimination assays. Our data showed that gut microbial profiles between groups significantly differed in beta-diversity but not in alpha-diversity indices. Enriched Fusobacterium and Escherichia_Shigella, and depleted Lachnospira were found in the F2-F4 group versus the F0-F1 group. Compared to F0-F1, the F2-F4 group had elevated plasma surrogate markers of gut epithelial permeability and bacterial translocation. The bacterial genera, PNPLA3 polymorphisms, old age, and diabetes were independently associated with advanced fibrosis in multivariable analyses. Using the Random Forest classifier, the gut microbial signature of three genera could differentiate the groups with high diagnostic accuracy (AUC of 0.93). These results indicated that the imbalance of enriched pathogenic genera and decreased beneficial bacteria, in association with several clinical and genetic factors, were potential contributors to the pathogenesis and progression of MASLD.
Collapse
Affiliation(s)
- Nantawat Satthawiwat
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.S.); (T.J.); (N.C.)
- Doctor of Philosophy Program in Medical Biochemistry, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Thananya Jinato
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.S.); (T.J.); (N.C.)
| | - Sawannee Sutheeworapong
- Systems Biology and Bioinformatics Research Unit, Pilot Plant Development and Training Institute, King Mongkut’s University of Technology Thonburi, Bangkok 10150, Thailand;
| | - Natthaporn Tanpowpong
- Department of Radiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Natthaya Chuaypen
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.S.); (T.J.); (N.C.)
| | - Pisit Tangkijvanich
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (N.S.); (T.J.); (N.C.)
| |
Collapse
|
7
|
Wu T, Zeng Z, Yu Y. Role of Probiotics in Gut Microbiome and Metabolome in Non-Alcoholic Fatty Liver Disease Mouse Model: A Comparative Study. Microorganisms 2024; 12:1020. [PMID: 38792849 PMCID: PMC11124503 DOI: 10.3390/microorganisms12051020] [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: 04/11/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver condition worldwide. Numerous studies conducted recently have demonstrated a connection between the dysbiosis of the development of NAFLD and gut microbiota. Rebuilding a healthy gut ecology has been proposed as a strategy involving the use of probiotics. The purpose of this work is to investigate and compare the function of probiotics Akkermansia muciniphila (A. muciniphila) and VSL#3 in NAFLD mice. Rodent NAFLD was modeled using a methionine choline-deficient diet (MCD) with/without oral probiotic delivery. Subsequently, qPCR, histological staining, and liver function tests were conducted. Mass spectrometry-based analysis and 16S rDNA gene sequencing were used to investigate the liver metabolome and gut microbiota. We found that while both A. muciniphila and VSL#3 reduced hepatic fat content, A. muciniphila outperformed VSL#3. Furthermore, probiotic treatment restored the β diversity of the gut flora and A. muciniphila decreased the abundance of pathogenic bacteria such as Ileibacterium valens. These probiotics altered the metabolism in MCD mice, especially the glycerophospholipid metabolism. In conclusion, our findings distinguished the role of A. muciniphila and VSL#3 in NAFLD and indicated that oral-gavage probiotics remodel gut microbiota and improve metabolism, raising the possibility of using probiotics in the cure of NAFLD.
Collapse
Affiliation(s)
| | - Zheng Zeng
- Department of Infectious Diseases, Peking University First Hospital, Beijing 100034, China;
| | - Yanyan Yu
- Department of Infectious Diseases, Peking University First Hospital, Beijing 100034, China;
| |
Collapse
|
8
|
Ghazanfar H, Javed N, Qasim A, Zacharia GS, Ghazanfar A, Jyala A, Shehi E, Patel H. Metabolic Dysfunction-Associated Steatohepatitis and Progression to Hepatocellular Carcinoma: A Literature Review. Cancers (Basel) 2024; 16:1214. [PMID: 38539547 PMCID: PMC10969013 DOI: 10.3390/cancers16061214] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 03/12/2024] [Accepted: 03/19/2024] [Indexed: 11/26/2024] Open
Abstract
The prevalence of metabolic-associated fatty liver disease (MAFLD) is increasing globally due to factors such as urbanization, obesity, poor nutrition, sedentary lifestyles, healthcare accessibility, diagnostic advancements, and genetic influences. Research on MAFLD and HCC risk factors, pathogenesis, and biomarkers has been conducted through a narrative review of relevant studies, with a focus on PubMed and Web of Science databases and exclusion criteria based on article availability and language. Steatosis marks the early stage of MASH advancement, commonly associated with factors of metabolic syndrome such as obesity and type 2 diabetes. Various mechanisms, including heightened lipolysis, hepatic lipogenesis, and consumption of high-calorie diets, contribute to the accumulation of lipids in the liver. Insulin resistance is pivotal in the development of steatosis, as it leads to the release of free fatty acids from adipose tissue. Natural compounds hold promise in regulating lipid metabolism and inflammation to combat these conditions. Liver fibrosis serves as a significant predictor of MASH progression and HCC development, underscoring the need to target fibrosis in treatment approaches. Risk factors for MASH-associated HCC encompass advanced liver fibrosis, older age, male gender, metabolic syndrome, genetic predispositions, and dietary habits, emphasizing the requirement for efficient surveillance and diagnostic measures. Considering these factors, it is important for further studies to determine the biochemical impact of these risk factors in order to establish targeted therapies that can prevent the development of HCC or reduce progression of MASH, indirectly decreasing the risk of HCC.
Collapse
Affiliation(s)
- Haider Ghazanfar
- Division of Gastroenterology, Department of Internal Medicine, BronxCare Health System, Bronx, NY 10457, USA (A.J.); (E.S.)
| | - Nismat Javed
- Department of Internal Medicine, BronxCare Health System, Bronx, NY 10457, USA (G.S.Z.)
| | - Abeer Qasim
- Department of Internal Medicine, BronxCare Health System, Bronx, NY 10457, USA (G.S.Z.)
| | - George Sarin Zacharia
- Department of Internal Medicine, BronxCare Health System, Bronx, NY 10457, USA (G.S.Z.)
| | - Ali Ghazanfar
- Department of Internal Medicine, Fauji Foundation Hospital, Rawalpindi 45000, Pakistan
| | - Abhilasha Jyala
- Division of Gastroenterology, Department of Internal Medicine, BronxCare Health System, Bronx, NY 10457, USA (A.J.); (E.S.)
| | - Elona Shehi
- Division of Gastroenterology, Department of Internal Medicine, BronxCare Health System, Bronx, NY 10457, USA (A.J.); (E.S.)
| | - Harish Patel
- Division of Gastroenterology, Department of Internal Medicine, BronxCare Health System, Bronx, NY 10457, USA (A.J.); (E.S.)
| |
Collapse
|
9
|
Yi YS. Pharmacological potential of ginseng and ginsenosides in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. J Ginseng Res 2024; 48:122-128. [PMID: 38465218 PMCID: PMC10920004 DOI: 10.1016/j.jgr.2023.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/01/2023] [Accepted: 11/10/2023] [Indexed: 03/12/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic liver disease characterized by hepatic fat accumulation, while nonalcoholic steatohepatitis (NASH) is an advanced form of NAFLD characterized by hepatic inflammation, fibrosis, and liver injury, resulting in liver cirrhosis and hepatocellular carcinoma (HCC). Given the evidence that ginseng and its major bioactive components, ginsenosides, have potent anti-adipogenic, anti-inflammatory, anti-oxidative, and anti-fibrogenic effects, the pharmacological effect of ginseng and ginsenosides on NAFLD and NASH is noteworthy. Furthermore, numerous studies have successfully demonstrated the protective effect of ginseng on these diseases, as well as the underlying mechanisms in animal disease models and cells, such as hepatocytes and macrophages. This review discusses recent studies that explore the pharmacological roles of ginseng and ginsenosides in NAFLD and NASH and highlights their potential as agents to prevent and treat NAFLD, NASH, and liver diseases caused by hepatic steatosis and inflammation.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, 154-42 Gwanggyosan-ro, Yeongtong-gu, Suwon, Republic of Korea
| |
Collapse
|
10
|
Qiu XX, Cheng SL, Liu YH, Li Y, Zhang R, Li NN, Li Z. Fecal microbiota transplantation for treatment of non-alcoholic fatty liver disease: Mechanism, clinical evidence, and prospect. World J Gastroenterol 2024; 30:833-842. [PMID: 38516241 PMCID: PMC10950639 DOI: 10.3748/wjg.v30.i8.833] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/08/2024] [Accepted: 01/23/2024] [Indexed: 02/26/2024] Open
Abstract
The population of non-alcoholic fatty liver disease (NAFLD) patients along with relevant advanced liver disease is projected to continue growing, because currently no medications are approved for treatment. Fecal microbiota transplantation (FMT) is believed a novel and promising therapeutic approach based on the concept of the gut-liver axis in liver disease. There has been an increase in the number of pre-clinical and clinical studies evaluating FMT in NAFLD treatment, however, existing findings diverge on its effects. Herein, we briefly summarized the mechanism of FMT for NAFLD treatment, reviewed randomized controlled trials for evaluating its efficacy in NAFLD, and proposed the prospect of future trials on FMT.
Collapse
Affiliation(s)
- Xiao-Xia Qiu
- Research and Education Department, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou 310022, Zhejiang Province, China
| | - Sheng-Li Cheng
- Anhui Provincial Hospital, The First Affiliated Hospital of University of Science and Technology of China, Hefei 230000, Anhui Province, China
| | - Yan-Hui Liu
- Department of Clinical Pharmacy, Anhui Provincial Children’s Hospital, Hefei 230000, Anhui Province, China
| | - Yu Li
- Department of Pharmacy, Taihe County People’s Hospital of Anhui Province, Fuyang 236600, Anhui Province, China
| | - Rui Zhang
- Department of Pharmacy, The Second People’s Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei 230000, Anhui Province, China
| | - Nan-Nan Li
- University of Science and Technology of China, The First Affiliated Hospital of University of Science and Technology of China, Hefei 230001, Anhui Province, China
| | - Zheng Li
- Jiangsu Engineering Research Center of Cardiovascular Drugs Targeting Endothelial Cells, College of Health Sciences, School of Life Sciences, Jiangsu Normal University, Xuzhou 221000, Jiangsu Province, China
| |
Collapse
|
11
|
Gruzdev SK, Podoprigora IV, Gizinger OA. Immunology of gut microbiome and liver in non-alcoholic fatty liver disease (NAFLD): mechanisms, bacteria, and novel therapeutic targets. Arch Microbiol 2024; 206:62. [PMID: 38216746 DOI: 10.1007/s00203-023-03752-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 01/14/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the world. Most important contributors to its development are diet and obesity. Gut microbiome's importance for immune system and inflammatory pathways more widely accepted as an important component in NAFLD and other liver diseases' pathogenesis. In this article we review potential mechanisms of microbiome alteration of local and systemic immune responses leading to NAFLD's development, and how can modulate them for the treatment. Our review mentions different immune system pathways and microorganisms regulating metabolism, liver inflammation and fibrosis. We specifically point out TLR-4 as a potential key immune pathway activated by bacterial lipopolysaccharides producing pro-inflammatory cytokines in NAFLD. Also, we discuss three endotoxin-producing strains (Enterobacter cloacae B29, Escherichia coli PY102, Klebsiella pneumoniae A7) that can promote NAFLD development via TLR4-dependent immune response activation in animal models and how they potentially contribute to disease progression in humans. Additionally, we discuss their other immune and non-immune mechanisms contributing to NAFLD pathogenesis. In the end we point out gut microbiome researches' future perspective in NAFLD as a potential new target for both diagnostic and treatment.
Collapse
Affiliation(s)
- Stanislav Konstantinovich Gruzdev
- Department of Microbiology V.S. Kiktenko, Medical Institute, Peoples' Friendship University of Russia, Miklukho-Maklaya Str. 6, Moscow, 117198, Russia.
| | - Irina Viktorovna Podoprigora
- Department of Microbiology V.S. Kiktenko, Medical Institute, Peoples' Friendship University of Russia, Miklukho-Maklaya Str. 6, Moscow, 117198, Russia
| | - Oksana Anatolievna Gizinger
- Department of Microbiology V.S. Kiktenko, Medical Institute, Peoples' Friendship University of Russia, Miklukho-Maklaya Str. 6, Moscow, 117198, Russia
| |
Collapse
|
12
|
Toshida K, Itoh S, Kosai‐Fujimoto Y, Ishikawa T, Nakayama Y, Tsutsui Y, Iseda N, Izumi T, Bekki Y, Yoshiya S, Toshima T, Nakamuta M, Yoshizumi T. Association of gut microbiota with portal vein pressure in patients with liver cirrhosis undergoing living donor liver transplantation. JGH Open 2023; 7:982-989. [PMID: 38162858 PMCID: PMC10757484 DOI: 10.1002/jgh3.13018] [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: 09/28/2023] [Revised: 11/12/2023] [Accepted: 11/22/2023] [Indexed: 01/03/2024]
Abstract
Background and Aim Many recent studies have shown a relationship between various systemic diseases and the gut microbiota (GM), with the gut-liver axis receiving particular attention. In contrast, no report has comprehensively shown the effects of GM on the pathophysiology of patients undergoing living donor liver transplantation (LDLT). Method We enrolled 16 recipients who underwent LDLT for liver cirrhosis, and 17 donors constituted the reference group. We examined the differences in GM between recipients and donors. We also examined the relationships between GM, short-chain fatty acids, and portal vein pressure (PVP) in recipients. Results There was no significant difference in alpha-diversity between the recipients and donors, but there was variation in beta-diversity among the recipients. The abundance of the phylum Bacteroidetes was significantly higher in recipients than in donors (P = 0.016), and it was positively correlated with PVP (r = 0.511, P = 0.043). Propionic acid, which is a component of short-chain fatty acids, was positively correlated with PVP (r = 0.544, P = 0.0295), the phylum Bacteroidetes (r = 0.677, P = 0.004), and total bilirubin concentration (r = 0.501, P = 0.048). Propionic acid was negatively correlated with serum albumin concentration (r = -0.482, P = 0.043). Conclusion Our findings suggest relationships between fecal Bacteroidetes levels, propionic acid concentrations, and PVP in patients with liver cirrhosis undergoing LDLT.
Collapse
Affiliation(s)
- Katsuya Toshida
- Department of Surgery and Science, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Shinji Itoh
- Department of Surgery and Science, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yukiko Kosai‐Fujimoto
- Department of Surgery and Science, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Takuma Ishikawa
- Department of Surgery and Science, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yuki Nakayama
- Department of Surgery and Science, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yuriko Tsutsui
- Department of Surgery and Science, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Norifumi Iseda
- Department of Surgery and Science, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Takuma Izumi
- Department of Surgery and Science, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yuki Bekki
- Department of Surgery and Science, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Shohei Yoshiya
- Department of Surgery and Science, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Takeo Toshima
- Department of Surgery and Science, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Makoto Nakamuta
- Department of Gastroenterology, Kyushu Medical CenterNational Hospital OrganizationFukuokaJapan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| |
Collapse
|
13
|
Crane H, Gofton C, Sharma A, George J. MAFLD: an optimal framework for understanding liver cancer phenotypes. J Gastroenterol 2023; 58:947-964. [PMID: 37470858 PMCID: PMC10522746 DOI: 10.1007/s00535-023-02021-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 07/05/2023] [Indexed: 07/21/2023]
Abstract
Hepatocellular carcinoma has a substantial global mortality burden which is rising despite advancements in tackling the traditional viral risk factors. Metabolic (dysfunction) associated fatty liver disease (MAFLD) is the most prevalent liver disease, increasing in parallel with the epidemics of obesity, diabetes and systemic metabolic dysregulation. MAFLD is a major factor behind this sustained rise in HCC incidence, both as a single disease entity and often via synergistic interactions with other liver diseases. Mechanisms behind MAFLD-related HCC are complex but is crucially underpinned by systemic metabolic dysregulation with variable contributions from interacting disease modifiers related to environment, genetics, dysbiosis and immune dysregulation. MAFLD-related HCC has a distinct clinical presentation, most notably its common occurrence in non-cirrhotic liver disease. This is just one of several major challenges to effective surveillance programmes. The response of MAFLD-related HCC to immune-checkpoint therapy is currently controversial, and is further complicated by the high prevalence of MAFLD in individuals with HCC from viral aetiologies. In this review, we highlight the current data on epidemiology, clinical characteristics, outcomes and screening controversies. In addition, concepts that have arisen because of the MAFLD paradigm such as HCC in MAFLD/NAFLD non-overlapping groups, dual aetiology tumours and MAFLD sub-phenotypes is reviewed.
Collapse
Affiliation(s)
- Harry Crane
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia.
- Department of Gastroenterology and Hepatology, Royal North Shore Hospital, 1 Reserve Road, St Leonards, New South Wales, Australia.
| | - Cameron Gofton
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia
- Department of Gastroenterology and Hepatology, Royal North Shore Hospital, 1 Reserve Road, St Leonards, New South Wales, Australia
| | - Ankur Sharma
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, 6 Verdun Street, Nedlands, Perth, WA, 6009, Australia
- Curtin Medical School, Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth, WA, 6102, Australia
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
14
|
Nonalcoholic steatohepatitis-related hepatocellular carcinoma: pathogenesis and treatment. Nat Rev Gastroenterol Hepatol 2023:10.1038/s41575-023-00754-7. [PMID: 36932227 DOI: 10.1038/s41575-023-00754-7] [Citation(s) in RCA: 131] [Impact Index Per Article: 65.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/03/2023] [Indexed: 03/19/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD), including its more severe manifestation, nonalcoholic steatohepatitis (NASH), has a global prevalence of 20-25% and is a major public health problem. Its incidence is increasing in parallel to the rise in obesity, diabetes and metabolic syndrome. Progression from NASH to NASH-related hepatocellular carcinoma (HCC) (~2% of cases per year) is influenced by many factors, including the tissue and immune microenvironment, germline mutations in PNPLA3, and the microbiome. NASH-HCC has unique molecular and immune traits compared with other aetiologies of HCC and is equally prevalent in men and women. Comorbidities associated with NASH, such as obesity and diabetes mellitus, can prevent the implementation of potentially curative therapies in certain patients; nonetheless, outcomes are similar in patients who receive treatment. NASH-HCC at the early to intermediate stages is managed with surgery and locoregional therapies, whereas advanced HCC is treated with systemic therapies, including anti-angiogenic therapies and immune-checkpoint inhibitors. In this Review, we present the latest knowledge of the pathogenic mechanisms and clinical management of NASH-HCC. We discuss data highlighting the controversy over varying responses to immune-checkpoint inhibitors according to underlying aetiology and suggest that the future of NASH-HCC management lies in improved surveillance, targeted combination therapies to overcome immune evasion, and identifying biomarkers to recognize treatment responders.
Collapse
|
15
|
Saldarriaga OA, Krishnan S, Wanninger TG, Oneka M, Rao A, Bao D, Arroyave E, Gosnell J, Kueht M, Moghe A, Millian D, Jiao J, Sanchez JI, Spratt H, Beretta L, Stevenson HL. Patients with fibrosis from non-alcoholic steatohepatitis have heterogeneous intrahepatic macrophages and therapeutic targets. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.16.23285924. [PMID: 36865099 PMCID: PMC9980226 DOI: 10.1101/2023.02.16.23285924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Background and Aims In clinical trials for reducing fibrosis in NASH patients, therapeutics that target macrophages have had variable results. We evaluated intrahepatic macrophages in patients with non-alcoholic steatohepatitis to determine if fibrosis influenced phenotypes and expression of CCR2 and Galectin-3. Approach & Results We used nCounter to analyze liver biopsies from well-matched patients with minimal (n=12) or advanced (n=12) fibrosis to determine which macrophage-related genes would be significantly different. Known therapy targets (e.g., CCR2 and Galectin-3) were significantly increased in patients with cirrhosis.However, several genes (e.g., CD68, CD16, and CD14) did not show significant differences, and CD163, a marker of pro-fibrotic macrophages was significantly decreased with cirrhosis. Next, we analyzed patients with minimal (n=6) or advanced fibrosis (n=5) using approaches that preserved hepatic architecture by multiplex-staining with anti-CD68, Mac387, CD163, CD14, and CD16. Spectral data were analyzed using deep learning/artificial intelligence to determine percentages and spatial relationships. This approach showed patients with advanced fibrosis had increased CD68+, CD16+, Mac387+, CD163+, and CD16+CD163+ populations. Interaction of CD68+ and Mac387+ populations was significantly increased in patients with cirrhosis and enrichment of these same phenotypes in individuals with minimal fibrosis correlated with poor outcomes. Evaluation of a final set of patients (n=4) also showed heterogenous expression of CD163, CCR2, Galectin-3, and Mac387, and significant differences were not dependent on fibrosis stage or NAFLD activity. Conclusions Approaches that leave hepatic architecture intact, like multispectral imaging, may be paramount to developing effective treatments for NASH. In addition, understanding individual differences in patients may be required for optimal responses to macrophage-targeting therapies.
Collapse
|
16
|
Abstract
Sucrose, the primary circulating sugar in plants, contains equal amounts of fructose and glucose. The latter is the predominant circulating sugar in animals and thus the primary fuel source for various tissue and cell types in the body. Chronic excessive energy intake has, however, emerged as a major driver of obesity and associated pathologies including nonalcoholic fatty liver diseases (NAFLD) and the more severe nonalcoholic steatohepatitis (NASH). Consumption of a high-caloric, western-style diet induces gut dysbiosis and inflammation resulting in leaky gut. Translocation of gut-derived bacterial content promotes hepatic inflammation and ER stress, and when either or both of these are combined with steatosis, it can cause NASH. Here, we review the metabolic links between diet-induced changes in the gut and NASH. Furthermore, therapeutic interventions for the treatment of obesity and liver metabolic diseases are also discussed with a focus on restoring the gut-liver axis.
Collapse
|
17
|
Abenavoli L, Maurizi V, Rinninella E, Tack J, Di Berardino A, Santori P, Rasetti C, Procopio AC, Boccuto L, Scarpellini E. Fecal Microbiota Transplantation in NAFLD Treatment. Medicina (B Aires) 2022; 58:medicina58111559. [PMID: 36363516 PMCID: PMC9695159 DOI: 10.3390/medicina58111559] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Introduction: Gut microbiota is not only a taxonomic biologic ecosystem but is also involved in human intestinal and extra-intestinal functions such as immune system modulation, nutrient absorption and digestion, as well as metabolism regulation. The latter is strictly linked to non-alcoholic fatty liver disease (NAFLD) pathophysiology. Materials and methods: We reviewed the literature on the definition of gut microbiota, the concepts of “dysbiosis” and “eubiosis”, their role in NAFLD pathogenesis, and the data on fecal microbiota transplantation (FMT) in these patients. We consulted the main medical databases using the following keywords, acronyms, and their associations: gut microbiota, eubiosis, dysbiosis, bile acids, NAFLD, and FMT. Results: Gut microbiota qualitative and quantitative composition is different in healthy subjects vs. NALFD patients. This dysbiosis is associated with and involved in NAFLD pathogenesis and evolution to non-acoholic steatohepatitis (NASH), liver cirrhosis, and hepatocellular carcinoma (HCC). In detail, microbial-driven metabolism of bile acids (BAs) and interaction with hepatic and intestinal farnesoid nuclear X receptor (FXR) have shown a determinant role in liver fat deposition and the development of fibrosis. Over the use of pre- or probiotics, FMT has shown preclinical and initial clinical promising results in NAFLD treatment through re-modulation of microbial dysbiosis. Conclusions: Promising clinical data support a larger investigation of gut microbiota dysbiosis reversion through FMT in NAFLD using randomized clinical trials to design precision-medicine treatments for these patients at different disease stages.
Collapse
Affiliation(s)
- Ludovico Abenavoli
- Department of Health Sciences, University “Magna Græcia”, 88100 Catanzaro, Italy
| | - Valentina Maurizi
- Internal Medicine Residency Program, Università Politecnica delle Marche, 60121 Ancona, Italy
| | - Emanuele Rinninella
- Clinical Nutrition Unit, Fondazione Policlinico A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Jan Tack
- T.A.R.G.I.D., Gasthuisberg University Hospital, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Arianna Di Berardino
- Clinical Nutrition and Internal Medicine Unit, “Madonna del Soccorso” General Hospital, 63074 San Benedetto del Tronto, Italy
| | - Pierangelo Santori
- Hepatology and Internal Medicine Unit, “Madonna del Soccorso” General Hospital, 63074 San Benedetto del Tronto, Italy
| | - Carlo Rasetti
- Clinical Nutrition and Internal Medicine Unit, “Madonna del Soccorso” General Hospital, 63074 San Benedetto del Tronto, Italy
- Hepatology and Internal Medicine Unit, “Madonna del Soccorso” General Hospital, 63074 San Benedetto del Tronto, Italy
| | | | - Luigi Boccuto
- Healthcare Genetics and Genomics Doctoral Program, School of Nursing, College of Behavioral, Social and Health Sciences, Clemson University, 105 Sikes Hall, Clemson, SC 29631, USA
| | - Emidio Scarpellini
- T.A.R.G.I.D., Gasthuisberg University Hospital, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
- Clinical Nutrition and Internal Medicine Unit, “Madonna del Soccorso” General Hospital, 63074 San Benedetto del Tronto, Italy
- Correspondence: ; Tel.: +3907-3579-3301; Fax: +3907-3579-3306
| |
Collapse
|
18
|
Fiaschini N, Negroni A, Palone F, Vitali R, Colantoni E, Laudadio I, Mancuso M, Cucchiara S, Stronati L. Colonic inflammation accelerates the progression of liver disease: A protective role of dipotassium glycyrrhizate. Dig Liver Dis 2022; 54:1084-1093. [PMID: 34903499 DOI: 10.1016/j.dld.2021.11.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND The incidence of non-alcoholic fatty liver disease (NAFLD) and its more severe and progressive form, non-alcoholic steatohepatitis (NASH) is increasing worldwide. Gut inflammation seems to concur to the pathogenesis of NASH. No drugs are currently approved for NASH treatment. AIMS To investigate if inflamed gut directly contributes to the progression of NASH through gut epithelial and vascular barrier impairment and to evaluate the efficacy of dipotassium glycyrrhizate (DPG) to improve the liver disease. METHODS A NASH model was set up by feeding mice, for 8 and 13 weeks, with high fat diet with high fructose and glucose (HFD-FG) supplemented periodically with dextran sulfate sodium (DSS) in drinking water. A group was also treated with DPG by gavage. Histological, immunohistochemical and molecular analysis were performed. RESULTS DSS-induced colitis increased steatosis, inflammatory (IL-6, TNFα, NLRP3, MCP-1) as well as fibrotic (TGF-β, α-SMA) mediator expression in HFD-FG mice. Beneficial effect of DPG was associated with restoration of intestinal epithelial and vascular barriers, evaluated respectively by ZO-1 and PV-1 expression, that are known to limit bacterial translocation. CONCLUSION Colonic inflammation strongly contributes to the progression of NASH, likely by favouring bacterial translocation. DPG treatment could represent a novel strategy to reduce liver injury.
Collapse
Affiliation(s)
- Noemi Fiaschini
- Maternal Infantile and Urological Sciences Department, Sapienza University, Rome, Italy
| | - Anna Negroni
- Division of Health Protection Technologies, ENEA, Rome, Italy.
| | | | - Roberta Vitali
- Division of Health Protection Technologies, ENEA, Rome, Italy
| | | | - Ilaria Laudadio
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | | | - Salvatore Cucchiara
- Maternal Infantile and Urological Sciences Department, Sapienza University, Rome, Italy
| | - Laura Stronati
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| |
Collapse
|
19
|
Ivnitsky JJ, Schäfer TV, Rejniuk VL, Vakunenkova OA. Secondary Dysfunction of the Intestinal Barrier in the Pathogenesis of Complications of Acute Poisoning. J EVOL BIOCHEM PHYS+ 2022; 58:1075-1098. [PMID: 36061072 PMCID: PMC9420239 DOI: 10.1134/s0022093022040123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 11/22/2022]
Abstract
The last decade has been marked by an exponential increase
in the number of publications on the physiological role of the normal
human gut microbiota. The idea of a symbiotic relationship between
the human organism and normal microbiota of its gastrointestinal
tract has been firmly established as an integral part of the current
biomedical paradigm. However, the type of this symbiosis varies
from mutualism to parasitism and depends on the functional state
of the host organism. Damage caused to the organism by external
agents can lead to the emergence of conditionally pathogenic properties
in the normal gut microbiota, mediated by humoral factors and affecting
the outcome of exogenous exposure. Among the substances produced
by symbiotic microbiota, there are an indefinite number of compounds
with systemic toxicity. Some occur in the intestinal chyme in potentially
lethal amounts in the case they enter the bloodstream quickly. The quick
entry of potential toxicants is prevented by the intestinal barrier
(IB), a set of structural elements separating the intestinal chyme
from the blood. Hypothetically, severe damage to the IB caused by
exogenous toxicants can trigger a leakage and subsequent systemic
redistribution of toxic substances of bacterial origin. Until recently,
the impact of such a redistribution on the outcome of acute exogenous
poisoning remained outside the view of toxicology. The present review
addresses causal relationships between the secondary dysfunction
of the IB and complications of acute poisoning. We characterize
acute systemic toxicity of such waste products of the normal gut microflora
as ammonia and endotoxins, and demonstrate their involvement in
the formation of such complications of acute poisoning as shock,
sepsis, cerebral insufficiency and secondary lung injuries. The
principles of assessing the functional state of the IB and the approaches
to its protection in acute poisoning are briefly considered.
Collapse
Affiliation(s)
- Ju. Ju. Ivnitsky
- Golikov Research Clinical Center of Toxicology, Federal Medical Biological Agency, St. Petersburg, Russia
| | - T. V. Schäfer
- State Scientific Research Test Institute of Military Medicine, Ministry of Defense of the Russian Federation, St. Petersburg, Russia
| | - V. L. Rejniuk
- Golikov Research Clinical Center of Toxicology, Federal Medical Biological Agency, St. Petersburg, Russia
| | - O. A. Vakunenkova
- Golikov Research Clinical Center of Toxicology, Federal Medical Biological Agency, St. Petersburg, Russia
| |
Collapse
|
20
|
Musazadeh V, Roshanravan N, Dehghan P, Ahrabi SS. Effect of Probiotics on Liver Enzymes in Patients With Non-alcoholic Fatty Liver Disease: An Umbrella of Systematic Review and Meta-Analysis. Front Nutr 2022; 9:844242. [PMID: 35677540 PMCID: PMC9169800 DOI: 10.3389/fnut.2022.844242] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 05/02/2022] [Indexed: 12/20/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become prevalent in recent decades, especially in developed countries; yet the approaches for preventing and treating NAFLD are not clear. This study aimed to summarize meta-analyses of randomized controlled trials that examined the effects of probiotics on NAFLD. We systematically searched PubMed, Scopus, Embase, Web of Science, and Cochrane Central Library databases up to August 2021. All Meta-analysis studies assessing the effect of probiotics on liver function tests [alanine aminotransferase (ALT), aspartate aminotransferase (AST), and Gamma-glutamyl transferase (GGT)] were included. Meta-analysis was conducted using a random-effects model. Sensitivity and subgroup analyses were also performed. The umbrella study covered ten eligible studies involving 5,162 individuals. Beneficial effects of probiotics supplementation were revealed on ALT (ES = −10.54 IU/L; 95% CI: −12.70, −8.39; p < 0.001; I2 = 60.9%, p = 0.006), AST (ES = −10.19 IU/L, 95%CI: −13.08, −7.29, p < 0.001; I2 = 79.8%, p < 0.001), and GGT (ES = −5.88 IU/L, 95% CI: −7.09, −4.67, p = 0.009; I2 = 0.0%, p = 0.591) levels. Probiotics have ameliorating effects on ALT, AST, and GGT levels in patients with NAFLD. Overall, Probiotics could be recommended as an adjuvant therapeutic method for the management of NAFLD.
Collapse
Affiliation(s)
- Vali Musazadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Community Nutrition, School of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Roshanravan
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Dehghan
- Faculty of Nutrition and Food Science, Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- *Correspondence: Parvin Dehghan,
| | - Sana Sedgh Ahrabi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
21
|
Wan H, Wang Y, Zhang H, Zhang K, Chen Y, Chen C, Zhang W, Xia F, Wang N, Lu Y. Chronic lead exposure induces fatty liver disease associated with the variations of gut microbiota. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 232:113257. [PMID: 35104782 DOI: 10.1016/j.ecoenv.2022.113257] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/06/2022] [Accepted: 01/26/2022] [Indexed: 05/02/2023]
Abstract
BACKGROUND Lead (Pb) has been suggested as an endocrine-disrupting chemical. However, few studies have investigated the association between chronic Pb exposure and fatty liver disease. OBJECTIVES We aimed to investigate the association of chronic Pb exposure with fatty liver disease and whether the variations of the gut microbiota involve in the mechanism of the fatty liver disease induced by chronic Pb exposure. METHODS We conducted a cross-sectional study of 3066 rural participants in East China. Blood lead level (BLL) was detected, and abdominal ultrasonography was used to diagnose hepatic steatosis. Both the definition of non-alcoholic fatty liver disease (NAFLD) and metabolic dysfunction-associated fatty liver disease (MAFLD) were used. Wistar rats were randomly divided into two groups and each group was exposed to 0 or 0.05% w/v Pb through drinking water for 28 weeks. The relevant parameters of hepatic lipid metabolism and gut microbiota were analyzed. RESULTS In humans, after adjusting for potential confounders, the odds of having NAFLD and MAFLD were significantly increased by 54% and 52% in the participants in the fourth BLL quartile (OR 1.54, 95% CI 1.24, 1.91 and OR 1.52, 95% CI 1.22, 1.89). In the rats, chronic Pb exposure induced the increased visceral fat, hepatic steatosis, and dysbiosis of the gut microbiota, including the decrease of richness, diversity, evenness and phylogenetic diversity of the gut microbiota and the significant alternations of the gut microbiota composition, particularly, the decrease of the relative abundance of Coprococcus and Oscillospira at the genus level. CONCLUSIONS Chronic Pb exposure could induce fatty liver disease, which may be associated with the variations of the gut microbiota.
Collapse
Affiliation(s)
- Heng Wan
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Institute and Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Yuying Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Haojie Zhang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Kun Zhang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yi Chen
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Chi Chen
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Wen Zhang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Fangzhen Xia
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Ningjian Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Institute and Department of Endocrinology and Metabolism, Huangpu Branch, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.
| | - Yingli Lu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Institute and Department of Endocrinology and Metabolism, Huangpu Branch, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.
| |
Collapse
|
22
|
Understanding the Role of the Gut Microbiome and Microbial Metabolites in Non-Alcoholic Fatty Liver Disease: Current Evidence and Perspectives. Biomolecules 2021; 12:biom12010056. [PMID: 35053205 PMCID: PMC8774162 DOI: 10.3390/biom12010056] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/24/2021] [Accepted: 12/30/2021] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide. NAFLD begins as a relatively benign hepatic steatosis which can evolve to non-alcoholic steatohepatitis (NASH); the risk of cirrhosis and hepatocellular carcinoma (HCC) increases when fibrosis is present. NAFLD represents a complex process implicating numerous factors—genetic, metabolic, and dietary—intertwined in a multi-hit etiopathogenetic model. Recent data have highlighted the role of gut dysbiosis, which may render the bowel more permeable, leading to increased free fatty acid absorption, bacterial migration, and a parallel release of toxic bacterial products, lipopolysaccharide (LPS), and proinflammatory cytokines that initiate and sustain inflammation. Although gut dysbiosis is present in each disease stage, there is currently no single microbial signature to distinguish or predict which patients will evolve from NAFLD to NASH and HCC. Using 16S rRNA sequencing, the majority of patients with NAFLD/NASH exhibit increased numbers of Bacteroidetes and differences in the presence of Firmicutes, resulting in a decreased F/B ratio in most studies. They also present an increased proportion of species belonging to Clostridium, Anaerobacter, Streptococcus, Escherichia, and Lactobacillus, whereas Oscillibacter, Flavonifaractor, Odoribacter, and Alistipes spp. are less prominent. In comparison to healthy controls, patients with NASH show a higher abundance of Proteobacteria, Enterobacteriaceae, and Escherichia spp., while Faecalibacterium prausnitzii and Akkermansia muciniphila are diminished. Children with NAFLD/NASH have a decreased proportion of Oscillospira spp. accompanied by an elevated proportion of Dorea, Blautia, Prevotella copri, and Ruminococcus spp. Gut microbiota composition may vary between population groups and different stages of NAFLD, making any conclusive or causative claims about gut microbiota profiles in NAFLD patients challenging. Moreover, various metabolites may be involved in the pathogenesis of NAFLD, such as short-chain fatty acids, lipopolysaccharide, bile acids, choline and trimethylamine-N-oxide, and ammonia. In this review, we summarize the role of the gut microbiome and metabolites in NAFLD pathogenesis, and we discuss potential preventive and therapeutic interventions related to the gut microbiome, such as the administration of probiotics, prebiotics, synbiotics, antibiotics, and bacteriophages, as well as the contribution of bariatric surgery and fecal microbiota transplantation in the therapeutic armamentarium against NAFLD. Larger and longer-term prospective studies, including well-defined cohorts as well as a multi-omics approach, are required to better identify the associations between the gut microbiome, microbial metabolites, and NAFLD occurrence and progression.
Collapse
|
23
|
Febbraio MA, Karin M. "Sweet death": Fructose as a metabolic toxin that targets the gut-liver axis. Cell Metab 2021; 33:2316-2328. [PMID: 34619076 PMCID: PMC8665123 DOI: 10.1016/j.cmet.2021.09.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/30/2021] [Accepted: 09/09/2021] [Indexed: 02/07/2023]
Abstract
Glucose and fructose are closely related simple sugars, but fructose has been associated more closely with metabolic disease. Until the 1960s, the major dietary source of fructose was fruit, but subsequently, high-fructose corn syrup (HFCS) became a dominant component of the Western diet. The exponential increase in HFCS consumption correlates with the increased incidence of obesity and type 2 diabetes mellitus, but the mechanistic link between these metabolic diseases and fructose remains tenuous. Although dietary fructose was thought to be metabolized exclusively in the liver, evidence has emerged that it is also metabolized in the small intestine and leads to intestinal epithelial barrier deterioration. Along with the clinical manifestations of hereditary fructose intolerance, these findings suggest that, along with the direct effect of fructose on liver metabolism, the gut-liver axis plays a key role in fructose metabolism and pathology. Here, we summarize recent studies on fructose biology and pathology and discuss new opportunities for prevention and treatment of diseases associated with high-fructose consumption.
Collapse
Affiliation(s)
- Mark A Febbraio
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.
| | - Michael Karin
- Department of Pharmacology, School of Medicine, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
24
|
Liptak R, Gromova B, Gardlik R. Fecal Microbiota Transplantation as a Tool for Therapeutic Modulation of Non-gastrointestinal Disorders. Front Med (Lausanne) 2021; 8:665520. [PMID: 34557498 PMCID: PMC8452915 DOI: 10.3389/fmed.2021.665520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/13/2021] [Indexed: 12/14/2022] Open
Abstract
Fecal microbiota transplantation has been primarily investigated as a therapeutic tool for a number of gut disorders. Optimistic results from clinical studies on Clostridium difficile infection, inflammatory bowel disease and irritable bowel syndrome have stimulated the expansion of possible indications in which FMT might represent a game changing approach. Microbial dysbiosis was shown in a number of non-gastrointestinal disorders. Moreover, FMT was proven to be effective in therapy of numerous animal models of disease. However, only a proportion of these disorders have been addressed in clinical studies using FMT. These include obesity, non-alcoholic fatty liver disease, cardiovascular inflammation and neurological disorders such as autism, depression and Parkinson's disease. Results from preclinical and clinical studies also outlined possible molecular mechanisms that contribute to alleviation of the disease. These range from increasing the circulating levels of microbial metabolites (trimethylamine N-oxide, lipopolysaccharide, short chain fatty acids) to stimulation of the enteric nervous system. Several methodological shortcomings are still to be addressed; however, positive results of the clinical studies indicate that further investigation of FMT as a therapeutic tool for non-gastrointestinal disorders can be expected in upcoming years.
Collapse
Affiliation(s)
- Robert Liptak
- Institute of Physiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia.,Emergency Department, University Hospital in Bratislava, Bratislava, Slovakia
| | - Barbora Gromova
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Roman Gardlik
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| |
Collapse
|
25
|
Mohamad Nor MH, Ayob N, Mokhtar NM, Raja Ali RA, Tan GC, Wong Z, Shafiee NH, Wong YP, Mustangin M, Nawawi KNM. The Effect of Probiotics (MCP ® BCMC ® Strains) on Hepatic Steatosis, Small Intestinal Mucosal Immune Function, and Intestinal Barrier in Patients with Non-Alcoholic Fatty Liver Disease. Nutrients 2021; 13:3192. [PMID: 34579068 PMCID: PMC8468225 DOI: 10.3390/nu13093192] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 12/25/2022] Open
Abstract
Treatment for non-alcoholic fatty liver disease (NAFLD) currently consists of lifestyle modifications such as a low-fat diet, weight loss, and exercise. The gut microbiota forms part of the gut-liver axis and serves as a potential target for NAFLD treatment. We investigated the effect of probiotics on hepatic steatosis, fibrosis, and biochemical blood tests in patients with NAFLD. At the small intestinal mucosal level, we examined the effect of probiotics on the expression of CD4+ and CD8+ T lymphocytes, as well as the tight junction protein zona occluden-1 (ZO-1). This was a randomized, double-blind, placebo-controlled trial involving ultrasound-diagnosed NAFLD patients (n = 39) who were supplemented with either a probiotics sachet (MCP® BCMC® strains) or a placebo for a total of 6 months. Multi-strain probiotics (MCP® BCMC® strains) containing six different Lactobacillus and Bifidobacterium species at a concentration of 30 billion CFU were used. There were no significant changes at the end of the study in terms of hepatic steatosis (probiotics: -21.70 ± 42.6 dB/m, p = 0.052 vs. placebo: -10.72 ± 46.6 dB/m, p = 0.29) and fibrosis levels (probiotics: -0.25 ± 1.77 kPa, p = 0.55 vs. placebo: -0.62 ± 2.37 kPa, p = 0.23) as measured by transient elastography. Likewise, no significant changes were found for both groups for the following parameters: LiverFAST analysis (steatosis, fibrosis and inflammation scores), alanine aminotransferase, total cholesterol, triglycerides, and fasting glucose. In the immunohistochemistry (IHC) analysis, no significant expression changes were seen for CD4+ T lymphocytes in either group (probiotics: -0.33 ± 1.67, p = 0.35 vs. placebo: 0.35 ± 3.25, p = 0.63). However, significant reductions in the expression of CD8+ T lymphocytes (-7.0 ± 13.73, p = 0.04) and ZO-1 (Z-score = -2.86, p = 0.04) were found in the placebo group, but no significant changes in the probiotics group. In this pilot study, the use of probiotics did not result in any significant clinical improvement in NAFLD patients. However, at the microenvironment level (i.e., the small intestinal mucosa), probiotics seemed to be able to stabilize the mucosal immune function and to protect NAFLD patients against increased intestinal permeability. Therefore, probiotics might have a complementary role in treating NAFLD. Further studies with larger sample sizes, a longer duration, and different probiotic strains are needed to evaluate the real benefit of probiotics in NAFLD.
Collapse
Affiliation(s)
- Mohamad Hizami Mohamad Nor
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (M.H.M.N.); (R.A.R.A.); (Z.W.)
| | - Nurainina Ayob
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (N.A.); (N.M.M.)
| | - Norfilza M. Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (N.A.); (N.M.M.)
- GUT Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Raja Affendi Raja Ali
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (M.H.M.N.); (R.A.R.A.); (Z.W.)
- GUT Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Geok Chin Tan
- GUT Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (Y.P.W.); (M.M.)
| | - Zhiqin Wong
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (M.H.M.N.); (R.A.R.A.); (Z.W.)
- GUT Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Nor Hamizah Shafiee
- Dietetics Programme, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Yin Ping Wong
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (Y.P.W.); (M.M.)
| | - Muaatamarulain Mustangin
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (Y.P.W.); (M.M.)
| | - Khairul Najmi Muhammad Nawawi
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (M.H.M.N.); (R.A.R.A.); (Z.W.)
- GUT Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
26
|
Zhang Y, Li JX, Zhang Y, Wang YL. Intestinal microbiota participates in nonalcoholic fatty liver disease progression by affecting intestinal homeostasis. World J Clin Cases 2021; 9:6654-6662. [PMID: 34447812 PMCID: PMC8362529 DOI: 10.12998/wjcc.v9.i23.6654] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/25/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic liver disease with a pathogenesis that has not been fully elucidated. With the development of the theory of the gut-liver axis and the deepening of related research, the role of the intestinal tract in the pathogenesis of NAFLD has been investigated more. Intestinal microbiota, intestinal metabolites, and intestinal epithelial and immune-based barriers constitute the intestinal environment, which uses crosstalk to maintain the homeostasis of the intestinal environment. This paper reviews the progress in the study of intestinal microbiota, intestinal environment, and NAFLD and suggests that repair of intestinal functional balance may be a new idea for early prevention and intervention of NAFLD.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Gastroenterology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing 100078, China
| | - Jun-Xiang Li
- Department of Gastroenterology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing 100078, China
| | - Yan Zhang
- Department of Gastroenterology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing 100078, China
| | - Yun-Liang Wang
- Department of Gastroenterology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing 100078, China
| |
Collapse
|
27
|
Hupa-Breier KL, Dywicki J, Hartleben B, Wellhöner F, Heidrich B, Taubert R, Mederacke YSE, Lieber M, Iordanidis K, Manns MP, Wedemeyer H, Hardtke-Wolenski M, Jaeckel E. Dulaglutide Alone and in Combination with Empagliflozin Attenuate Inflammatory Pathways and Microbiome Dysbiosis in a Non-Diabetic Mouse Model of NASH. Biomedicines 2021; 9:biomedicines9040353. [PMID: 33808404 PMCID: PMC8066839 DOI: 10.3390/biomedicines9040353] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/22/2021] [Accepted: 03/25/2021] [Indexed: 12/15/2022] Open
Abstract
Dysregulation of glucose homeostasis plays a major role in the pathogenesis of non-alcoholic steatohepatitis (NASH) as it activates proinflammatory and profibrotic processes. Beneficial effects of antiglycemic treatments such as GLP-1 agonist or SGLT-2 inhibitor on NASH in patients with diabetes have already been investigated. However, their effect on NASH in a non-diabetic setting remains unclear. With this aim, we investigated the effect of long-acting GLP1-agonist dulaglutide and SGLT-2 inhibitor empagliflozin and their combination in a non-diabetic mouse model of NASH. C57BL/6 mice received a high-fat-high-fructose (HFHC) diet with a surplus of cholesterol for 16 weeks. After 12 weeks of diet, mice were treated with either dulaglutide, empagliflozin or their combination. Dulaglutide alone and in combination with empagliflozin led to significant weight loss, improved glucose homeostasis and diminished anti-inflammatory and anti-fibrotic pathways. Combination of dulaglutide and empagliflozin further decreased MoMFLy6CHigh and CD4+Foxp3+ T cells. No beneficial effects for treatment with empagliflozin alone could be shown. While no effect of dulaglutide or its combination with empaglifozin on hepatic steatosis was evident, these data demonstrate distinct anti-inflammatory effects of dulaglutide and their combination with empagliflozin in a non-diabetic background, which could have important implications for further treatment of NASH.
Collapse
Affiliation(s)
- Katharina Luise Hupa-Breier
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; (J.D.); (F.W.); (B.H.); (R.T.); (Y.S.M.); (M.L.); (K.I.); (M.P.M.); (H.W.); (M.H.-W.); (E.J.)
- Correspondence: ; Tel.: +49-(0)-511-532-6992
| | - Janine Dywicki
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; (J.D.); (F.W.); (B.H.); (R.T.); (Y.S.M.); (M.L.); (K.I.); (M.P.M.); (H.W.); (M.H.-W.); (E.J.)
| | - Björn Hartleben
- Department of Pathology, Hannover Medical School, 30625 Hannover, Germany;
| | - Freya Wellhöner
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; (J.D.); (F.W.); (B.H.); (R.T.); (Y.S.M.); (M.L.); (K.I.); (M.P.M.); (H.W.); (M.H.-W.); (E.J.)
| | - Benjamin Heidrich
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; (J.D.); (F.W.); (B.H.); (R.T.); (Y.S.M.); (M.L.); (K.I.); (M.P.M.); (H.W.); (M.H.-W.); (E.J.)
| | - Richard Taubert
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; (J.D.); (F.W.); (B.H.); (R.T.); (Y.S.M.); (M.L.); (K.I.); (M.P.M.); (H.W.); (M.H.-W.); (E.J.)
| | - Young-Seon Elisabeth Mederacke
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; (J.D.); (F.W.); (B.H.); (R.T.); (Y.S.M.); (M.L.); (K.I.); (M.P.M.); (H.W.); (M.H.-W.); (E.J.)
| | - Maren Lieber
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; (J.D.); (F.W.); (B.H.); (R.T.); (Y.S.M.); (M.L.); (K.I.); (M.P.M.); (H.W.); (M.H.-W.); (E.J.)
| | - Konstantinos Iordanidis
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; (J.D.); (F.W.); (B.H.); (R.T.); (Y.S.M.); (M.L.); (K.I.); (M.P.M.); (H.W.); (M.H.-W.); (E.J.)
| | - Michael P. Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; (J.D.); (F.W.); (B.H.); (R.T.); (Y.S.M.); (M.L.); (K.I.); (M.P.M.); (H.W.); (M.H.-W.); (E.J.)
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; (J.D.); (F.W.); (B.H.); (R.T.); (Y.S.M.); (M.L.); (K.I.); (M.P.M.); (H.W.); (M.H.-W.); (E.J.)
| | - Matthias Hardtke-Wolenski
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; (J.D.); (F.W.); (B.H.); (R.T.); (Y.S.M.); (M.L.); (K.I.); (M.P.M.); (H.W.); (M.H.-W.); (E.J.)
- Department of Gastroenterology and Hepatology, Essen University Hospital, University Duisburg-Essen, 45147 Essen, Germany
| | - Elmar Jaeckel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; (J.D.); (F.W.); (B.H.); (R.T.); (Y.S.M.); (M.L.); (K.I.); (M.P.M.); (H.W.); (M.H.-W.); (E.J.)
| |
Collapse
|
28
|
Coupling Machine Learning and Lipidomics as a Tool to Investigate Metabolic Dysfunction-Associated Fatty Liver Disease. A General Overview. Biomolecules 2021; 11:biom11030473. [PMID: 33810079 PMCID: PMC8004861 DOI: 10.3390/biom11030473] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/08/2021] [Accepted: 03/18/2021] [Indexed: 12/15/2022] Open
Abstract
Hepatic biopsy is the gold standard for staging nonalcoholic fatty liver disease (NAFLD). Unfortunately, accessing the liver is invasive, requires a multidisciplinary team and is too expensive to be conducted on large segments of the population. NAFLD starts quietly and can progress until liver damage is irreversible. Given this complex situation, the search for noninvasive alternatives is clinically important. A hallmark of NAFLD progression is the dysregulation in lipid metabolism. In this context, recent advances in the area of machine learning have increased the interest in evaluating whether multi-omics data analysis performed on peripheral blood can enhance human interpretation. In the present review, we show how the use of machine learning can identify sets of lipids as predictive biomarkers of NAFLD progression. This approach could potentially help clinicians to improve the diagnosis accuracy and predict the future risk of the disease. While NAFLD has no effective treatment yet, the key to slowing the progression of the disease may lie in predictive robust biomarkers. Hence, to detect this disease as soon as possible, the use of computational science can help us to make a more accurate and reliable diagnosis. We aimed to provide a general overview for all readers interested in implementing these methods.
Collapse
|