1
|
Deslande M, Puig-Castellvi F, Castro-Dionicio I, Pacheco-Tapia R, Raverdy V, Caiazzo R, Lassailly G, Leloire A, Andrikopoulos P, Kahoul Y, Zaïbi N, Toussaint B, Oger F, Gambardella N, Lefebvre P, Derhourhi M, Amanzougarene S, Staels B, Pattou F, Froguel P, Bonnefond A, Dumas ME. Intrahepatic levels of microbiome-derived hippurate associates with improved metabolic dysfunction-associated steatotic liver disease. Mol Metab 2025; 92:102090. [PMID: 39746606 PMCID: PMC11772989 DOI: 10.1016/j.molmet.2024.102090] [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: 08/21/2024] [Revised: 12/24/2024] [Accepted: 12/25/2024] [Indexed: 01/04/2025] Open
Abstract
OBJECTIVE Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterised by lipid accumulation in the liver and is often associated with obesity and type 2 diabetes. The gut microbiome recently emerged as a significant player in liver metabolism and health. Hippurate, a host-microbial co-metabolite has been associated with human gut microbial gene richness and with metabolic health. However, its role on liver metabolism and homeostasis is poorly understood. METHODS We characterised liver biospies from 318 patients with obesity using RNAseq and metabolomics in liver and plasma to derive associations among hepatic hippurate, hepatic gene expression and MASLD and phenotypes. To test a potential beneficial role for hippurate in hepatic insulin resistance, we profile the metabolome of (IHH) using ultra-high-performance liquid chromatography coupled to high-resolution tandem mass spectrometry (UHPLC-MS/MS), and characterised intracellular triglyceride accumulation and glucose internalisation after a 24 h insulin exposure. RESULTS We first report significant associations among MASLD traits, plasma and hepatic hippurate. Further analysis of the hepatic transcriptome shows that liver and plasma hippurate are inversely associated with MASLD, implicating lipid metabolism and regulation of inflammatory responses pathways. Hippurate treatment inhibits lipid accumulation and rescues insulin resistance induced by 24-hour chronic insulin in IHH. Hippurate also improves hepatocyte metabolic profiles by increasing the abundance of metabolites involved in energy homeostasis that are depleted by chronic insulin treatment while decreasing those involved in inflammation. CONCLUSIONS Altogether, our results further highlight hippurate as a mechanistic marker of metabolic health, by its ability to improve metabolic homeostasis as a postbiotic candidate.
Collapse
Affiliation(s)
- Maxime Deslande
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Francesc Puig-Castellvi
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France; Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom
| | - Inés Castro-Dionicio
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France; Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom
| | - Romina Pacheco-Tapia
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France; Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom
| | - Violeta Raverdy
- INSERM U1190, Institut Pasteur de Lille, University of Lille, Lille University Hospital, 59045, Lille, France
| | - Robert Caiazzo
- INSERM U1190, Institut Pasteur de Lille, University of Lille, Lille University Hospital, 59045, Lille, France
| | - Guillaume Lassailly
- INSERM U1011 Institut Pasteur de Lille, University of Lille, Lille University Hospital, 59045, Lille, France
| | - Audrey Leloire
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Petros Andrikopoulos
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom
| | - Yasmina Kahoul
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Nawel Zaïbi
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Bénédicte Toussaint
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Frédérik Oger
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Nicolas Gambardella
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Philippe Lefebvre
- INSERM U1011 Institut Pasteur de Lille, University of Lille, Lille University Hospital, 59045, Lille, France
| | - Mehdi Derhourhi
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Souhila Amanzougarene
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Bart Staels
- INSERM U1011 Institut Pasteur de Lille, University of Lille, Lille University Hospital, 59045, Lille, France
| | - François Pattou
- INSERM U1190, Institut Pasteur de Lille, University of Lille, Lille University Hospital, 59045, Lille, France
| | - Philippe Froguel
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France; Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom
| | - Amélie Bonnefond
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France; Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom
| | - Marc-Emmanuel Dumas
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France; Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom; The Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, H3A 0G1, Canada.
| |
Collapse
|
2
|
Jiang QR, Zeng DW. Gut microbiota shifts in hepatitis B-related portal hypertension after transjugular intrahepatic portosystemic shunt: Mechanistic and clinical implications. World J Gastroenterol 2025; 31:100752. [PMID: 39839897 PMCID: PMC11684156 DOI: 10.3748/wjg.v31.i3.100752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/07/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
In this article, we provide commentary on the recent article by Zhao et al. We focus on the shifts in the gut microbiota of patients with hepatitis B virus (HBV)-associated cirrhosis/portal hypertension (PH) following transjugular intrahepatic portosystemic shunt (TIPS) and the implications for understanding the mechanisms, diagnosis, and treatment. By comparing the gut microbiota composition and dynamic changes before and after TIPS in patients with and without hepatic encephalopathy, the authors found an increase in non-probiotic bacteria in those who developed hepatic encephalopathy post-TIPS, with Morganella species present only in the hepatic encephalopathy group. The gut microbiota changes post-TIPS among patients without the occurrence of hepatic encephalopathy suggest potential therapeutic benefits through prophylactic microbiome therapies. Furthermore, the specific gut microbiota alterations may hold promise to predict the risk of hepatic encephalopathy in individuals undergoing TIPS for HBV-related PH. Despite these promising findings, future studies are needed to address limitations, including a small sample size, a relatively short evaluation period for gut microbiota alterations, the absence of data on dynamic alterations in gut microbiota post-TIPS and their correlation with blood ammonia levels, and the lack of validation in animal models. In conclusion, Zhao et al's study has shed new light on the link of gut microbiota with post-TIPS hepatic encephalopathy, potentially through the intricate gut-liver axis, and has important clinical implications for improving the management of patients with HBV-related PH.
Collapse
Affiliation(s)
- Qi-Rong Jiang
- Department of Hepatology, Hepatology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, Fujian Province, China
| | - Da-Wu Zeng
- Department of Infectious Diseases, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350004, Fujian Province, China
| |
Collapse
|
3
|
Vega-Abellaneda S, Dopazo C, Yañez F, Soler Z, Xie Z, Canalda-Baltrons A, Pons-Tarín M, Bilbao I, Manichanh C. Microbiome composition recovery after liver transplantation correlates with initial liver disease severity and antibiotics treatment. Am J Transplant 2024; 24:1623-1633. [PMID: 38556088 DOI: 10.1016/j.ajt.2024.03.038] [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: 12/20/2023] [Revised: 03/02/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
Liver transplantation (LT) is crucial for end-stage liver disease, but it is linked to infection risks. Pathobionts, microorganisms potentially harmful under specific conditions, can cause complications posttransplant. Monitoring such pathogens in fecal samples can be challenging and therefore remains underexplored post-LT. This study aimed to analyze the gut microbiome before and after LT, tracking pathobionts and correlating clinical data. The study involved 17 liver transplant recipients, 17 healthy relatives (spouses), and 13 donors. Gut samples collected pretranplantation and posttransplantation underwent bacterial and fungal profiling through DNA sequencing. Quantitative polymerase chain reaction was used to assess microbial load. Statistical analyses included alpha and beta diversity measures, differential abundance analysis, and correlation tests between microbiome and clinical parameters. Microbiome analysis revealed dynamic changes in diversity posttransplant. Notably, high-severity patients showed persistent and greater dysbiosis during the first months post-LT compared with low-severity patients, partly due to an antibiotic treatment pre-LT. The analysis identified a higher proportion of pathogens such as Escherichia coli/Shigella flexneri in high-severity cases posttransplant. Furthermore, butyrate producers including Roseburia intestinalis, Anaerostipes hadrus, and Eubacterium coprostanoligenes were positively correlated with levels of albumin. This study offers valuable insights into post-LT microbiome changes, shedding light on the need for tailored prophylactic treatment post-LT.
Collapse
Affiliation(s)
- Sara Vega-Abellaneda
- Microbiome Lab, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Cristina Dopazo
- Department of HPB Surgery and Transplants, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autónoma de Barcelona, Barcelona, Spain; CIBER of Hepatic and Digestive Diseases (CIBERehd), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Francisca Yañez
- Microbiome Lab, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Zaida Soler
- Microbiome Lab, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Zixuan Xie
- Microbiome Lab, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Aleix Canalda-Baltrons
- Microbiome Lab, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Marc Pons-Tarín
- Microbiome Lab, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Itxarone Bilbao
- Department of HPB Surgery and Transplants, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autónoma de Barcelona, Barcelona, Spain; CIBER of Hepatic and Digestive Diseases (CIBERehd), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| | - Chaysavanh Manichanh
- Microbiome Lab, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain; CIBER of Hepatic and Digestive Diseases (CIBERehd), Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
4
|
Llorente C. The Imperative for Innovative Enteric Nervous System-Intestinal Organoid Co-Culture Models: Transforming GI Disease Modeling and Treatment. Cells 2024; 13:820. [PMID: 38786042 PMCID: PMC11119846 DOI: 10.3390/cells13100820] [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: 03/15/2024] [Revised: 04/29/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
This review addresses the need for innovative co-culture systems integrating the enteric nervous system (ENS) with intestinal organoids. The breakthroughs achieved through these techniques will pave the way for a transformative era in gastrointestinal (GI) disease modeling and treatment strategies. This review serves as an introduction to the companion protocol paper featured in this journal. The protocol outlines the isolation and co-culture of myenteric and submucosal neurons with small intestinal organoids. This review provides an overview of the intestinal organoid culture field to establish a solid foundation for effective protocol application. Remarkably, the ENS surpasses the number of neurons in the spinal cord. Referred to as the "second brain", the ENS orchestrates pivotal roles in GI functions, including motility, blood flow, and secretion. The ENS is organized into myenteric and submucosal plexuses. These plexuses house diverse subtypes of neurons. Due to its proximity to the gut musculature and its cell type complexity, there are methodological intricacies in studying the ENS. Diverse approaches such as primary cell cultures, three-dimensional (3D) neurospheres, and induced ENS cells offer diverse insights into the multifaceted functionality of the ENS. The ENS exhibits dynamic interactions with the intestinal epithelium, the muscle layer, and the immune system, influencing epithelial physiology, motility, immune responses, and the microbiome. Neurotransmitters, including acetylcholine (ACh), serotonin (5-HT), and vasoactive intestinal peptide (VIP), play pivotal roles in these intricate interactions. Understanding these dynamics is imperative, as the ENS is implicated in various diseases, ranging from neuropathies to GI disorders and neurodegenerative diseases. The emergence of organoid technology presents an unprecedented opportunity to study ENS interactions within the complex milieu of the small and large intestines. This manuscript underscores the urgent need for standardized protocols and advanced techniques to unravel the complexities of the ENS and its dynamic relationship with the gut ecosystem. The insights gleaned from such endeavors hold the potential to revolutionize GI disease modeling and treatment paradigms.
Collapse
Affiliation(s)
- Cristina Llorente
- Department of Medicine, University of California San Diego, MC0063, 9500 Gilman Drive, La Jolla, CA 92093, USA
| |
Collapse
|
5
|
Silva-Caso W, Carrillo-Ng H, Aguilar-Luis MA, Tarazona-Castro Y, Valle LJD, Tinco-Valdez C, Palomares-Reyes C, Urteaga N, Bazán-Mayra J, del Valle-Mendoza J. Parasitosis by Fasciola hepatica and Variations in Gut Microbiota in School-Aged Children from Peru. Microorganisms 2024; 12:371. [PMID: 38399775 PMCID: PMC10891680 DOI: 10.3390/microorganisms12020371] [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: 12/24/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
(1) Background: Human fascioliasis is considered an endemic and hyper-endemic disease in the Peruvian Andean valleys. Our objective was to determine variations in the composition of the gut microbiota among children with Fasciola hepatica and children who do not have this parasitosis. (2) Method: A secondary analysis was performed using fecal samples stored in our biobank. The samples were collected as part of an epidemiological Fasciola hepatica cross-sectional study in children from 4 through 14 years old from a community in Cajamarca, Peru. (3) Results: In a comparison of the bacterial genera that make up the intestinal microbiota between the F. hepatica positive and negative groups, it was found that there are significant differences in the determination of Lactobacillus (p = 0.010, CI: 8.5-61.4), Bacteroides (p = 0.020, CI: 18.5-61.4), Clostridium (p < 0.001, CI: 3.5-36.0), and Bifidobacterium (p = 0.018, CI: 1.1-28.3), with each of these genera being less frequent in children parasitized with F. hepatica. (4) Conclusions: These results show that F. hepatica may be associated with direct or indirect changes in the bacterial population of the intestinal microbiota, particularly affecting three bacterial genera.
Collapse
Affiliation(s)
- Wilmer Silva-Caso
- Research and Innovation Centre, Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru; (H.C.-N.); (M.A.A.-L.); (Y.T.-C.); (C.T.-V.); (C.P.-R.)
- Instituto de Investigación Nutricional, Lima 15024, Peru
- Escuela de Medicina, Facultad de Ciencias de la Salud, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
| | - Hugo Carrillo-Ng
- Research and Innovation Centre, Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru; (H.C.-N.); (M.A.A.-L.); (Y.T.-C.); (C.T.-V.); (C.P.-R.)
- Instituto de Investigación Nutricional, Lima 15024, Peru
| | - Miguel Angel Aguilar-Luis
- Research and Innovation Centre, Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru; (H.C.-N.); (M.A.A.-L.); (Y.T.-C.); (C.T.-V.); (C.P.-R.)
- Instituto de Investigación Nutricional, Lima 15024, Peru
- Escuela de Medicina, Facultad de Ciencias de la Salud, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
| | - Yordi Tarazona-Castro
- Research and Innovation Centre, Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru; (H.C.-N.); (M.A.A.-L.); (Y.T.-C.); (C.T.-V.); (C.P.-R.)
- Instituto de Investigación Nutricional, Lima 15024, Peru
| | - Luis J. Del Valle
- Barcelona Research Center for Multiscale Science and Engineering, Departament d’Enginyeria Química, EEBE, Universitat Politècnica de Catalunya (UPC), 08019 Barcelona, Spain;
| | - Carmen Tinco-Valdez
- Research and Innovation Centre, Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru; (H.C.-N.); (M.A.A.-L.); (Y.T.-C.); (C.T.-V.); (C.P.-R.)
| | - Carlos Palomares-Reyes
- Research and Innovation Centre, Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru; (H.C.-N.); (M.A.A.-L.); (Y.T.-C.); (C.T.-V.); (C.P.-R.)
- Escuela de Medicina, Facultad de Ciencias de la Salud, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
| | - Numan Urteaga
- Puesto de Salud Callancas, Dirección Regional de Salud Cajamarca (DIRESA), Cajamarca 60101, Peru;
| | - Jorge Bazán-Mayra
- Laboratorio Regional de Cajamarca, Dirección Regional de Salud de Cajamarca (DIRESA), Cajamarca 60101, Peru;
| | - Juana del Valle-Mendoza
- Research and Innovation Centre, Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru; (H.C.-N.); (M.A.A.-L.); (Y.T.-C.); (C.T.-V.); (C.P.-R.)
- Instituto de Investigación Nutricional, Lima 15024, Peru
- Escuela de Medicina, Facultad de Ciencias de la Salud, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
| |
Collapse
|
6
|
Chi X, Cheng DY, Sun X, Liu SA, Wang RB, Chen Q, Xing HC. Efficacy of Biejiajian Pill on Intestinal Microbiota in Patients with Hepatitis B Cirrhosis/Liver Fibrosis: A Randomized Double-Blind Controlled Trial. Chin J Integr Med 2023; 29:771-781. [PMID: 37222832 DOI: 10.1007/s11655-023-3542-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2022] [Indexed: 05/25/2023]
Abstract
OBJECTIVE To analyze the efficacy of Biejiajian Pill (BJJP) on intestinal microbiota in patients with hepatitis B cirrhosis/liver fibrosis, and explore its relationship with liver fibrosis. METHODS This was a prospective, randomized double-blind controlled trial. Using the stratified block randomization method, 35 patients with hepatitis B liver cirrhosis/liver fibrosis were randomly assigned (1:1) to receive entecavir (0.5 mg/d) combined with BJJP (3 g/time, 3 times a day) or placebo (simulator as control, SC group, simulator 3 g/time, 3 times a day) for 48 weeks. Blood and stool samples were collected from patients at baseline and week 48 of treatment, respectively. Liver and renal functions as well as hematological indices were detected. Fecal samples were analyzed by 16S rDNA V3-V4 high-throughput sequencing, and intestinal microbiota changes in both groups before and after treatment were compared, and their correlations with liver fibrosis were analyzed. RESULTS Compared with the SC group, there was no significant difference in liver function, renal function and hematology indices in the BJJP group, however, the improvement rate of liver fibrosis was higher in the BJJP group (94.4% vs. 64.7%, P=0.041). Principal coordinate analysis (PCoA) based on weighted Unifrac distance showed significant differences in intestinal microbiota community diversity before and after BJJP treatment (P<0.01 and P=0.003), respectively. After 48 weeks' treatment, the abundance levels of beneficial bacteria (Bifidobacteria, Lactobacillus, Faecalibacterium and Blautia) increased, whereas the abundance levels of potential pathogenic bacteria, including Escherichia coli, Bacteroides, Ruminococcus, Parabacteroides and Prevotella decreased, among which Ruminococcus and Parabacteroides were significantly positively correlated with degree of liver fibrosis (r=0.34, P=0.04; r=0.38, P=0.02), respectively. The microbiota in the SC group did not change significantly throughout the whole process of treatment. CONCLUSION BJJP had a certain regulatory effect on intestinal microbiota of patients with hepatitis B cirrhosis/liver fibrosis (ChiCTR1800016801).
Collapse
Affiliation(s)
- Xin Chi
- Center of Liver Diseases Division 3, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- National Center for Infectious Diseases, Beijing, 100015, China
| | - Dan-Ying Cheng
- Center of Liver Diseases Division 3, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
- National Center for Infectious Diseases, Beijing, 100015, China.
- Peking University Ditan Teaching Hospital, Beijing, 100015, China.
| | - Xiu Sun
- Center of Liver Diseases Division 3, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- National Center for Infectious Diseases, Beijing, 100015, China
| | - Shun-Ai Liu
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- National Center for Infectious Diseases, Beijing, 100015, China
| | - Rong-Bing Wang
- National Center for Infectious Diseases, Beijing, 100015, China
- Central of Integrated Chinese and Western Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Qin Chen
- China Traditional Chinese Medicine Holdings Co. Limited, Guangzhou, 528303, China
| | - Hui-Chun Xing
- Center of Liver Diseases Division 3, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- National Center for Infectious Diseases, Beijing, 100015, China
- Peking University Ditan Teaching Hospital, Beijing, 100015, China
| |
Collapse
|
7
|
Protective Effects of Clinacanthus nutans (Burm.f.) Lindau Aqueous Extract on HBV Mouse Model by Modulating Gut Microbiota and Liver Metabolomics. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:5625222. [PMID: 36636608 PMCID: PMC9831714 DOI: 10.1155/2023/5625222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/04/2022] [Accepted: 12/13/2022] [Indexed: 01/14/2023]
Abstract
Background Clinacanthus nutans (Burm.f.) Lindau (C. nutans) has been used in the therapy of hepatitis B (HB) and is effective; however, the mechanism of action has not been elucidated. Objective To investigate the protective effects of C. nutans aqueous extract on the hepatitis B virus (HBV) mouse model based on correlation analysis between gut microbiota and liver metabolomics. Materials and Methods We firstly constructed the animal model by high-pressure injection of pcDNA3.1(+)/HBV plasmid into the tail vein and treated it with C. nutans. The biomarkers and inflammatory cytokines of HB were detected by enzyme-linked immunosorbent assay and quantitative PCR; the Illumina-MiSeq platform was used for investigating gut microbiota; the LC-MS/MS method was utilized on screening liver tissue metabolites; multiomics joint analysis was performed using the R program. Results Compared with the modeling group, C. nutans significantly decreased the expression levels of HBsAg, IL-1β, TNF-α(P < 0.05) in the serum, and cccDNA (P < 0.05) in the liver tissues of mice. C. nutans dramatically reduced the ratio of Firmicutes and Bacteroidetes (P < 0.05) and significantly declined the proportion of Lactobacillaceae and Lactobacillus(P < 0.05), dramatically increasing the relative abundance of Bacteroidales_S24-7_group, Rikenellaceae, and Alistipes(P < 0.05); LC-MS/MS analysis results showed that C. nutans dramatically upregulate hippuric acid, L-histidine, trehalose, D-threitol, and stachyose and downregulate uridine 5'-diphosphate, cholic acid, trimethylamine N-oxide, CDP-ethanolamine, and phosphorylcholine (P < 0.05). The correlation analysis revealed that C. nutans affects the related metabolite levels of hippuric acid and cholic acid through the modulation of crucial bacteria (Alistipes) (P < 0.01), exerting specific anti-inflammatory effects. Conclusion These results suggest that C. nutans exerts protective effects in HBV model mice, showing the therapeutic potential for anti-HBV infection.
Collapse
|
8
|
Muñoz L, Caparrós E, Albillos A, Francés R. The shaping of gut immunity in cirrhosis. Front Immunol 2023; 14:1139554. [PMID: 37122743 PMCID: PMC10141304 DOI: 10.3389/fimmu.2023.1139554] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Abstract
Cirrhosis is the common end-stage of chronic liver diseases of different etiology. The altered bile acids metabolism in the cirrhotic liver and the increase in the blood-brain barrier permeability, along with the progressive dysbiosis of intestinal microbiota, contribute to gut immunity changes, from compromised antimicrobial host defense to pro-inflammatory adaptive responses. In turn, these changes elicit a disruption in the epithelial and gut vascular barriers, promoting the increased access of potential pathogenic microbial antigens to portal circulation, further aggravating liver disease. After summarizing the key aspects of gut immunity during homeostasis, this review is intended to update the contribution of liver and brain metabolites in shaping the intestinal immune status and, in turn, to understand how the loss of homeostasis in the gut-associated lymphoid tissue, as present in cirrhosis, cooperates in the advanced chronic liver disease progression. Finally, several therapeutic approaches targeting the intestinal homeostasis in cirrhosis are discussed.
Collapse
Affiliation(s)
- Leticia Muñoz
- Departamento de Medicina y Especialidades Médicas, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Esther Caparrós
- Grupo de Inmunobiología Hepática e Intestinal, Departamento Medicina Clínica, Universidad Miguel Hernández, San Juan, Spain
- Instituto de Investigación Sanitaria ISABIAL, Hospital General Universitario de Alicante, Alicante, Spain
| | - Agustín Albillos
- Departamento de Medicina y Especialidades Médicas, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Gastroenterología y Hepatología, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- *Correspondence: Agustín Albillos, ; Rubén Frances,
| | - Rubén Francés
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
- Grupo de Inmunobiología Hepática e Intestinal, Departamento Medicina Clínica, Universidad Miguel Hernández, San Juan, Spain
- Instituto de Investigación Sanitaria ISABIAL, Hospital General Universitario de Alicante, Alicante, Spain
- Instituto de Investigación, Desarrollo e Innovación en Biotecnologiía Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Elche, Spain
- *Correspondence: Agustín Albillos, ; Rubén Frances,
| |
Collapse
|
9
|
Miao Y, Zhang Q, Yuan Z, Wang J, Xu Y, Chai Y, Du M, Yu Q, Zhang L, Jiang Z. Proteomics analysis reveals novel insights into the mechanism of hepatotoxicity induced by Tripterygium wilfordii multiglycoside in mice. Front Pharmacol 2022; 13:1032741. [DOI: 10.3389/fphar.2022.1032741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/07/2022] [Indexed: 11/18/2022] Open
Abstract
Tripterygium wilfordii multiglycoside (GTW), extracted and purified from the peeled roots of T. wilfordii Hook.f. (TwHF), is a well-known traditional Chinese medicine and applied to various autoimmune diseases clinically. However, it has been reported to cause severe liver injury. At present, the mechanism underlying GTW-induced hepatotoxicity remain poorly defined. Here, we evaluated the effects of GTW on mouse liver and elucidated the associated mechanisms via label-free proteomics combined with bioinformatics analysis. Male C57BL/6J mice were randomly divided into normal group, a low-dose GTW (70 mg/kg) group and a high-dose GTW (140 mg/kg) group. After 1-week administration, GTW dose-dependently induced hepatotoxicity. Further analysis showed that GTW could act on the intestinal immune network for IgA production pathway, which plays an important role in maintaining intestinal homeostasis and influences the crosstalk between gut and liver. Western blots confirmed that GTW could decrease pIgR protein expression in the liver and ileum, and, as a result, the secretion of IgA into gut lumen was reduced. Further validation showed that intestinal barrier integrity was impaired in GTW-treated mice, promoting bacteria transferring to the liver and triggering proinflammatory response. Our study demonstrated that gut-liver axis may play a vital part in the progression of GTW-induced hepatotoxicity, which provides guidance for basic research and clinical application of GTW.
Collapse
|
10
|
Age-Related NAFLD: The Use of Probiotics as a Supportive Therapeutic Intervention. Cells 2022; 11:cells11182827. [PMID: 36139402 PMCID: PMC9497179 DOI: 10.3390/cells11182827] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/26/2022] [Accepted: 09/08/2022] [Indexed: 11/24/2022] Open
Abstract
Human aging, a natural process characterized by structural and physiological changes, leads to alterations of homeostatic mechanisms, decline of biological functions, and subsequently, the organism becomes vulnerable to external stress or damage. In fact, the elderly population is prone to develop diseases due to deterioration of physiological and biological systems. With aging, the production of reactive oxygen species (ROS) increases, and this causes lipid, protein, and DNA damage, leading to cellular dysfunction and altered cellular processes. Indeed, oxidative stress plays a key role in the pathogenesis of several chronic disorders, including hepatic diseases, such as non-alcoholic fatty liver disease (NAFLD). NAFLD, the most common liver disorder in the Western world, is characterized by intrahepatic lipid accumulation; is highly prevalent in the aging population; and is closely associated with obesity, insulin resistance, hypertension, and dyslipidemia. Among the risk factors involved in the pathogenesis of NAFLD, the dysbiotic gut microbiota plays an essential role, leading to low-grade chronic inflammation, oxidative stress, and production of various toxic metabolites. The intestinal microbiota is a dynamic ecosystem of microbes involved in the maintenance of physiological homeostasis; the alteration of its composition and function, during aging, is implicated in different liver diseases. Therefore, gut microbiota restoration might be a complementary approach for treating NAFLD. The administration of probiotics, which can relieve oxidative stress and elicit several anti-aging properties, could be a strategy to modify the composition and restore a healthy gut microbiota. Indeed, probiotics could represent a valid supplement to prevent and/or help treating some diseases, such as NAFLD, thus improving the already available pharmacological intervention. Moreover, in aging, intervention of prebiotics and fecal microbiota transplantation, as well as probiotics, will provide novel therapeutic approaches. However, the relevant research is limited, and several scientific research works need to be done in the near future to confirm their efficacy.
Collapse
|
11
|
Beyoğlu D, Idle JR. The gut microbiota - a vehicle for the prevention and treatment of hepatocellular carcinoma. Biochem Pharmacol 2022; 204:115225. [PMID: 35998677 DOI: 10.1016/j.bcp.2022.115225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) arises principally against a background of cirrhosis and these two diseases are responsible globally for over 2 million deaths a year. There are few treatment options for liver cirrhosis and HCC, so it is vital to arrest these pathologies early in their development. To do so, we propose dietary and therapeutic solutions that involve the gut microbiota and its consequences. Integrated dietary, environmental and intrinsic signals result in a bidirectional connection between the liver and the gut with its microbiota, known as the gut-liver axis. Numerous lifestyle factors can result in dysbiosis with a change in the functional composition and metabolic activity of the microbiota. A panoply of metabolites can be produced by the microbiota, including ethanol, secondary bile acids, trimethylamine, indole, quinolone, phenazine and their derivatives and the quorum sensor acyl homoserine lactones that may contribute to HCC but have yet to be fully investigated. Gram-negative bacteria can activate the pattern recognition receptor toll-like receptor 4 (TLR4) in the liver leading to nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling, which can contribute to HCC initiation and progression. The goal in preventing HCC should be to ensure a healthy gut microbiota using probiotic supplements containing beneficial bacteria and prebiotic plant fibers such as oligosaccharides that stimulate their growth. The clinical development of TLR4 antagonists is urgently needed to counteract the pathological effects of dysbiosis on the liver and other organs. Further nutrigenomic studies are required to understand better how the diet influences the gut microbiota and its adverse effects on the liver.
Collapse
Affiliation(s)
- Diren Beyoğlu
- Arthur G. Zupko Institute for Systems Pharmacology and Pharmacogenomics, Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, New York 11201, USA
| | - Jeffrey R Idle
- Arthur G. Zupko Institute for Systems Pharmacology and Pharmacogenomics, Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, New York 11201, USA.
| |
Collapse
|
12
|
Wang T, Li XJ, Qin LH, Liang X, Xue HH, Guo J, Li SF, Zhang LW. Better detoxifying effect of ripe forsythiae fructus over green forsythiae fructus and the potential mechanisms involving bile acids metabolism and gut microbiota. Front Pharmacol 2022; 13:987695. [PMID: 36034807 PMCID: PMC9417252 DOI: 10.3389/fphar.2022.987695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Forsythiae Fructus (FF), the fruit of Forsythia suspensa (Thunb.) Vahl. (Lianqiao), is one of the most fundamental herbs in Traditional Chinese Medicines (TCM), mainly due to its heat-clearing and detoxifying effects. There are two types of FF, the greenish fruits that start to ripen (GF) and the yellow fruits that are fully ripe (RF), called “Qingqiao” and “Laoqiao” referred to the Chinese Pharmacopoeia, respectively. It undergoes a complex series of changes during the maturation of FF. However, the clinical uses and preparation of phytopharmaceuticals of FF have not been distinguished to date. Moreover, there is limited information on the study of the difference in pharmacological activity between RF and GF. In this study, a rat model of bile duct ligation (BDL)-induced cholestasis was used to compare the differences in their effects. RF was found to have better results than GF in addressing toxic bile acids (BAs) accumulation and related pathological conditions caused by BDL. The underlying mechanism may be related to the interventions of gut microbiota. The results of the present study suggest that the better detoxifying effect of RF than GF may be indirectly exerted through the regulation of gut microbiota and thus the improvement of BAs metabolism.
Collapse
Affiliation(s)
- Tao Wang
- Institute of Molecule Science, Modern Research Center for Traditional Chinese Medicine, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
- Department of Pharmacy, Changzhi Medical College, Changzhi, China
| | - Xu-Jiong Li
- Department of Physiology, Changzhi Medical College, Changzhi, China
- *Correspondence: Xu-Jiong Li, ; Li-Wei Zhang,
| | - Ling-Hao Qin
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xue Liang
- Institute of Molecule Science, Modern Research Center for Traditional Chinese Medicine, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
| | - Huan-Huan Xue
- Institute of Molecule Science, Modern Research Center for Traditional Chinese Medicine, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
| | - Jing Guo
- Institute of Molecule Science, Modern Research Center for Traditional Chinese Medicine, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
| | - Shi-Fei Li
- Institute of Molecule Science, Modern Research Center for Traditional Chinese Medicine, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
| | - Li-Wei Zhang
- Institute of Molecule Science, Modern Research Center for Traditional Chinese Medicine, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
- *Correspondence: Xu-Jiong Li, ; Li-Wei Zhang,
| |
Collapse
|
13
|
Yang AM, Lin CY, Liu SH, Syu GD, Sun HJ, Lee KC, Lin HC, Hou MC. Saccharomyces Boulardii Ameliorates Non-alcoholic Steatohepatitis in Mice Induced by a Methionine-Choline-Deficient Diet Through Gut-Liver Axis. Front Microbiol 2022; 13:887728. [PMID: 35814685 PMCID: PMC9260146 DOI: 10.3389/fmicb.2022.887728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is affecting people worldwide. Changes in the intestinal microbiome are crucial to NASH. A previous study showed that eradicating intestinal fungi ameliorates NASH; however, the role of intestinal fungi in the development of NASH remains unclear. Saccharomyces boulardii (SB), a dietary supplement yeast, has been reported to restore the integrity of the intestine. Here, we tested the effect of SB in the treatment of NASH. For this study, we fed eight-week-old C57/BL6 male mice either a methionine-choline deficient (MCD) diet or a normal chow diet (NCD) for eight weeks. Half of the MCD diet-fed mice were gavaged with SB (5 mg/day) once daily. The remainder of the NCD–fed mice were gavaged with normal saline as a control. The MCD diet-fed mice on SB supplement showed better liver function, less hepatic steatosis, and decreased inflammation. Both hepatic inflammatory gene expression and fibrogenic gene expression were suppressed in mice with SB gavage. Intestinal damage caused by the MCD diet was tampered with, intestine inflammation decreased, and gut permeability improved in mice that had been given the SB supplement. Deep sequencing of the fecal microbiome showed a potentially increased beneficial gut microbiota and increased microbiota diversity in the SB-supplemented mice. The SB supplement maintains gut integrity, increases microbial diversity, and increases the number of potentially beneficial gut microbiota. Thus, the SB supplement attenuates gut leakage and exerts a protective effect against NASH. Our results provide new insight into the prevention of NASH.
Collapse
Affiliation(s)
- An-Ming Yang
- Department of Internal Medicine, En Chu Kong Hospital, New Taipei City, Taiwan
- Department of Nursing, Yuanpei University of Medical Technology, Hsinchu, Taiwan
- *Correspondence: An-Ming Yang,
| | - Chien-Yu Lin
- Department of Internal Medicine, En Chu Kong Hospital, New Taipei City, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Shih-Hao Liu
- Division of Pathology, En Chu Kong Hospital, New Taipei City, Taiwan
| | - Guan-Da Syu
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - Hao-Jhe Sun
- Department of Medicine, National Yang Ming Chiao Tung University School of Medicine, Taipei City, Taiwan
| | - Kuei-Chuan Lee
- Department of Medicine, National Yang Ming Chiao Tung University School of Medicine, Taipei City, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei City, Taiwan
- Kuei-Chuan Lee,
| | - Han-Chieh Lin
- Department of Medicine, National Yang Ming Chiao Tung University School of Medicine, Taipei City, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei City, Taiwan
| | - Ming-Chih Hou
- Department of Medicine, National Yang Ming Chiao Tung University School of Medicine, Taipei City, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei City, Taiwan
| |
Collapse
|
14
|
Wang L, Chen W, Tian Y, Duan X, Yuan Y, Wang N, Xu C, Liu X, Liu Z. Preventive Effects of Sesamol on Deep‐frying Oil‐induced Liver Metabolism Disorders by Altering Gut Microbiota and Protecting Gut Barrier Integrity. Mol Nutr Food Res 2022; 66:e2101122. [DOI: 10.1002/mnfr.202101122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/07/2022] [Indexed: 11/10/2022]
Affiliation(s)
- Lei Wang
- College of Food Science and Engineering Northwest A&F University Yangling 712100 China
| | - Weixuan Chen
- College of Food Science and Engineering Northwest A&F University Yangling 712100 China
| | - Yujie Tian
- College of Food Science and Engineering Northwest A&F University Yangling 712100 China
| | - Xiaorong Duan
- College of Food Science and Engineering Northwest A&F University Yangling 712100 China
| | - Yi Yuan
- College of Food Science and Engineering Northwest A&F University Yangling 712100 China
| | - Na Wang
- College of Food Science and Technology Henan Agricultural University Zhengzhou 450002 China
- Zhengzhou Key Laboratory of Nutrition and Health Food Zhengzhou 450002 China
| | - Chao Xu
- College of Food Science and Technology Henan Agricultural University Zhengzhou 450002 China
- Zhengzhou Key Laboratory of Nutrition and Health Food Zhengzhou 450002 China
| | - Xuebo Liu
- College of Food Science and Engineering Northwest A&F University Yangling 712100 China
| | - Zhigang Liu
- College of Food Science and Engineering Northwest A&F University Yangling 712100 China
- Department of Food Science Cornell University Ithaca New York 14853 United States
| |
Collapse
|
15
|
Brown MD, Shinn LM, Reeser G, Browning M, Schwingel A, Khan NA, Holscher HD. Fecal and soil microbiota composition of gardening and non-gardening families. Sci Rep 2022; 12:1595. [PMID: 35102166 PMCID: PMC8804003 DOI: 10.1038/s41598-022-05387-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 12/30/2021] [Indexed: 02/07/2023] Open
Abstract
Historically, humans have interacted with soils, which contain a rich source of microorganisms. Fruit and vegetable gardening is the primary interaction humans have with soil today. Animal research reveals that soil microorganisms can be transferred to the rodent intestine. However, studies on fecal and soil microbial changes associated with gardening in humans are lacking. The current case-controlled cohort study aimed to characterize the fecal and soil microbiota of gardening families (n = 10) and non-gardening (control) families (n = 9). Families included two adults and one child (5-18 years) for a total of 56 participants. All participants provided a fecal sample, soil sample, and diet history questionnaires before the gardening season (April) and during the peak of the gardening season (August). Healthy Eating Index (HEI-2015) scores and nutrient analysis were performed. Fecal and soil DNA were extracted and amplified. Sequence data were then processed and analyzed. Peak season gardening families tended to have greater fecal operational features, a greater Faith's Phylogenetic Diversity score, greater fiber intake, and higher abundances of fiber fermenting bacteria than peak control families. Soil endemic microbes were also shared with gardening participant's fecal samples. This study revealed that the fecal microbiota of gardening families differs from non-gardening families, and that there are detectable changes in the fecal microbial community of gardeners and their family members over the course of the gardening season. Additional research is necessary to determine if changes induced by gardening on the gut microbiota contribute to human health.
Collapse
Affiliation(s)
- Marina D Brown
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Leila M Shinn
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Ginger Reeser
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Matthew Browning
- Parks, Recreation, and Tourism Management, Clemson University, Clemson, SC, USA
| | - Andiara Schwingel
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Naiman A Khan
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Family Resiliency Center, University of Illinois, Urbana, IL, USA
| | - Hannah D Holscher
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Family Resiliency Center, University of Illinois, Urbana, IL, USA.
| |
Collapse
|
16
|
Juanola O, Hassan M, Kumar P, Yilmaz B, Keller I, Simillion C, Engelmann C, Tacke F, Dufour JF, De Gottardi A, Moghadamrad S. Intestinal microbiota drives cholestasis-induced specific hepatic gene expression patterns. Gut Microbes 2022; 13:1-20. [PMID: 33847205 PMCID: PMC8049203 DOI: 10.1080/19490976.2021.1911534] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Intestinal microbiota regulates multiple host metabolic and immunological processes. Consequently, any difference in its qualitative and quantitative composition is susceptible to exert significant effects, in particular along the gut-liver axis. Indeed, recent findings suggest that such changes modulate the severity and the evolution of a wide spectrum of hepatobiliary disorders. However, the mechanisms linking intestinal microbiota and the pathogenesis of liver disease remain largely unknown. In this work, we investigated how a distinct composition of the intestinal microbiota, in comparison with germ-free conditions, may lead to different outcomes in an experimental model of acute cholestasis. Acute cholestasis was induced in germ-free (GF) and altered Schaedler's flora (ASF) colonized mice by common bile duct ligation (BDL). Studies were performed 5 days after BDL and hepatic histology, gene expression, inflammation, lipids metabolism, and mitochondrial functioning were evaluated in normal and cholestatic mice. Differences in plasma concentration of bile acids (BA) were evaluated by UHPLC-HRMS. The absence of intestinal microbiota was associated with significant aggravation of hepatic bile infarcts after BDL. At baseline, we found the absence of gut microbiota induced altered expression of genes involved in the metabolism of fatty and amino acids. In contrast, acute cholestasis induced altered expression of genes associated with extracellular matrix, cell cycle, autophagy, activation of MAPK, inflammation, metabolism of lipids, and mitochondrial functioning pathways. Ductular reactions, cell proliferation, deposition of collagen 1 and autophagy were increased in the presence of microbiota after BDL whereas GF mice were more susceptible to hepatic inflammation as evidenced by increased gene expression levels of osteopontin, interleukin (IL)-1β and activation of the ERK/MAPK pathway as compared to ASF colonized mice. Additonally, we found that the presence of microbiota provided partial protection to the mitochondrial functioning and impairment in the fatty acid metabolism after BDL. The concentration of the majority of BA markedly increased after BDL in both groups without remarkable differences according to the hygiene status of the mice. In conclusion, acute cholestasis induced more severe liver injury in GF mice compared to mice with limited intestinal bacterial colonization. This protective effect was associated with different hepatic gene expression profiles mostly related to tissue repair, metabolic and immune functions. Our findings suggest that microbial-induced differences may impact the course of cholestasis and modulate liver injury, offering a background for novel therapies based on the modulation of the intestinal microbiota.
Collapse
Affiliation(s)
- Oriol Juanola
- Translational Research Laboratory, Gastroenterology and Hepatology, Ente Ospedaliero Cantonale, Università Della Svizzera Italiana, Lugano, Switzerland
| | - Mohsin Hassan
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Pavitra Kumar
- Hepatology, Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Bahtiyar Yilmaz
- Gastroenterology, Department for Biomedical Research, University of Bern, Bern, Switzerland,University Clinic of Visceral Surgery and Medicine, Inselspital, Bern, Switzerland
| | - Irene Keller
- Department for Biomedical Research and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Cédric Simillion
- Department for Biomedical Research and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Cornelius Engelmann
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany,Institute for Liver and Digestive Health, University College London, London, UK,Berlin Institute of Health (BIH), Berlin, Germany
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jean-François Dufour
- Hepatology, Department for Biomedical Research, University of Bern, Bern, Switzerland,University Clinic of Visceral Surgery and Medicine, Inselspital, Bern, Switzerland
| | - Andrea De Gottardi
- Translational Research Laboratory, Gastroenterology and Hepatology, Ente Ospedaliero Cantonale, Università Della Svizzera Italiana, Lugano, Switzerland,Hepatology, Department for Biomedical Research, University of Bern, Bern, Switzerland,University Clinic of Visceral Surgery and Medicine, Inselspital, Bern, Switzerland
| | - Sheida Moghadamrad
- Translational Research Laboratory, Gastroenterology and Hepatology, Ente Ospedaliero Cantonale, Università Della Svizzera Italiana, Lugano, Switzerland,Hepatology, Department for Biomedical Research, University of Bern, Bern, Switzerland,University Clinic of Visceral Surgery and Medicine, Inselspital, Bern, Switzerland,CONTACT Sheida Moghadamrad Translational Research Laboratory, Gastroenterology and Hepatology, Ente Ospedaliero Cantonale, Università Della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
17
|
Derbak MA, Vorobets VV, Koval GM, Nikolska OІ, Ustych OV, Hechko MM, Ilko AV. ASSESSMENT OF COLON MICROBIOCENOSIS DISORDERS IN PATIENTS WITH CHRONIC HEPATITIS C. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2022; 75:2334-2338. [PMID: 36472257 DOI: 10.36740/wlek202210104] [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: 06/17/2023]
Abstract
OBJECTIVE The aim: To investigate the peculiarities of colon microbiocenosis disorders in patients with chronic hepatitis C. PATIENTS AND METHODS Materials and methods: 142 patients with CHC were under observation, determination of the degree of liver fibrosis (FibroMax), bacteriological examination of stools and pancreatic elastase was performed. RESULTS Results: It was found that 59.2% of patients with CHC had gut dysbiosis (DB), of which 61.9% had increased body weight. Intestinal microbiocenosis disorders were manifested by constipation in 57.1% of patients, diarrhea in 31% of patients, and alternating constipation and diarrhea in 11.9% of patients. Bacteriologically, gut dysbiosis was character¬ized by suppression of the growth of normal microflora: Escherichia coli in 47.6%, bifidobacteria in 61.9%, lactobacilli in 53.6%, complete absence of bifidobacteria in 20.2% of cases. In patients with CHC combined with DB deep stages of liver fibrosis (F2-3 and F3-4) are registered 3.6 times more often compared to patients without intestinal dysbiosis (53.6% versus 24.1% and 11.9% versus 3.4%). The degree of gut DB increased in proportion to the stage of liver fibrosis (p<0.05). 32.1% of patients with CHC with dysbiosis were diagnosed with exocrine insufficiency of the pancreas. CONCLUSION Conclusions: Gut dysbiosis occurs more often in CHC patients with increased body weight and is characterized by constipation in 59.2% of patients. Intestinal microbiocenosis is characterized by suppression of the growth of normal microflora. In 32.1% of CHC patients with intestinal dysbiosis, according to the results of the pancreatic elastase-1 test, pancreatic exocrine insufficiency of various degrees was found.
Collapse
|
18
|
Kassa Y, Million Y, Gedefie A, Moges F. Alteration of Gut Microbiota and Its Impact on Immune Response in Patients with Chronic HBV Infection: A Review. Infect Drug Resist 2021; 14:2571-2578. [PMID: 34262302 PMCID: PMC8274626 DOI: 10.2147/idr.s305901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic hepatitis B virus infection is a source of substantial global health problems, particularly in economically underdeveloped and/or developing countries. It is the primary cause of severe liver disorders such as liver fibrosis, cirrhosis, and hepatocellular carcinoma. The liver is connected by the bile duct to the small intestine that carries bile produced in the liver to the intestine. The liver is the initial organ exposed to materials originating from the gut including dietary compounds, bacteria, and their products. Human intestines harbor a wide diversity of the community of microbes which are collectively termed as gut microbiota. In chronic infection with the hepatitis B virus, microbial alteration of the gut is a source of systemic immune activation. Besides, gut permeability is altered in hepatitis B virus-infected patients with an increased bacterial translocation and endotoxin load in the portal vein that caused toll-like receptor activation in the liver, which facilitates immune-mediated liver injury. Toll-like receptors further triggered the host-wide inflammatory response by inducing signaling cascades such as nuclear factor-kappa B-linked pathways and by accelerating cytokine secretion like tumor necrosis factor-alpha, which evokes chronic inflammation and leads to liver lesion formation, fibrosis progression, and cirrhosis and hepatocellular carcinoma development. In conclusion, changes in intestinal flora play an important role in encouraging the production of chronic infection with the hepatitis B virus. Therefore, careful attention should be given to the maintenance of intestinal microecology of patients which can provide a sound foundation for the treatment of chronic infection with the hepatitis B virus.
Collapse
Affiliation(s)
- Yeshimebet Kassa
- Department of Medical Laboratory Sciences, College of Medicine and Health Science, Wollo University, Dessie, Ethiopia
| | - Yihenew Million
- Department of Medical Microbiology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Alemu Gedefie
- Department of Medical Laboratory Sciences, College of Medicine and Health Science, Wollo University, Dessie, Ethiopia
| | - Feleke Moges
- Department of Medical Microbiology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
19
|
Galán MG, Cian RE, Albarracín M, López-Oliva Muñoz ME, Weisstaub A, Zuleta A, Drago SR. Refined sorghum flours precooked by extrusion enhance the integrity of the colonic mucosa barrier and promote a hepatic antioxidant environment in growing Wistar rats. Food Funct 2021; 11:7638-7650. [PMID: 32966464 DOI: 10.1039/d0fo01160f] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The effects of precooked-refined sorghum flour consumption on antioxidant status, lipid profile, and colonic and bone health were evaluated. Twenty-four male Wistar rats were fed with control diet (C), or red or white precooked-refined sorghum based diets (SD) for 60 days. The intake of SD was lower than that of C, but the efficiency of all diets was similar. Rats fed with SD showed lower feces excretion, cecal pH and enzyme activities (β-glucosidase, β-glucuronidase and mucinase) than C. White SD improved intestinal architecture, cell proliferation and apoptosis, upregulated ZO1 and occludin tight junction proteins and stimulated goblet cell differentiation, enhancing the integrity of the mucosa barrier in both proximal and distal colonic mucosa in a better way than red SD. Consumption of SD significantly decreased serum triglyceride levels compared with the C diet. The mineral content of the right femur was not different among diets. The liver enzyme activities (superoxide dismutase, catalase, glutathione reductase, and glutathione peroxidase) did not show differences among diets. Liver reducing power and reduced glutathione/oxidize glutathione ratio were higher for animals consuming SD than C. It can be concluded that the consumption of precooked refined sorghum flours still has beneficial effects for health, mainly at the colonic level, despite the lower phenolics and fibre contents of refined flours with respect to whole grain flours.
Collapse
Affiliation(s)
- María Gimena Galán
- Instituto de Tecnología de Alimentos, CONICET, FIQ - UNL, 1° de Mayo 3250, (3000), Santa Fe, Argentina.
| | - Raúl Esteban Cian
- Instituto de Tecnología de Alimentos, CONICET, FIQ - UNL, 1° de Mayo 3250, (3000), Santa Fe, Argentina.
| | - Micaela Albarracín
- Instituto de Tecnología de Alimentos, CONICET, FIQ - UNL, 1° de Mayo 3250, (3000), Santa Fe, Argentina.
| | | | - Adriana Weisstaub
- Departamento de Bromatología y Nutrición, Facultad de Farmacia y Bioquímica, (UBA), Junín 956, CABA, Argentina
| | - Angela Zuleta
- Departamento de Bromatología y Nutrición, Facultad de Farmacia y Bioquímica, (UBA), Junín 956, CABA, Argentina
| | - Silvina Rosa Drago
- Instituto de Tecnología de Alimentos, CONICET, FIQ - UNL, 1° de Mayo 3250, (3000), Santa Fe, Argentina.
| |
Collapse
|
20
|
Bennett H, Troutman TD, Sakai M, Glass CK. Epigenetic Regulation of Kupffer Cell Function in Health and Disease. Front Immunol 2021; 11:609618. [PMID: 33574817 PMCID: PMC7870864 DOI: 10.3389/fimmu.2020.609618] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/08/2020] [Indexed: 12/13/2022] Open
Abstract
Kupffer cells, the resident macrophages of the liver, comprise the largest pool of tissue macrophages in the body. Within the liver sinusoids Kupffer cells perform functions common across many tissue macrophages including response to tissue damage and antigen presentation. They also engage in specialized activities including iron scavenging and the uptake of opsonized particles from the portal blood. Here, we review recent studies of the epigenetic pathways that establish Kupffer cell identity and function. We describe a model by which liver-environment specific signals induce lineage determining transcription factors necessary for differentiation of Kupffer cells from bone-marrow derived monocytes. We conclude by discussing how these lineage determining transcription factors (LDTFs) drive Kupffer cell behavior during both homeostasis and disease, with particular focus on the relevance of Kupffer cell LDTF pathways in the setting of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis.
Collapse
Affiliation(s)
- Hunter Bennett
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Ty D Troutman
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Mashito Sakai
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States.,Department of Biochemistry & Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States.,Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
21
|
Proteomic Analysis of the Protective Effect of Early Heat Exposure against Chronic Heat Stress in Broilers. Animals (Basel) 2020; 10:ani10122365. [PMID: 33321873 PMCID: PMC7764366 DOI: 10.3390/ani10122365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/09/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Heat stress affects the livestock industry, especially in poultry. Screening for metabolic changes after early and chronic heat exposure in poultry would be beneficial in resolving the production issues. In this study, we identified differentially expressed proteins that affected early heat exposure during chronic heat stress. Chronic heat stress affected 277 proteins, of which 95 differed in expression by early heat exposure. Differentially expressed proteins were related to actin metabolism and also involved in carbohydrate and carbon metabolism. According to our results, early heat exposed liver of broilers activates the different physiological mechanisms for protection from later heat stress. Abstract The increasing trend of global warming has affected the livestock industry through the heat stress, especially in poultry. Therefore, a better understanding of the mechanisms of heat stress in poultry would be helpful for maintaining the poultry production. Three groups were designed to determine early heat stress effects during chronic heat stress: CC, raised at a comfortable temperature; CH, chronic heat exposure at 35 °C for 21–35 days continuously; and HH, early heat exposure at 40 °C for 24 h at 5 days old with 35 °C temperature for 21–35 days continuously. In this study, proteome analysis was carried out to identify differentially expressed proteins in the liver tissue of broilers under chronic and early heat exposure. There were eight differentially expressed proteins from early heat stress during chronic heat exposure, which were related to actin metabolism. According to KEGG (Kyoto encyclopedia of genes and genomes) analysis, the proteins involved in carbohydrate metabolism were expressed to promote the metabolism of carbohydrates under chronic heat stress. Early heat reduced the heat stress-induced expression changes of select proteins. Our study has shown that early heat exposure suggests that the liver of broilers has various physiological mechanisms for regulating homeostasis to aid heat resistance.
Collapse
|
22
|
Hasan Siddiqui S, Kang D, Park J, Choi HW, Shim K. Acute Heat Stress Induces the Differential Expression of Heat Shock Proteins in Different Sections of the Small Intestine of Chickens Based on Exposure Duration. Animals (Basel) 2020; 10:ani10071234. [PMID: 32708054 PMCID: PMC7401550 DOI: 10.3390/ani10071234] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
In this study, we examined the protein and gene expression of heat shock proteins (HSPs) in different sections of the small intestine of chickens. In total, 300 one-day-old Ross 308 broiler chicks were randomly allocated to the control and treatment groups. The treatment group was divided into four subgroups, according to the duration of acute heat exposure (3, 6, 12, and 24 h). The influence of heat stress on the protein and gene expression of HSP70, HSP60, and HSP47 in different sections of the small intestine of chickens was determined. The protein expression of HSP70 and HSP60 was significantly higher at 6 h in the duodenum and jejunum and 12 h in the ileum. The HSP47 protein expression was significantly higher at 3 h in the duodenum and ileum and at 6 h in the jejunum. The gene expression levels of HSP70, HSP60, and HSP47 were significantly higher at the 3 h treatment group than the control group in the duodenum, jejunum, and ileum. The glutamate pyruvate transaminase and glutamate oxaloacetate transaminase levels were significantly higher at 12 and 24 h in the serum of the blood. Acute heat stress affected the expression of intestinal proteins and genes in chickens, until the induction of heat tolerance.
Collapse
Affiliation(s)
- Sharif Hasan Siddiqui
- Department of Animal Biotechnology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju 54896, Korea; (S.H.S.); (D.K.); (J.P.)
| | - Darae Kang
- Department of Animal Biotechnology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju 54896, Korea; (S.H.S.); (D.K.); (J.P.)
| | - Jinryong Park
- Department of Animal Biotechnology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju 54896, Korea; (S.H.S.); (D.K.); (J.P.)
| | - Hyun Woo Choi
- Department of Animal Science, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju 54896, Korea
- Correspondence: (H.W.C.); (K.S.); Tel.: +82-10-3169-9417 (H.W.C.); +82-10-3889-1003 (K.S.)
| | - Kwanseob Shim
- Department of Animal Biotechnology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju 54896, Korea; (S.H.S.); (D.K.); (J.P.)
- Correspondence: (H.W.C.); (K.S.); Tel.: +82-10-3169-9417 (H.W.C.); +82-10-3889-1003 (K.S.)
| |
Collapse
|
23
|
Stadlbauer V, Komarova I, Klymiuk I, Durdevic M, Reisinger A, Blesl A, Rainer F, Horvath A. Disease severity and proton pump inhibitor use impact strongest on faecal microbiome composition in liver cirrhosis. Liver Int 2020; 40:866-877. [PMID: 31943691 PMCID: PMC7187411 DOI: 10.1111/liv.14382] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/13/2019] [Accepted: 01/07/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Compositional changes of the faecal microbiome in cirrhosis are well described and have been associated with complications and prognosis. However, it is less well known, which disease or treatment-related factors affect microbiome composition most distinctively. METHODS 16S rDNA sequencing data of 88 cirrhotic outpatients were investigated. Factors influencing microbiome composition were analysed by univariate and multivariate redundancy analysis. The association of the identified factors with changes in diversity and taxonomic composition was studied in depth using analysis of composition of microbiome, LDA-effect size and least absolute shrinkage and selection operator regularized regression. RESULTS Disease severity and aetiology, proton pump inhibitor (PPI) use, nutritional status, age and C-reactive protein are significant explanatory variables for faecal microbiome composition in liver cirrhosis. Despite some taxonomic overlaps especially between disease severity and PPI use, we could show that the effects of disease severity, aetiology, PPI use and age are independent factors influencing microbiome composition also in subgroup analyses. CONCLUSION Our cross sectional system biology study identifies disease severity, aetiology, PPI use and age as independent factors that influence microbiome composition in liver cirrhosis. In chronic diseases with high morbidity, such as liver cirrhosis, precise patient metadata documentation is of utmost importance in microbiome analysis. Further studies with a higher sample size are necessary to validate this finding. TRIAL REGISTRATION NUMBER NCT01607528.
Collapse
Affiliation(s)
- Vanessa Stadlbauer
- Division of Gastroenterology and HepatologyDepartment of Internal MedicineMedical University of GrazGrazAustria,Center for Biomarker Research in Medicine (CBmed)GrazAustria
| | - Irina Komarova
- Division of Gastroenterology and HepatologyDepartment of Internal MedicineMedical University of GrazGrazAustria
| | - Ingeborg Klymiuk
- Center for Medical ResearchMedical University of GrazGrazAustria
| | - Marija Durdevic
- Center for Medical ResearchMedical University of GrazGrazAustria,Institute of PathologyMedical University of GrazGrazAustria
| | - Alexander Reisinger
- Intensive Care UnitDepartment of Internal MedicineMedical University of GrazGrazAustria
| | - Andreas Blesl
- Division of Gastroenterology and HepatologyDepartment of Internal MedicineMedical University of GrazGrazAustria
| | - Florian Rainer
- Division of Gastroenterology and HepatologyDepartment of Internal MedicineMedical University of GrazGrazAustria
| | - Angela Horvath
- Division of Gastroenterology and HepatologyDepartment of Internal MedicineMedical University of GrazGrazAustria,Center for Biomarker Research in Medicine (CBmed)GrazAustria
| |
Collapse
|
24
|
Metabolomic Analysis of the Liver of a Dextran Sodium Sulfate-Induced Acute Colitis Mouse Model: Implications of the Gut-Liver Connection. Cells 2020; 9:cells9020341. [PMID: 32024178 PMCID: PMC7072179 DOI: 10.3390/cells9020341] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/21/2020] [Accepted: 01/29/2020] [Indexed: 01/18/2023] Open
Abstract
The incidence of ulcerative colitis (UC) is increasing worldwide, and it has become a growing problem in Asia. Previous research on UC has focused on serum, plasma, urine, gut tissues, and fecal metabolic profiling, but a comprehensive investigation into the correlation between the severity of colitis and changes in liver metabolism is still lacking. Since the liver and gut exchange nutrients and metabolites through a complex network, intestinal diseases can affect both the liver and other organs. In the present study, concentration-dependent dextran sodium sulfate (DSS)-induced ulcerative colitis was employed to examine changes in liver metabolism using a proton nuclear magnetic resonance spectroscopy (1H-NMR)-and ultra-performance liquid chromatography time of flight mass spectroscopy (UPLC-TOF MS)-based metabolomics study. Using the multivariate statistical analysis method orthogonal projections to latent structures discriminant analysis (OPLS-DA), changes in metabolites depending on the DSS dose could be clearly distinguished. Specifically, hepatic metabolites involved in one-carbon metabolism, carnitine-related metabolism, and nucleotide synthesis were found to be affected by intestinal inflammation, implying the existence of a metabolic connection between the gut and liver. We are currently investigating the significance of this metabolic condition in UC.
Collapse
|
25
|
Vakhrushev YM, Suchkova EV, Lukashevich AP. [Non - alcoholic fatty liver disease and enteral insufficiency: comorbidity of their development]. TERAPEVT ARKH 2019; 91:84-89. [PMID: 32598594 DOI: 10.26442/00403660.2019.12.000134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Indexed: 01/10/2023]
Abstract
The article reflects current literature data on the epidemiology and risk factors of non - alcoholic fatty liver disease. An important aspect is the description of the modern views of combined lesions of the hepatobiliary tract and small intestine. Disorders of the intestinal microbiota play a special role in the development of non - alcoholic fatty liver disease. The value of enterohepatic circulation of bile acids in the development of intestinal and liver diseases was shown. It seems relevant to further study the comorbidity of the development of non - alcoholic fatty liver disease and enteropathy for the development of pathogenetically substantiated therapy.
Collapse
|
26
|
Cabrera-Rubio R, Patterson AM, Cotter PD, Beraza N. Cholestasis induced by bile duct ligation promotes changes in the intestinal microbiome in mice. Sci Rep 2019; 9:12324. [PMID: 31444478 PMCID: PMC6707139 DOI: 10.1038/s41598-019-48784-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 07/22/2019] [Indexed: 02/08/2023] Open
Abstract
Increasing evidence point to the relevance of intestinal disfunction and changes in the microbiome composition during chronic liver disease. More specifically, recent studies have highlighted that cholestatic diseases associate with a reduction in the microbiome diversity in patients. Still, the dynamics of the changes in the microbiome composition observed, as well as their implication in contributing to the pathogenesis of this disease remain largely undefined. Hence, experimental mouse models resembling the human pathogenesis are crucial to move forward our understanding on the mechanisms underpinning cholestatic disease and to enable the development of effective therapeutics. Our results show that the bile duct ligation (BDL) experimental model of cholestasis leads to rapid and significant changes in the microbiome diversity, with more than 100 OTUs being significantly different in faecal samples obtained from WT mice at 3 days and 7 days after BDL when compared to control animals. Changes in the microbial composition in mice after BDL included the enrichment of Akkermansia, Prevotella, Bacteroides and unclassified Ruminococcaceae in parallel with a drastic reduction of the presence of Faecalibacterium prausnitzii. In conclusion, our results support that bile duct ligation induces changes in the microbiome that partly resemble the gut microbial changes observed during human cholestatic disease.
Collapse
Affiliation(s)
- Raul Cabrera-Rubio
- Teagasc Food Research Centre, Moorepark, Fermoy, Co, Cork, Ireland.,APC Microbiome Institute, University College Cork, Co, Cork, Ireland
| | - Angela M Patterson
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute, Norwich Research Park, Norwich, UK
| | - Paul D Cotter
- Teagasc Food Research Centre, Moorepark, Fermoy, Co, Cork, Ireland.,APC Microbiome Institute, University College Cork, Co, Cork, Ireland
| | - Naiara Beraza
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute, Norwich Research Park, Norwich, UK.
| |
Collapse
|
27
|
Iacob S, Iacob DG. Infectious Threats, the Intestinal Barrier, and Its Trojan Horse: Dysbiosis. Front Microbiol 2019; 10:1676. [PMID: 31447793 PMCID: PMC6692454 DOI: 10.3389/fmicb.2019.01676] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023] Open
Abstract
The ecosystem of the gut microbiota consists of diverse intestinal species with multiple metabolic and immunologic activities and it is closely connected with the intestinal epithelia and mucosal immune response, with which it builds a complex barrier against intestinal pathogenic bacteria. The microbiota ensures the integrity of the gut barrier through multiple mechanisms, either by releasing antibacterial molecules (bacteriocins) and anti-inflammatory short-chain fatty acids or by activating essential cell receptors for the immune response. Experimental studies have confirmed the role of the intestinal microbiota in the epigenetic modulation of the gut barrier through posttranslational histone modifications and regulatory mechanisms induced by epithelial miRNA in the epithelial lumen. Any quantitative or functional changes of the intestinal microbiota, referred to as dysbiosis, alter the immune response, decrease epithelial permeability and destabilize intestinal homeostasis. Consequently, the overgrowth of pathobionts (Staphylococcus, Pseudomonas, and Escherichia coli) favors intestinal translocations with Gram negative bacteria or their endotoxins and could trigger sepsis, septic shock, secondary peritonitis, or various intestinal infections. Intestinal infections also induce epithelial lesions and perpetuate the risk of bacterial translocation and dysbiosis through epithelial ischemia and pro-inflammatory cytokines. Furthermore, the decline of protective anaerobic bacteria (Bifidobacterium and Lactobacillus) and inadequate release of immune modulators (such as butyrate) affects the release of antimicrobial peptides, de-represses microbial virulence factors and alters the innate immune response. As a result, intestinal germs modulate liver pathology and represent a common etiology of infections in HIV immunosuppressed patients. Antibiotic and antiretroviral treatments also promote intestinal dysbiosis, followed by the selection of resistant germs which could later become a source of infections. The current article addresses the strong correlations between the intestinal barrier and the microbiota and discusses the role of dysbiosis in destabilizing the intestinal barrier and promoting infectious diseases.
Collapse
Affiliation(s)
- Simona Iacob
- Infectious Diseases Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.,National Institute of Infectious Diseases "Prof. Dr. Matei Balş", Bucharest, Romania
| | - Diana Gabriela Iacob
- Infectious Diseases Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
28
|
Ni J, Huang R, Zhou H, Xu X, Li Y, Cao P, Zhong K, Ge M, Chen X, Hou B, Yu M, Peng B, Li Q, Zhang P, Gao Y. Analysis of the Relationship Between the Degree of Dysbiosis in Gut Microbiota and Prognosis at Different Stages of Primary Hepatocellular Carcinoma. Front Microbiol 2019; 10:1458. [PMID: 31293562 PMCID: PMC6603198 DOI: 10.3389/fmicb.2019.01458] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/11/2019] [Indexed: 12/22/2022] Open
Abstract
Gut microbiota dysbiosis is closely associated with primary hepatocellular carcinoma (HCC). Recent studies have evaluated the early diagnosis of primary HCC through analysis of gut microbiota dysbiosis. However, the relationship between the degree of dysbiosis and the prognosis of primary HCC remains unclear. Because primary HCC is accompanied by dysbiosis and dysbiosis usually increases the circulatory concentrations of endotoxin and other harmful bacterial substances, which further increases liver damage, we hypothesized that level of dysbiosis associated with primary HCC increases with the stage of cancer progression. To test this hypothesis, we introduced a more integrated index referred to as the degree of dysbiosis (Ddys ); and we investigated Ddys of the gut microbiota with the development of primary HCC through high-throughput sequencing of 16S rRNA gene amplicons. Our results showed that compared with healthy individuals, patients with primary HCC showed increased pro-inflammatory bacteria in their fecal microbiota. The Ddys increased significantly in patients with primary HCC compared with that in healthy controls. Moreover, there was a tendency for the Ddys to increase with the development of primary HCC, although no significant difference was detected between different stages of primary HCC. Our findings provide important insights into the use of gut microbiota analysis during the treatment of primary HCC.
Collapse
Affiliation(s)
- Jiajia Ni
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Rong Huang
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, China
| | - Huifang Zhou
- Department of Clinical Laboratory, First People's Hospital of Kashi, Kashgar, China
| | - Xiaoping Xu
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Yang Li
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Peihua Cao
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Kebo Zhong
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Mei Ge
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Xiaoxia Chen
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Baohua Hou
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Min Yu
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Baogang Peng
- Department of Hepatic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiao Li
- Department of Hepatic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Peng Zhang
- Department of Organ Transplantation, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yi Gao
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| |
Collapse
|
29
|
Jindal A, Jagdish RK. Sarcopenia: Ammonia metabolism and hepatic encephalopathy. Clin Mol Hepatol 2019; 25:270-279. [PMID: 31006226 PMCID: PMC6759436 DOI: 10.3350/cmh.2019.0015] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 02/07/2019] [Indexed: 12/15/2022] Open
Abstract
Sarcopenia (loss of muscle mass and/or strength) frequently complicates liver cirrhosis and adversely affects the quality of life; cirrhosis related liver decompensation and significantly decreases wait-list and post-liver transplantation survival. The main therapeutic strategies to improve or reverse sarcopenia include dietary interventions (supplemental calorie and protein intake), increased physical activity (supervised resistance and endurance exercises), hormonal therapy (testosterone), and ammonia lowering agents (L-ornithine L-aspartate, branch chain amino acids) as well as mechanistic approaches that target underlying molecular and metabolic abnormalities. Besides other factors, hyperammonemia has recently gained attention and increase sarcopenia by various mechanisms including increased expression of myostatin, increased phosphorylation of eukaryotic initiation factor 2a, cataplerosis of α ketoglutarate, mitochondrial dysfunction, increased reactive oxygen species that decrease protein synthesis and increased autophagy-mediated proteolysis. Sarcopenia contributes to frailty and increases the risk of minimal and overt hepatic encephalopathy.
Collapse
Affiliation(s)
- Ankur Jindal
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Rakesh Kumar Jagdish
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| |
Collapse
|
30
|
González-González M, Díaz-Zepeda C, Eyzaguirre-Velásquez J, González-Arancibia C, Bravo JA, Julio-Pieper M. Investigating Gut Permeability in Animal Models of Disease. Front Physiol 2019; 9:1962. [PMID: 30697168 PMCID: PMC6341294 DOI: 10.3389/fphys.2018.01962] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/31/2018] [Indexed: 12/12/2022] Open
Abstract
A growing number of investigations report the association between gut permeability and intestinal or extra-intestinal disorders under the basis that translocation of gut luminal contents could affect tissue function, either directly or indirectly. Still, in many cases it is unknown whether disruption of the gut barrier is a causative agent or a consequence of these conditions. Adequate experimental models are therefore required to further understand the pathophysiology of health disorders associated to gut barrier disruption and to develop and test pharmacological treatments. Here, we review the current animal models that display enhanced intestinal permeability, and discuss (1) their suitability to address mechanistic questions, such as the association between gut barrier alterations and disease and (2) their validity to test potential treatments for pathologies that are characterized by enhanced intestinal permeability.
Collapse
Affiliation(s)
- Marianela González-González
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Camilo Díaz-Zepeda
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Johana Eyzaguirre-Velásquez
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Camila González-Arancibia
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Javier A Bravo
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Marcela Julio-Pieper
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| |
Collapse
|
31
|
Schneider KM, Mohs A, Kilic K, Candels LS, Elfers C, Bennek E, Schneider LB, Heymann F, Gassler N, Penders J, Trautwein C. Intestinal Microbiota Protects against MCD Diet-Induced Steatohepatitis. Int J Mol Sci 2019; 20:308. [PMID: 30646522 PMCID: PMC6358781 DOI: 10.3390/ijms20020308] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/14/2018] [Accepted: 01/08/2019] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease in western countries, with a continuously rising incidence. Gut-liver communication and microbiota composition have been identified as critical drivers of the NAFLD progression. Hence, it has been shown that microbiota depletion can ameliorate high-fat diet or western-diet induced experimental Non-alcoholic steatohepatitis (NASH). However, its functional implications in the methionine-choline dietary model, remain incompletely understood. Here, we investigated the physiological relevance of gut microbiota in methionine-choline deficient (MCD) diet induced NASH. Experimental liver disease was induced by 8 weeks of MCD feeding in wild-type (WT) mice, either with or without commensal microbiota depletion, by continuous broad-spectrum antibiotic (AB) treatment. MCD diet induced steatohepatitis was accompanied by a reduced gut microbiota diversity, indicating intestinal dysbiosis. MCD treatment prompted macroscopic shortening of the intestine, as well as intestinal villi in histology. However, gut microbiota composition of MCD-treated mice, neither resembled human NASH, nor did it augment the intestinal barrier integrity or intestinal inflammation. In the MCD model, AB treatment resulted in increased steatohepatitis activity, compared to microbiota proficient control mice. This phenotype was driven by pronounced neutrophil infiltration, while AB treatment only slightly increased monocyte-derived macrophages (MoMF) abundance. Our data demonstrated the differential role of gut microbiota, during steatohepatitis development. In the context of MCD induced steatohepatitis, commensal microbiota was found to be hepatoprotective.
Collapse
Affiliation(s)
- Kai Markus Schneider
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany.
| | - Antje Mohs
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany.
| | - Konrad Kilic
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany.
| | - Lena Susanna Candels
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany.
| | - Carsten Elfers
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany.
| | - Eveline Bennek
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany.
| | - Lukas Ben Schneider
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany.
| | - Felix Heymann
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany.
| | - Nikolaus Gassler
- Department of Pathology, Klinikum Braunschweig, 38118 Braunschweig, Germany.
| | - John Penders
- Department of Medical Microbiology, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands.
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany.
| |
Collapse
|
32
|
Zhang R, Pan Z, Wang X, Shen M, Zhou J, Fu Z, Jin Y. Short-term propamocarb exposure induces hepatic metabolism disorder associated with gut microbiota dysbiosis in adult male zebrafish. Acta Biochim Biophys Sin (Shanghai) 2019; 51:88-96. [PMID: 30544157 DOI: 10.1093/abbs/gmy153] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/03/2018] [Indexed: 12/30/2022] Open
Abstract
Propamocarb (PM) is a pesticide that is widely used to protect cucumbers and other plants from downy mildew. Recently, some studies indicated that PM exposure had potential toxic effects in animals. In this study, adult male zebrafish were exposed to 100 and 1000 μg/l PM for 7 days to assess its effects on metabolism and the gut microbiota. We observed a significant decrease in triglyceride (TG) in the livers of zebrafish that were exposed to 1000 μg/l PM for 7 days. At the same time, some genes related to glycolysis and lipid metabolism in the livers of zebrafish, including hexokinase-1 (HK1), pyruvate kinase (PK), acyl-CoA oxidase (Aco), peroxisome proliferator activated receptor alpha (Ppar-α), apolipoprotein A-IV-like (Apo), Acetyl CoA carboxylase-1 (Acc1), diacylglycerol acyltransferase (Dgat), and fatty acid synthase (Fas), were also decreased significantly after PM exposure. Based on GC-MS metabolomics analysis, a total of 48 metabolites changed significantly in the 1000 μg/l PM treatment group in comparison with the control group. These altered metabolites were mainly associated with the glycolysis, amino acid metabolism, and lipid metabolism pathways. Interestingly, we further found that the 1000 μg/l PM treatment group also showed significant elevations in Proteobacteria, Bacteroidetes, and Firmicutes at the phylum level. Sequencing of the 16S rRNA gene in the V3-V4 region also showed a significant change in the abundance and diversity of the gut microbiota in the 1000 μg/l PM treatment group. Our results indicated that exposure to PM for a short time could induce hepatic metabolic disorders and gut microbiota dysbiosis in adult male zebrafish.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Zihong Pan
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Xiaoyu Wang
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Manlu Shen
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Jiajie Zhou
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Zhengwei Fu
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Yuanxiang Jin
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
33
|
Emile SH. Predictive Factors for Intestinal Transmural Necrosis in Patients with Acute Mesenteric Ischemia. World J Surg 2018; 42:2364-2372. [PMID: 29387956 DOI: 10.1007/s00268-018-4503-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Acute mesenteric ischemia (AMI) is a serious and potentially fatal condition. No definite parameter can predict transmural bowel necrosis in patients with AMI to justify early surgical intervention. The current study aimed to identify the clinical, laboratory, and radiologic parameters that can successfully predict the onset of intestinal transmural necrosis in patients with AMI. PATIENTS AND METHODS Records of patients with AMI in the period of January 2013 to October 2017 were reviewed. Clinical parameters as patients' symptoms, vital signs, and signs of peritonitis along with the results of laboratory and radiologic investigations were analyzed to identify predictive factors for intestinal transmural necrosis using binary logistic regression analysis. RESULTS One hundred and one patients (70 males) with mean age of 55 years were included. Venous occlusion was the cause of AMI in 78 (77.3%) patients and arterial occlusion in 23 (22.7%) patients. Twenty-two patients completed conservative treatment successfully, whereas 79 patients required exploratory laparotomy. On laparotomy, six patients were found to have viable bowel. Overall, 28 patients had viable bowel and 73 had bowel necrosis. The significant independent predictors for transmural bowel necrosis were mesenteric arterial occlusion (OR: 26.5, p = 0.02), leukocytosis (OR: 1.3, p < 0.0001), acidosis (OR: 3.8, p = 0.04), free intraperitoneal fluid (OR: 4.21, p = 0.005), and combined portal vein and SMV thrombosis in CT scan (OR: 3.4, p = 0.026). CONCLUSION The independent predictors for transmural bowel necrosis were mesenteric arterial occlusion, leukocytosis, acidosis, free intraperitoneal fluid, and combined portal vein and SMV thrombosis in CT scan.
Collapse
Affiliation(s)
- Sameh Hany Emile
- General Surgery Department, Mansoura Faculty of Medicine, Mansoura University Hospitals, Elgomhuoria Street, Mansoura City, Egypt.
| |
Collapse
|
34
|
Huang R, Li T, Ni J, Bai X, Gao Y, Li Y, Zhang P, Gong Y. Different Sex-Based Responses of Gut Microbiota During the Development of Hepatocellular Carcinoma in Liver-Specific Tsc1-Knockout Mice. Front Microbiol 2018; 9:1008. [PMID: 29867896 PMCID: PMC5964185 DOI: 10.3389/fmicb.2018.01008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 04/30/2018] [Indexed: 12/12/2022] Open
Abstract
Gut microbial dysbiosis is correlated with the development of hepatocellular carcinoma (HCC). Therefore, analyzing the changing patterns in gut microbiota during HCC development, especially before HCC occurrence, is essential for the diagnosis and prevention of HCC based on gut microbial composition. However, these changing patterns in HCC are poorly understood, especially considering the sex differences in HCC incidence and mortality. Here, with an aim to determine the relationship between gut microbiota and HCC development in both sexes, and to screen potential microbial biomarkers for HCC diagnosis, we studied the changing patterns in the gut microbiota from mice of both sexes with liver-specific knockout of Tsc1 (LTsc1KO) that spontaneously developed HCC by 9–10 months of age and compared them to the patterns observed in their wide-type Tsc1fl/fl cohorts using high-throughput sequencing. Using the LTsc1KO model, we were able to successfully exclude the continuing influence of diet on the gut microbiota. Based on gut microbial composition, the female LTsc1KO mice exhibited gut microbial disorder earlier than male LTsc1KO mice during the development of HCC. Our findings also indicated that the decrease in the relative abundance of anaerobic bacteria and the increase in the relative abundance of facultative anaerobic bacteria can be used as risk indexes of female HCC, but would be invalid for male HCC. Most of the changes in the gut bacteria were different between female and male LTsc1KO mice. In particular, the increased abundances of Allobaculum, Erysipelotrichaceae, Neisseriaceae, Sutterella, Burkholderiales, and Prevotella species have potential for use as risk indicators of female HCC, and the increased abundances of Paraprevotella, Paraprevotellaceae, and Prevotella can probably be applied as risk indicators of male HCC. These relationships between the gut microbiota and HCC discovered in the present study may serve as a platform for the identification of potential targets for the diagnosis and prevention of HCC in the future.
Collapse
Affiliation(s)
- Rong Huang
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Ting Li
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Jiajia Ni
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Xiaochun Bai
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Yi Gao
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Yang Li
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Peng Zhang
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Yan Gong
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| |
Collapse
|
35
|
Mohamadkhani A. On the potential role of intestinal microbial community in hepatocarcinogenesis in chronic hepatitis B. Cancer Med 2018; 7:3095-3100. [PMID: 29761927 PMCID: PMC6051233 DOI: 10.1002/cam4.1550] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/17/2018] [Accepted: 04/19/2018] [Indexed: 12/12/2022] Open
Abstract
The chronic infection of hepatitis B virus (HBV) is the most potent risk factor for the development of cirrhosis and hepatocellular carcinoma (HCC). The association of intestinal microbiota alteration with progressive liver disease has been investigated in recent studies. Overgrowth of potentially pathogenic bacteria of gram‐negative species and, in particular, a significant increase in the fecal count of Escherichia coli (E. coli) are characterized in the presence of HCC. This study was conducted to describe the characteristics of the intestinal microbiota related to the presence of HCC in HBV‐carrier patients. The available literature indicates the colonization of E. coli as principal source of portal vein lipopolysaccharide (LPS), in the gut may contribute to the carcinogenesis process by inducing chronic inflammation. This understanding could help to predict the clinical outcomes in HBV‐carrier patients and innovative strategies to reduce the virulence of liver disease from intestinal dysbiosis.
Collapse
Affiliation(s)
- Ashraf Mohamadkhani
- Liver and Pancreatobiliary Disease Research Center, Digestive Disease Research institute, Shariati Hospital, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
36
|
Mohandas S, Vairappan B. Role of pregnane X-receptor in regulating bacterial translocation in chronic liver diseases. World J Hepatol 2017; 9:1210-1226. [PMID: 29184608 PMCID: PMC5696604 DOI: 10.4254/wjh.v9.i32.1210] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 09/21/2017] [Accepted: 10/30/2017] [Indexed: 02/06/2023] Open
Abstract
Bacterial translocation (BT) has been impeccably implicated as a driving factor in the pathogenesis of a spectrum of chronic liver diseases (CLD). Scientific evidence accumulated over the last four decades has implied that the disease pathologies in CLD and BT are connected as a loop in the gut-liver axis and exacerbate each other. Pregnane X receptor (PXR) is a ligand-activated transcription factor and nuclear receptor that is expressed ubiquitously along the gut-liver-axis. PXR has been intricately associated with the regulation of various mechanisms attributed in causing BT. The importance of PXR as the mechanistic linker molecule in the gut-liver axis and its role in regulating bacterial interactions with the host in CLD has not been explored. PubMed was used to perform an extensive literature search using the keywords PXR and bacterial translocation, PXR and chronic liver disease including cirrhosis. In an adequate expression state, PXR acts as a sensor for bile acid dysregulation and bacterial derived metabolites, and in response shapes the immune profile beneficial to the host. Activation of PXR could be therapeutic in CLD as it counter-regulates endotoxin mediated inflammation and maintains the integrity of intestinal epithelium. This review mainly focuses PXR function and its regulation in BT in the context of chronic liver diseases.
Collapse
Affiliation(s)
- Sundhar Mohandas
- Liver Diseases Research Lab, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Dhanvantari Nagar, Pondicherry 605006, India
| | - Balasubramaniyan Vairappan
- Liver Diseases Research Lab, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Dhanvantari Nagar, Pondicherry 605006, India
| |
Collapse
|
37
|
Wang J, Wang Y, Zhang X, Liu J, Zhang Q, Zhao Y, Peng J, Feng Q, Dai J, Sun S, Zhao Y, Zhao L, Zhang Y, Hu Y, Zhang M. Gut Microbial Dysbiosis Is Associated with Altered Hepatic Functions and Serum Metabolites in Chronic Hepatitis B Patients. Front Microbiol 2017; 8:2222. [PMID: 29180991 PMCID: PMC5693892 DOI: 10.3389/fmicb.2017.02222] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 10/30/2017] [Indexed: 12/20/2022] Open
Abstract
Chronic hepatitis B (CHB) is a global epidemic disease that results from hepatitis B virus (HBV) infection and may progress to severe liver failure, including liver fibrosis, cirrhosis and hepatocellular carcinoma. Previous evidence has indicated that the dysbiosis of gut microbiota occurs after liver virus infection and is associated with severe liver disease. The aim of this study is to elucidate the compositional and functional characteristics of the gut microbiota in early-stage CHB and to understand their influence on disease progression. We investigated the gut microbial composition of stool samples from 85 CHB patients with low Child-Pugh scores and 22 healthy controls using the Illumina MiSeq sequencing platform. Furthermore, the serum metabolome of 40 subjects was measured by gas chromatography mass spectrometry. Compared with the controls, significant alteration in the gut microbiota was observed in the CHB patients; 5 operational taxonomic units (OTUs) belonging to Actinomyces, Clostridium sensu stricto, unclassified Lachnospiraceae and Megamonas were increased, and 27 belonging to Alistipes, Asaccharobacter, Bacteroides, Butyricimonas, Clostridium IV, Escherichia/Shigella, Parabacteroides, Ruminococcus, unclassified Bacteria, unclassified Clostridiales, Unclassified Coriobacteriaceae, unclassified Enterobacteriaceae, unclassified Lachnospiraceae and unclassified Ruminococcaceae were decreased. The inferred metagenomic information of gut microbiota in CHB showed 21 enriched and 17 depleted KEGG level-2 pathways. Four OTUs, OTU38 (Streptococcus), OTU124 (Veillonella), OTU224 (Streptococcus), and OTU55 (Haemophilus), had high correlations with hosts' hepatic function indices and 10 serum metabolites, including phenylalanine and tyrosine, which are aromatic amino acids that play pathogenic roles in liver disease. In particular, these 4 OTUs were significantly higher in patients with higher Child-Pugh scores, who also showed diminished phenylalanine and tryptophan metabolisms in the inferred gut metagenomic functions. These compositional and functional changes in the gut microbiota in early-stage CHB patients suggest the potential contributions of gut microbiota to the progression of CHB, and thus provide new insight into gut microbiota-targeted interventions to improve the prognosis of this disease.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Wang
- Center for Traditional Chinese Medicine and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xu Zhang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Jiaqi Liu
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Qianpeng Zhang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Zhao
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jinghua Peng
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qin Feng
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianye Dai
- Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Shujun Sun
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China.,Unimicro (Shanghai) Technologics Co., Ltd., Shanghai, China
| | - Yufeng Zhao
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Liping Zhao
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yongyu Zhang
- Center for Traditional Chinese Medicine and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Traditional Dai Medicine, West Yunnan University of Applied Sciences, Jinghong, Yunnan, China
| | - Yiyang Hu
- Institute of Liver Disease, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Menghui Zhang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
38
|
Wang Q, Wang B, Saxena V, Miles L, Tiao J, Mortensen JE, Nathan JD. The gut-liver axis: impact of a mouse model of small-bowel bacterial overgrowth. J Surg Res 2017; 221:246-256. [PMID: 29229136 DOI: 10.1016/j.jss.2017.08.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 08/07/2017] [Accepted: 08/30/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND The mechanisms by which intestinal bacteria impact liver diseases remain poorly understood. The aim of this study was to develop a mouse model of small-bowel bacterial overgrowth and to determine its impact on hepatobiliary injury. MATERIALS AND METHODS A jejunal self-filling blind loop (SFBL) was created in C57BL/6 mice. Three weeks after surgery, the mice were euthanized, and bacterial cultures of luminal content of the loop and extraintestinal tissues were performed. Liver and jejunum were collected for histological grading of inflammation and injury. Serum liver biochemistry assays were performed. Hepatobiliary transporter mRNA expression in liver was measured by quantitative real-time polymerase chain reaction. Bile and blood were collected for measurement of total bile acids, phospholipid, and cholesterol. Mice undergoing jejunal transection and reanastomosis and laparotomy only served as control groups. RESULTS SFBL induced a dramatic increase in intraluminal bacterial counts, mesenteric lymph node bacterial translocation, and evidence of jejunal and hepatobiliary injury. Significant reductions in hepatic expression of hepatobiliary transporters involved in biliary canalicular export and basolateral uptake were observed in SFBL mice. SFBL resulted in a significant increase in biliary total bile acid concentration, decreases in bile phospholipid and cholesterol output, and an increase in the bile acid/phospholipid ratio. CONCLUSIONS We have developed a reproducible mouse model of small-bowel bacterial overgrowth with evidence of liver inflammation, altered hepatobiliary transporter expression, and alterations in bile composition. This model may help to elucidate the mechanisms by which gut-derived bacterial factors impact the liver and contribute to the exacerbation of liver diseases and biliary injury.
Collapse
Affiliation(s)
- Qingqing Wang
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Bin Wang
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Vijay Saxena
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Lili Miles
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Josh Tiao
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Joel E Mortensen
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jaimie D Nathan
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| |
Collapse
|
39
|
Enteric dysbiosis-linked gut barrier disruption triggers early renal injury induced by chronic high salt feeding in mice. Exp Mol Med 2017; 49:e370. [PMID: 28857085 PMCID: PMC5579512 DOI: 10.1038/emm.2017.122] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 03/14/2017] [Accepted: 03/20/2017] [Indexed: 12/12/2022] Open
Abstract
Chronic high-salt diet-associated renal injury is a key risk factor for the development of hypertension. However, the mechanism by which salt triggers kidney damage is poorly understood. Our study investigated how high salt (HS) intake triggers early renal injury by considering the ‘gut-kidney axis’. We fed mice 2% NaCl in drinking water continuously for 8 weeks to induce early renal injury. We found that the ‘quantitative’ and ‘qualitative’ levels of the intestinal microflora were significantly altered after chronic HS feeding, which indicated the occurrence of enteric dysbiosis. In addition, intestinal immunological gene expression was impaired in mice with HS intake. Gut permeability elevation and enteric bacterial translocation into the kidney were detected after chronic HS feeding. Gut bacteria depletion by non-absorbable antibiotic administration restored HS loading-induced gut leakiness, renal injury and systolic blood pressure elevation. The fecal microbiota from mice fed chronic HS could independently cause gut leakiness and renal injury. Our current work provides a novel insight into the mechanism of HS-induced renal injury by investigating the role of the intestine with enteric bacteria and gut permeability and clearly illustrates that chronic HS loading elicited renal injury and dysfunction that was dependent on the intestine.
Collapse
|
40
|
Dembek A, Laggai S, Kessler SM, Czepukojc B, Simon Y, Kiemer AK, Hoppstädter J. Hepatic interleukin-6 production is maintained during endotoxin tolerance and facilitates lipid accumulation. Immunobiology 2017; 222:786-796. [DOI: 10.1016/j.imbio.2017.01.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 11/23/2016] [Accepted: 01/17/2017] [Indexed: 02/08/2023]
|
41
|
Altered gut microbiome in a mouse model of Gulf War Illness causes neuroinflammation and intestinal injury via leaky gut and TLR4 activation. PLoS One 2017; 12:e0172914. [PMID: 28328972 PMCID: PMC5362211 DOI: 10.1371/journal.pone.0172914] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/10/2017] [Indexed: 02/06/2023] Open
Abstract
Many of the symptoms of Gulf War Illness (GWI) that include neurological abnormalities, neuroinflammation, chronic fatigue and gastrointestinal disturbances have been traced to Gulf War chemical exposure. Though the association and subsequent evidences are strong, the mechanisms that connect exposure to intestinal and neurological abnormalities remain unclear. Using an established rodent model of Gulf War Illness, we show that chemical exposure caused significant dysbiosis in the gut that included increased abundance of phylum Firmicutes and Tenericutes, and decreased abundance of Bacteroidetes. Several gram negative bacterial genera were enriched in the GWI-model that included Allobaculum sp. Altered microbiome caused significant decrease in tight junction protein Occludin with a concomitant increase in Claudin-2, a signature of a leaky gut. Resultant leaching of gut caused portal endotoxemia that led to upregulation of toll like receptor 4 (TLR4) activation in the small intestine and the brain. TLR4 knock out mice and mice that had gut decontamination showed significant decrease in tyrosine nitration and inflammatory mediators IL1β and MCP-1 in both the small intestine and frontal cortex. These events signified that gut dysbiosis with simultaneous leaky gut and systemic endotoxemia-induced TLR4 activation contributes to GW chemical-induced neuroinflammation and gastrointestinal disturbances.
Collapse
|
42
|
Wesolowski SR, El Kasmi KC, Jonscher KR, Friedman JE. Developmental origins of NAFLD: a womb with a clue. Nat Rev Gastroenterol Hepatol 2017; 14:81-96. [PMID: 27780972 PMCID: PMC5725959 DOI: 10.1038/nrgastro.2016.160] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Changes in the maternal environment leading to an altered intrauterine milieu can result in subtle insults to the fetus, promoting increased lifetime disease risk and/or disease acceleration in childhood and later in life. Particularly worrisome is that the prevalence of NAFLD is rapidly increasing among children and adults, and is being diagnosed at increasingly younger ages, pointing towards an early-life origin. A wealth of evidence, in humans and non-human primates, suggests that maternal nutrition affects the placenta and fetal tissues, leading to persistent changes in hepatic metabolism, mitochondrial function, the intestinal microbiota, liver macrophage activation and susceptibility to NASH postnatally. Deleterious exposures in utero include fetal hypoxia, increased nutrient supply, inflammation and altered gut microbiota that might produce metabolic clues, including fatty acids, metabolites, endotoxins, bile acids and cytokines, which prime the infant liver for NAFLD in a persistent manner and increase susceptibility to NASH. Mechanistic links to early disease pathways might involve shifts in lipid metabolism, mitochondrial dysfunction, pioneering gut microorganisms, macrophage programming and epigenetic changes that alter the liver microenvironment, favouring liver injury. In this Review, we discuss how maternal, fetal, neonatal and infant exposures provide developmental clues and mechanisms to help explain NAFLD acceleration and increased disease prevalence. Mechanisms identified in clinical and preclinical models suggest important opportunities for prevention and intervention that could slow down the growing epidemic of NAFLD in the next generation.
Collapse
Affiliation(s)
| | - Karim C. El Kasmi
- Department of Pediatrics, Section of Gastroenterology, Hepatology and Nutrition, University of Colorado
| | | | - Jacob E. Friedman
- Department of Pediatrics, Section of Neonatology, University of Colorado,Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado, Anschutz Medical Campus, 12801 East 17th Avenue, MS 8106, Aurora, Colorado 80045, USA
| |
Collapse
|
43
|
Doycheva I, Leise MD, Watt KD. The Intestinal Microbiome and the Liver Transplant Recipient: What We Know and What We Need to Know. Transplantation 2016; 100:61-8. [PMID: 26647107 DOI: 10.1097/tp.0000000000001008] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The intestinal microbiome and immune system are in close symbiotic relationship in health. Gut microbiota plays a role in many chronic liver diseases and cirrhosis. However, alterations in the gut microbiome after liver transplantation and the implications for liver transplant recipients are not well understood and rely mainly on experimental animal studies. Recent advances in molecular techniques have identified that increased intestinal permeability, decreased beneficial bacteria, and increased pathogenic species may play important roles in the early posttransplant period. The associations between microbiota perturbation and postliver transplant infections and acute rejection are evolving. The link with metabolic syndrome, obesity, and cardiac disease in the general population require translation into the transplant recipient. This review focuses on our current knowledge of the known and potential interaction of the microbiome in the liver transplant recipient. Future human studies focused on microbiota changes in liver transplant patients are warranted and expected.
Collapse
Affiliation(s)
- Iliana Doycheva
- 1 Division of Gastroenterology and Hepatology, Medical University, Sofia, Bulgaria. 2 Division of Gastroenterology and Hepatology, Mayo Clinic Transplant Center, Rochester, MN
| | | | | |
Collapse
|
44
|
Arnal ME, Lallès JP. Gut epithelial inducible heat-shock proteins and their modulation by diet and the microbiota. Nutr Rev 2016; 74:181-97. [PMID: 26883882 DOI: 10.1093/nutrit/nuv104] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The epidemic of metabolic diseases has raised questions about the interplay between the human diet and the gut and its microbiota. The gut has two vital roles: nutrient absorption and intestinal barrier function. Gut barrier defects are involved in many diseases. Excess energy intake disturbs the gut microbiota and favors body entry of microbial compounds that stimulate chronic metabolic inflammation. In this context, the natural defense mechanisms of gut epithelial cells and the potential to boost them nutritionally warrant further study. One such important defense system is the activation of inducible heat-shock proteins (iHSPs) which protect the gut epithelium against oxidative stress and inflammation. Importantly, various microbial components can induce the expression of iHSPs. This review examines gut epithelial iHSPs as the main targets of microbial signals and nutrients and presents data on diseases involving disturbances of gut epithelial iHSPs. In addition, a broad literature analysis of dietary modulation of gut epithelial iHSPs is provided. Future research aims should include the identification of gut microbes that can optimize gut-protective iHSPs and the evaluation of iHSP-mediated health benefits of nutrients and food components.
Collapse
Affiliation(s)
- Marie-Edith Arnal
- M.E. Arnal and J.P. Lallès are with the Institut National de la Recherche Agronomique (INRA), Human Nutrition Division, Clermont-Ferrand, France. J.P. Lallès is with the Centre de Recherche en Nutrition Humaine Ouest, Nantes, France
| | - Jean-Paul Lallès
- M.E. Arnal and J.P. Lallès are with the Institut National de la Recherche Agronomique (INRA), Human Nutrition Division, Clermont-Ferrand, France. J.P. Lallès is with the Centre de Recherche en Nutrition Humaine Ouest, Nantes, France.
| |
Collapse
|
45
|
Nyima T, Müller M, Hooiveld GJEJ, Morine MJ, Scotti M. Nonlinear transcriptomic response to dietary fat intake in the small intestine of C57BL/6J mice. BMC Genomics 2016; 17:106. [PMID: 26861690 PMCID: PMC4748552 DOI: 10.1186/s12864-016-2424-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 02/02/2016] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND A high caloric diet, in conjunction with low levels of physical activity, promotes obesity. Many studies are available regarding the relation between dietary saturated fats and the etiology of obesity, but most focus on liver, muscle and white adipose tissue. Furthermore, the majority of transcriptomic studies seek to identify linear effects of an external stimulus on gene expression, although such an assumption does not necessarily hold. Our work assesses the dose-dependent effects of dietary fat intake on differential gene expression in the proximal, middle and distal sections of the small intestine in C57BL/6J mice. Gene expression is analyzed in terms of either linear or nonlinear responses to fat intake. RESULTS The highest number of differentially expressed genes was observed in the middle section. In all intestine sections, most of the identified processes exhibited a linear response to increasing fat intake. The relative importance of logarithmic and exponential responses was higher in the proximal and distal sections, respectively. Functional enrichment analysis highlighted a constantly linear regulation of acute-phase response along the whole small intestine, with up-regulation of Serpina1b. The study of gene expression showed that exponential down-regulation of cholesterol transport in the middle section is coupled with logarithmic up-regulation of cholesterol homeostasis. A shift from linear to exponential response was observed in genes involved in the negative regulation of caspase activity, from middle to distal section (e.g., Birc5, up-regulated). CONCLUSIONS The transcriptomic signature associated with inflammatory processes preserved a linear response in the whole small intestine (e.g., up-regulation of Serpina1b). Processes related to cholesterol homeostasis were particularly active in the middle small intestine and only the highest fat intake down-regulated cholesterol transport and efflux (with a key role played by the down-regulation of ATP binding cassette transporters). Characterization of nonlinear patterns of gene expression triggered by different levels of dietary fat is an absolute novelty in intestinal studies. This approach helps identifying which processes are overloaded (i.e., positive, logarithmic regulation) or arrested (i.e., negative, exponential regulation) in response to excessive fat intake, and can shed light on the relationships linking lipid intake to obesity and its associated molecular disturbances.
Collapse
Affiliation(s)
- Tenzin Nyima
- The Microsoft Research - University of Trento Centre for Computational and Systems Biology, Rovereto, Italy.
| | - Michael Müller
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands. .,Norwich Medical School, University of East Anglia, Norwich, UK.
| | - Guido J E J Hooiveld
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands.
| | - Melissa J Morine
- The Microsoft Research - University of Trento Centre for Computational and Systems Biology, Rovereto, Italy.
| | - Marco Scotti
- The Microsoft Research - University of Trento Centre for Computational and Systems Biology, Rovereto, Italy. .,GEOMAR Helmholtz Centre for Ocean Research, Kiel, Germany.
| |
Collapse
|
46
|
Márquez M, Fernández Gutiérrez del Álamo C, Girón-González JA. Gut epithelial barrier dysfunction in human immunodeficiency virus-hepatitis C virus coinfected patients: Influence on innate and acquired immunity. World J Gastroenterol 2016; 22:1433-1448. [PMID: 26819512 PMCID: PMC4721978 DOI: 10.3748/wjg.v22.i4.1433] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 10/11/2015] [Accepted: 11/13/2015] [Indexed: 02/06/2023] Open
Abstract
Even in cases where viral replication has been controlled by antiretroviral therapy for long periods of time, human immunodeficiency virus (HIV)-infected patients have several non-acquired immunodeficiency syndrome (AIDS) related co-morbidities, including liver disease, cardiovascular disease and neurocognitive decline, which have a clear impact on survival. It has been considered that persistent innate and acquired immune activation contributes to the pathogenesis of these non-AIDS related diseases. Immune activation has been related with several conditions, remarkably with the bacterial translocation related with the intestinal barrier damage by the HIV or by hepatitis C virus (HCV)-related liver cirrhosis. Consequently, increased morbidity and mortality must be expected in HIV-HCV coinfected patients. Disrupted gut barrier lead to an increased passage of microbial products and to an activation of the mucosal immune system and secretion of inflammatory mediators, which in turn might increase barrier dysfunction. In the present review, the intestinal barrier structure, measures of intestinal barrier dysfunction and the modifications of them in HIV monoinfection and in HIV-HCV coinfection will be considered. Both pathogenesis and the consequences for the progression of liver disease secondary to gut microbial fragment leakage and immune activation will be assessed.
Collapse
|
47
|
Schneider KM, Bieghs V, Heymann F, Hu W, Dreymueller D, Liao L, Frissen M, Ludwig A, Gassler N, Pabst O, Latz E, Sellge G, Penders J, Tacke F, Trautwein C. CX3CR1 is a gatekeeper for intestinal barrier integrity in mice: Limiting steatohepatitis by maintaining intestinal homeostasis. Hepatology 2015; 62:1405-1416. [PMID: 26178698 DOI: 10.1002/hep.27982] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 07/09/2015] [Indexed: 12/13/2022]
Abstract
UNLABELLED Nonalcoholic fatty liver disease is seen as the hepatic manifestation of the metabolic syndrome and represents the most common liver disease in Western societies. The G protein-coupled chemokine receptor CX3CR1 plays a central role in several metabolic syndrome-related disease manifestations and is involved in maintaining intestinal homeostasis. Because diet-induced intestinal dysbiosis is a driver for nonalcoholic fatty liver disease, we hypothesized that CX3CR1 may influence the development of steatohepatitis. In two independent models of diet-induced steatohepatitis (high-fat diet and methionine/choline-deficient diet), CX3CR1 protected mice from excessive hepatic steatosis and inflammation, as well as systemic glucose intolerance. Lack of Cx3cr1 expression was associated with significantly altered intestinal microbiota composition, which was linked to an impaired intestinal barrier. Concomitantly, endotoxin levels in portal serum and inflammatory macrophages in liver were increased in Cx3cr1-/- mice, indicating an increased inflammatory response. Depletion of intestinal microbiota by administration of broad-spectrum antibiotics suppressed the number of infiltrating macrophages and promoted macrophage polarization in liver. Consequently, antibiotic-treated mice demonstrated a marked improvement of steatohepatitis. CONCLUSION Microbiota-mediated activation of the innate immune responses through CX3CR1 is crucial for controlling steatohepatitis progression, which recognizes CX3CR1 as an essential gatekeeper in this scenario.
Collapse
Affiliation(s)
- Kai Markus Schneider
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - Veerle Bieghs
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - Felix Heymann
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - Wei Hu
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - Daniela Dreymueller
- Institute of Pharmacology and Toxicology, Medical Faculty, University Hospital, RWTH Aachen, Aachen, Germany
| | - Lijun Liao
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - Mick Frissen
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - Andreas Ludwig
- Institute of Pharmacology and Toxicology, Medical Faculty, University Hospital, RWTH Aachen, Aachen, Germany
| | - Nikolaus Gassler
- Institute of Pathology, University Hospital, RWTH Aachen, Aachen, Germany
| | - Oliver Pabst
- Institute of Molecular Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital, University of Bonn, Bonn, Germany
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Gernot Sellge
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - John Penders
- Department of Medical Microbiology, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Frank Tacke
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| |
Collapse
|
48
|
Demir M, Lang S, Steffen HM. Nonalcoholic fatty liver disease - current status and future directions. J Dig Dis 2015; 16:541-57. [PMID: 26406351 DOI: 10.1111/1751-2980.12291] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 09/23/2015] [Accepted: 09/23/2015] [Indexed: 12/11/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as the most common chronic liver disease worldwide with a reported prevalence ranging 6-33%, depending on the studied populations. It encompasses a spectrum of liver manifestations ranging from simple steatosis (also known as nonalcoholic fatty liver, NAFL) to nonalcoholic steatohepatitis (NASH), fibrosis and cirrhosis, which may ultimately progress to hepatocellular carcinoma. NAFLD is strongly associated with the components of metabolic syndrome, mainly obesity and type 2 diabetes mellitus. NAFLD patients are at increased risk of liver-related as well as cardiovascular mortality. Current paradigm suggests a benign course for NAFL whereas NASH is considered to be the progressive phenotype. Although previously under-recognized accumulating evidence suggests that NAFL may also progress, suggesting a higher number of patients at risk than previously appreciated. Liver biopsy remains the gold standard for definitive diagnosis, but the majority of patients can be diagnosed accurately by noninvasive methods. Approved therapies for NAFLD are still lacking and lifestyle modifications aiming at weight loss remain the mainstay of NAFLD treatment. Intensive research could identify insulin resistance, lipotoxicity and dysbiosis of the gut microbiota as major pathophysiological mechanisms, leading to the development of promising targeted therapies which are currently investigated in clinical trials. In this review we summarized the current knowledge of NAFLD epidemiology, natural history, diagnosis, pathogenesis and treatment and considered future directions.
Collapse
Affiliation(s)
- Münevver Demir
- Clinic for Gastroenterology and Hepatology, University Hospital of Cologne, Cologne, Germany
| | - Sonja Lang
- Clinic for Gastroenterology and Hepatology, University Hospital of Cologne, Cologne, Germany
| | - Hans-Michael Steffen
- Clinic for Gastroenterology and Hepatology, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
49
|
Gut microbiota modulate the immune effect against hepatitis B virus infection. Eur J Clin Microbiol Infect Dis 2015; 34:2139-47. [PMID: 26272175 DOI: 10.1007/s10096-015-2464-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/24/2015] [Indexed: 12/11/2022]
Abstract
The immunological mechanisms by which hepatitis B virus (HBV) initiates and maintains acute or chronic infection, even the formation of cirrhosis and hepatocellular carcinoma, are still undefined. An increasing number of studies have shown that intestinal flora regulate immune homeostasis, and, thus, protect the immunologic function against hepatitis virus infection. In this article, we discuss gut microbiota and its potential immune effects against HBV infection. It may provide a novel insight into the pathogenesis of HBV infection, as well as a potential therapeutic target to HBV-related disease.
Collapse
|
50
|
Wang L, Llorente C, Hartmann P, Yang AM, Chen P, Schnabl B. Methods to determine intestinal permeability and bacterial translocation during liver disease. J Immunol Methods 2015; 421:44-53. [PMID: 25595554 PMCID: PMC4451427 DOI: 10.1016/j.jim.2014.12.015] [Citation(s) in RCA: 189] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 12/04/2014] [Indexed: 02/08/2023]
Abstract
Liver disease is often times associated with increased intestinal permeability. A disruption of the gut barrier allows microbial products and viable bacteria to translocate from the intestinal lumen to extraintestinal organs. The majority of the venous blood from the intestinal tract is drained into the portal circulation, which is part of the dual hepatic blood supply. The liver is therefore the first organ in the body to encounter not only absorbed nutrients, but also gut-derived bacteria and pathogen associated molecular patterns (PAMPs). Chronic exposure to increased levels of PAMPs has been linked to disease progression during early stages and to infectious complications during late stages of liver disease (cirrhosis). It is therefore important to assess and monitor gut barrier dysfunction during hepatic disease. We review methods to assess intestinal barrier disruption and discuss advantages and disadvantages. We will in particular focus on methods that we have used to measure increased intestinal permeability and bacterial translocation in experimental liver disease models.
Collapse
Affiliation(s)
- Lirui Wang
- Department of Medicine, University of California San Diego, La Jolla, CA, United States; Department of Medicine, VA San Diego Healthcare System, San Diego, CA, United States
| | - Cristina Llorente
- Department of Medicine, University of California San Diego, La Jolla, CA, United States; Department of Medicine, VA San Diego Healthcare System, San Diego, CA, United States
| | - Phillipp Hartmann
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - An-Ming Yang
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Peng Chen
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, United States; Department of Medicine, VA San Diego Healthcare System, San Diego, CA, United States.
| |
Collapse
|