1
|
Velasco-Aburto S, Llama-Palacios A, Sánchez MC, Ciudad MJ, Collado L. Nutritional Approach to Small Intestinal Bacterial Overgrowth: A Narrative Review. Nutrients 2025; 17:1410. [PMID: 40362719 PMCID: PMC12073203 DOI: 10.3390/nu17091410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/19/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Small intestinal bacterial overgrowth (SIBO) is a functional digestive disorder whose incidence has been acknowledged by several medical associations, such as the American Gastroenterological Association. It is estimated that between 14% and 40% of patients diagnosed with irritable bowel syndrome also have SIBO, highlighting the importance of accurate diagnosis to enable effective treatment plans. Nutrition and diet therapy play a pivotal role in SIBO management, not only in alleviating symptoms but also in preventing relapses. The objective of this review is to gather updated information on dietary management for SIBO to define the role of the dietitian and determine the most suitable nutritional therapy based on scientific evidence. The review will encompass various strategies, ranging from specific diets to dietary supplements, as well as the potential contribution of dietary treatment to improving SIBO.
Collapse
Affiliation(s)
- Sol Velasco-Aburto
- Department of Medicine, Faculty of Medicine, University Complutense, 28040 Madrid, Spain; (S.V.-A.); (A.L.-P.); (M.C.S.)
| | - Arancha Llama-Palacios
- Department of Medicine, Faculty of Medicine, University Complutense, 28040 Madrid, Spain; (S.V.-A.); (A.L.-P.); (M.C.S.)
- GINTRAMIS Research Group (Translational Research Group on Microbiota and Health), Faculty of Medicine, University Complutense, 28040 Madrid, Spain
| | - María Carmen Sánchez
- Department of Medicine, Faculty of Medicine, University Complutense, 28040 Madrid, Spain; (S.V.-A.); (A.L.-P.); (M.C.S.)
- GINTRAMIS Research Group (Translational Research Group on Microbiota and Health), Faculty of Medicine, University Complutense, 28040 Madrid, Spain
| | - María José Ciudad
- Department of Medicine, Faculty of Medicine, University Complutense, 28040 Madrid, Spain; (S.V.-A.); (A.L.-P.); (M.C.S.)
- GINTRAMIS Research Group (Translational Research Group on Microbiota and Health), Faculty of Medicine, University Complutense, 28040 Madrid, Spain
| | - Luis Collado
- Department of Medicine, Faculty of Medicine, University Complutense, 28040 Madrid, Spain; (S.V.-A.); (A.L.-P.); (M.C.S.)
- GINTRAMIS Research Group (Translational Research Group on Microbiota and Health), Faculty of Medicine, University Complutense, 28040 Madrid, Spain
| |
Collapse
|
2
|
Silva BCD, Ramos GP, Barros LL, Ramos AFP, Domingues G, Chinzon D, Passos MDCF. DIAGNOSIS AND TREATMENT OF SMALL INTESTINAL BACTERIAL OVERGROWTH: AN OFFICIAL POSITION PAPER FROM THE BRAZILIAN FEDERATION OF GASTROENTEROLOGY. ARQUIVOS DE GASTROENTEROLOGIA 2025; 62:e24107. [PMID: 39968993 PMCID: PMC12043196 DOI: 10.1590/s0004-2803.24612024-107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 10/31/2024] [Indexed: 02/20/2025]
Abstract
BACKGROUND Small intestinal bacterial overgrowth (SIBO) is a condition characterized by an abnormal increase in bacterial population in the small intestine, leading to symptoms such as bloating, abdominal pain, distension, diarrhea, and eventually malabsorption. The diagnosis and management of SIBO remain challenging due to overlapping symptoms with other gastrointestinal disorders such as inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), and coeliac disease. OBJECTIVE This article aims to review current evidence on the diagnosis and treatment of SIBO, with a focus on strategies suitable for the Brazilian healthcare system. METHODS A comprehensive literature review was performed, focusing on clinical guidelines, randomized controlled trials, and cohort studies concerning SIBO. Diagnostic methods, including breath tests and direct aspiration techniques, were critically analyzed. Treatment approaches, including antibiotics, dietary modifications, and probiotics, were reviewed. The recommendations were formulated based on a panel of gastroenterologists, members of the Brazilian Federation of Gastroenterology (FBG), with approval from the majority of the members. RESULTS Breath tests using glucose and lactulose remain the most commonly used non-invasive diagnostic tools, though they are subject to limitations such as false positives and false negatives. Treatment with rifaximin is effective in most cases of SIBO, while systemic antibiotics like metronidazole and ciprofloxacin are alternatives. Probiotics and dietary interventions, particularly low FODMAP diets, can complement antibiotic therapy. Long-term follow-up is essential due to the recurrence rate, which is common in SIBO patients. CONCLUSION Standardizing SIBO diagnosis and treatment in Brazil is essential to reduce diagnostic delays and optimize care, especially given the disparities and heterogeneity in clinical practice across the country. This article provides evidence-based recommendations to guide clinical practice. Further research is needed to refine diagnostic methods, explore novel treatment strategies, and better understand the specific characteristics of the Brazilian population.
Collapse
Affiliation(s)
| | | | - Luisa Leite Barros
- Faculdade de Medicina da Universidade de São Paulo, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | | | - Gerson Domingues
- Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Décio Chinzon
- Faculdade de Medicina da Universidade de São Paulo, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | | |
Collapse
|
3
|
Alcedo González J, Estremera-Arévalo F, Cobián Malaver J, Santos Vicente J, Alcalá-González LG, Naves J, Barba Orozco E, Barber Caselles C, Serrano-Falcón B, Accarino Garaventa A, Alonso-Cotoner C, Serra Pueyo J. Common questions and rationale answers about the intestinal bacterial overgrowth syndrome (SIBO). GASTROENTEROLOGIA Y HEPATOLOGIA 2025; 48:502216. [PMID: 38852778 DOI: 10.1016/j.gastrohep.2024.502216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/15/2024] [Accepted: 05/24/2024] [Indexed: 06/11/2024]
Abstract
The recognition and treatment of intestinal bacterial overgrowth syndrome are matters of controversy. The symptoms that have guided the search for the disorder suffer from lack of specificity, especially in the absence of well-defined predisposing factors. The accuracy of diagnostic procedures has been questioned and the proposed therapies achieve generally low effectiveness figures, with large differences between available studies. It is also unknown whether the normalization of tests is really a guarantee of cure. Within this framework of uncertainty, and in order to contribute to the guidance and homogenization of medical practice, a group of experts from the AEG and ASENEM have formulated the key questions on the management of this pathology and have provided answers to them, in accordance with the available scientific evidence. In addition, they have drawn up statements based on the conclusions of the review and have voted on them individually to reflect the degree of consensus for each statement.
Collapse
Affiliation(s)
- Javier Alcedo González
- Servicio de Aparato Digestivo, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria (IIS) Aragón, Zaragoza, España.
| | - Fermín Estremera-Arévalo
- Servicio de Aparato Digestivo, Hospital Universitario de Navarra, Navarrabiomed, Universidad Pública de Navarra - IdiSNA, Navarra, España
| | | | - Javier Santos Vicente
- Laboratorio de Neuro-Inmuno-Gastroenterología, Unidad de Investigación de Aparato Digestivo, Institut de Recerca (VHIR), Servicio de Aparato Digestivo, Hospital Universitario Vall d'Hebron, Barcelona, España; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Instituto de Salud Carlos III, Madrid, España
| | | | - Juan Naves
- Servicio de Aparato Digestivo, Hospital del Mar, Barcelona, España
| | - Elizabeth Barba Orozco
- Unidad de Neurogastroenterología y Motilidad, Hospital Clínic de Barcelona, Barcelona, España; Departamento de Gastroenterología, Universidad de Barcelona, Barcelona, España
| | | | - Blanca Serrano-Falcón
- Servicio de Aparato Digestivo, Hospital Universitario Virgen de las Nieves, Granada, España
| | | | - Carmen Alonso-Cotoner
- Laboratorio de Neuro-Inmuno-Gastroenterología, Unidad de Investigación de Aparato Digestivo, Institut de Recerca (VHIR), Servicio de Aparato Digestivo, Hospital Universitario Vall d'Hebron, Barcelona, España; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Instituto de Salud Carlos III, Madrid, España; Facultad de Medicina, Universidad Autónoma de Barcelona, Barcelona, España
| | - Jordi Serra Pueyo
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Instituto de Salud Carlos III, Madrid, España; Área de Aparato Digestivo, Hospital Universitario Vall d'Hebron, Barcelona, España
| |
Collapse
|
4
|
Martyniak A, Wójcicka M, Rogatko I, Piskorz T, Tomasik PJ. A Comprehensive Review of the Usefulness of Prebiotics, Probiotics, and Postbiotics in the Diagnosis and Treatment of Small Intestine Bacterial Overgrowth. Microorganisms 2025; 13:57. [PMID: 39858825 PMCID: PMC11768010 DOI: 10.3390/microorganisms13010057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Small intestinal bacterial overgrowth (SIBO) is a disorder characterized by the excessive growth of bacteria in the small intestine. Bacterial overgrowth disrupts the bacterial balance and can lead to abdominal pain, weight loss, and gastrointestinal symptoms, including bloating, diarrhea, and malabsorption. SIBO is widespread in the population. There are two main methods for diagnosing SIBO: breath tests and bacterial culture. The most commonly used method is a breath test, which enables the division of SIBO into the following three types: hydrogen-dominant (H-SIBO), methane-dominant (CH4-SIBO), and hydrogen/methane-dominant (H/CH4-SIBO). This comprehensive review aims to present the current knowledge on the use of prebiotics, probiotics, and postbiotics in the context of SIBO. For this purpose, medical databases such as MEDLINE (PubMed) and Scopus were analyzed using specific keywords and their combinations. This review is based on research studies no older than 10 years old and those using only human models. In summary, clinical studies have shown that the efficacy of SIBO therapy can be increased by combining antibiotics with probiotics, especially in vulnerable patients such as children and pregnant women. The further development of diagnostic methods, such as point of care testing (POCT) and portable devices, and a better understanding of the mechanisms of biotics action are needed to treat SIBO more effectively and improve the quality of life of patients.
Collapse
Affiliation(s)
- Adrian Martyniak
- Department of Clinical Biochemistry, Pediatric Institute, Faculty of Medicine, Jagiellonian University Medical College, 30-663 Krakow, Poland; (A.M.); (M.W.); (I.R.)
| | - Magdalena Wójcicka
- Department of Clinical Biochemistry, Pediatric Institute, Faculty of Medicine, Jagiellonian University Medical College, 30-663 Krakow, Poland; (A.M.); (M.W.); (I.R.)
| | - Iwona Rogatko
- Department of Clinical Biochemistry, Pediatric Institute, Faculty of Medicine, Jagiellonian University Medical College, 30-663 Krakow, Poland; (A.M.); (M.W.); (I.R.)
| | - Tomasz Piskorz
- Chair in Gynecology and Obstetrics, Faculty of Medicine, Jagiellonian University Medical College, 31-008 Krakow, Poland;
| | - Przemysław J. Tomasik
- Department of Clinical Biochemistry, Pediatric Institute, Faculty of Medicine, Jagiellonian University Medical College, 30-663 Krakow, Poland; (A.M.); (M.W.); (I.R.)
| |
Collapse
|
5
|
Matar A, Damianos JA, Jencks KJ, Camilleri M. Intestinal Barrier Impairment, Preservation, and Repair: An Update. Nutrients 2024; 16:3494. [PMID: 39458489 PMCID: PMC11509958 DOI: 10.3390/nu16203494] [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: 09/19/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Our objective was to review published studies of the intestinal barrier and permeability, the deleterious effects of dietary components (particularly fat), the impact of altered intestinal permeability in disease models and human diseases, the role of the microbiome and epigenomics in control of barrier function, and the opportunities to restore normal barrier function with dietary interventions and products of the microbiota. METHODS We conducted a literature review including the following keywords alone or in combination: intestinal barrier, permeability, microbiome, epigenomics, diet, irritable bowel syndrome, inflammatory bowel disease, probiotics. RESULTS Intestinal permeability is modified by a diet including fat, which increases permeability, and nutrients such as fiber, glutamine, zinc, vitamin D, polyphenols, emulsifiers, and anthocyanins, which decrease permeability. There is significant interaction of the microbiome and barrier function, including the inflammatory of luminal/bacterial antigens, and anti-inflammatory effects of commensals or probiotics and their products, including short-chain fatty acids. Epigenomic modification of barrier functions are best illustrated by effects on junction proteins or inflammation. Detailed documentation of the protective effects of diet, probiotics, prebiotics, and microbiota is provided. CONCLUSION intestinal permeability is a critical factor in protection against gastrointestinal diseases and is impacted by nutrients that preserve or heal and repair the barrier and nurture anti-inflammatory effects.
Collapse
Affiliation(s)
| | | | | | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA; (A.M.); (J.A.D.)
| |
Collapse
|
6
|
Pan Y, Li J, Fan Z, Chen Y, Huang X, Wu D. New Insights into Chronic Pancreatitis: Potential Mechanisms Related to Probiotics. Microorganisms 2024; 12:1760. [PMID: 39338435 PMCID: PMC11434092 DOI: 10.3390/microorganisms12091760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
Chronic pancreatitis is a progressive fibroinflammatory disorder with no currently satisfactory treatment. Emerging evidence suggests an association between gut microbial dysbiosis and chronic pancreatitis. Although direct causative evidence is lacking, it is hypothesized that the gut microbiota may play a pivotal role in modulating pancreatic function via the gut-pancreas axis. Thus, modulating the gut microbiota through the administration of probiotics or prebiotics may alleviate pancreatic disorders. In this review, we first propose the potential mechanisms by which specific probiotics or prebiotics may ameliorate chronic pancreatitis, including the alleviation of small intestinal bacterial overgrowth (SIBO), the facilitation of short-chain fatty acids' (SCFAs) production, and the activation of glucagon-like peptide-1 receptors (GLP-1Rs) in the pancreas. Since there are currently no probiotics or prebiotics used for the treatment of chronic pancreatitis, we discuss research in other disease models that have used probiotics or prebiotics to modulate pancreatic endocrine and exocrine functions and prevent pancreatic fibrosis. This provides indirect evidence for their potential application in the treatment of chronic pancreatitis. We anticipate that this research will stimulate further investigation into the gut-pancreas axis and the potential therapeutic value of probiotics and prebiotics in chronic pancreatitis.
Collapse
Affiliation(s)
- Yingyu Pan
- Department of Gastroenterology, State Key Laborotary of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jianing Li
- Department of Gastroenterology, State Key Laborotary of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Zhengyang Fan
- Department of Gastroenterology, State Key Laborotary of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yonghao Chen
- Department of Gastroenterology, State Key Laborotary of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xiaoxuan Huang
- Department of Gastroenterology, State Key Laborotary of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Dong Wu
- Department of Gastroenterology, State Key Laborotary of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
7
|
Knez E, Kadac-Czapska K, Grembecka M. The importance of food quality, gut motility, and microbiome in SIBO development and treatment. Nutrition 2024; 124:112464. [PMID: 38657418 DOI: 10.1016/j.nut.2024.112464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 03/18/2024] [Accepted: 04/04/2024] [Indexed: 04/26/2024]
Abstract
The prevalence of small intestinal bacterial overgrowth (SIBO) is rising worldwide, particularly in nations with high rates of urbanization. Irritable bowel syndrome, inflammatory bowel illnesses, and nonspecific dysmotility are strongly linked to SIBO. Moreover, repeated antibiotic therapy promotes microorganisms' overgrowth through the development of antibiotic resistance. The primary cause of excessive fermentation in the small intestine is a malfunctioning gastrointestinal motor complex, which results in the gut's longer retention of food residues. There are anatomical and physiological factors affecting the functioning of the myoelectric motor complex. Except for them, diet conditions the activity of gastrointestinal transit. Indisputably, the Western type of nutrition is unfavorable. Some food components have greater importance in the functioning of the gastrointestinal motor complex than others. Tryptophan, an essential amino acid and precursor of the serotonin hormone, accelerates intestinal transit, and gastric emptying, similarly to fiber and polyphenols. Additionally, the effect of food on the microbiome is important, and diet should prevent bacterial overgrowth and exhibit antimicrobial effects against pathogens. Therefore, knowledge about proper nutrition is essential to prevent the development and recurrence of SIBO. Since the scientific world was unsure whether there was a long-term or potential solution for SIBO until quite recently, research on a number of the topics included in the article should be performed. The article aimed to summarize current knowledge about proper nutrition after SIBO eradication and the prevention of recurrent bacterial overgrowth. Moreover, a connection was found between diet, gut dysmotility, and SIBO.
Collapse
Affiliation(s)
- Eliza Knez
- Department of Bromatology, Medical University of Gdańsk, Gdańsk, Poland
| | | | | |
Collapse
|
8
|
Sivaprasadan S, Anila KN, Nair K, Mallick S, Biswas L, Valsan A, Praseedom RK, Nair BKG, Sudhindran S. Microbiota and Gut-Liver Axis: An Unbreakable Bond? Curr Microbiol 2024; 81:193. [PMID: 38805045 DOI: 10.1007/s00284-024-03694-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/08/2024] [Indexed: 05/29/2024]
Abstract
The gut microbiota, amounting to approximately 100 trillion (1014) microbes represents a genetic repertoire that is bigger than the human genome itself. Evidence on bidirectional interplay between human and microbial genes is mounting. Microbiota probably play vital roles in diverse aspects of normal human metabolism, such as digestion, immune modulation, and gut endocrine function, as well as in the genesis and progression of many human diseases. Indeed, the gut microbiota has been most closely linked to various chronic ailments affecting the liver, although concrete scientific data are sparse. In this narrative review, we initially discuss the basic epidemiology of gut microbiota and the factors influencing their initial formation in the gut. Subsequently, we delve into the gut-liver axis and the evidence regarding the link between gut microbiota and the genesis or progression of various liver diseases. Finally, we summarise the recent research on plausible ways to modulate the gut microbiota to alter the natural history of liver disease.
Collapse
Affiliation(s)
- Saraswathy Sivaprasadan
- Department of Gastrointestinal Surgery and Solid Organ Transplantation, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, India
| | - K N Anila
- Department of Gastrointestinal Surgery and Solid Organ Transplantation, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Krishnanunni Nair
- Department of Gastrointestinal Surgery and Solid Organ Transplantation, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Shweta Mallick
- Department of Gastrointestinal Surgery and Solid Organ Transplantation, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Lalitha Biswas
- Amrita School of Nanosciences and Molecular Medicine, Kochi, India
| | - Arun Valsan
- Department of Hepatology & Gastroenterology, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, India
| | | | | | - Surendran Sudhindran
- Department of Gastrointestinal Surgery and Solid Organ Transplantation, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, India.
| |
Collapse
|
9
|
Chi X, Sun X, Cheng D, Liu S, Q. Pan C, Xing H. Intestinal microbiome-targeted therapies improve liver function in alcohol-related liver disease by restoring bifidobacteria: a systematic review and meta-analysis. Front Pharmacol 2024; 14:1274261. [PMID: 38259268 PMCID: PMC10800551 DOI: 10.3389/fphar.2023.1274261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/05/2023] [Indexed: 01/24/2024] Open
Abstract
Objective: To systematically evaluate the efficacy of intestinal microbiome-targeted therapies (MTTs) in alcohol-related liver disease (ALD). Methods: With pre-specified keywords and strategies, we searched databases including Cochrane Library, PubMed, EMBASE, CNKI, Wanfang Data, and Weipu for RCTs on intestinal MTTs in ALD patients from January 2000 to May 2021. Two researchers independently conducted literature screening, data extraction, and quality evaluation according to the eligible criteria. Outcomes of interest included the effects of intestinal MTTs on ALT, AST, GGT, TBIL, TNF-α, IL-6, intestinal Escherichia coli, and Bifidobacteria when compared to the control group. Pooled data were compiled and analyzed with Revman 5.4 software. Results: Among 5 RCTs included with 456 ALD patients who received probiotics, the therapeutic pooled effects in the experimental group were the followings: ALT (MD = -7.16.95% CI: 10.71∼-3.60; p < 0.0001)、AST (MD = -25.11.95% CI: 30.57∼-19.47; p < 0.00001)、GGT (MD = -6.72.95% CI: 11.91∼-1.53; p = 0.01)、IL-6(SMD = -0.82.95% CI: 1.10∼-0.54; p < 0.00001), which were significantly better than those in the placebo or standard treatment group respectively, while the difference of TBIL (SMD = -0.06, 95%CI: 0.29-0.16; p = 0.59), TNF-α(SMD = -0.53.95% CI: 1.57-0.50; p = 0.31)in the two groups was not significant. After intestinal MTT treatment, the number of intestinal Bifidobacteria increased significantly (MD = 0.79.95% CI: 0.00-1.58; p = 0.05)in the experimental group. However, there were no significant changes in the number of E. coli in both groups (SMD = -0.29.95% CI: 0.92-0.34; p = 0.36). Conclusion: Intestinal MTTs can significantly improve liver function, associated with the increase of intestinal Bifidobacteria, which may be beneficial to ALD. Systematic Review Registration: https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021246067, Identifier CRD42021246067.
Collapse
Affiliation(s)
- Xin Chi
- Center of Liver Diseases Division, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Diseases, Beijing, China
| | - Xiu Sun
- Center of Liver Diseases Division, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Diseases, Beijing, China
| | - Danying Cheng
- Center of Liver Diseases Division, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Diseases, Beijing, China
- Peking University Ditan Teaching Hospital, Beijing, China
| | - Shunai Liu
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Diseases, Beijing, China
| | - Calvin Q. Pan
- Center of Liver Diseases Division, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Diseases, Beijing, China
- Division of Gastroenterology and Hepatology, NYU Langone Health, New York University School of Medicine, New York, NY, United States
| | - Huichun Xing
- Center of Liver Diseases Division, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Diseases, Beijing, China
- Peking University Ditan Teaching Hospital, Beijing, China
| |
Collapse
|
10
|
Maslennikov R, Alieva A, Poluektova E, Zharikov Y, Suslov A, Letyagina Y, Vasileva E, Levshina A, Kozlov E, Ivashkin V. Sarcopenia in cirrhosis: Prospects for therapy targeted to gut microbiota. World J Gastroenterol 2023; 29:4236-4251. [PMID: 37545638 PMCID: PMC10401661 DOI: 10.3748/wjg.v29.i27.4236] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/25/2023] [Accepted: 06/21/2023] [Indexed: 07/13/2023] Open
Abstract
Decreased muscle mass and function, also known as sarcopenia, is common in patients with cirrhosis and is associated with a poor prognosis. Although the pathogenesis of this disorder has not been fully elucidated, a disordered gut-muscle axis probably plays an important role. Decreased barrier function of the gut and liver, gut dysbiosis, and small intestinal bacterial overgrowth (SIBO) can lead to increased blood levels of ammonia, lipopolysaccharides, pro-inflammatory mediators, and myostatin. These factors have complex negative effects on muscle mass and function. Drug interventions that target the gut microbiota (long-term use of rifaximin, lactulose, lactitol, or probiotics) positively affect most links of the compromised gut-muscle axis in patients with cirrhosis by decreasing the levels of hyperammonemia, bacterial translocation, and systemic inflammation and correcting gut dysbiosis and SIBO. However, although these drugs are promising, they have not yet been investigated in randomized controlled trials specifically for the treatment and prevention of sarcopenia in patients with cirrhosis. No data exist on the effects of fecal transplantation on most links of gut-muscle axis in cirrhosis; however, the results of animal experimental studies are promising.
Collapse
Affiliation(s)
- Roman Maslennikov
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow 119435, Russia
- The Scientific Community for Human Microbiome Research, Moscow 119435, Russia
| | - Aliya Alieva
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow 119435, Russia
| | - Elena Poluektova
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow 119435, Russia
- The Scientific Community for Human Microbiome Research, Moscow 119435, Russia
| | - Yury Zharikov
- Department of Human Anatomy and Histology, Sechenov University, Moscow 119435, Russia
| | - Andrey Suslov
- Department of Human Anatomy and Histology, Sechenov University, Moscow 119435, Russia
| | - Yana Letyagina
- Department of Human Anatomy and Histology, Sechenov University, Moscow 119435, Russia
| | - Ekaterina Vasileva
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow 119435, Russia
| | - Anna Levshina
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow 119435, Russia
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergy, Sechenov University, Moscow 119991, Russia
| | - Evgenii Kozlov
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergy, Sechenov University, Moscow 119991, Russia
| | - Vladimir Ivashkin
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow 119435, Russia
- The Scientific Community for Human Microbiome Research, Moscow 119435, Russia
| |
Collapse
|
11
|
Kaufmann B, Seyfried N, Hartmann D, Hartmann P. Probiotics, prebiotics, and synbiotics in nonalcoholic fatty liver disease and alcohol-associated liver disease. Am J Physiol Gastrointest Liver Physiol 2023; 325:G42-G61. [PMID: 37129252 PMCID: PMC10312326 DOI: 10.1152/ajpgi.00017.2023] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/26/2023] [Accepted: 04/26/2023] [Indexed: 05/03/2023]
Abstract
The use of probiotics, prebiotics, and synbiotics has become an important therapy in numerous gastrointestinal diseases in recent years. Modifying the gut microbiota, this therapeutic approach helps to restore a healthy microbiome. Nonalcoholic fatty liver disease and alcohol-associated liver disease are among the leading causes of chronic liver disease worldwide. A disrupted intestinal barrier, microbial translocation, and an altered gut microbiome metabolism, or metabolome, are crucial in the pathogenesis of these chronic liver diseases. As pro-, pre-, and synbiotics modulate these targets, they were identified as possible new treatment options for liver disease. In this review, we highlight the current findings on clinical and mechanistic effects of this therapeutic approach in nonalcoholic fatty liver disease and alcohol-associated liver disease.
Collapse
Affiliation(s)
- Benedikt Kaufmann
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Nick Seyfried
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Daniel Hartmann
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Phillipp Hartmann
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States
| |
Collapse
|
12
|
Sun J, Zhang M, Liu W, Liu Y, Zhang D, Fan X, Zhang J, Li T, Lu M. Evaluation of the effectiveness and mechanism of action of the Chang-Kang-Fang formula combined with bifid triple viable capsules on diarrhea-predominant irritable bowel syndrome. Front Microbiol 2023; 14:1160783. [PMID: 37440881 PMCID: PMC10333534 DOI: 10.3389/fmicb.2023.1160783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/23/2023] [Indexed: 07/15/2023] Open
Abstract
Introduction The Chang-Kang-Fang (CKF) formula, a traditional Chinese herbal formula, can decrease serotonin (5-HT) levels and treat irritable bowel syndrome (IBS). Probiotics have a better synergistic effect on diarrhea-predominant IBS (IBS-D) when combined with 5-HT3 receptor antagonists. The present study aimed to elucidate the efficacy and the mechanisms of action of the CKF formula combined with bifid triple viable capsules (PFK) against IBS-D. Methods The rat models of IBS-D were induced by gavage with senna decoction plus restraint stress. The CKF formula, PFK and their combination were administered to the rats. Their effects were evaluated based on general condition of the rats and the AWR score. The levels of 5-HT and fos protein in the colon and hippocampus were measured by immunohistochemistry. The levels of SP and VIP, as well as ZO-1 and occludin in the colon, were determined by enzyme-linked immunosorbent assay and immunohistochemistry. The intestinal microbiota in faeces was analyzed by 16S rRNA high-throughput sequencing. Results The results showed that the oral CKF formula combined with PFK (CKF + PFK) could significantly relieve the symptoms of IBS-D, including elevating the weight rate and decreasing the AWR score. Compared with the MC group, administration of CKF + PFK significantly reduced the expression of fos in the colon and hippocampus and that of 5-HT, SP and VIP in the colon and increased the levels of 5-HT in the hippocampus and ZO-1 and occludin in the colon. The above indexes exhibited statistical significance in the CKF + PFK group relative to those in the other groups. Moreover, treatment with CKF + PFK improved the diversity of intestinal microbiota and the abundance of Firmicutes, Lachnospiraceae and Ruminococcaceae but decreased those of Bacteroidetes and Prevotellaceae. Conclusions The CKF formula combined with PFK may have a synergistic effect on IBS-D by slowing gastrointestinal motility, lowering visceral hypersensitivity, enhancing the intestinal barrier function and modulating the composition of intestinal microbiota.
Collapse
Affiliation(s)
- Jing Sun
- Department of Central laboratory, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, China
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Mengqiu Zhang
- School of Third Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Suqian Hospital of Traditional Chinese Medicine, Suqian, China
| | - Wei Liu
- School of Third Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Youqian Liu
- Department of Gastroenterology, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Dongjian Zhang
- School of Third Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinyu Fan
- Department of Gastroenterology, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jian Zhang
- School of Third Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tian Li
- Department of Central laboratory, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Min Lu
- School of Third Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
13
|
Mishra G, Singh P, Molla M, Yimer YS, Dinda SC, Chandra P, Singh BK, Dagnew SB, Assefa AN, Ewunetie A. Harnessing the potential of probiotics in the treatment of alcoholic liver disorders. Front Pharmacol 2023; 14:1212742. [PMID: 37361234 PMCID: PMC10287977 DOI: 10.3389/fphar.2023.1212742] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
In the current scenario, prolonged consumption of alcohol across the globe is upsurging an appreciable number of patients with the risk of alcohol-associated liver diseases. According to the recent report, the gut-liver axis is crucial in the progression of alcohol-induced liver diseases, including steatosis, steatohepatitis, fibrosis, cirrhosis, and hepatocellular carcinoma. Despite several factors associated with alcoholic liver diseases, the complexity of the gut microflora and its great interaction with the liver have become a fascinating area for researchers due to the high exposure of the liver to free radicals, bacterial endotoxins, lipopolysaccharides, inflammatory markers, etc. Undoubtedly, alcohol-induced gut microbiota imbalance stimulates dysbiosis, disrupts the intestinal barrier function, and trigger immune as well as inflammatory responses which further aggravate hepatic injury. Since currently available drugs to mitigate liver disorders have significant side effects, hence, probiotics have been widely researched to alleviate alcohol-associated liver diseases and to improve liver health. A broad range of probiotic bacteria like Lactobacillus, Bifidobacteria, Escherichia coli, Sacchromyces, and Lactococcus are used to reduce or halt the progression of alcohol-associated liver diseases. Several underlying mechanisms, including alteration of the gut microbiome, modulation of intestinal barrier function and immune response, reduction in the level of endotoxins, and bacterial translocation, have been implicated through which probiotics can effectively suppress the occurrence of alcohol-induced liver disorders. This review addresses the therapeutic applications of probiotics in the treatment of alcohol-associated liver diseases. Novel insights into the mechanisms by which probiotics prevent alcohol-associated liver diseases have also been elaborated.
Collapse
Affiliation(s)
- Garima Mishra
- Pharmaceutical Chemistry Unit, Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Pradeep Singh
- Pharmaceutical Chemistry Unit, Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Mulugeta Molla
- Pharmacology and Toxicology Unit, Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Yohannes Shumet Yimer
- Social Pharmacy Unit, Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | | | - Phool Chandra
- Department of Pharmacology, Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad, India
| | | | - Samuel Berihun Dagnew
- Clinical Pharmacy Unit, Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Abraham Nigussie Assefa
- Social Pharmacy Unit, Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Amien Ewunetie
- Pharmacology and Toxicology Unit, Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| |
Collapse
|
14
|
Kiecka A, Szczepanik M. Proton pump inhibitor-induced gut dysbiosis and immunomodulation: current knowledge and potential restoration by probiotics. Pharmacol Rep 2023:10.1007/s43440-023-00489-x. [PMID: 37142877 PMCID: PMC10159235 DOI: 10.1007/s43440-023-00489-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/06/2023]
Abstract
Proton pump inhibitors (PPIs) are the most commonly prescribed drugs for the treatment of non-erosive reflux disease (NERD), ulcers associated with non-steroidal anti-inflammatory drugs (NSAIDs), esophagitis, peptic ulcer disease (PUD), Zollinger-Ellison syndrome (ZES), gastroesophageal reflux disease (GERD), non-ulcer dyspepsia, and Helicobacter pylori eradication therapy. The drugs have the effect of inhibiting acid production in the stomach. According to research, PPIs can affect the composition of gut microbiota and modulate the immune response. Recently, there has been a problem with the over-prescription of such drugs. Although PPIs do not have many side effects, their long-term use can contribute to small intestinal bacterial overgrowth (SIBO) or C. difficile and other intestinal infections. Probiotic supplementation during PPIs therapy may provide some hope in the reduction of emerging therapy side effects. This review aims to present the most important effects of long-term PPI use and provides critical insights into the role of probiotic intervention in PPI therapy.
Collapse
Affiliation(s)
- Aneta Kiecka
- Chair of Biomedical Sciences, Institute of Physiotherapy, Faculty of Health Sciences, Jagiellonian University Medical College, Kopernika 7a, 31-034, Kraków, Poland.
| | - Marian Szczepanik
- Chair of Biomedical Sciences, Institute of Physiotherapy, Faculty of Health Sciences, Jagiellonian University Medical College, Kopernika 7a, 31-034, Kraków, Poland
| |
Collapse
|
15
|
Zheng Y, Zhang Z, Tang P, Wu Y, Zhang A, Li D, Wang CZ, Wan JY, Yao H, Yuan CS. Probiotics fortify intestinal barrier function: a systematic review and meta-analysis of randomized trials. Front Immunol 2023; 14:1143548. [PMID: 37168869 PMCID: PMC10165082 DOI: 10.3389/fimmu.2023.1143548] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 04/10/2023] [Indexed: 05/13/2023] Open
Abstract
Background Probiotics play a vital role in treating immune and inflammatory diseases by improving intestinal barrier function; however, a comprehensive evaluation is missing. The present study aimed to explore the impact of probiotics on the intestinal barrier and related immune function, inflammation, and microbiota composition. A systematic review and meta-analyses were conducted. Methods Four major databases (PubMed, Science Citation Index Expanded, CENTRAL, and Embase) were thoroughly searched. Weighted mean differences were calculated for continuous outcomes with corresponding 95% confidence intervals (CIs), heterogeneity among studies was evaluated utilizing I2 statistic (Chi-Square test), and data were pooled using random effects meta-analyses. Results Meta-analysis of data from a total of 26 RCTs (n = 1891) indicated that probiotics significantly improved gut barrier function measured by levels of TER (MD, 5.27, 95% CI, 3.82 to 6.72, P < 0.00001), serum zonulin (SMD, -1.58, 95% CI, -2.49 to -0.66, P = 0.0007), endotoxin (SMD, -3.20, 95% CI, -5.41 to -0.98, P = 0.005), and LPS (SMD, -0.47, 95% CI, -0.85 to -0.09, P = 0.02). Furthermore, probiotic groups demonstrated better efficacy over control groups in reducing inflammatory factors, including CRP, TNF-α, and IL-6. Probiotics can also modulate the gut microbiota structure by boosting the enrichment of Bifidobacterium and Lactobacillus. Conclusion The present work revealed that probiotics could improve intestinal barrier function, and alleviate inflammation and microbial dysbiosis. Further high-quality RCTs are warranted to achieve a more definitive conclusion. Clinical trial registration https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=281822, identifier CRD42021281822.
Collapse
Affiliation(s)
- Yanfei Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zengliang Zhang
- Traditional Chinese Medicine College, Inner Mongolia Medical University, Inner Mongolia, China
| | - Ping Tang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuqi Wu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Anqi Zhang
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Delong Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chong-Zhi Wang
- Tang Center for Herbal Medicine Research, The University of Chicago, Chicago, IL, United States
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, IL, United States
| | - Jin-Yi Wan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Jin-Yi Wan, ; Haiqiang Yao,
| | - Haiqiang Yao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Jin-Yi Wan, ; Haiqiang Yao,
| | - Chun-Su Yuan
- Tang Center for Herbal Medicine Research, The University of Chicago, Chicago, IL, United States
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
16
|
Gupta H, Kim SH, Kim SK, Han SH, Kwon HC, Suk KT. Beneficial Shifts in Gut Microbiota by Lacticaseibacillus rhamnosus R0011 and Lactobacillus helveticus R0052 in Alcoholic Hepatitis. Microorganisms 2022; 10:microorganisms10071474. [PMID: 35889193 PMCID: PMC9319967 DOI: 10.3390/microorganisms10071474] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 02/01/2023] Open
Abstract
Gut microbiota performs indispensable functions in the pathophysiology of alcoholic hepatitis (AH). We investigated the effects of Lacticaseibacillus rhamnosus R0011 and Lactobacillus helveticus for gut microbial restoration toward eubiosis in patients with AH. A multicenter, double-blind, and randomized trial was conducted. Probiotics (n = 44) and placebo (n = 45) groups received, during 7 days, L. rhamnosus R0011/L. helveticus R0052 at 120 mg/day and placebo. All patients were hospitalized to ensure abstinence. Liver function, lipopolysaccharide level, and stool analysis were evaluated in patients before and after 7 days of treatment. At baseline, the dominant bacteria were Gram-negative in both groups which decreased after the probiotics treatment and exhibited a significant reduction in lipopolysaccharide level (p < 0.001). The probiotics ameliorated the Child−Pugh scores (p < 0.001). Furthermore, the probiotics group showed a decline in the levels of alanine aminotransferase and gamma-glutamyltranspeptidase (p < 0.05). The probiotics changed the gut microbial composition at various taxonomical levels. The proportion of Bacteroidetes (147%) was increased after 7 days of probiotics supplementation while Proteobacteria (30%) and Fusobacteria (0%) were decreased. Administration of L. rhamnosus R0011 and L. helveticus R0052 conceivably associated with restoration of gut microbiome in AH patients and improved AH by modulating the gut−liver axis.
Collapse
Affiliation(s)
- Haripriya Gupta
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, Korea; (H.G.); (S.K.K.)
| | - Sung Hun Kim
- Korea Institute of Science and Technology, Gangneung 25451, Korea;
| | - Seul Ki Kim
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, Korea; (H.G.); (S.K.K.)
| | - Sang Hak Han
- Department of Pathology, Hallym University College of Medicine, Chuncheon 24252, Korea;
| | - Hak Cheol Kwon
- Korea Institute of Science and Technology, Gangneung 25451, Korea;
- Correspondence: (H.C.K.); (K.T.S.)
| | - Ki Tae Suk
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, Korea; (H.G.); (S.K.K.)
- Correspondence: (H.C.K.); (K.T.S.)
| |
Collapse
|
17
|
Rode J, Edebol Carlman HMT, König J, Repsilber D, Hutchinson AN, Thunberg P, Andersson P, Persson J, Kiselev A, Lathrop Stern L, Salomon B, Mohammed AA, Labus JS, Brummer RJ. Probiotic Mixture Containing Lactobacillus helveticus, Bifidobacterium longum and Lactiplantibacillus plantarum Affects Brain Responses Toward an Emotional Task in Healthy Subjects: A Randomized Clinical Trial. Front Nutr 2022; 9:827182. [PMID: 35571902 PMCID: PMC9104811 DOI: 10.3389/fnut.2022.827182] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 04/05/2022] [Indexed: 01/04/2023] Open
Abstract
Background Evidence from preclinical studies suggests that probiotics affect brain function via the microbiome-gut-brain axis, but evidence in humans remains limited. Objective The present proof-of-concept study investigated if a probiotic product containing a mixture of Bifidobacterium longum R0175, Lactobacillus helveticus R0052 and Lactiplantibacillus plantarum R1012 (in total 3 × 109 CFU/day) affected functional brain responses in healthy subjects during an emotional attention task. Design In this double-blinded, randomized, placebo-controlled crossover study (Clinicaltrials.gov, NCT03615651), 22 healthy subjects (24.2 ± 3.4 years, 6 males/16 females) were exposed to a probiotic intervention and a placebo for 4 weeks each, separated by a 4-week washout period. Subjects underwent functional magnetic resonance imaging while performing an emotional attention task after each intervention period. Differential brain activity and functional connectivity were assessed. Results Altered brain responses were observed in brain regions implicated in emotional, cognitive and face processing. Increased activation in the orbitofrontal cortex, a region that receives extensive sensory input and in turn projects to regions implicated in emotional processing, was found after probiotic intervention compared to placebo using a cluster-based analysis of functionally defined areas. Significantly reduced task-related functional connectivity was observed after the probiotic intervention compared to placebo. Fecal microbiota composition was not majorly affected by probiotic intervention. Conclusion The probiotic intervention resulted in subtly altered brain activity and functional connectivity in healthy subjects performing an emotional task without major effects on the fecal microbiota composition. This indicates that the probiotic effects occurred via microbe-host interactions on other levels. Further analysis of signaling molecules could give possible insights into the modes of action of the probiotic intervention on the gut-brain axis in general and brain function specifically. The presented findings further support the growing consensus that probiotic supplementation influences brain function and emotional regulation, even in healthy subjects. Future studies including patients with altered emotional processing, such as anxiety or depression symptoms are of great interest. Clinical Trial Registration [http://clinicaltrials.gov/], identifier [NCT03615651].
Collapse
Affiliation(s)
- Julia Rode
- Nutrition-Gut-Brain Interactions Research Center, Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
- *Correspondence: Julia Rode,
| | - Hanna M. T. Edebol Carlman
- Nutrition-Gut-Brain Interactions Research Center, Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Julia König
- Nutrition-Gut-Brain Interactions Research Center, Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Dirk Repsilber
- Nutrition-Gut-Brain Interactions Research Center, Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Ashley N. Hutchinson
- Nutrition-Gut-Brain Interactions Research Center, Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Per Thunberg
- Department of Radiology and Medical Physics, Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Pernilla Andersson
- Center for Lifespan Developmental Research (LEADER), Faculty of Humanities and Social Sciences, School of Law, Psychology and Social Work, Örebro University, Örebro, Sweden
| | - Jonas Persson
- Center for Lifespan Developmental Research (LEADER), Faculty of Humanities and Social Sciences, School of Law, Psychology and Social Work, Örebro University, Örebro, Sweden
| | - Andrey Kiselev
- Center for Applied Autonomous Sensor Systems, Faculty for Business, Science and Engineering, School of Natural Science and Technology, Örebro University, Örebro, Sweden
| | - Lori Lathrop Stern
- Global Medical Innovation, Pfizer Consumer Healthcare, Madison, NJ, United States
| | - Benita Salomon
- Nutrition-Gut-Brain Interactions Research Center, Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Ahmed Abdulilah Mohammed
- Department of Radiology and Medical Physics, Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Jennifer S. Labus
- Integrative Bioinformatics and Biostatistics Core, Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Robert J. Brummer
- Nutrition-Gut-Brain Interactions Research Center, Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
18
|
Chen B, Huang H, Pan CQ. The role of gut microbiota in hepatitis B disease progression and treatment. J Viral Hepat 2022; 29:94-106. [PMID: 34415656 DOI: 10.1111/jvh.13595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 01/05/2023]
Abstract
Current therapeutic interventions can only suppress hepatitis B virus (HBV) replication or reduce complications without a cure. Therefore, further development of new treatment methods is critical for the global eradication of HBV. Accumulating evidence suggests that the liver and gut share an interconnected relationship referred to as the 'Gut-Liver Axis', where exchanges happen bi-directionally. The gut itself is the host to a unique microbiota profile which has metabolic, immunological, neurological and nutritional functions. Gut microbiota is not only constantly intersecting with the liver but also associated with hepatic injury when dysbiosis occurs. In recent years, there has been increased interest in gut microbiota and its implications on liver disease treatment. Progress has been made in understanding the complex relationship between chronic hepatitis B (CHB) and gut microbiota. New investigative techniques such as colony-free sequencing enabled new perspectives into this field. Mouse models and human studies revealed that HBV infection is associated with significant alteration of gut microbiota, which differ depending on the stage of CHB disease progression. Different mechanisms of the hepatic injury from gut microbiota dysbiosis have also been proposed based on findings of increased intestinal permeability to toxins, disruption of normal bacterial metabolism, and colonization of the gut by oral microbiota. New treatment methods targeting gut microbiota in CHB, such as probiotics and faecal microbiota transplant, have also gained promising results in recent years. The current review recapitulated the most recent investigations into the relationship between gut microbiota and CHB to provide research directions towards the new therapeutic target of CHB.
Collapse
Affiliation(s)
- Bryan Chen
- University of California, Los Angeles, California, USA
| | - Harry Huang
- Stony Brook University, Stony Brook, New York, USA
| | - Calvin Q Pan
- Center of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Division of Gastroenterology and Hepatology, Department of Medicine, NYU Langone Health, NYU School of Medicine, New York, New York, USA
| |
Collapse
|
19
|
Helsley RN, Miyata T, Kadam A, Varadharajan V, Sangwan N, Huang EC, Banerjee R, Brown AL, Fung KK, Massey WJ, Neumann C, Orabi D, Osborn LJ, Schugar RC, McMullen MR, Bellar A, Poulsen KL, Kim A, Pathak V, Mrdjen M, Anderson JT, Willard B, McClain CJ, Mitchell M, McCullough AJ, Radaeva S, Barton B, Szabo G, Dasarathy S, Garcia-Garcia JC, Rotroff DM, Allende DS, Wang Z, Hazen SL, Nagy LE, Brown JM. Gut microbial trimethylamine is elevated in alcohol-associated hepatitis and contributes to ethanol-induced liver injury in mice. eLife 2022; 11:e76554. [PMID: 35084335 PMCID: PMC8853661 DOI: 10.7554/elife.76554] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 12/31/2021] [Indexed: 11/13/2022] Open
Abstract
There is mounting evidence that microbes residing in the human intestine contribute to diverse alcohol-associated liver diseases (ALD) including the most deadly form known as alcohol-associated hepatitis (AH). However, mechanisms by which gut microbes synergize with excessive alcohol intake to promote liver injury are poorly understood. Furthermore, whether drugs that selectively target gut microbial metabolism can improve ALD has never been tested. We used liquid chromatography tandem mass spectrometry to quantify the levels of microbe and host choline co-metabolites in healthy controls and AH patients, finding elevated levels of the microbial metabolite trimethylamine (TMA) in AH. In subsequent studies, we treated mice with non-lethal bacterial choline TMA lyase (CutC/D) inhibitors to blunt gut microbe-dependent production of TMA in the context of chronic ethanol administration. Indices of liver injury were quantified by complementary RNA sequencing, biochemical, and histological approaches. In addition, we examined the impact of ethanol consumption and TMA lyase inhibition on gut microbiome structure via 16S rRNA sequencing. We show the gut microbial choline metabolite TMA is elevated in AH patients and correlates with reduced hepatic expression of the TMA oxygenase flavin-containing monooxygenase 3 (FMO3). Provocatively, we find that small molecule inhibition of gut microbial CutC/D activity protects mice from ethanol-induced liver injury. CutC/D inhibitor-driven improvement in ethanol-induced liver injury is associated with distinct reorganization of the gut microbiome and host liver transcriptome. The microbial metabolite TMA is elevated in patients with AH, and inhibition of TMA production from gut microbes can protect mice from ethanol-induced liver injury.
Collapse
Affiliation(s)
- Robert N Helsley
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology, and Nutrition, College of Medicine, University of KentuckyLexingtonUnited States
| | - Tatsunori Miyata
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Anagha Kadam
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Venkateshwari Varadharajan
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Naseer Sangwan
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Emily C Huang
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Rakhee Banerjee
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Amanda L Brown
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Kevin K Fung
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - William J Massey
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Chase Neumann
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Danny Orabi
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Lucas J Osborn
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Rebecca C Schugar
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Megan R McMullen
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Annette Bellar
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Kyle L Poulsen
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Adam Kim
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Vai Pathak
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Marko Mrdjen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - James T Anderson
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Belinda Willard
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Craig J McClain
- Department of Medicine, University of LouisvilleLouisvilleUnited States
| | - Mack Mitchell
- Department of Internal Medicine, University of Texas Southwestern Medical CenterDallasUnited States
| | - Arthur J McCullough
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Svetlana Radaeva
- National Institute on Alcohol Abuse and AlcoholismBethesdaUnited States
| | - Bruce Barton
- Department of Population and Quantitative Health Sciences, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Gyongyi Szabo
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical SchoolBostonUnited States
| | - Srinivasan Dasarathy
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | | | - Daniel M Rotroff
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Daniela S Allende
- Department of Anatomical Pathology, Cleveland ClinicClevelandUnited States
| | - Zeneng Wang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Stanley L Hazen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Cardiovascular Medicine, Heart and Vascular and Thoracic Institute, Cleveland ClinicClevelandUnited States
| | - Laura E Nagy
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Jonathan Mark Brown
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| |
Collapse
|
20
|
Milosevic I, Russo E, Vujovic A, Barac A, Stevanovic O, Gitto S, Amedei A. Microbiota and viral hepatitis: State of the art of a complex matter. World J Gastroenterol 2021; 27:5488-5501. [PMID: 34588747 PMCID: PMC8433613 DOI: 10.3748/wjg.v27.i33.5488] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/26/2021] [Accepted: 07/21/2021] [Indexed: 02/06/2023] Open
Abstract
Changes in gut microbiota influence both the gut and liver, which are strictly connected by the so-called "gut-liver axis". The gut microbiota acts as a major determinant of this relationship in the onset and clinical course of liver diseases. According to the results of several studies, gut dysbiosis is linked to viral hepatitis, mainly hepatitis C virus and hepatitis B virus infection. Gut bacteria-derived metabolites and cellular components are key molecules that affect liver function and modulate the pathology of viral hepatitis. Recent studies showed that the gut microbiota produces various molecules, such as peptidoglycans, lipopolysaccharides, DNA, lipoteichoic acid, indole-derivatives, bile acids, and trimethylamine, which are translocated to the liver and interact with liver immune cells causing pathological effects. Therefore, the existence of crosstalk between the gut microbiota and the liver and its implications on host health and pathologic status are essential factors impacting the etiology and therapeutic approach. Concrete mechanisms behind the pathogenic role of gut-derived components on the pathogenesis of viral hepatitis remain unclear and not understood. In this review, we discuss the current findings of research on the bidirectional relationship of the components of gut microbiota and the progression of liver diseases and viral hepatitis and vice versa. Moreover, this paper highlights the current therapeutic and preventive strategies, such as fecal transplantation, used to restore the gut microbiota composition and so improve host health.
Collapse
Affiliation(s)
- Ivana Milosevic
- Clinic for Infectious and Tropical Diseases, Clinical Centre of Serbia Faculty of Medicine, University of Belgrade, Belgrade 101801, Serbia
| | - Edda Russo
- Department of Experimental and Clinical Medicine, University of Florence, Firenze 50100, Italy
| | - Ankica Vujovic
- Clinic for Infectious and Tropical Diseases, Clinical Centre of Serbia Faculty of Medicine, University of Belgrade, Belgrade 101801, Serbia
| | - Aleksandra Barac
- Clinic for Infectious and Tropical Diseases, Clinical Centre of Serbia Faculty of Medicine, University of Belgrade, Belgrade 101801, Serbia
| | - Olja Stevanovic
- Clinic for Infectious and Tropical Diseases, Clinical Centre of Serbia Faculty of Medicine, University of Belgrade, Belgrade 101801, Serbia
| | - Stefano Gitto
- Department of Experimental and Clinical Medicine, University of Florence, Firenze 50100, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Firenze 50100, Italy
| |
Collapse
|
21
|
Lu YX, Chang YZ, Liang P, Yang CQ. Effect of Additional Clostridium butyricum on the Intestinal Flora of Chronic Hepatitis B Patients Treated with Entecavir. Infect Dis Ther 2021; 10:1519-1530. [PMID: 34132991 PMCID: PMC8322241 DOI: 10.1007/s40121-021-00463-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/22/2021] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION To explore the influence of intestinal flora on the occurrence, development and antiviral therapy of chronic hepatitis B (CHB), 16S rDNA amplification sequencing was performed to investigate the intestinal flora in CHB patients treated with entecavir (ETV) and Clostridium butyricum (CB). METHODS CHB patients were divided into the ETV group (treatment with ETV alone) and ETV + CB group (treatment with ETV and CB). After 8-week treatment, feces samples were collected and processed for 16S rDNA amplicon sequencing; blood samples were collected for the biochemical, immunologic and virologic evaluations, which were compared between groups. RESULTS ETV treatment for 8 weeks significantly decreased the serum levels of alanine aminotransferase (ALT), interleukin-6 (IL-6), IL-8, tumor necrosis factor-α (TNF-α) and HBV DNA compared to those before treatment, but there were no marked differences between the ETV group and ETV + CB group. The intestinal flora changed significantly in the CHB patients after ETV + CB treatment: there were marked differences in 13 unique species before treatment and 4 unique species after ETV + CB treatment; at the phylum level, the top five bacteria with significant difference between patients before treatment and ETV + CB patients were Firmicutes, Actinobacteria, Cyanobacteria, Euryarchaeota and Synergistetes. There were significant differences in 25 unique species in the ETV group and 4 unique species in the ETV + CB group; at the phylum level, the top five bacteria with significant difference between ETV patients and ETV + CB patients were Actinobacteria, Fusobacteria, Proteobacteria, Saccharibacteria and Synergistetes. CONCLUSION ETV treatment improves the serum biochemical, immunologic and virologic variables, but additional CB fails to further improve these variables. Of note, additional CB affects the intestinal flora in the CHB patients treated with ETV.
Collapse
Affiliation(s)
- Yu-Xia Lu
- Department of Gastroenterology and Hepatology, Department of Infectious Diseases, Institution of Digestive Diseases, Tongji Hospital, Tongji University School of Medicine, 389 Xin Cun Road, Shanghai, 200065, China
| | - Yi-Zhong Chang
- Department of Gastroenterology and Hepatology, Department of Infectious Diseases, Institution of Digestive Diseases, Tongji Hospital, Tongji University School of Medicine, 389 Xin Cun Road, Shanghai, 200065, China
| | - Ping Liang
- Department of Laboratory Medicine, Tongji Hospital, Tongji University School of Medicine, 389 Xin Cun Road, Shanghai, 200065, China
| | - Chang-Qing Yang
- Department of Gastroenterology and Hepatology, Department of Infectious Diseases, Institution of Digestive Diseases, Tongji Hospital, Tongji University School of Medicine, 389 Xin Cun Road, Shanghai, 200065, China.
| |
Collapse
|
22
|
Regulating Intestinal Microbiota in the Prevention and Treatment of Alcohol-Related Liver Disease. Can J Gastroenterol Hepatol 2020; 2020:6629196. [PMID: 33381475 PMCID: PMC7759392 DOI: 10.1155/2020/6629196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/29/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023] Open
Abstract
When alcohol-related liver disease occurs, the number and composition ratio of intestinal microorganisms will accordingly change. The alcohol-induced changes in the intestinal microbiota play a pivotal role in the process of developing the alcohol-related liver disease through the translocation of microbial products due to increased intestinal permeability. In recent years, therapeutic interventions with a concentration on regulating intestinal microbiota have been conducted for patients with alcohol-related liver disease. We aimed to provide a critical review and updates on the prevention and treatment of alcohol-related liver disease through regulating intestinal microbiota. A literature search was performed on the PubMed database for studies published in English about the therapeutic intervention with microbiota using animal models and patients with alcohol-related liver disease (1/2010-4/2020). The accumulating pieces of evidence suggest that the therapeutic use of probiotics, prebiotics, antibiotics, phages, or fecal microbial transplantation may have several influences on alcohol-related liver disease patients. Emergent data unveiled that these interventions can further regulate the composition of intestinal microbiota, minimize the negative impact of microbiota on the liver, and prevent disease progression from mild to severe alcoholic hepatitis, fibrosis, cirrhosis, or even liver cancer. The current review provides updates on the advances of therapeutic interventions with the effects of regulating intestinal microbiota on patients who have alcohol-related liver disease. In addition, the data gaps and research directions on further exploration of the role of intestinal microbiota for the management of the alcohol-related liver disease are also discussed.
Collapse
|
23
|
Plaza-Díaz J, Solís-Urra P, Rodríguez-Rodríguez F, Olivares-Arancibia J, Navarro-Oliveros M, Abadía-Molina F, Álvarez-Mercado AI. The Gut Barrier, Intestinal Microbiota, and Liver Disease: Molecular Mechanisms and Strategies to Manage. Int J Mol Sci 2020; 21:8351. [PMID: 33171747 PMCID: PMC7664383 DOI: 10.3390/ijms21218351] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/31/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
Liver disease encompasses pathologies as non-alcoholic fatty liver disease, non-alcoholic steatohepatitis, alcohol liver disease, hepatocellular carcinoma, viral hepatitis, and autoimmune hepatitis. Nowadays, underlying mechanisms associating gut permeability and liver disease development are not well understood, although evidence points to the involvement of intestinal microbiota and their metabolites. Animal studies have shown alterations in Toll-like receptor signaling related to the leaky gut syndrome by the action of bacterial lipopolysaccharide. In humans, modifications of the intestinal microbiota in intestinal permeability have also been related to liver disease. Some of these changes were observed in bacterial species belonging Roseburia, Streptococcus, and Rothia. Currently, numerous strategies to treat liver disease are being assessed. This review summarizes and discusses studies addressed to determine mechanisms associated with the microbiota able to alter the intestinal barrier complementing the progress and advancement of liver disease, as well as the main strategies under development to manage these pathologies. We highlight those approaches that have shown improvement in intestinal microbiota and barrier function, namely lifestyle changes (diet and physical activity) and probiotics intervention. Nevertheless, knowledge about how such modifications are beneficial is still limited and specific mechanisms involved are not clear. Thus, further in-vitro, animal, and human studies are needed.
Collapse
Affiliation(s)
- Julio Plaza-Díaz
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada;
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18071 Granada, Spain
| | - Patricio Solís-Urra
- Faculty of Education and Social Sciences, Universidad Andres Bello, Viña del Mar 2531015, Chile;
| | - Fernando Rodríguez-Rodríguez
- IRyS Research Group, School of Physical Education, Pontificia Universidad Católica de Valparaíso, Valparaíso 2374631, Chile; (F.R.-R.); (J.O.-A.)
| | - Jorge Olivares-Arancibia
- IRyS Research Group, School of Physical Education, Pontificia Universidad Católica de Valparaíso, Valparaíso 2374631, Chile; (F.R.-R.); (J.O.-A.)
- Escuela de Pedagogía en Educación Física, Facultad de Educación, Universidad de las Américas, Santiago 8370035, Chile
| | - Miguel Navarro-Oliveros
- BioCritic. Group for Biomedical Research in Critical Care Medicine, 47005 Valladolid, Spain;
| | - Francisco Abadía-Molina
- Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. 18016 Armilla, Granada, Spain;
- Department of Cell Biology, School of Sciences, University of Granada, 18071 Granada, Spain
| | - Ana I. Álvarez-Mercado
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18071 Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. 18016 Armilla, Granada, Spain;
| |
Collapse
|
24
|
Nickles MA, Hasan A, Shakhbazova A, Wright S, Chambers CJ, Sivamani RK. Alternative Treatment Approaches to Small Intestinal Bacterial Overgrowth: A Systematic Review. J Altern Complement Med 2020; 27:108-119. [PMID: 33074705 DOI: 10.1089/acm.2020.0275] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: Broad-spectrum antibiotics are the first-line treatment for small intestinal bacterial overgrowth (SIBO). However, many antibiotics have a considerable side-effect profile and SIBO commonly reoccurs after successful eradication with antibiotics. Alternative therapies such as probiotics, therapeutic diets, and herbal medicines have been used to individualize SIBO management, particularly in recalcitrant cases. Objectives: The objective of this review is to evaluate the role of alternative therapies in SIBO treatment. Data Sources: EMBASE, MEDLINE, and the Cochrane Central Register were systematically searched for clinical studies evaluating alternative therapies in the management of SIBO. Study Eligibility Criteria: Human studies in which an alternative intervention was used to treat SIBO were included. Alternative interventions were defined as an intervention that included a probiotic supplement, herbal preparation, or a dietary change. Randomized controlled trials (RCTs), nonrandomized clinical trials with or without a control, and crossover studies were included. Study Appraisal: The following information was extracted from the selected studies: study type, study participants, SIBO subtype, intervention, comparison, outcome measures, relevant results, relevant side effects, and Jadad score. Results: Eight studies met inclusion criteria. The studies evaluated probiotics (n = 5), therapeutic diet (n = 1), and herbal medicines (n = 2). Among these studies, there were four RCTs, two open-label single-arm studies, one randomized, double-blind crossover study, and one two-arm open-label study with crossover. Main results are summarized. Limitations: There may be studies not captured by the defined search criteria. Additionally, studies used different methodologies in both breath testing and measurement of clinical symptoms, making it difficult to draw conclusions on SIBO eradication and symptom improvement across studies. Conclusions and Implications: Our findings suggest preliminary evidence for a role of alternative therapies in the treatment of SIBO. However, robust clinical trials are generally lacking. Existing studies tend to be small and lack standardized formulations of treatment. Breath testing protocols and clinical symptom measurement greatly varied between studies. Large-scale, randomized, placebo-controlled trials are needed to further evaluate the best way to utilize alternative therapies in the treatment of SIBO.
Collapse
Affiliation(s)
- Melissa A Nickles
- College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Aliza Hasan
- Department of Dermatology, University of California-Davis, Sacramento, CA, USA
| | | | | | - Cynthia J Chambers
- College of Medicine, California Northstate University, Elk Grove, CA, USA.,Pacific Skin Institute, Sacramento, CA, USA.,Zen Dermatology, Sacramento, CA, USA
| | - Raja K Sivamani
- Department of Dermatology, University of California-Davis, Sacramento, CA, USA.,College of Medicine, California Northstate University, Elk Grove, CA, USA.,Pacific Skin Institute, Sacramento, CA, USA.,Zen Dermatology, Sacramento, CA, USA.,Department of Biological Sciences, California State University, Sacramento, CA, USA
| |
Collapse
|
25
|
Abstract
Small intestinal bacterial overgrowth (SIBO) is a common, yet underrecognized, problem. Its prevalence is unknown because SIBO requires diagnostic testing. Although abdominal bloating, gas, distension, and diarrhea are common symptoms, they do not predict positive diagnosis. Predisposing factors include proton-pump inhibitors, opioids, gastric bypass, colectomy, and dysmotility. Small bowel aspirate/culture with growth of 10-10 cfu/mL is generally accepted as the "best diagnostic method," but it is invasive. Glucose or lactulose breath testing is noninvasive but an indirect method that requires further standardization and validation for SIBO. Treatment, usually with antibiotics, aims to provide symptom relief through eradication of bacteria in the small intestine. Limited numbers of controlled studies have shown systemic antibiotics (norfloxacin and metronidazole) to be efficacious. However, 15 studies have shown rifaximin, a nonsystemic antibiotic, to be effective against SIBO and well tolerated. Through improved awareness and scientific rigor, the SIBO landscape is poised for transformation.
Collapse
|
26
|
Pérez-Matute P, Íñiguez M, Villanueva-Millán MJ, Recio-Fernández E, Vázquez AM, Sánchez SC, Morano LE, Oteo JA. Short-term effects of direct-acting antiviral agents on inflammation and gut microbiota in hepatitis C-infected patients. Eur J Intern Med 2019; 67:47-58. [PMID: 31221551 DOI: 10.1016/j.ejim.2019.06.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 06/09/2019] [Accepted: 06/10/2019] [Indexed: 02/07/2023]
Abstract
Liver damage is associated with gut dysbiosis. New direct-acting antiviral agents (DAAs) are able to eradicate hepatitis C virus (HCV) from the body. However, the short and medium-term effects of DAAs at gut level before advanced liver damage occurs have not been evaluated yet. Thus, we investigated the impact of HCV and DAAs on gut microbiota composition (GM) and systemic inflammation. To achieve this objective, twenty-three non HCV-infected controls and 22 HCV-infected patients were recruited. Only non-cirrhotic patients (fibrosis stage 0-3) were included to avoid the direct impact of cirrhosis and portal hypertension on gut. The HCV-groups were evaluated before the treatment, after completing DAAs treatment and after 3 months. Fecal bacterial 16S rDNA was ultrasequenced and several biochemical/metabolic/inflammatory parameters were quantified. HCV infection was accompanied by a significant increase in TNFα plasma levels. DAAs were able to reduce this increase, especially in lower fibrosis grades. HCV infection was not accompanied by dramatic changes in α-diversity and was not recovered after HCV negativization, although a complete restoration was observed in lower fibrosis degrees. Six phyla, 15 genera and 9 bacterial species resulted differentially abundant among the groups. These differences were almost blunted with lower fibrosis. In summary, neither the usage of DAAs nor 3 months in sustained viral response were able to counteract the changes induced by HCV at gut level. The partial restoration observed in inflammation and α-diversity was only observed in low fibrosis degrees. Thus, it is urgent to begin treatment with DAAs as soon as possible.
Collapse
Affiliation(s)
- Patricia Pérez-Matute
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Logroño, (La Rioja), Spain.
| | - María Íñiguez
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Logroño, (La Rioja), Spain.
| | - María J Villanueva-Millán
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Logroño, (La Rioja), Spain.
| | - Emma Recio-Fernández
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Logroño, (La Rioja), Spain.
| | | | - Sheila Castro Sánchez
- Fundación Biomédica Galicia Sur, Instituto de Investigación Sanitaria Galicia Sur, Vigo, (Galicia), Spain.
| | - Luís E Morano
- Fundación Biomédica Galicia Sur, Instituto de Investigación Sanitaria Galicia Sur, Vigo, (Galicia), Spain; Infectious Diseases Department, Hospital Universitario Álvaro Cunqueiro, Vigo, (Galicia), Spain.
| | - José A Oteo
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Logroño, (La Rioja), Spain; Infectious Diseases Department, Hospital Universitario San Pedro, Logroño, (La Rioja), Spain.
| |
Collapse
|
27
|
Schwenger KJ, Clermont-Dejean N, Allard JP. The role of the gut microbiome in chronic liver disease: the clinical evidence revised. JHEP Rep 2019; 1:214-226. [PMID: 32039372 PMCID: PMC7001555 DOI: 10.1016/j.jhepr.2019.04.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 04/08/2019] [Accepted: 04/27/2019] [Indexed: 02/07/2023] Open
Abstract
Recent research has suggested a role for the intestinal microbiota in the pathogenesis and potential treatment of a wide range of liver diseases. The intestinal microbiota and bacterial products may contribute to the development of liver diseases through multiple mechanisms including increased intestinal permeability, chronic systemic inflammation, production of short-chain fatty acids and changes in metabolism. This suggests a potential role for pre-, pro- and synbiotic products in the prevention or treatment of some liver diseases. In addition, there is emerging evidence on the effects of faecal microbial transplant. Herein, we discuss the relationship between the intestinal microbiota and liver diseases, as well as reviewing intestinal microbiota-based treatment options that are currently being investigated.
Collapse
Affiliation(s)
- Katherine Jp Schwenger
- Toronto General Hospital, University Health Network, Toronto, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | | | - Johane P Allard
- Toronto General Hospital, University Health Network, Toronto, Canada.,Department of Medicine, University of Toronto, Toronto, Canada.,Department of Nutritional Sciences, University of Toronto, Toronto, Canada
| |
Collapse
|
28
|
Hong M, Han DH, Hong J, Kim DJ, Suk KT. Are Probiotics Effective in Targeting Alcoholic Liver Diseases? Probiotics Antimicrob Proteins 2019; 11:335-347. [PMID: 29687200 DOI: 10.1007/s12602-018-9419-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Alcoholic liver disease (ALD) encompasses a broad spectrum of disorders including steatosis, steatohepatitis, fibrosis, and cirrhosis. Despite intensive research in the last two decades, there is currently no Food and Drug Administration-approved therapy for treating ALD. Several studies have demonstrated the importance of the gut-liver axis and gut microbiome on the pathogenesis of ALD. Alcohol may induce intestinal dysbiosis and increased intestinal permeability, which in turn result in increased levels of pathogen-associated molecular patterns such as lipopolysaccharide (LPS) and translocation of microbial products from the gut to the liver (bacterial translocation). LPS is an inflammatory signal that activates toll-like receptor 4 on Kupffer cells, contributing to the inflammation observed in ALD. Recently, probiotics have been shown to be effective in reducing or preventing the progression of ALD. A potential mechanism is that the probiotics transforms the composition of intestinal microbiota, which leads to reductions in alcohol-induced dysbiosis, intestinal permeability, bacterial translocation, endotoxemia, and consequently, the development of ALD. While transformation of intestinal microbiota by probiotics appears to be a promising therapeutic strategy for the treatment of intestinal barrier dysfunction, there is a scarcity of research that studies probiotics in the context of ALD. In this review, we discuss the potential therapeutic applications of probiotics in the treatment of ALD.
Collapse
Affiliation(s)
- Meegun Hong
- Department of Internal Medicine, Hallym University Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Gyo-dong, Chuncheon, 24253, South Korea
| | - Dae Hee Han
- Department of Internal Medicine, Hallym University Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Gyo-dong, Chuncheon, 24253, South Korea
| | - Jitaek Hong
- Department of Internal Medicine, Hallym University Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Gyo-dong, Chuncheon, 24253, South Korea
| | - Dong Joon Kim
- Department of Internal Medicine, Hallym University Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Gyo-dong, Chuncheon, 24253, South Korea
| | - Ki Tae Suk
- Department of Internal Medicine, Hallym University Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Gyo-dong, Chuncheon, 24253, South Korea.
| |
Collapse
|
29
|
Wang W, Xu Y, Jiang C, Gao Y. Advances in the treatment of severe alcoholic hepatitis. Curr Med Res Opin 2019; 35:261-273. [PMID: 29781336 DOI: 10.1080/03007995.2018.1479247] [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] [Indexed: 10/16/2022]
Abstract
Severe alcoholic hepatitis (SAH) is a costly and worldwide public health issue with high morbidity and mortality. Specific effective treatments for SAH have yet to be established. The aim of the present article is to review the current knowledge of the pathogenesis, assessment and treatment options in patients with SAH. To date, alcohol abstinence and enteral nutrition are the recommended first-line treatments. Although corticosteroids remain the preferred therapy for certain patients with a modified Maddrey discriminant function level greater than 54, they only improve short-term survival rates. New research focuses on liver inflammation, liver regeneration, the gut-liver axis, human induced pluripotent stem cells and extracorporeal albumin dialysis. Liver transplantation is considered the last medical option for patients with SAH who are nonresponsive to other medical treatments.
Collapse
Affiliation(s)
- Wenjun Wang
- a Department of Hepatology , First Hospital of Jilin University, Jilin University , Jilin , China
| | - Ying Xu
- a Department of Hepatology , First Hospital of Jilin University, Jilin University , Jilin , China
| | - Chang Jiang
- a Department of Hepatology , First Hospital of Jilin University, Jilin University , Jilin , China
| | - Yanhang Gao
- a Department of Hepatology , First Hospital of Jilin University, Jilin University , Jilin , China
| |
Collapse
|
30
|
The Immunologic Role of Gut Microbiota in Patients with Chronic HBV Infection. J Immunol Res 2018; 2018:2361963. [PMID: 30148173 PMCID: PMC6083645 DOI: 10.1155/2018/2361963] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 05/15/2018] [Accepted: 05/29/2018] [Indexed: 02/07/2023] Open
Abstract
Hepatitis B can cause acute or chronic liver damage due to hepatitis B virus (HBV) infection. Cirrhosis or hepatocellular carcinoma (HCC) caused by chronic HBV infection often leads to increased mortality. However, the gut and liver have the same embryonic origin; therefore, a close relationship must exist in terms of anatomy and function, and the gut microbiota plays an important role in host metabolic and immune modulation. It is believed that structural changes in the gut microbiota, bacterial translocation, and the resulting immune injury may affect the occurrence and development of liver inflammation caused by chronic HBV infection based on the in-depth cognition of the concept of the “gut-liver axis” and the progress in intestinal microecology. This review aims to summarize and discuss the immunologic role of the gut microbiota in chronic HBV infection.
Collapse
|
31
|
Investigation into complementary and integrative medicine practitioners' clinical experience of intestinal permeability: A cross-sectional survey. Complement Ther Clin Pract 2018; 31:200-209. [DOI: 10.1016/j.ctcp.2018.02.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 01/22/2018] [Accepted: 02/21/2018] [Indexed: 12/12/2022]
|
32
|
Zhang J, Wang C, Wang J, Zhang F. Relationship between intestinal flora and inflammatory factors in patients with nonalcoholic steatohepatitis. Exp Ther Med 2018; 15:723-726. [PMID: 29399077 PMCID: PMC5772052 DOI: 10.3892/etm.2017.5490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 09/12/2017] [Indexed: 01/12/2023] Open
Abstract
This study was conducted to analyze the change in intestinal flora of patients with nonalcoholic steatohepatitis and its correlation to the levels of the inflammatory cytokines interleukin-10 (IL-10) and IL-17. We selected 90 patients that were diagnosed with and treated for nonalcoholic steatohepatitis as the patient group and 80 healthy cases as the control group. We then compared the intestinal flora in the subject feces and the intestinal colonization resistance (B/E, Bifidobacterium to Enterobacter) of both groups. Using RT-PCR, we also detected IL-10 and IL-17 mRNA levels in the peripheral blood mononuclear cells of both groups. Furthermore, we used the ELISA method to determine serum IL-10 and IL-17 levels in order to explore the correlation between IL-10, IL-17 and B/E. The number of Bifidobacterium and Lactobacillus were significantly lower in the patient group than the control group (P<0.05), while Enterobacter and Enterococcus pathogenic bacteria were significantly higher in the patient group than the control group (P<0.05). The B/E value was lower in the patient group than the control group (P<0.05). The relative expression of IL-10 and IL-17 mRNA in the patient group was significantly higher than in the control group (P<0.05). In the patient group, the serum IL-10 levels were 1.17±0.15 pg/ml, which is significantly higher than the control group serum IL-10 levels which were 0.32±0.04 pg/ml (P<0.05). The serum IL-17 levels in the patient groups were 0.96±0.11 pg/ml, which was significantly higher than the control group, which had an average of 0.28±0.01 pg/ml serum IL-17 levels (P<0.05). Pearson's correlation analysis showed that the change of B/E value of intestinal flora in the patients group were negatively correlated with serum IL-10 (r=−0.546, P<0.05), and negatively correlated with serum IL-17 (r=−0.535, P<0.05). Therefore, compared to healthy people, the expression of IL-10 and IL-17 in the peripheral blood of patients with non-alcoholic fatty liver is high. The changes in intestinal flora in patients with nonalcoholic steatohepatitis are closely related to the changes of serum IL-10 and IL-17 levels, and they are involved in the development of nonalcoholic steatohepatitis.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Liver Disease, Xuzhou Infectious Disease Hospital, Xuzhou, Jiangsu 221000, P.R. China
| | - Chunying Wang
- Department of Liver Disease, Xuzhou Infectious Disease Hospital, Xuzhou, Jiangsu 221000, P.R. China
| | - Ji Wang
- Department of Liver Disease, Xuzhou Infectious Disease Hospital, Xuzhou, Jiangsu 221000, P.R. China
| | - Fengchi Zhang
- Department of Liver Disease, Xuzhou Infectious Disease Hospital, Xuzhou, Jiangsu 221000, P.R. China
| |
Collapse
|
33
|
Mokkala K, Pussinen P, Houttu N, Koivuniemi E, Vahlberg T, Laitinen K. The impact of probiotics and n-3 long-chain polyunsaturated fatty acids on intestinal permeability in pregnancy: a randomised clinical trial. Benef Microbes 2018; 9:199-208. [PMID: 29345158 DOI: 10.3920/bm2017.0072] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A disruption in intestinal barrier integrity may predispose individuals to metabolic aberrations, particularly during the vulnerable period of pregnancy. We investigated whether intestinal permeability, as measured by serum zonulin concentration, changes over the duration of pregnancy and whether this change is reflected in lipopolysaccharide (LPS) activity. Second, we tested in a randomised double-blind placebo controlled clinical trial the impact of consuming dietary probiotics and/or long chain polyunsaturated fatty acid (LC-PUFA) supplements in lowering serum zonulin concentration and LPS activity. The probiotic supplement was a combination of two bacteria, Bifidobacterium animalis ssp. lactis 420 and Lactobacillus rhamnosus HN001. This study included 200 overweight pregnant women participating in an on-going study; participants were randomised to consume either (1) probiotics, (2) LC-PUFA, (3) probiotics and LC-PUFA, or (4) placebo for each supplement. Blood samples were obtained at early, the baseline, and late pregnancy (mean 14 and 35 weeks of gestation, respectively). Serum zonulin concentration increased from early (mean (standard deviation): 62.7 (12.9) ng/ml) to late pregnancy by 5.3 (95%CI 3.7-6.9) ng/ml, and LPS activity increased from (0.16 (0.04) EU/ml) by 0.04 (95%CI 0.03-0.05) EU/ml. No differences among the intervention groups were detected in the change from early to late pregnancy in serum zonulin concentration (P=0.8) or LPS activity (P=0.2). The change in serum zonulin concentration during the pregnancy was associated with the weeks of follow up (r=0.25, P<0.001). Serum LPS activity was correlated with higher maternal weight gain (r=0.19, P=0.008). As a conclusion, intestinal permeability increased with the progression of pregnancy in overweight and obese women and was reflected in LPS activity. No efficacy of supplementation with probiotics and/or LC-PUFA was demonstrated in pregnancy-induced changes in serum zonulin concentration or LPS activity.
Collapse
Affiliation(s)
- K Mokkala
- 1 Institute of Biomedicine, University of Turku, 20014 Turku, Finland
| | - P Pussinen
- 2 Oral and Maxillofacial Diseases, University of Helsinki and University Hospital of Helsinki, 00014 Helsinki, Finland
| | - N Houttu
- 1 Institute of Biomedicine, University of Turku, 20014 Turku, Finland
| | - E Koivuniemi
- 1 Institute of Biomedicine, University of Turku, 20014 Turku, Finland.,3 Turku University Hospital, Department of Obstetrics and Gynaecology, 20014 Turku, Finland
| | - T Vahlberg
- 4 Department of Clinical Medicine, Biostatistics, University of Turku and Turku University Hospital, 20014 Turku, Finland
| | - K Laitinen
- 1 Institute of Biomedicine, University of Turku, 20014 Turku, Finland
| |
Collapse
|
34
|
Abstract
The ageing trajectory is plastic and can be slowed down by lifestyle factors, including good nutrition, adequate physical activity and avoidance of smoking. In humans, plant-based diets such as the Mediterranean dietary pattern are associated with healthier ageing and lower risk of age-related disease, whereas obesity accelerates ageing and increases the likelihood of most common complex diseases including CVD, T2D, dementia, musculoskeletal diseases and several cancers. As yet, there is only weak evidence in humans about the molecular mechanisms through which dietary factors modulate ageing but evidence from cell systems and animal models suggest that it is probable that better dietary choices influence all 9 hallmarks of ageing. It seems likely that better eating patterns retard ageing in at least two ways including (i) by reducing pervasive damaging processes such as inflammation, oxidative stress/redox changes and metabolic stress and (ii) by enhancing cellular capacities for damage management and repair. From a societal perspective, there is an urgent imperative to discover, and to implement, cost-effective lifestyle (especially dietary) interventions which enable each of us to age well, i.e. to remain physically and socially active and independent and to minimise the period towards the end of life when individuals suffer from frailty and multi-morbidity.
Collapse
Affiliation(s)
- Fiona C Malcomson
- Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - John C Mathers
- Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
35
|
Effect of probiotics and synbiotics consumption on serum concentrations of liver function test enzymes: a systematic review and meta-analysis. Eur J Nutr 2017; 57:2037-2053. [PMID: 29119235 DOI: 10.1007/s00394-017-1568-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 10/14/2017] [Indexed: 02/07/2023]
Abstract
PURPOSE The gut-liver interaction suggests that modification of gut bacterial flora using probiotics and synbiotics may improve liver function. This systematic review and meta-analysis aimed to clarify the effect of probiotics and synbiotics consumption on the serum concentration of liver function enzymes. METHODS PubMed (MEDLINE), Cumulative Index to Nursing and Allied Health Literature, and Cochrane Library (Central) were searched from 1980 to August 2017 for studies where adults consumed probiotics and/or synbiotics in controlled trials and changes in liver function enzymes were examined. RESULTS A total of 17 studies (19 trials) were included in the meta-analysis. Random effects meta-analyses were applied. Probiotics and synbiotics significantly reduced serum alanine aminotransferase [- 8.05 IU/L, 95% confidence interval (CI) - 13.07 to - 3.04; p = 0.002]; aspartate aminotransferase (- 7.79 IU/L, 95% CI: - 13.93 to - 1.65; p = 0.02) and gamma-glutamyl transpeptidase (- 8.40 IU/L, 95% CI - 12.61 to - 4.20; p < 0.001). Changes in the serum concentration of alkaline phosphatase and albumin did not reach a statistically significant level. Changes to bilirubin levels were in favour of the control group (0.95 μmol/L, 95% CI 0.48-1.42; p < 0.001). Subgroup analysis suggested the existence of liver disease at baseline, synbiotics supplementation and duration of supplementation ≥ 8 weeks resulted in more pronounced improvement in liver function enzymes than their counterparts. CONCLUSIONS Probiotics and synbiotics may be suggested as supplements to improve serum concentration of liver enzymes, especially when synbiotics administered for a period ≥ 8 weeks and in individuals with liver disease.
Collapse
|
36
|
Song SC, An YM, Shin JH, Chung MJ, Seo JG, Kim E. Beneficial effects of a probiotic blend on gastrointestinal side effects induced by leflunomide and amlodipine in a rat model. Benef Microbes 2017; 8:801-808. [DOI: 10.3920/bm2016.0231] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Patients with metabolic disorders frequently suffer from side effects induced by long-term oral medications. The present study using a rat model system indicated that leflunomide (LF) and amlodipine (AMD), the active ingredients contained in the medications for rheumatoid arthritis and hypertension, respectively, appeared to induce various bowel problems including constipation and inflammation. In the small and large intestine, LF increased the expression of tumour necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6 compared to the null control and AMD increased the expression of both TNF-α and IL-1β, although its effect on IL-6 was only increased in the large intestine. It is noteworthy that the probiotic blend tested was found to alleviate intestinal complications caused by LF and AMD. Analysis of the gut microbiota revealed that AMD induced compositional changes in the gut microbiota. Namely, members of the phylum Bacteroidetes, which constituted only about 0.3% of the microbiota in the null control, made up more than 10% of the total composition in the AMD-administered rats. Interestingly, the probiotic blend was also found to normaliSe the gut microbiota.
Collapse
Affiliation(s)
- S.-C. Song
- Department of Systems Biology, Yonsei University, 323, SB140, Yonsei-ro 50, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Y.-M. An
- Department of Systems Biology, Yonsei University, 323, SB140, Yonsei-ro 50, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - J.-H. Shin
- R&D Center, Cell Biotech Co., Ltd., 50, Aegibong-ro 409 beon-gil, Wolgot-myeon, Gimpo-si, Gyeonggi-do 10003, Republic of Korea
| | - M.-J. Chung
- R&D Center, Cell Biotech Co., Ltd., 50, Aegibong-ro 409 beon-gil, Wolgot-myeon, Gimpo-si, Gyeonggi-do 10003, Republic of Korea
| | - J.-G. Seo
- R&D Center, Cell Biotech Co., Ltd., 50, Aegibong-ro 409 beon-gil, Wolgot-myeon, Gimpo-si, Gyeonggi-do 10003, Republic of Korea
| | - E. Kim
- Department of Systems Biology, Yonsei University, 323, SB140, Yonsei-ro 50, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
37
|
Shin JH, Nam MH, Lee H, Lee JS, Kim H, Chung MJ, Seo JG. Amelioration of obesity-related characteristics by a probiotic formulation in a high-fat diet-induced obese rat model. Eur J Nutr 2017; 57:2081-2090. [DOI: 10.1007/s00394-017-1481-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 06/04/2017] [Indexed: 12/20/2022]
|
38
|
Singh RK, Chang HW, Yan D, Lee KM, Ucmak D, Wong K, Abrouk M, Farahnik B, Nakamura M, Zhu TH, Bhutani T, Liao W. Influence of diet on the gut microbiome and implications for human health. J Transl Med 2017; 15:73. [PMID: 28388917 PMCID: PMC5385025 DOI: 10.1186/s12967-017-1175-y] [Citation(s) in RCA: 1589] [Impact Index Per Article: 198.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 03/21/2017] [Indexed: 02/06/2023] Open
Abstract
Recent studies have suggested that the intestinal microbiome plays an important role in modulating risk of several chronic diseases, including inflammatory bowel disease, obesity, type 2 diabetes, cardiovascular disease, and cancer. At the same time, it is now understood that diet plays a significant role in shaping the microbiome, with experiments showing that dietary alterations can induce large, temporary microbial shifts within 24 h. Given this association, there may be significant therapeutic utility in altering microbial composition through diet. This review systematically evaluates current data regarding the effects of several common dietary components on intestinal microbiota. We show that consumption of particular types of food produces predictable shifts in existing host bacterial genera. Furthermore, the identity of these bacteria affects host immune and metabolic parameters, with broad implications for human health. Familiarity with these associations will be of tremendous use to the practitioner as well as the patient.
Collapse
Affiliation(s)
- Rasnik K. Singh
- University of California, Los Angeles, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095 USA
| | - Hsin-Wen Chang
- Department of Dermatology, University of California, San Francisco, 2340 Sutter St. Room N431, Box 0808, San Francisco, CA 94115 USA
| | - Di Yan
- Department of Dermatology, University of California, San Francisco, 2340 Sutter St. Room N431, Box 0808, San Francisco, CA 94115 USA
| | - Kristina M. Lee
- Department of Dermatology, University of California, San Francisco, 2340 Sutter St. Room N431, Box 0808, San Francisco, CA 94115 USA
| | - Derya Ucmak
- Department of Dermatology, University of California, San Francisco, 2340 Sutter St. Room N431, Box 0808, San Francisco, CA 94115 USA
| | - Kirsten Wong
- Department of Dermatology, University of California, San Francisco, 2340 Sutter St. Room N431, Box 0808, San Francisco, CA 94115 USA
| | - Michael Abrouk
- University of California, Irvine, School of Medicine, Irvine, CA 92697 USA
| | | | - Mio Nakamura
- Department of Dermatology, University of California, San Francisco, 2340 Sutter St. Room N431, Box 0808, San Francisco, CA 94115 USA
| | - Tian Hao Zhu
- University of Southern California Keck School of Medicine, Los Angeles, CA 90033 USA
| | - Tina Bhutani
- Department of Dermatology, University of California, San Francisco, 2340 Sutter St. Room N431, Box 0808, San Francisco, CA 94115 USA
| | - Wilson Liao
- Department of Dermatology, University of California, San Francisco, 2340 Sutter St. Room N431, Box 0808, San Francisco, CA 94115 USA
| |
Collapse
|
39
|
Probiotics for Preventing and Treating Small Intestinal Bacterial Overgrowth: A Meta-Analysis and Systematic Review of Current Evidence. J Clin Gastroenterol 2017; 51:300-311. [PMID: 28267052 DOI: 10.1097/mcg.0000000000000814] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The present study conducted a meta-analysis and systematic review of current evidence to assess the efficacy of probiotics in preventing or treating small intestinal bacterial overgrowth (SIBO). Relevant studies from PubMed, Embase, and the Cochrane Central Register of Controlled Trials, until May 2016, were assimilated. The prevention efficacy was assessed by the incidence of SIBO in the probiotic group, and the treatment efficacy by the SIBO decontamination rate, reduction in H2 concentration, and symptom improvement. The relative risk (RR) and weighted mean difference (WMD) were used as effect measures and the random-effects model used for meta-analysis. A total of 14 full-text articles and 8 abstracts were included for the systematic review, and 18 studies were eligible for data synthesis. Patients on probiotic usage showed an insignificant trend toward low SIBO incidence [RR=0.54; 95% confidence intervals (CI), 0.19-1.52; P=0.24]. The pooled SIBO decontamination rate was 62.8% (51.5% to 72.8%). The probiotics group showed a significantly higher SIBO decontamination rate than the nonprobiotic group (RR=1.61; 95% CI, 1.19-2.17; P<0.05). Also, the H2 concentration was significantly reduced among probiotic users (WMD=-36.35 ppm; 95% CI, -44.23 to -28.47 ppm; P<0.05). Although probiotics produced a marked decrease in the abdominal pain scores (WMD=-1.17; 95% CI, -2.30 to -0.04; P<0.05), it did not significantly reduce the daily stool frequency (WMD=-0.09; 95% CI, -0.47 to 0.29). Therefore, the present findings indicated that probiotics supplementation could effectively decontaminate SIBO, decrease H2 concentration, and relieve abdominal pain, but were ineffective in preventing SIBO.
Collapse
|
40
|
Abstract
BACKGROUND Hepatic encephalopathy is a disorder of brain function as a result of liver failure or portosystemic shunt or both. Both hepatic encephalopathy (clinically overt) and minimal hepatic encephalopathy (not clinically overt) significantly impair patient's quality of life and daily functioning, and represent a significant burden on healthcare resources. Probiotics are live micro-organisms, which when administered in adequate amounts, may confer a health benefit on the host. OBJECTIVES To determine the beneficial and harmful effects of probiotics in any dosage, compared with placebo or no intervention, or with any other treatment for people with any grade of acute or chronic hepatic encephalopathy. This review did not consider the primary prophylaxis of hepatic encephalopathy. SEARCH METHODS We searched The Cochrane Hepato-Biliary Group Controlled Trials Register, CENTRAL, MEDLINE, Embase, Science Citation Index Expanded, conference proceedings, reference lists of included trials, and the World Health Organization International Clinical Trials Registry Platform until June 2016. SELECTION CRITERIA We included randomised clinical trials that compared probiotics in any dosage with placebo or no intervention, or with any other treatment in people with hepatic encephalopathy. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by The Cochrane Collaboration. We conducted random-effects model meta-analysis due to obvious heterogeneity of participants and interventions. We defined a P value of 0.05 or less as significant. We expressed dichotomous outcomes as risk ratio (RR) and continuous outcomes as mean difference (MD) with 95% confidence intervals (CI). MAIN RESULTS We included 21 trials with 1420 participants, of these, 14 were new trials. Fourteen trials compared a probiotic with placebo or no treatment, and seven trials compared a probiotic with lactulose. The trials used a variety of probiotics; the most commonly used group of probiotic was VSL#3, a proprietary name for a group of eight probiotics. Duration of administration ranged from 10 days to 180 days. Eight trials declared their funding source, of which six were independently funded and two were industry funded. The remaining 13 trials did not disclose their funding source. We classified 19 of the 21 trials at high risk of bias.We found no effect on all-cause mortality when probiotics were compared with placebo or no treatment (7 trials; 404 participants; RR 0.58, 95% CI 0.23 to 1.44; low-quality evidence). No-recovery (as measured by incomplete resolution of symptoms) was lower for participants treated with probiotic (10 trials; 574 participants; RR 0.67, 95% CI 0.56 to 0.79; moderate-quality evidence). Adverse events were lower for participants treated with probiotic than with no intervention when considering the development of overt hepatic encephalopathy (10 trials; 585 participants; RR 0.29, 95% CI 0.16 to 0.51; low-quality evidence), but effects on hospitalisation and change of/or withdrawal from treatment were uncertain (hospitalisation: 3 trials, 163 participants; RR 0.67, 95% CI 0.11 to 4.00; very low-quality evidence; change of/or withdrawal from treatment: 9 trials, 551 participants; RR 0.70, 95% CI 0.46 to 1.07; very low-quality evidence). Probiotics may slightly improve quality of life compared with no intervention (3 trials; 115 participants; results not meta-analysed; low-quality evidence). Plasma ammonia concentration was lower for participants treated with probiotic (10 trials; 705 participants; MD -8.29 μmol/L, 95% CI -13.17 to -3.41; low-quality evidence). There were no reports of septicaemia attributable to probiotic in any trial.When probiotics were compared with lactulose, the effects on all-cause mortality were uncertain (2 trials; 200 participants; RR 5.00, 95% CI 0.25 to 102.00; very low-quality evidence); lack of recovery (7 trials; 430 participants; RR 1.01, 95% CI 0.85 to 1.21; very low-quality evidence); adverse events considering the development of overt hepatic encephalopathy (6 trials; 420 participants; RR 1.17, 95% CI 0.63 to 2.17; very low-quality evidence); hospitalisation (1 trial; 80 participants; RR 0.33, 95% CI 0.04 to 3.07; very low-quality evidence); intolerance leading to discontinuation (3 trials; 220 participants; RR 0.35, 95% CI 0.08 to 1.43; very low-quality evidence); change of/or withdrawal from treatment (7 trials; 490 participants; RR 1.27, 95% CI 0.88 to 1.82; very low-quality evidence); quality of life (results not meta-analysed; 1 trial; 69 participants); and plasma ammonia concentration overall (6 trials; 325 participants; MD -2.93 μmol/L, 95% CI -9.36 to 3.50; very low-quality evidence). There were no reports of septicaemia attributable to probiotic in any trial. AUTHORS' CONCLUSIONS The majority of included trials suffered from a high risk of systematic error ('bias') and a high risk of random error ('play of chance'). Accordingly, we consider the evidence to be of low quality. Compared with placebo or no intervention, probiotics probably improve recovery and may lead to improvements in the development of overt hepatic encephalopathy, quality of life, and plasma ammonia concentrations, but probiotics may lead to little or no difference in mortality. Whether probiotics are better than lactulose for hepatic encephalopathy is uncertain because the quality of the available evidence is very low. High-quality randomised clinical trials with standardised outcome collection and data reporting are needed to further clarify the true efficacy of probiotics.
Collapse
Affiliation(s)
- Rohan Dalal
- Sydney Medical School, Westmead Hospital, Sydney, Australia
| | - Richard G McGee
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW, Australia, 2145
| | - Stephen M Riordan
- Gastrointestinal and Liver Unit, The Prince of Wales, Barker St, Randwick, Australia, NSW 2031
| | - Angela C Webster
- Sydney School of Public Health, The University of Sydney, Edward Ford Building A27, Sydney, NSW, Australia, 2006
| |
Collapse
|
41
|
Bluemel S, Williams B, Knight R, Schnabl B. Precision medicine in alcoholic and nonalcoholic fatty liver disease via modulating the gut microbiota. Am J Physiol Gastrointest Liver Physiol 2016; 311:G1018-G1036. [PMID: 27686615 PMCID: PMC5206291 DOI: 10.1152/ajpgi.00245.2016] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/25/2016] [Indexed: 02/08/2023]
Abstract
Alcoholic liver disease (ALD) and nonalcoholic fatty liver disease (NAFLD) represent a major health burden in industrialized countries. Although alcohol abuse and nutrition play a central role in disease pathogenesis, preclinical models support a contribution of the gut microbiota to ALD and NAFLD. This review describes changes in the intestinal microbiota compositions related to ALD and NAFLD. Findings from in vitro, animal, and human studies are used to explain how intestinal pathology contributes to disease progression. This review summarizes the effects of untargeted microbiome modifications using antibiotics and probiotics on liver disease in animals and humans. While both affect humoral inflammation, regression of advanced liver disease or mortality has not been demonstrated. This review further describes products secreted by Lactobacillus- and microbiota-derived metabolites, such as fatty acids and antioxidants, that could be used for precision medicine in the treatment of liver disease. A better understanding of host-microbial interactions is allowing discovery of novel therapeutic targets in the gut microbiota, enabling new treatment options that restore the intestinal ecosystem precisely and influence liver disease. The modulation options of the gut microbiota and precision medicine employing the gut microbiota presented in this review have excellent prospects to improve treatment of liver disease.
Collapse
Affiliation(s)
- Sena Bluemel
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Brandon Williams
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Rob Knight
- Departments of Pediatrics and Computer Science and Engineering, University of California San Diego, La Jolla, California; and
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, California;
- Veterans Affairs San Diego Healthcare System, San Diego, California
| |
Collapse
|
42
|
Byun S, Lim T, Lim Y, Seo J, Chung M. In vivo effects of s-pantoprazole, polaprenzinc, and probiotic blend on chronic small intestinal injury induced by indomethacin. Benef Microbes 2016; 7:731-737. [DOI: 10.3920/bm2016.0029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Treatment and prevention methods for non-steroidal anti-inflammatory drug-induced enteropathy have not yet been established. We tested the preventive effects of s-pantoprazole sodium trihydrate (PAN), polaprezinc (PZ), and probiotics on an indomethacin (Indo)-induced small intestinal injury in a rat model. Rats were randomised into 6 groups to receive: normal saline (control), Indo (6 mg/kg), PZ plus Indo, PAN plus Indo, or probiotics plus Indo (at 108 and 109 cfu/head) for 2 weeks. We measured body weight, food intake, severity of small intestinal damage, haemoglobin (Hb) levels in the small intestinal fluid, intestinal inflammatory cytokines, and a few groups of faecal bacteria. The experimental groups were found to have the following survival rates: 0% for the Indo, PZ, and PAN groups; 50% for both probiotic groups; and 100% for control. Treatment with probiotics of different concentrations reduced small intestinal lesion scores and intestinal fluid Hb as compared with the Indo group, while these parameters did not reduce in the PZ and PAN groups. The anti-inflammatory marker interleukin 10 increased in both probiotic groups. Analysis of a few groups of faecal bacteria revealed that Indo-induced a significant increase in Gram-negative bacteria and decreases in Bifidobacterium and Lactobacillus. Similar changes were also observed in the PZ and PAN groups. However, opposite effects were found in both probiotic groups. The use of probiotics appeared to be beneficial in preventing Indo-induced chronic small intestinal injury.
Collapse
Affiliation(s)
- S.J. Byun
- Department of Internal Medicine, Ilsan Hospital, College of Medicine, Dongguk University, Goyang 410-773, Republic of Korea
| | - T.J. Lim
- Research and Development Center, Cell Biotech Co. Ltd., Gimpo, Gyeonggi 157-030, Republic of Korea
| | - Y.J. Lim
- Department of Internal Medicine, Ilsan Hospital, College of Medicine, Dongguk University, Goyang 410-773, Republic of Korea
| | - J.G. Seo
- Research and Development Center, Cell Biotech Co. Ltd., Gimpo, Gyeonggi 157-030, Republic of Korea
| | - M.J. Chung
- Research and Development Center, Cell Biotech Co. Ltd., Gimpo, Gyeonggi 157-030, Republic of Korea
| |
Collapse
|
43
|
Ponziani FR, Gerardi V, Gasbarrini A. Diagnosis and treatment of small intestinal bacterial overgrowth. Expert Rev Gastroenterol Hepatol 2016; 10:215-27. [PMID: 26636484 DOI: 10.1586/17474124.2016.1110017] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A huge number of bacteria are hosted in the gastrointestinal tract, following a gradient increasing towards the colon. Gastric acid secretion and intestinal clearance provide the qualitative and quantitative partitioning of intestinal bacteria; small intestinal bacteria overgrowth (SIBO) occurs when these barrier mechanisms fail. Diagnosis of SIBO is challenging due to the low specificity of symptoms, the frequent association with other diseases of the gastrointestinal tract and the absence of optimal objective diagnostic tests. The therapeutic approach to SIBO is oriented towards resolving predisposing conditions, and is supported by antibiotic treatment to restore the normal small intestinal microflora and by modifications of dietary habits for symptomatic relief. In the near future, metagenomics and metabolomics will help to overcome the uncertainties of SIBO diagnosis and the pitfalls of therapeutic management, allowing the design of a personalized strategy based on the direct insight into the small intestinal microbial community.
Collapse
Affiliation(s)
| | - Viviana Gerardi
- a Internal Medicine and Gastroenterology , Agostino Gemelli Hospital , Rome , Italy
| | - Antonio Gasbarrini
- a Internal Medicine and Gastroenterology , Agostino Gemelli Hospital , Rome , Italy
| |
Collapse
|
44
|
Gloor GB, Reid G. Compositional analysis: a valid approach to analyze microbiome high-throughput sequencing data. Can J Microbiol 2016; 62:692-703. [PMID: 27314511 DOI: 10.1139/cjm-2015-0821] [Citation(s) in RCA: 213] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A workshop held at the 2015 annual meeting of the Canadian Society of Microbiologists highlighted compositional data analysis methods and the importance of exploratory data analysis for the analysis of microbiome data sets generated by high-throughput DNA sequencing. A summary of the content of that workshop, a review of new methods of analysis, and information on the importance of careful analyses are presented herein. The workshop focussed on explaining the rationale behind the use of compositional data analysis, and a demonstration of these methods for the examination of 2 microbiome data sets. A clear understanding of bioinformatics methodologies and the type of data being analyzed is essential, given the growing number of studies uncovering the critical role of the microbiome in health and disease and the need to understand alterations to its composition and function following intervention with fecal transplant, probiotics, diet, and pharmaceutical agents.
Collapse
Affiliation(s)
- Gregory B Gloor
- a Department of Biochemistry, Western University, London, Ontario, Canada.,b Canadian Center for Human Microbiome and Probiotic Research, Lawson Health Research Institute, London, Ontario, Canada
| | - Gregor Reid
- b Canadian Center for Human Microbiome and Probiotic Research, Lawson Health Research Institute, London, Ontario, Canada.,c Department of Microbiology and Immunology, and Department of Surgery, Western University, London, Ontario, Canada
| |
Collapse
|
45
|
Shin JH, Lee JS, Seo JG. Assessment of cell adhesion, cell surface hydrophobicity, autoaggregation, and lipopolysaccharide-binding properties of live and heat-killed Lactobacillus acidophilus CBT LA1. ACTA ACUST UNITED AC 2015. [DOI: 10.7845/kjm.2015.5029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|