51
|
Liu J, Yang D, Wang X, Asare PT, Zhang Q, Na L, Shao L. Gut Microbiota Targeted Approach in the Management of Chronic Liver Diseases. Front Cell Infect Microbiol 2022; 12:774335. [PMID: 35444959 PMCID: PMC9014089 DOI: 10.3389/fcimb.2022.774335] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
The liver is directly connected to the intestines through the portal vein, which enables the gut microbiota and gut-derived products to influence liver health. There is accumulating evidence of decreased gut flora diversity and alcohol sensitivity in patients with various chronic liver diseases, including non-alcoholic/alcoholic liver disease, chronic hepatitis virus infection, primary sclerosing cholangitis and liver cirrhosis. Increased intestinal mucosal permeability and decline in barrier function were also found in these patients. Followed by bacteria translocation and endotoxin uptake, these will lead to systemic inflammation. Specific microbiota and microbiota-derived metabolites are altered in various chronic liver diseases studies, but the complex interaction between the gut microbiota and liver is missing. This review article discussed the bidirectional relationship between the gut and the liver, and explained the mechanisms of how the gut microbiota ecosystem alteration affects the pathogenesis of chronic liver diseases. We presented gut-microbiota targeted interventions that could be the new promising method to manage chronic liver diseases.
Collapse
Affiliation(s)
- Jing Liu
- Department of Research, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital; The College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Dakai Yang
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xiaojing Wang
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Paul Tetteh Asare
- Human and Animal Health Unit, Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Qingwen Zhang
- Department of Research, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital; The College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Lixin Na
- Department of Research, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital; The College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Lei Shao
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
- *Correspondence: Lei Shao,
| |
Collapse
|
52
|
Luo Y, Fang Q, Lai Y, Lei H, Zhang D, Niu H, Wang R, Song C. Polysaccharides from the leaves of Polygonatum sibiricum Red. regulate the gut microbiota and affect the production of short-chain fatty acids in mice. AMB Express 2022; 12:35. [PMID: 35312878 PMCID: PMC8938542 DOI: 10.1186/s13568-022-01376-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/13/2022] [Indexed: 12/20/2022] Open
Abstract
Polysaccharides from the rhizome of Polygonatum sibiricum display a variety of biological activities, including the regulation of intestinal microbiota, but the polysaccharides from the leaves of P. sibiricum have not been studied extensively. Here, we extracted crude polysaccharides from the leaves of P. sibiricum and further separated and purified them to study the effects of P. sibiricum polysaccharides (PsPs) on intestinal microbes and short-chain fatty acids (SCFAs). The PsPs had a total sugar content of 97.48% and a monosaccharide composition comprising mannose, rhamnose, galacturonic acid, glucose, xylose, and arabinose, with molar ratios of 6.6:15.4:4.5:8.8:40.7:24, respectively. The effects of PsPs on intestinal microflora in mice were also studied, with 16S sequencing results showing an increase in the relative abundance of Firmicutes and a decrease in Bacteroidetes at the phylum level. The abundance of Lactobacillus increased, while those of Lachnospiraceae and Bacteroides reduced (at the genus level) by PsPs treatment. The composition of microbes changed. Levels of SCFAs in the PsPs group were significantly increased compared with control mice, including acetic acid, propionic acid, and butyric acid. These results suggest that PsPs can act as prebiotics, regulating the intestinal tract probiotics.
Collapse
|
53
|
Liu Z, Zhang J, Zhao Q, Wen A, Li L, Zhang Y. The regulating effect of Tibet Opuntia ficus-indica (Linn.) Mill. polysaccharides on the intestinal flora of cyclophosphamide-induced immunocompromised mice. Int J Biol Macromol 2022; 207:570-579. [PMID: 35292280 DOI: 10.1016/j.ijbiomac.2022.03.039] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/23/2022] [Accepted: 03/07/2022] [Indexed: 12/18/2022]
Abstract
The stem of Opuntia species, a traditional medicinal plant, is widely used as food and functional raw material because of its rich polysaccharide content. There have been many studies on the immune function of polysaccharides from Opuntia stem, but only few have examined this function with respect to intestinal microbes. In this study, the effects of different concentrations of Opuntia stem polysaccharides on the immunity and intestinal microflora of cyclophosphamide (CTX)-induced immunocompromised mice were explored. The results showed that Tibet Opuntia ficus-indica (Linn.) Mill. polysaccharides (ODPs) could effectively increase the white blood cells (WBC) count index of mice and improve their thymus and spleen indices, while effectively promoting the secretion of IL-4, IL-1β, TNF-α and IFN-γ, with these effects being dependent on the concentration of crude polysaccharides. The intake of ODPs significantly regulated the relative abundance of Lactobacillus, Bacteroides and Akkermansia, and the new dominant intestinal bacterial species were Deferribacteres, Actinomycetes, Firmicutes, Tenericutes, Actinomycetes and Pasteurella. In addition, the ODPs could effectively enhance the metabolic level of lysine synthesis and decomposition, regulate the gene expression level after immune disorders, and enhance the overall health of the immunodeficient mice.
Collapse
Affiliation(s)
- Zhendong Liu
- Food Science College, Tibet Agriculture & Animal Husbandry University, Nyingchi 860000, China; The Provincial and Ministerial Co-founded Collaborative Innovation Center for R & D in Tibet Characteristic Agricultural and Animal Husbandry Resources, Nyingchi 860000, China
| | - Jinchao Zhang
- Food Science College, Tibet Agriculture & Animal Husbandry University, Nyingchi 860000, China; The Provincial and Ministerial Co-founded Collaborative Innovation Center for R & D in Tibet Characteristic Agricultural and Animal Husbandry Resources, Nyingchi 860000, China
| | - Qian Zhao
- Food Science College, Tibet Agriculture & Animal Husbandry University, Nyingchi 860000, China
| | - Aomei Wen
- Food Science College, Tibet Agriculture & Animal Husbandry University, Nyingchi 860000, China
| | - Liang Li
- Food Science College, Tibet Agriculture & Animal Husbandry University, Nyingchi 860000, China.
| | - Yu Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
54
|
Chen L, Zhu Y, Hou X, Yang L, Chu H. The Role of Gut Bacteria and Fungi in Alcohol-Associated Liver Disease. Front Med (Lausanne) 2022; 9:840752. [PMID: 35308525 PMCID: PMC8927088 DOI: 10.3389/fmed.2022.840752] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
Cirrhosis and liver cancer caused by alcohol-associated liver disease (ALD) are serious threats to people's health. In addition to hepatic cell apoptosis and liver inflammation caused by oxidative stress during alcohol metabolism, intestinal microbiota disorders are also involved in the onset and development of ALD. Ethanol and its' oxidative and non-oxidative metabolites, together with dysbiosis-caused-inflammation, destroys the intestinal barrier. Changes of several microbial metabolites, such as bile acids, short-chain fatty acids, and amino acid, are closely associated with gut dysbiosis in ALD. The alcohol-caused dysbiosis can further influence intestinal barrier-related proteins, such as mucin2, bile acid-related receptors, and aryl hydrocarbon receptor (AhR), and these abnormal changes also participate in the injury of the intestinal barrier and hepatic steatosis. Gut-derived bacteria, fungi, and their toxins, such as lipopolysaccharide (LPS) and β-glucan translocate into the liver through the damaged intestinal barrier and promote the progression of inflammation and fibrosis of ALD. Thus, the prevention of alcohol-induced disruption of intestinal permeability has a beneficial effect on ALD. Currently, multiple therapeutic treatments have been applied to restore the gut microbiota of patients with ALD. Fecal microbial transplantation, probiotics, antibiotics, and many other elements has already shown their ability of restoring the gut microbiota. Targeted approaches, such as using bacteriophages to remove cytolytic Enterococcus faecalis, and supplement with Lactobacillus, Bifidobacterium, or boulardii are also powerful therapeutic options for ALD.
Collapse
Affiliation(s)
- Liuying Chen
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yixin Zhu
- Department of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
55
|
Warner JB, Larsen IS, Hardesty JE, Song YL, Warner DR, McClain CJ, Sun R, Deng Z, Jensen BAH, Kirpich IA. Human Beta Defensin 2 Ameliorated Alcohol-Associated Liver Disease in Mice. Front Physiol 2022; 12:812882. [PMID: 35153819 PMCID: PMC8829467 DOI: 10.3389/fphys.2021.812882] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Alcohol-associated liver disease (ALD) is a prevalent liver disorder and significant global healthcare burden with limited effective therapeutic options. The gut-liver axis is a critical factor contributing to susceptibility to liver injury due to alcohol consumption. In the current study, we tested whether human beta defensin-2 (hBD-2), a small anti-microbial peptide, attenuates experimental chronic ALD. Male C57Bl/6J mice were fed an ethanol (EtOH)-containing diet for 6 weeks with daily administration of hBD-2 (1.2 mg/kg) by oral gavage during the final week. Two independent cohorts of mice with distinct baseline gut microbiota were used. Oral hBD-2 administration attenuated liver injury in both cohorts as determined by decreased plasma ALT activity. Notably, the degree of hBD-2-mediated reduction of EtOH-associated liver steatosis, hepatocellular death, and inflammation was different between cohorts, suggesting microbiota-specific mechanisms underlying the beneficial effects of hBD-2. Indeed, we observed differential mechanisms of hBD-2 between cohorts, which included an induction of hepatic and small intestinal IL-17A and IL-22, as well as an increase in T regulatory cell abundance in the gut and mesenteric lymph nodes. Lastly, hBD-2 modulated the gut microbiota composition in EtOH-fed mice in both cohorts, with significant decreases in multiple genera including Barnesiella, Parabacteroides, Akkermansia, and Alistipes, as well as altered abundance of several bacteria within the family Ruminococcaceae. Collectively, our results demonstrated a protective effect of hBD-2 in experimental ALD associated with immunomodulation and microbiota alteration. These data suggest that while the beneficial effects of hBD-2 on liver injury are uniform, the specific mechanisms of action are associated with baseline microbiota.
Collapse
Affiliation(s)
- Jeffrey B. Warner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Ida S. Larsen
- Québec Heart and Lung Institute (IUCPQ), Faculty of Medicine, Laval University, Québec city, QC, Canada
| | - Josiah E. Hardesty
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Ying L. Song
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Dennis R. Warner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Craig J. McClain
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States
- University of Louisville Alcohol Center, University of Louisville School of Medicine, Louisville, KY, United States
- University of Louisville Hepatobiology and Toxicology Center, University of Louisville School of Medicine, Louisville, KY, United States
- Robley Rex Veterans Medical Center, Louisville, KY, United States
| | - Rui Sun
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Zhongbin Deng
- University of Louisville Alcohol Center, University of Louisville School of Medicine, Louisville, KY, United States
- University of Louisville Hepatobiology and Toxicology Center, University of Louisville School of Medicine, Louisville, KY, United States
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
- Department of Surgery, University of Louisville, Louisville, KY, United States
| | - Benjamin A. H. Jensen
- Québec Heart and Lung Institute (IUCPQ), Faculty of Medicine, Laval University, Québec city, QC, Canada
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Irina A. Kirpich
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States
- University of Louisville Alcohol Center, University of Louisville School of Medicine, Louisville, KY, United States
- University of Louisville Hepatobiology and Toxicology Center, University of Louisville School of Medicine, Louisville, KY, United States
| |
Collapse
|
56
|
Crosstalk between Oxidative Stress and Inflammatory Liver Injury in the Pathogenesis of Alcoholic Liver Disease. Int J Mol Sci 2022; 23:ijms23020774. [PMID: 35054960 PMCID: PMC8775426 DOI: 10.3390/ijms23020774] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 02/06/2023] Open
Abstract
Alcoholic liver disease (ALD) is characterized by the injury, inflammation, and scarring in the liver owing to excessive alcohol consumption. Currently, ALD is a leading cause for liver transplantation. Therefore, extensive studies (in vitro, in experimental ALD models and in humans) are needed to elucidate pathological features and pathogenic mechanisms underlying ALD. Notably, oxidative changes in the liver have been recognized as a signature trait of ALD. Progression of ALD is linked to the generation of highly reactive free radicals by reactions involving ethanol and its metabolites. Furthermore, hepatic oxidative stress promotes tissue injury and, in turn, stimulates inflammatory responses in the liver, forming a pathological loop that promotes the progression of ALD. Accordingly, accumulating further knowledge on the relationship between oxidative stress and inflammation may help establish a viable therapeutic approach for treating ALD.
Collapse
|
57
|
Mesona chinensis Benth polysaccharides alleviates liver injury by beneficial regulation of gut microbiota in cyclophosphamide-induced mice. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2021.07.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
58
|
He Q, Liu L, Liu C, Hua H, Zhao W, Zhu H, Cheng Y, Guo Y. Effect of polysaccharides from Tibetan turnip (Brassica rapa L.) on the gut microbiome in vitro fermentation and in vivo metabolism. Food Funct 2022; 13:3063-3076. [DOI: 10.1039/d1fo03821d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Tibetan turnip ( Brassica rapa L. ) polysaccharide (TTP) is an active ingredient and has been studied for many years due to its biological effect. There are few studies on...
Collapse
|
59
|
Park JW, Kim SE, Lee NY, Kim JH, Jung JH, Jang MK, Park SH, Lee MS, Kim DJ, Kim HS, Suk KT. Role of Microbiota-Derived Metabolites in Alcoholic and Non-Alcoholic Fatty Liver Diseases. Int J Mol Sci 2021; 23:426. [PMID: 35008852 PMCID: PMC8745242 DOI: 10.3390/ijms23010426] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/28/2021] [Accepted: 12/28/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic liver disease encompasses diseases that have various causes, such as alcoholic liver disease (ALD) and non-alcoholic fatty liver disease (NAFLD). Gut microbiota dysregulation plays a key role in the pathogenesis of ALD and NAFLD through the gut-liver axis. The gut microbiota consists of various microorganisms that play a role in maintaining the homeostasis of the host and release a wide number of metabolites, including short-chain fatty acids (SCFAs), peptides, and hormones, continually shaping the host's immunity and metabolism. The integrity of the intestinal mucosal and vascular barriers is crucial to protect liver cells from exposure to harmful metabolites and pathogen-associated molecular pattern molecules. Dysbiosis and increased intestinal permeability may allow the liver to be exposed to abundant harmful metabolites that promote liver inflammation and fibrosis. In this review, we introduce the metabolites and components derived from the gut microbiota and discuss their pathologic effect in the liver alongside recent advances in molecular-based therapeutics and novel mechanistic findings associated with the gut-liver axis in ALD and NAFLD.
Collapse
Affiliation(s)
- Ji-Won Park
- Department of Internal Medicine, Hallym University Sacred Heart Hospital of Hallym University Medical Center, 22, Gwanpyeong-ro 170 beon-gil, Dongan-gu, Anyang-si 14068, Korea; (J.-W.P.); (S.-E.K.)
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon-si 24252, Korea; (N.Y.L.); (J.-H.K.); (J.-H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (D.-J.K.)
| | - Sung-Eun Kim
- Department of Internal Medicine, Hallym University Sacred Heart Hospital of Hallym University Medical Center, 22, Gwanpyeong-ro 170 beon-gil, Dongan-gu, Anyang-si 14068, Korea; (J.-W.P.); (S.-E.K.)
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon-si 24252, Korea; (N.Y.L.); (J.-H.K.); (J.-H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (D.-J.K.)
| | - Na Young Lee
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon-si 24252, Korea; (N.Y.L.); (J.-H.K.); (J.-H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (D.-J.K.)
- Department of Internal Medicine, Chuncheon Sacred Heart Hospital of Hallym University Medical Center, 77, Sakju-ro, Chuncheon-si 24253, Korea
| | - Jung-Hee Kim
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon-si 24252, Korea; (N.Y.L.); (J.-H.K.); (J.-H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (D.-J.K.)
- Department of Internal Medicine, Dongtan Sacred Heart Hospital of Hallym University Medical Center, 7, Keunjaebong-gil, Hwaseong-si 445-907, Korea
| | - Jang-Han Jung
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon-si 24252, Korea; (N.Y.L.); (J.-H.K.); (J.-H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (D.-J.K.)
- Department of Internal Medicine, Dongtan Sacred Heart Hospital of Hallym University Medical Center, 7, Keunjaebong-gil, Hwaseong-si 445-907, Korea
| | - Myoung-Kuk Jang
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon-si 24252, Korea; (N.Y.L.); (J.-H.K.); (J.-H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (D.-J.K.)
- Department of Internal Medicine, Kangdong Sacred Heart Hospital of Hallym University Medical Center, 18, Cheonho-daero 173-gil, Gangdong-gu, Seoul 05355, Korea
| | - Sang-Hoon Park
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon-si 24252, Korea; (N.Y.L.); (J.-H.K.); (J.-H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (D.-J.K.)
- Department of Internal Medicine, Kangnam Sacred Heart Hospital of Hallym University Medical Center, 1, Singil-ro, Yeongdeungpo-gu, Seoul 07441, Korea
| | - Myung-Seok Lee
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon-si 24252, Korea; (N.Y.L.); (J.-H.K.); (J.-H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (D.-J.K.)
- Department of Internal Medicine, Kangnam Sacred Heart Hospital of Hallym University Medical Center, 1, Singil-ro, Yeongdeungpo-gu, Seoul 07441, Korea
| | - Dong-Joon Kim
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon-si 24252, Korea; (N.Y.L.); (J.-H.K.); (J.-H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (D.-J.K.)
- Department of Internal Medicine, Chuncheon Sacred Heart Hospital of Hallym University Medical Center, 77, Sakju-ro, Chuncheon-si 24253, Korea
| | - Hyoung-Su Kim
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon-si 24252, Korea; (N.Y.L.); (J.-H.K.); (J.-H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (D.-J.K.)
- Department of Internal Medicine, Kangdong Sacred Heart Hospital of Hallym University Medical Center, 18, Cheonho-daero 173-gil, Gangdong-gu, Seoul 05355, Korea
| | - Ki Tae Suk
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon-si 24252, Korea; (N.Y.L.); (J.-H.K.); (J.-H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (D.-J.K.)
- Department of Internal Medicine, Chuncheon Sacred Heart Hospital of Hallym University Medical Center, 77, Sakju-ro, Chuncheon-si 24253, Korea
| |
Collapse
|
60
|
Portincasa P, Bonfrate L, Khalil M, Angelis MD, Calabrese FM, D’Amato M, Wang DQH, Di Ciaula A. Intestinal Barrier and Permeability in Health, Obesity and NAFLD. Biomedicines 2021; 10:83. [PMID: 35052763 PMCID: PMC8773010 DOI: 10.3390/biomedicines10010083] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/20/2021] [Accepted: 12/28/2021] [Indexed: 02/07/2023] Open
Abstract
The largest surface of the human body exposed to the external environment is the gut. At this level, the intestinal barrier includes luminal microbes, the mucin layer, gastrointestinal motility and secretion, enterocytes, immune cells, gut vascular barrier, and liver barrier. A healthy intestinal barrier is characterized by the selective permeability of nutrients, metabolites, water, and bacterial products, and processes are governed by cellular, neural, immune, and hormonal factors. Disrupted gut permeability (leaky gut syndrome) can represent a predisposing or aggravating condition in obesity and the metabolically associated liver steatosis (nonalcoholic fatty liver disease, NAFLD). In what follows, we describe the morphological-functional features of the intestinal barrier, the role of major modifiers of the intestinal barrier, and discuss the recent evidence pointing to the key role of intestinal permeability in obesity/NAFLD.
Collapse
Affiliation(s)
- Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| | - Leonilde Bonfrate
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| | - Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Francesco Maria Calabrese
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Mauro D’Amato
- Gastrointestinal Genetics Lab, CIC bioGUNE-BRTA, 48160 Derio, Spain;
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - David Q.-H. Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York, NY 10461, USA;
| | - Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| |
Collapse
|
61
|
Singhal R, Donde H, Ghare S, Stocke K, Zhang J, Vadhanam M, Reddy S, Gobejishvili L, Chilton P, Joshi-Barve S, Feng W, McClain C, Hoffman K, Petrosino J, Vital M, Barve S. Decrease in acetyl-CoA pathway utilizing butyrate-producing bacteria is a key pathogenic feature of alcohol-induced functional gut microbial dysbiosis and development of liver disease in mice. Gut Microbes 2021; 13:1946367. [PMID: 34369304 PMCID: PMC8354657 DOI: 10.1080/19490976.2021.1946367] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Emerging research evidence has established the critical role of the gut-liver axis in the development of alcohol-associated liver disease (ALD). The present study employed 16S rRNA gene and whole genome shotgun (WGS) metagenomic analysis in combination with a revised microbial dataset to comprehensively detail the butyrate-producing microbial communities and the associated butyrate metabolic pathways affected by chronic ethanol feeding. Specifically, the data demonstrated that a decrease in several butyrate-producing bacterial genera belonging to distinct families within the Firmicutes phyla was a significant component of ethanol-induced dysbiosis. WGS analysis of total bacterial genomes encompassing butyrate synthesizing pathways provided the functional characteristics of the microbiome associated with butyrate synthesis. The data revealed that in control mice microbiome, the acetyl-coenzyme A (CoA) butyrate synthesizing pathway was the most prevalent and was significantly and maximally decreased by chronic ethanol feeding. Further WGS analysis i) validated the ethanol-induced decrease in the acetyl-CoA pathway by identifying the decrease in two critical genes but - (butyryl-CoA: acetate CoA transferase) and buk - (butyrate kinase) that encode the terminal condensing enzymes required for converting butyryl-CoA to butyrate and ii) detection of specific taxa of butyrate-producing bacteria containing but and buk genes. Notably, the administration of tributyrin (Tb) - a butyrate prodrug - significantly prevented ethanol-induced decrease in butyrate-producing bacteria, hepatic steatosis, inflammation, and injury. Taken together, our findings strongly suggest that the loss of butyrate-producing bacteria using the acetyl-CoA pathway is a significant pathogenic feature of ethanol-induced microbial dysbiosis and ALD and can be targeted for therapy.
Collapse
Affiliation(s)
- Richa Singhal
- University of Louisville Department of Medicine,University of Louisville Alcohol Research Center
| | - Hridgandh Donde
- University of Louisville Department of Medicine,University of Louisville Alcohol Research Center
| | - Smita Ghare
- University of Louisville Department of Medicine,University of Louisville Alcohol Research Center
| | - Kendall Stocke
- University of Louisville Department of Environmental and Occupational Health Science
| | - Jingwein Zhang
- University of Louisville Department of Medicine,University of Louisville Alcohol Research Center
| | - Manicka Vadhanam
- University of Louisville Department of Medicine,University of Louisville Alcohol Research Center
| | - Sreelatha Reddy
- University of Louisville Department of Medicine,University of Louisville Alcohol Research Center
| | - Leila Gobejishvili
- University of Louisville Department of Medicine,University of Louisville Alcohol Research Center
| | - Paula Chilton
- University of Louisville Department of Medicine,University of Louisville Alcohol Research Center
| | - Swati Joshi-Barve
- University of Louisville Department of Medicine,University of Louisville Alcohol Research Center
| | - Wenke Feng
- University of Louisville Department of Medicine,University of Louisville Alcohol Research Center
| | - Craig McClain
- University of Louisville Department of Medicine,University of Louisville Alcohol Research Center
| | - Kristi Hoffman
- Baylor College of Medicine Department of Molecular Virology and Microbiology,Baylor College of Medicine Center for Metagenomics and Microbiome Research
| | - Joseph Petrosino
- Baylor College of Medicine Department of Molecular Virology and Microbiology,Baylor College of Medicine Center for Metagenomics and Microbiome Research
| | - Marius Vital
- Hannover Medical School, Hanover, Germany,Helmholtz Center for Infection Research,Helmholtz Association of German Research Centers
| | - Shirish Barve
- University of Louisville Department of Medicine,University of Louisville Alcohol Research Center,University of Louisville Department of Pharmacology and Toxicology,CONTACT Dr. Shirish Barve Departments of Medicine and Pharmacology and Toxicology, University of Louisville Health Sciences Center, 505 S. Hancock St. CTR Bldg., Room 515, Louisville, KY40202, USA
| |
Collapse
|
62
|
Hu B, Liu C, Jiang W, Zhu H, Zhang H, Qian H, Zhang W. Chronic in vitro fermentation and in vivo metabolism: Extracellular polysaccharides from Sporidiobolus pararoseus regulate the intestinal microbiome of humans and mice. Int J Biol Macromol 2021; 192:398-406. [PMID: 34571128 DOI: 10.1016/j.ijbiomac.2021.09.127] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/04/2021] [Accepted: 09/19/2021] [Indexed: 11/17/2022]
Abstract
The fungus Sporidiobolus pararoseus not only produces carotenoids, but also produces bioactive extracellular polysaccharides (SPP). However, the relationship between SPP and the metabolism of gut microbiome is unclear. The aim of this study was to investigate the mechanism of SPP regulating intestinal health in vivo and in vitro. Results showed that SPP are nondigestible polysaccharides after the digestion with simulated stomach and small intestinal juice in vitro. After SPP was cultured in an in vitro intestinal simulation system for seven days, the concentration of short-chain fatty acids (SCFAs) increased; the microbial diversity changed; the relative abundance of Bifidobacterium and Streptococcus increased; and that of Escherichia Shigella and Lachnospiraceae NK4A136 decreased. In addition, metabolism of SPP by the mice colonic microbiome showed SPP decreased the relative abundance of Firmicutes and Bacteroidota, while the relative abundance of Verrucomicrobiota, Desulfobacterota, and Actinobacteriota increased. Finally, predicted Kyoto Encyclopedia of Genes and Genomes (KEGG) metabolism results also showed that SPP can enhance the metabolism of cofactors, vitamins, amino acids, starch, and sucrose. In conclusion, SPP can multiply the intestinal beneficial bacteria of humans and mice, promote the production of SCFAs and metabolism of amino acids, and promote intestinal health.
Collapse
Affiliation(s)
- Bin Hu
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Chang Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| | - Wenhao Jiang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| | - Hongkang Zhu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| | - Hui Zhang
- China Certification & Inspection Group Shanghai Co., Ltd, Shanghai 200120, China
| | - He Qian
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| | - Weiguo Zhang
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
63
|
Siddiqui MT, Cresci GAM. The Immunomodulatory Functions of Butyrate. J Inflamm Res 2021; 14:6025-6041. [PMID: 34819742 PMCID: PMC8608412 DOI: 10.2147/jir.s300989] [Citation(s) in RCA: 171] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/15/2021] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal (GI) system contains many different types of immune cells, making it a key immune organ system in the human body. In the last decade, our knowledge has substantially expanded regarding our understanding of the gut microbiome and its complex interaction with the gut immune system. Short chain fatty acids (SCFA), and specifically butyrate, play an important role in mediating the effects of the gut microbiome on local and systemic immunity. Gut microbial alterations and depletion of luminal butyrate have been well documented in the literature for a number of systemic and GI inflammatory disorders. Although a substantial knowledge gap exists requiring the need for further investigations to determine cause and effect, there is heightened interest in developing immunomodulatory therapies by means of reprogramming of gut microbiome or by supplementing its beneficial metabolites, such as butyrate. In the current review, we discuss the role of endogenous butyrate in the inflammatory response and maintaining immune homeostasis within the intestine. We also present the experimental models and human studies which explore therapeutic potential of butyrate supplementation in inflammatory conditions associated with butyrate depletion.
Collapse
Affiliation(s)
- Mohamed Tausif Siddiqui
- Department of Gastroenterology, Hepatology and Human Nutrition, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Gail A M Cresci
- Department of Gastroenterology, Hepatology and Human Nutrition, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Pediatric Gastroenterology, Cleveland Clinic, Cleveland, OH, 44195, USA
| |
Collapse
|
64
|
Gut Microbiota as the Link between Elevated BCAA Serum Levels and Insulin Resistance. Biomolecules 2021; 11:biom11101414. [PMID: 34680047 PMCID: PMC8533624 DOI: 10.3390/biom11101414] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 12/17/2022] Open
Abstract
The microbiota-harboring human gut is an exquisitely active ecosystem that has evolved in a constant symbiosis with the human host. It produces numerous compounds depending on its metabolic capacity and substrates availability. Diet is the major source of the substrates that are metabolized to end-products, further serving as signal molecules in the microbiota-host cross-talk. Among these signal molecules, branched-chain amino acids (BCAAs) has gained significant scientific attention. BCAAs are abundant in animal-based dietary sources; they are both produced and degraded by gut microbiota and the host circulating levels are associated with the risk of type 2 diabetes. This review aims to summarize the current knowledge on the complex relationship between gut microbiota and its functional capacity to handle BCAAs as well as the host BCAA metabolism in insulin resistance development. Targeting gut microbiota BCAA metabolism with a dietary modulation could represent a promising approach in the prevention and treatment of insulin resistance related states, such as obesity and diabetes.
Collapse
|
65
|
Pohl K, Moodley P, Dhanda AD. Alcohol's Impact on the Gut and Liver. Nutrients 2021; 13:nu13093170. [PMID: 34579046 PMCID: PMC8472839 DOI: 10.3390/nu13093170] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 12/13/2022] Open
Abstract
Alcohol is inextricably linked with the digestive system. It is absorbed through the gut and metabolised by hepatocytes within the liver. Excessive alcohol use results in alterations to the gut microbiome and gut epithelial integrity. It contributes to important micronutrient deficiencies including short-chain fatty acids and trace elements that can influence immune function and lead to liver damage. In some people, long-term alcohol misuse results in liver disease progressing from fatty liver to cirrhosis and hepatocellular carcinoma, and results in over half of all deaths from chronic liver disease, over half a million globally per year. In this review, we will describe the effect of alcohol on the gut, the gut microbiome and liver function and structure, with a specific focus on micronutrients and areas for future research.
Collapse
Affiliation(s)
- Keith Pohl
- South West Liver Unit, University Hospitals Plymouth NHS Trust, Plymouth PL6 8DH, UK; (K.P.); (P.M.)
- Hepatology Research Group, Faculty of Health, University of Plymouth, Plymouth PL4 8AA, UK
| | - Prebashan Moodley
- South West Liver Unit, University Hospitals Plymouth NHS Trust, Plymouth PL6 8DH, UK; (K.P.); (P.M.)
- Hepatology Research Group, Faculty of Health, University of Plymouth, Plymouth PL4 8AA, UK
| | - Ashwin D. Dhanda
- South West Liver Unit, University Hospitals Plymouth NHS Trust, Plymouth PL6 8DH, UK; (K.P.); (P.M.)
- Hepatology Research Group, Faculty of Health, University of Plymouth, Plymouth PL4 8AA, UK
- Correspondence: ; Tel.: +44-1752-432723
| |
Collapse
|
66
|
Abstract
Antifibrotic therapies for the treatment of liver fibrosis represent an unconquered area of drug development. The significant involvement of the gut microbiota as a driving force in a multitude of liver disease, be it pathogenesis or fibrotic progression, suggest that targeting the gut–liver axis, relevant signaling pathways, and/or manipulation of the gut’s commensal microbial composition and its metabolites may offer opportunities for biomarker discovery, novel therapies and personalized medicine development. Here, we review potential links between bacterial translocation and deficits of host-microbiome compartmentalization and liver fibrosis that occur in settings of advanced chronic liver disease. We discuss established and emerging therapeutic strategies, translated from our current knowledge of the gut–liver axis, targeted at restoring intestinal eubiosis, ameliorating hepatic fibrosis and rising portal hypertension that characterize and define the course of decompensated cirrhosis.
Collapse
|
67
|
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
|
68
|
Giraud J, Saleh M. Host-Microbiota Interactions in Liver Inflammation and Cancer. Cancers (Basel) 2021; 13:cancers13174342. [PMID: 34503151 PMCID: PMC8430654 DOI: 10.3390/cancers13174342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Hepatocellular carcinoma (HCC) is a difficult to treat liver cancer that generally arises in individuals suffering from alcoholic or non-alcoholic fatty liver diseases. Inflammation, tissue injury and fibrosis are important precursors of HCC. In this review, we explore the links between the microbiota, inflammation and carcinogenesis in the context of HCC. We discuss how the gut and liver communicate and how microbial molecules, including structural components and metabolites, elicit inflammation and tumorigenesis in the liver. A better understanding of microbiota-dependent mechanisms of liver cancer development might lead to novel microbial-based therapeutic approaches. Abstract Hepatocellular carcinoma (HCC) is a classical inflammation-promoted cancer that occurs in a setting of liver diseases, including nonalcoholic fatty liver disease (NAFLD) or alcoholic liver disease (ALD). These pathologies share key characteristics, notably intestinal dysbiosis, increased intestinal permeability and an imbalance in bile acids, choline, fatty acids and ethanol metabolites. Translocation of microbial- and danger-associated molecular patterns (MAMPs and DAMPs) from the gut to the liver elicits profound chronic inflammation, leading to severe hepatic injury and eventually HCC progression. In this review, we first describe how the gut and the liver communicate and discuss mechanisms by which the intestinal microbiota elicit hepatic inflammation and HCC. We focus on the role of microbial products, e.g., MAMPs, host inflammatory effectors and host–microbiome-derived metabolites in tumor-promoting mechanisms, including cell death and senescence. Last, we explore the potential of harnessing the microbiota to treat liver diseases and HCC.
Collapse
Affiliation(s)
- Julie Giraud
- ImmunoConcEpT, CNRS, UMR 5164, University of Bordeaux, F-33000 Bordeaux, France;
| | - Maya Saleh
- ImmunoConcEpT, CNRS, UMR 5164, University of Bordeaux, F-33000 Bordeaux, France;
- Department of Medicine, McGill University, Montreal, QC H3G 0B1, Canada
- Correspondence:
| |
Collapse
|
69
|
Guo Y, Chen X, Gong P, Chen F, Cui D, Wang M. Advances in the
in vitro
digestion and fermentation of polysaccharides. Int J Food Sci Technol 2021. [DOI: 10.1111/ijfs.15308] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Yuxi Guo
- School of Food and Biological Engineering Shaanxi University of Science & Technology Xi'an 710021 China
| | - Xuefeng Chen
- School of Food and Biological Engineering Shaanxi University of Science & Technology Xi'an 710021 China
- Shaanxi Research Institute of Agricultural Product Processing Technology Xi'an 710021 China
| | - Pin Gong
- School of Food and Biological Engineering Shaanxi University of Science & Technology Xi'an 710021 China
| | - Fuxin Chen
- School of Chemistry and Chemical Engineering Xi’an University of Science and Technology Xi’an 710054 China
| | - Dandan Cui
- School of Food and Biological Engineering Shaanxi University of Science & Technology Xi'an 710021 China
| | - Mengrao Wang
- School of Food and Biological Engineering Shaanxi University of Science & Technology Xi'an 710021 China
| |
Collapse
|
70
|
Liu C, Du P, Guo Y, Xie Y, Yu H, Yao W, Cheng Y, Qian H. Extraction, characterization of aloe polysaccharides and the in-depth analysis of its prebiotic effects on mice gut microbiota. Carbohydr Polym 2021; 261:117874. [DOI: 10.1016/j.carbpol.2021.117874] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/15/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023]
|
71
|
Li R, Mao Z, Ye X, Zuo T. Human Gut Microbiome and Liver Diseases: From Correlation to Causation. Microorganisms 2021; 9:1017. [PMID: 34066850 PMCID: PMC8151257 DOI: 10.3390/microorganisms9051017] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/25/2021] [Accepted: 04/30/2021] [Indexed: 02/06/2023] Open
Abstract
The important role of human gut microbiota in liver diseases has long been recognized as dysbiosis and the translocation of certain microbes from the gut to liver. With the development of high-throughput DNA sequencing, the complexity and integrity of the gut microbiome in the whole spectrum of liver diseases is emerging. Specific patterns of gut microbiota have been identified in liver diseases with different causes, including alcoholic, non-alcoholic, and virus induced liver diseases, or even at different stages, ranging from steatohepatitis, fibrosis, cirrhosis, to hepatocellular carcinoma. At the same time, the mechanism of how microbiota contributes to liver diseases goes beyond the traditional function of the gut-liver axis which could lead to liver injury and inflammation. With the application of proteomics, metabolomics, and modern molecular technologies, more microbial metabolites and the complicated interaction of microbiota with host immunity come into our understanding in the liver pathogenesis. Germ-free animal models serve as a workhorse to test the function of microbiota and their derivatives in liver disease models. Here, we review the current evidence on the relationship between gut microbiota and liver diseases, and the mechanisms underlying this phenotype. In addition to original liver diseases, gut microbiota might also affect liver injury in systemic disorders involving multiple organs, as in the case of COVID-19 at a severe state. A better understanding of the gut microbial contribution to liver diseases might help us better benefit from this guest-host relationship and pave the way for novel therapies.
Collapse
Affiliation(s)
- Rui Li
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan 430070, China;
| | - Zhengsheng Mao
- Department of Neurology, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China;
| | - Xujun Ye
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan 430070, China;
| | - Tao Zuo
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou 510000, China
| |
Collapse
|
72
|
Zhang T, Li J, Liu CP, Guo M, Gao CL, Zhou LP, Long Y, Xu Y. Butyrate ameliorates alcoholic fatty liver disease via reducing endotoxemia and inhibiting liver gasdermin D-mediated pyroptosis. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:873. [PMID: 34164507 PMCID: PMC8184481 DOI: 10.21037/atm-21-2158] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/13/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Alcoholic fatty liver disease (AFLD) is characterized by hepatic steatosis and carries an elevated risk of cirrhosis and hepatocellular carcinoma. However, the mechanism of AFLD has not been elucidated thoroughly, and there are still no efficient therapies in clinic. Notably, butyrate, one kind of short-chain fatty acids produced by gut microbiota, has been shown to improve methionine-choline-deficient diet-induced non-alcoholic steatohepatitis. And our previous study found that butyrate ameliorated endotoxemia in db/db mice. In this study, we aimed to explore the role of butyrate in the development of AFLD. METHODS C57BL/6 mice were treated with saline (normal control), alcohol with or without butyrate by gavage for 6 months. AFLD was evaluated by the levels of serum alcohol, aspartate aminotransferase (AST), alanine transaminase (ALT), triglyceride (TG) and intrahepatic TG. And the histology and inflammation in liver and colon were analyzed using hematoxylin-eosin (H&E) staining, immunohistochemistry and western blot. In addition, gut microbiota composition was analyzed using the V3-V4 regions of the bacterial 16S ribosomal RNA gene by sequence. Furthermore, we performed in vitro experiment to verify the role of butyrate in hepatocyte by western blot and transmission electron microscopy. RESULTS We found that butyrate ameliorated alcohol-induced hepatic steatosis and inflammation. Furthermore, chronic alcohol feeding induced dysbiosis and dysfunction of the gut microbiota, disrupted the intestinal barrier, and increased serum endotoxin levels. Meanwhile, butyrate improved the intestinal barrier disruption and endotoxemia induced by alcohol, but did not significantly alleviate the microbiome dysfunction. Mechanistically, butyrate ameliorated AFLD by inhibiting gasdermin D (GSDMD)-mediated pyroptosis. CONCLUSIONS In summary, we found butyrate ameliorated alcoholic fatty liver by down-regulating GSDMD-mediated pyroptosis. We speculate that butyrate improves AFLD mainly by maintaining intestinal barrier function and alleviating gut leakage. These findings suggest that butyrate may have the potential to serve as a novel treatment for AFLD.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Jun Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Cui-Ping Liu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Man Guo
- Department of Endocrinology and Metabolism, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chen-Lin Gao
- Department of Endocrinology and Metabolism, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lu-Ping Zhou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yang Long
- Department of Endocrinology and Metabolism, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China
| | - Yong Xu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| |
Collapse
|
73
|
Salavrakos M, Leclercq S, De Timary P, Dom G. Microbiome and substances of abuse. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:110113. [PMID: 32971216 DOI: 10.1016/j.pnpbp.2020.110113] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 12/12/2022]
Abstract
There is a growing amount of evidence showing a reciprocal relation between the gut microbiota and the brain. Substance use disorders (SUD), which are a major cause of preventable morbidity and mortality worldwide, have an influence on the gut microbiota and on the gut-brain axis. The communication between the microbiota and the brain exists through different pathways: (1) the immune response elicited by bacterial products, coupled with alterations of the intestinal barrier allowing these products to enter the bloodstream, (2) the direct and indirect effects of bacterial metabolites such as short chain fatty acids (SCFAs) or tryptophan on the brain, (3) and the hypothalamic-pituitary-adrenal (HPA) axis, whose peripheral afferents can be influenced by the microbiota, and can in turn activate microglia. Among substances of abuse, alcohol has been the subject of the greatest number of studies in this field. In some but not all patients suffering from alcohol-use-disorder (AUD), alcohol alters the composition of the gut microbiota and the permeability of the intestinal barrier, directly and through dysbiosis. It has also been well demonstrated that alcohol induces a peripheral inflammation; it is still unclear whether it induces a central inflammation, as there are contradictory results in human studies. In animal studies, it has been shown that neuroinflammation increases during alcohol withdrawal. Literature on opioids and stimulants is less numerous. Chronic morphine intake induces dysbiosis, increased intestinal permeability and a probable neuroinflammation, which could explain symptoms such as tolerance, hyperalgesia and deficit in reward behavior. Cocaine induces a dysbiosis and conversely the microbiome can modulate the behavioral response to stimulant drugs. Tobacco cessation is associated with an increase in microbiota diversity. Taken together, the findings of our narrative literature review suggest a bidirectional influence in the pathogenesis of substance use disorders.
Collapse
Affiliation(s)
- M Salavrakos
- Target Journal Progress in Neuropsychopharmacology and Biological Psychiatry, Belgium
| | - S Leclercq
- Target Journal Progress in Neuropsychopharmacology and Biological Psychiatry, Belgium
| | - P De Timary
- Target Journal Progress in Neuropsychopharmacology and Biological Psychiatry, Belgium
| | - G Dom
- Target Journal Progress in Neuropsychopharmacology and Biological Psychiatry, Belgium.
| |
Collapse
|
74
|
Khan A, Ding Z, Ishaq M, Bacha AS, Khan I, Hanif A, Li W, Guo X. Understanding the Effects of Gut Microbiota Dysbiosis on Nonalcoholic Fatty Liver Disease and the Possible Probiotics Role: Recent Updates. Int J Biol Sci 2021; 17:818-833. [PMID: 33767591 PMCID: PMC7975705 DOI: 10.7150/ijbs.56214] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is leading chronic liver syndrome worldwide. Gut microbiota dysbiosis significantly contributes to the pathogenesis and severity of NAFLD. However, its role is complex and even unclear. Treatment of NAFLD through chemotherapeutic agents have been questioned because of their side effects on health. In this review, we highlighted and discussed the current understanding on the importance of gut microbiota, its dysbiosis and its effects on the gut-liver axis and gut mucosa. Further, we discussed key mechanisms involved in gut dysbiosis to provide an outline of its role in progression to NAFLD and liver cirrhosis. In addition, we also explored the potential role of probiotics as a treatment approach for the prevention and treatment of NAFLD. Based on the latest findings, it is evident that microbiota targeted interventions mostly the use of probiotics have shown promising effects and can possibly alleviate the gut microbiota dysbiosis, regulate the metabolic pathways which in turn inhibit the progression of NAFLD through the gut-liver axis. However, very limited studies in humans are available on this issue and suggest further research work to identify a specific core microbiome association with NAFLD and to discover its mechanism of pathogenesis, which will help to enhance the therapeutic potential of probiotics to NAFLD.
Collapse
Affiliation(s)
- Ashiq Khan
- School of Life Sciences, Probiotics and Biological Feed Research Centre, Lanzhou University, Lanzhou 730000, PR China
- Department of Microbiology, Balochistan University of Information Technology Engineering & Management Sciences Quetta 87300, Pakistan
| | - Zitong Ding
- School of Life Sciences, Probiotics and Biological Feed Research Centre, Lanzhou University, Lanzhou 730000, PR China
| | - Muhammad Ishaq
- School of Life Sciences, Probiotics and Biological Feed Research Centre, Lanzhou University, Lanzhou 730000, PR China
| | - Ali Sher Bacha
- School of Life Sciences, Probiotics and Biological Feed Research Centre, Lanzhou University, Lanzhou 730000, PR China
| | - Israr Khan
- School of Life Sciences, Institute of Microbiology Lanzhou University, Lanzhou 730000, PR China
| | - Anum Hanif
- School of Life Sciences, Probiotics and Biological Feed Research Centre, Lanzhou University, Lanzhou 730000, PR China
| | - Wenyuan Li
- School of Life Sciences, Probiotics and Biological Feed Research Centre, Lanzhou University, Lanzhou 730000, PR China
| | - Xusheng Guo
- School of Life Sciences, Probiotics and Biological Feed Research Centre, Lanzhou University, Lanzhou 730000, PR China
| |
Collapse
|
75
|
Kwong EK, Puri P. Gut microbiome changes in Nonalcoholic fatty liver disease & alcoholic liver disease. Transl Gastroenterol Hepatol 2021; 6:3. [PMID: 33409398 DOI: 10.21037/tgh.2020.02.18] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 02/11/2020] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD) are some of the most common liver diseases worldwide. The human gut microbiome is dynamic and shifts in bacterial composition have been implicated in many diseases. Studies have shown that there is a shift in bacterial overgrowth favoring pro-inflammatory mediators in patients with advanced disease progression such as cirrhosis. Further investigation demonstrated that the transplantation of gut microbiota from advanced liver disease patients can reproduce severe liver inflammation and injury in mice. Various techniques in manipulating the gut microbiota have been attempted including fecal transplantation and probiotics. This review focuses on the changes in the gut microbiota as well as emerging lines of microbiome work with respect to NAFLD and ALD.
Collapse
Affiliation(s)
- Eric K Kwong
- Department of Microbiology and Immunology, McGuire VA Medical Center, Richmond, VA, USA
| | - Puneet Puri
- Section of Gastroenterology, Hepatology and Nutrition, McGuire VA Medical Center, Richmond, VA, USA.,Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
76
|
Liu C, Du P, Cheng Y, Guo Y, Hu B, Yao W, Zhu X, Qian H. Study on fecal fermentation characteristics of aloe polysaccharides in vitro and their predictive modeling. Carbohydr Polym 2021; 256:117571. [PMID: 33483068 DOI: 10.1016/j.carbpol.2020.117571] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/14/2020] [Accepted: 12/25/2020] [Indexed: 02/08/2023]
Abstract
Aloe polysaccharides (APs) are well-known plant polysaccharides, but little is known about their digestion and fermentation characteristics in vitro. In this study, the molecular weight of APs had no significant changes after gastric and intestinal digestion. During the fecal fermentation, the content of volatiles and pH value decreased continuously, while the short-chain fatty acids (SCFAs) concentration increased significantly. Additionally, the abundance of the microbiota associated with the metabolism of SCFAs was increased, including Prevotella, Catenibacterium, Lachnospiraceae, and Coprococcus, while the harmful microbiota was decreased, like Escherichia-Shigella, and Veillonella. Moreover, bioinformatics analysis indicated that APs boosted fructose and mannose metabolism, and the gene expressions of enzymes, containing mannose-6-phosphate isomerase [EC:5.3.1.8]. Structural equation modeling also highlighted that SCFAs-producing microbiota were primary degraders of APs, suggesting APs may facilitate the manufacture of functional foods with the purpose of maintaining intestinal health.
Collapse
Affiliation(s)
- Chang Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, and International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China
| | - Peng Du
- Air Force Medical Center, PLA, Beijing, 100142. China
| | - Yuliang Cheng
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, and International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China
| | - Yahui Guo
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, and International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China
| | - Bin Hu
- School of Biotechnology, Jiangnan University, Jiangnan University, Wuxi, 214122, China
| | - Weirong Yao
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, and International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China
| | - Xuan Zhu
- School of Food Science and Bioengineering, Zhejiang Gongshang University, No. 18 Xuezheng Str, Hangzhou, Zhejiang Province, 310018, China.
| | - He Qian
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, and International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
77
|
Hartmann P, Schnabl B. New Developments in Microbiome in Alcohol-Associated and Nonalcoholic Fatty Liver Disease. Semin Liver Dis 2021; 41:87-102. [PMID: 33957682 PMCID: PMC8163568 DOI: 10.1055/s-0040-1719174] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Alcohol-associated liver disease (ALD) and nonalcoholic fatty liver disease (NAFLD) are important causes of morbidity and mortality worldwide. The intestinal microbiota is involved in the development and progression of both ALD and NAFLD. Here we describe associated changes in the intestinal microbiota, and we detail randomized clinical trials in ALD and NAFLD which evaluate treatments modulating the intestinal microbiome including fecal microbiota transplantation, probiotics, prebiotics, synbiotics, and antibiotics. Finally, we discuss precision medicine approaches targeting the intestinal microbiome to ameliorate ALD and NAFLD.
Collapse
Affiliation(s)
- Phillipp Hartmann
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA;,Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA;,Department of Medicine, VA San Diego Healthcare System, San Diego, CA 92161, USA.,Corresponding Author: Bernd Schnabl, MD, Department of Medicine, University of California, San Diego, Biomedical Research Facility 2 (BRF2), Room 4A22, 9500 Gilman Drive, MC0063, La Jolla, CA 92093, Phone: +1 858-822-5311, Fax: +1 858-822-5370,
| |
Collapse
|
78
|
Bruellman R, Llorente C. A Perspective Of Intestinal Immune-Microbiome Interactions In Alcohol-Associated Liver Disease. Int J Biol Sci 2021; 17:307-327. [PMID: 33390852 PMCID: PMC7757023 DOI: 10.7150/ijbs.53589] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
Uncovering the intricacies of the gut microbiome and how it interacts with the host immune system has opened up pathways in the search for the treatment of disease conditions. Alcohol-associated liver disease is a major cause of death worldwide. Research has shed light on the breakdown of the protective gut barriers, translocation of gut microbes to the liver and inflammatory immune response to microbes all contributing to alcohol-associated liver disease. This knowledge has opened up avenues for alternative therapies to alleviate alcohol-associated liver disease based on the interaction of the commensal gut microbiome as a key player in the regulation of the immune response. This review describes the relevance of the intestinal immune system, the gut microbiota, and specialized and non-specialized intestinal cells in the regulation of intestinal homeostasis. It also reflects how these components are altered during alcohol-associated liver disease and discusses new approaches for potential future therapies in alcohol-associated liver disease.
Collapse
Affiliation(s)
- Ryan Bruellman
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Cristina Llorente
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
79
|
Lamas-Paz A, Morán L, Peng J, Salinas B, López-Alcántara N, Sydor S, Vilchez-Vargas R, Asensio I, Hao F, Zheng K, Martín-Adrados B, Moreno L, Cogolludo A, Gómez del Moral M, Bechmann L, Martínez-Naves E, Vaquero J, Bañares R, Nevzorova YA, Cubero FJ. Intestinal Epithelial Cell-Derived Extracellular Vesicles Modulate Hepatic Injury via the Gut-Liver Axis During Acute Alcohol Injury. Front Pharmacol 2020; 11:603771. [PMID: 33408632 PMCID: PMC7779758 DOI: 10.3389/fphar.2020.603771] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022] Open
Abstract
Binge drinking, i.e., heavy episodic drinking in a short time, has recently become an alarming societal problem with negative health impact. However, the harmful effects of acute alcohol injury in the gut-liver axis remain elusive. Hence, we focused on the physiological and pathological changes and the underlying mechanisms of experimental binge drinking in the context of the gut-liver axis. Eight-week-old mice with a C57BL/6 background received a single dose (p.o.) of ethanol (EtOH) [6 g/kg b.w.] as a preclinical model of acute alcohol injury. Controls received a single dose of PBS. Mice were sacrificed 8 h later. In parallel, HepaRGs and Caco-2 cells, human cell lines of differentiated hepatocytes and intestinal epithelial cells intestinal epithelial cells (IECs), respectively, were challenged in the presence or absence of EtOH [0-100 mM]. Extracellular vesicles (EVs) isolated by ultracentrifugation from culture media of IECs were added to hepatocyte cell cultures. Increased intestinal permeability, loss of zonula occludens-1 (ZO-1) and MUCIN-2 expression, and alterations in microbiota-increased Lactobacillus and decreased Lachnospiraceae species-were found in the large intestine of mice exposed to EtOH. Increased TUNEL-positive cells, infiltration of CD11b-positive immune cells, pro-inflammatory cytokines (e.g., tlr4, tnf, il1β), and markers of lipid accumulation (Oil Red O, srbep1) were evident in livers of mice exposed to EtOH, particularly in females. In vitro experiments indicated that EVs released by IECs in response to ethanol exerted a deleterious effect on hepatocyte viability and lipid accumulation. Overall, our data identified a novel mechanism responsible for driving hepatic injury in the gut-liver axis, opening novel avenues for therapy.
Collapse
Affiliation(s)
- Arantza Lamas-Paz
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Laura Morán
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
- Servicio de Aparato Digestivo del Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Jin Peng
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Beatriz Salinas
- Servicio de Aparato Digestivo del Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- Bioengineering and Aerospace Engineering Department, Universidad Carlos III de Madrid, Madrid, Spain
- Centro de Investigación Biomédico en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Nuria López-Alcántara
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
| | - Svenja Sydor
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, Bochum, Germany
| | - Ramiro Vilchez-Vargas
- Department of Gastroenterology, Hepatology, and Infectious Diseases, Otto von Guericke University Hospital Magdeburg, Magdeburg, Germany
| | - Iris Asensio
- Servicio de Aparato Digestivo del Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centre for Biomedical Research, Network on Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
| | - Fengjie Hao
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
- Department of General Surgery, Hepatobiliary Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kang Zheng
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Beatriz Martín-Adrados
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Laura Moreno
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, Bochum, Germany
- Department of Pharmacology and Toxicology, Complutense University School of Medicine and Centre for Biomedical Research, Network on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Angel Cogolludo
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, Bochum, Germany
- Department of Pharmacology and Toxicology, Complutense University School of Medicine and Centre for Biomedical Research, Network on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Manuel Gómez del Moral
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
- Department of Cell Biology, Complutense University School of Medicine, Madrid, Spain
| | - Lars Bechmann
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, Bochum, Germany
| | - Eduardo Martínez-Naves
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Javier Vaquero
- Servicio de Aparato Digestivo del Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centre for Biomedical Research, Network on Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
| | - Rafael Bañares
- Servicio de Aparato Digestivo del Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centre for Biomedical Research, Network on Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
| | - Yulia A. Nevzorova
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| |
Collapse
|
80
|
Immunological mechanisms and therapeutic targets of fatty liver diseases. Cell Mol Immunol 2020; 18:73-91. [PMID: 33268887 PMCID: PMC7852578 DOI: 10.1038/s41423-020-00579-3] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023] Open
Abstract
Alcoholic liver disease (ALD) and nonalcoholic fatty liver disease (NAFLD) are the two major types of chronic liver disease worldwide. Inflammatory processes play key roles in the pathogeneses of fatty liver diseases, and continuous inflammation promotes the progression of alcoholic steatohepatitis (ASH) and nonalcoholic steatohepatitis (NASH). Although both ALD and NAFLD are closely related to inflammation, their respective developmental mechanisms differ to some extent. Here, we review the roles of multiple immunological mechanisms and therapeutic targets related to the inflammation associated with fatty liver diseases and the differences in the progression of ASH and NASH. Multiple cell types in the liver, including macrophages, neutrophils, other immune cell types and hepatocytes, are involved in fatty liver disease inflammation. In addition, microRNAs (miRNAs), extracellular vesicles (EVs), and complement also contribute to the inflammatory process, as does intertissue crosstalk between the liver and the intestine, adipose tissue, and the nervous system. We point out that inflammation also plays important roles in promoting liver repair and controlling bacterial infections. Understanding the complex regulatory process of disrupted homeostasis during the development of fatty liver diseases may lead to the development of improved targeted therapeutic intervention strategies.
Collapse
|
81
|
Microbiota reprogramming for treatment of alcohol-related liver disease. Transl Res 2020; 226:26-38. [PMID: 32687975 PMCID: PMC7572584 DOI: 10.1016/j.trsl.2020.07.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/01/2020] [Accepted: 07/13/2020] [Indexed: 02/08/2023]
Abstract
In the past decade knowledge has expanded regarding the importance of the gut microbiota in maintaining intestinal homeostasis and overall health. During this same time, we have also gained appreciation for the role of the gut-liver axis in the development of liver diseases. Alcohol overconsumption is one of the leading causes of liver failure globally. However, not all people with alcohol use disorder progress to advanced stages of liver disease. With advances in technology to investigate the gut microbiome and metabolome, we are now beginning to delineate alcohol's effects on the gut microbiome in relation to liver disease. This review presents our current understanding on the role of the gut microbiota during alcohol exposure, and various therapeutic attempts that have been made to reprogram the gut microbiota with the goal of alleviating alcoholic-related liver disease.
Collapse
|
82
|
Jiang XW, Li YT, Ye JZ, Lv LX, Yang LY, Bian XY, Wu WR, Wu JJ, Shi D, Wang Q, Fang DQ, Wang KC, Wang QQ, Lu YM, Xie JJ, Li LJ. New strain of Pediococcus pentosaceus alleviates ethanol-induced liver injury by modulating the gut microbiota and short-chain fatty acid metabolism. World J Gastroenterol 2020; 26:6224-6240. [PMID: 33177795 PMCID: PMC7596634 DOI: 10.3748/wjg.v26.i40.6224] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/08/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Intestinal dysbiosis has been shown to be associated with the pathogenesis of alcoholic liver disease (ALD), which includes changes in the microbiota composition and bacterial overgrowth, but an effective microbe-based therapy is lacking. Pediococcus pentosaceus (P. pentosaceus) CGMCC 7049 is a newly isolated strain of probiotic that has been shown to be resistant to ethanol and bile salts. However, further studies are needed to determine whether P. pentosaceus exerts a protective effect on ALD and to elucidate the potential mechanism.
AIM To evaluate the protective effect of the probiotic P. pentosaceus on ethanol-induced liver injury in mice.
METHODS A new ethanol-resistant strain of P. pentosaceus CGMCC 7049 was isolated from healthy adults in our laboratory. The chronic plus binge model of experimental ALD was established to evaluate the protective effects. Twenty-eight C57BL/6 mice were randomly divided into three groups: The control group received a pair-fed control diet and oral gavage with sterile phosphate buffered saline, the EtOH group received a ten-day Lieber-DeCarli diet containing 5% ethanol and oral gavage with phosphate buffered saline, and the P. pentosaceus group received a 5% ethanol Lieber-DeCarli diet but was treated with P. pentosaceus. One dose of isocaloric maltose dextrin or ethanol was administered by oral gavage on day 11, and the mice were sacrificed nine hours later. Blood and tissue samples (liver and gut) were harvested to evaluate gut barrier function and liver injury-related parameters. Fresh cecal contents were collected, gas chromatography–mass spectrometry was used to measure short-chain fatty acid (SCFA) concentrations, and the microbiota composition was analyzed using 16S rRNA gene sequencing.
RESULTS The P. pentosaceus treatment improved ethanol-induced liver injury, with lower alanine aminotransferase, aspartate transaminase and triglyceride levels and decreased neutrophil infiltration. These changes were accompanied by decreased levels of endotoxin and inflammatory cytokines, including interleukin-5, tumor necrosis factor-α, granulocyte colony-stimulating factor, keratinocyte-derived protein chemokine, macrophage inflammatory protein-1α and monocyte chemoattractant protein-1. Ethanol feeding resulted in intestinal dysbiosis and gut barrier disruption, increased relative abundance of potentially pathogenic Escherichia and Staphylococcus, and the depletion of SCFA-producing bacteria, such as Prevotella, Faecalibacterium, and Clostridium. In contrast, P. pentosaceus administration increased the microbial diversity, restored the relative abundance of Lactobacillus, Pediococcus, Prevotella, Clostridium and Akkermansia and increased propionic acid and butyric acid production by modifying SCFA-producing bacteria. Furthermore, the levels of the tight junction protein ZO-1, mucin proteins (mucin [MUC]-1, MUC-2 and MUC-4) and the antimicrobial peptide Reg3β were increased after probiotic supplementation.
CONCLUSION Based on these results, the new strain of P. pentosaceus alleviated ethanol-induced liver injury by reversing gut microbiota dysbiosis, regulating intestinal SCFA metabolism, improving intestinal barrier function, and reducing circulating levels of endotoxin and proinflammatory cytokines and chemokines. Thus, this strain is a potential probiotic treatment for ALD.
Collapse
Affiliation(s)
- Xian-Wan Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Ya-Ting Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Jian-Zhong Ye
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Long-Xian Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Li-Ya Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Xiao-Yuan Bian
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Wen-Rui Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Jing-Jing Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Ding Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Qing Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Dai-Qiong Fang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Kai-Cen Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Qiang-Qiang Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Yan-Meng Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Jiao-Jiao Xie
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Lan-Juan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
83
|
Guo W, Liu J, Yang Y, Ma H, Gong Q, Kan X, Ran X, Cao Y, Wang J, Fu S, Hu G. Rumen-bypassed tributyrin alleviates heat stress by reducing the inflammatory responses of immune cells. Poult Sci 2020; 100:348-356. [PMID: 33357699 PMCID: PMC7772712 DOI: 10.1016/j.psj.2020.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/06/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022] Open
Abstract
Heat stress (HS) in summer will seriously affect the health and performance of dairy cows. To alleviate the injury to dairy cows caused by HS, we added the rumen-bypassed tributyrin to the feed. We determined whether cows were in a heat-stressed environment by testing the temperature humidity index in the morning, at noon, and in the evening. The detection of anal temperature and respiratory frequency further proved the HS state of the dairy cows. The quantificational real time PCR results showed that tributyrin could significantly reduce the relative expression of tumor necrosis factor α, interleukin 1β, and Interleukin 6. Western blot results showed that tributyrin could alleviate the lymphocyte inflammatory response by inhibiting the mitogen-activated protein kinase and nuclear factor-кB signaling pathways. To further detect the effect of tributyrin on HS in dairy cows, routine biochemical and blood tests were carried out. The results showed that the contents of aspartate aminotransferase, total bilirubin, creatinine, albumin, and globulin were significantly reduced by tributyrin. The results showed that tributyrin could significantly alleviate the liver and kidney injury induced by heat stress in dairy cows. Moreover, tributyrin could also significantly reduce the numbers of intermediate cells and increase the level of hemoglobin. Tributyrin could also improve the performance of dairy cows. These results suggested that tributylglycerol may have a positive effect on breast health of dairy cows. In conclusion, these results indicated that tributyrin could relieve HS and increase the production performance of dairy cows by reducing the inflammatory responses of lymphocytes.
Collapse
Affiliation(s)
- Wenjin Guo
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Juxiong Liu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuanxi Yang
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China
| | - He Ma
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Qian Gong
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xingchi Kan
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xin Ran
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yu Cao
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jianfa Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163000, China
| | - Shoupeng Fu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Guiqiu Hu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China
| |
Collapse
|
84
|
Dvorak Z, Klapholz M, Burris TP, Willing BP, Gioiello A, Pellicciari R, Galli F, March J, O'Keefe SJ, Sartor RB, Kim CH, Levy M, Mani S. Weak Microbial Metabolites: a Treasure Trove for Using Biomimicry to Discover and Optimize Drugs. Mol Pharmacol 2020; 98:343-349. [PMID: 32764096 PMCID: PMC7485585 DOI: 10.1124/molpharm.120.000035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022] Open
Abstract
For decades, traditional drug discovery has used natural product and synthetic chemistry approaches to generate libraries of compounds, with some ending as promising drug candidates. A complementary approach has been to adopt the concept of biomimicry of natural products and metabolites so as to improve multiple drug-like features of the parent molecule. In this effort, promiscuous and weak interactions between ligands and receptors are often ignored in a drug discovery process. In this Emerging Concepts article, we highlight microbial metabolite mimicry, whereby parent metabolites have weak interactions with their receptors that then have led to discrete examples of more potent and effective drug-like molecules. We show specific examples of parent-metabolite mimics with potent effects in vitro and in vivo. Furthermore, we show examples of emerging microbial ligand-receptor interactions and provide a context in which these ligands could be improved as potential drugs. A balanced conceptual advance is provided in which we also acknowledge potential pitfalls-hyperstimulation of finely balanced receptor-ligand interactions could also be detrimental. However, with balance, we provide examples of where this emerging concept needs to be tested. SIGNIFICANCE STATEMENT: Microbial metabolite mimicry is a novel way to expand on the chemical repertoire of future drugs. The emerging concept is now explained using specific examples of the discovery of therapeutic leads from microbial metabolites.
Collapse
Affiliation(s)
- Zdenek Dvorak
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Max Klapholz
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Thomas P Burris
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Benjamin P Willing
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Antimo Gioiello
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Roberto Pellicciari
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Francesco Galli
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - John March
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Stephen J O'Keefe
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - R Balfour Sartor
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Chang H Kim
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Maayan Levy
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Sridhar Mani
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| |
Collapse
|
85
|
Eom T, Ko G, Kim KC, Kim JS, Unno T. Dendropanax morbifera Leaf Extracts Improved Alcohol Liver Injury in Association with Changes in the Gut Microbiota of Rats. Antioxidants (Basel) 2020; 9:antiox9100911. [PMID: 32987739 PMCID: PMC7598590 DOI: 10.3390/antiox9100911] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022] Open
Abstract
This study evaluated the protective effects of Dendropanax morbifera leaf (DML) extracts in the liver due to excessive ethanol consumption. Our results showed that the ethanol extract had better antioxidant activity than the water extract, likely due to the higher levels of total flavonoid and phenolic compounds in the former. We found that the main phenolic acid was chlorogenic acid and the major flavonoid was rutin. Results from the animal model experiment showed concentration-dependent liver protection with the distilled water extract showing better liver protection than the ethanol extract. Gut microbiota dysbiosis induced by alcohol consumption was significantly shifted by DML extracts through increasing mainly Bacteroides and Allobaculum. Moreover, predicted metabolic activities of biosynthesis of beneficial monounsaturated fatty acids such as oleate and palmitoleate were enhanced. Our results suggest that these hepatoprotective effects are likely due to the increased activities of antioxidant enzymes and partially promoted by intestinal microbiota shifts.
Collapse
Affiliation(s)
- Taekil Eom
- Subtropical/Tropical Organism Gene Bank, SARI, Jeju National University, Jeju 63243, Korea;
| | - Gwangpyo Ko
- Faculty of Biotechnology, College of Agriculture & Life Sciences, SARI, Jeju National University, Jeju 63243, Korea;
| | - Kyeoung Cheol Kim
- Majors in Plant Resource and Environment, College of Agriculture & Life Sciences, SARI, Jeju National University, Jeju 63243, Korea; (K.C.K.); (J.-S.K.)
| | - Ju-Sung Kim
- Majors in Plant Resource and Environment, College of Agriculture & Life Sciences, SARI, Jeju National University, Jeju 63243, Korea; (K.C.K.); (J.-S.K.)
| | - Tatsuya Unno
- Subtropical/Tropical Organism Gene Bank, SARI, Jeju National University, Jeju 63243, Korea;
- Faculty of Biotechnology, College of Agriculture & Life Sciences, SARI, Jeju National University, Jeju 63243, Korea;
- Correspondence: ; Tel.: +82-64-754-3354; Fax: +82-64-756-3351
| |
Collapse
|
86
|
Sato FT, Yap YA, Crisma AR, Portovedo M, Murata GM, Hirabara SM, Ribeiro WR, Marcantonio Ferreira C, Cruz MM, Pereira JNB, Payolla TB, Guima SES, Thomas AM, Setubal JC, Alonso-Vale MIC, Santos MF, Curi R, Marino E, Vinolo MAR. Tributyrin Attenuates Metabolic and Inflammatory Changes Associated with Obesity through a GPR109A-Dependent Mechanism. Cells 2020; 9:E2007. [PMID: 32882837 PMCID: PMC7563536 DOI: 10.3390/cells9092007] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022] Open
Abstract
Obesity is linked with altered microbial short-chain fatty acids (SCFAs), which are a signature of gut dysbiosis and inflammation. In the present study, we investigated whether tributyrin, a prodrug of the SCFA butyrate, could improve metabolic and inflammatory profiles in diet-induced obese mice. Mice fed a high-fat diet for eight weeks were treated with tributyrin or placebo for another six weeks. We show that obese mice treated with tributyrin had lower body weight gain and an improved insulin responsiveness and glucose metabolism, partly via reduced hepatic triglycerides content. Additionally, tributyrin induced an anti-inflammatory state in the adipose tissue by reduction of Il-1β and Tnf-a and increased Il-10, Tregs cells and M2-macrophages. Moreover, improvement in glucose metabolism and reduction of fat inflammatory states associated with tributyrin treatment were dependent on GPR109A activation. Our results indicate that exogenous targeting of SCFA butyrate attenuates metabolic and inflammatory dysfunction, highlighting a potentially novel approach to tackle obesity.
Collapse
Affiliation(s)
- Fabio Takeo Sato
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas 13083007, Brazil; (F.T.S.); (M.P.)
| | - Yu Anne Yap
- Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia;
| | - Amanda Rabello Crisma
- Department of Clinical Analyses, Federal University of Paraná, Curitiba 80060000, Brazil;
| | - Mariana Portovedo
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas 13083007, Brazil; (F.T.S.); (M.P.)
| | - Gilson Masahiro Murata
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, Brazil; (G.M.M.); (T.B.P.); (R.C.)
| | - Sandro Massao Hirabara
- Interdisciplinary Postgraduate Program in Health Science, Cruzeiro do Sul University, São Paulo 01506000, Brazil; (S.M.H.); (J.N.B.P.)
| | - Willian Rodrigues Ribeiro
- Department of Pharmaceutics Sciences, Institute of Environmental Chemistry and Pharmaceutical Sciences, Federal University of São Paulo, Diadema 09972270, Brazil; (W.R.R.); (C.M.F.)
| | - Caroline Marcantonio Ferreira
- Department of Pharmaceutics Sciences, Institute of Environmental Chemistry and Pharmaceutical Sciences, Federal University of São Paulo, Diadema 09972270, Brazil; (W.R.R.); (C.M.F.)
| | - Maysa Mariana Cruz
- Department of Biological Sciences, Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema 09972270, Brazil; (M.M.C.); (M.I.C.A.-V.)
| | - Joice Naiara Bertaglia Pereira
- Interdisciplinary Postgraduate Program in Health Science, Cruzeiro do Sul University, São Paulo 01506000, Brazil; (S.M.H.); (J.N.B.P.)
| | - Tanyara Baliani Payolla
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, Brazil; (G.M.M.); (T.B.P.); (R.C.)
| | - Suzana Eiko Sato Guima
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508000, Brazil; (S.E.S.G.); (A.M.T.); (J.C.S.)
| | - Andrew Maltez Thomas
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508000, Brazil; (S.E.S.G.); (A.M.T.); (J.C.S.)
| | - João Carlos Setubal
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508000, Brazil; (S.E.S.G.); (A.M.T.); (J.C.S.)
| | - Maria Isabel Cardoso Alonso-Vale
- Department of Biological Sciences, Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema 09972270, Brazil; (M.M.C.); (M.I.C.A.-V.)
| | - Marinilce Fagundes Santos
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, Brazil;
| | - Rui Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, Brazil; (G.M.M.); (T.B.P.); (R.C.)
- Interdisciplinary Postgraduate Program in Health Science, Cruzeiro do Sul University, São Paulo 01506000, Brazil; (S.M.H.); (J.N.B.P.)
- Butantan Institute, São Paulo 05503900, Brazil
| | - Eliana Marino
- Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia;
| | - Marco A. R. Vinolo
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas 13083007, Brazil; (F.T.S.); (M.P.)
- Experimental Medicine Research Cluster (EMRC), University of Campinas (UNICAMP), Campinas 13083007, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP), Campinas 13083864, Brazil
| |
Collapse
|
87
|
Di Ciaula A, Baj J, Garruti G, Celano G, De Angelis M, Wang HH, Di Palo DM, Bonfrate L, Wang DQH, Portincasa P. Liver Steatosis, Gut-Liver Axis, Microbiome and Environmental Factors. A Never-Ending Bidirectional Cross-Talk. J Clin Med 2020; 9:2648. [PMID: 32823983 PMCID: PMC7465294 DOI: 10.3390/jcm9082648] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/07/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023] Open
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) is increasing worldwide and parallels comorbidities such as obesity, metabolic syndrome, dyslipidemia, and diabetes. Recent studies describe the presence of NAFLD in non-obese individuals, with mechanisms partially independent from excessive caloric intake. Increasing evidences, in particular, point towards a close interaction between dietary and environmental factors (including food contaminants), gut, blood flow, and liver metabolism, with pathways involving intestinal permeability, the composition of gut microbiota, bacterial products, immunity, local, and systemic inflammation. These factors play a critical role in the maintenance of intestinal, liver, and metabolic homeostasis. An anomalous or imbalanced gut microbial composition may favor an increased intestinal permeability, predisposing to portal translocation of microorganisms, microbial products, and cell wall components. These components form microbial-associated molecular patterns (MAMPs) or pathogen-associated molecular patterns (PAMPs), with potentials to interact in the intestine lamina propria enriched in immune cells, and in the liver at the level of the immune cells, i.e., Kupffer cells and stellate cells. The resulting inflammatory environment ultimately leads to liver fibrosis with potentials to progression towards necrotic and fibrotic changes, cirrhosis. and hepatocellular carcinoma. By contrast, measures able to modulate the composition of gut microbiota and to preserve gut vascular barrier might prevent or reverse NAFLD.
Collapse
Affiliation(s)
- Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (L.B.)
| | - Jacek Baj
- Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Gabriella Garruti
- Section of Endocrinology, Department of Emergency and Organ Transplantations, University of Bari “Aldo Moro” Medical School, Piazza G. Cesare 11, 70124 Bari, Italy;
| | - Giuseppe Celano
- Dipartimento di Scienze del Suolo, della Pianta e Degli Alimenti, Università degli Studi di Bari Aldo Moro, 70124 Bari, Italy; (G.C.); (M.D.A.)
| | - Maria De Angelis
- Dipartimento di Scienze del Suolo, della Pianta e Degli Alimenti, Università degli Studi di Bari Aldo Moro, 70124 Bari, Italy; (G.C.); (M.D.A.)
| | - Helen H. Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (H.H.W.); (D.Q.-H.W.)
| | - Domenica Maria Di Palo
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (L.B.)
- Dipartimento di Scienze del Suolo, della Pianta e Degli Alimenti, Università degli Studi di Bari Aldo Moro, 70124 Bari, Italy; (G.C.); (M.D.A.)
| | - Leonilde Bonfrate
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (L.B.)
| | - David Q-H Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (H.H.W.); (D.Q.-H.W.)
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (L.B.)
| |
Collapse
|
88
|
Gao B, Emami A, Nath S, Schnabl B. Microbial Products and Metabolites Contributing to Alcohol-Related Liver Disease. Mol Nutr Food Res 2020; 65:e2000023. [PMID: 32583604 DOI: 10.1002/mnfr.202000023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/01/2020] [Indexed: 02/06/2023]
Abstract
As a serious public health concern, alcohol-related liver disease is associated with dysregulations in the intestinal barrier function and the gut microbiota. The liver and gut communicate via the gut-liver axis, through which microbial products and metabolites translocate to the liver. Here, the current knowledge of various microbial products and metabolites which contribute to the alcohol-related liver diseases, including bile acids, indole-3-acetic acid, butyrate, long-chain fatty acids, endotoxin, cytolysin, β-glucan, and candidalysin is reviewed. Some of these might serve as therapeutic targets for alcohol-related liver disease.
Collapse
Affiliation(s)
- Bei Gao
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Atoosa Emami
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Shilpa Nath
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.,Department of Medicine, VA San Diego Healthcare System, San Diego, CA, 92093, USA
| |
Collapse
|
89
|
Swanson GR, Siskin J, Gorenz A, Shaikh M, Raeisi S, Fogg L, Forsyth C, Keshavarzian A. Disrupted diurnal oscillation of gut-derived Short chain fatty acids in shift workers drinking alcohol: Possible mechanism for loss of resiliency of intestinal barrier in disrupted circadian host. Transl Res 2020; 221:97-109. [PMID: 32376406 PMCID: PMC8136245 DOI: 10.1016/j.trsl.2020.04.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 03/30/2020] [Accepted: 04/07/2020] [Indexed: 02/08/2023]
Abstract
Microbiota derived short chain fatty acids (SCFAs) are produced by fermentation of nondigestible fiber, and are a key component in intestinal barrier homeostasis. Since the microbiome has diurnal fluctuations, we hypothesized that SCFAs in humans have a diurnal rhythm and their rhythmicity would be impacted by the host central circadian misalignment (night shift work) which would make intestinal barrier more susceptible to disruption by alcohol. To test this hypothesis, we studied 3 groups of subjects: patients with alcohol use disorder, but no liver disease (AD), healthy day workers (DW), and night workers (NW). All subjects were studied at baseline and then in DW and NW subjects after moderate daily alcohol (0.5 g/kg) for 7 days. Gut-derived plasma SCFAs showed a significant circadian oscillation by cosinor analysis in DW; however, SCFA in the AD and NW subjects lost 24-hour rhythmicity. Decrease in SCFA correlated with increased colonic permeability. Both chronic and moderate alcohol consumption for 1 week caused circadian disruption based on wrist actigraphy and urinary melatonin. Our study shows that (1) gut-derived plasma SCFAs have a diurnal rhythm in humans that is impacted by the central clock of the host; (2) moderate alcohol suppresses SCFAs which was associated with increased colonic permeability; and (3) less invasive urinary 6-SM correlated and rest-activity actigraphy correlated with plasma melatonin. Future studies are needed to examine the role circadian misalignment on gut derived SCFAs as possible mechanism for loss of intestinal barrier resiliency to injurious agents like alcohol.
Collapse
Affiliation(s)
- Garth R Swanson
- Department Digestive Diseases, Rush University Medical Center, Chicago, Illinois.
| | - Joel Siskin
- Department Digestive Diseases, Rush University Medical Center, Chicago, Illinois
| | - Annika Gorenz
- Department Digestive Diseases, Rush University Medical Center, Chicago, Illinois
| | - Maliha Shaikh
- Department Digestive Diseases, Rush University Medical Center, Chicago, Illinois
| | - Shohreh Raeisi
- Department Digestive Diseases, Rush University Medical Center, Chicago, Illinois
| | - Louis Fogg
- Community, Systems and Mental Health Nursing, Rush University, Chicago, Illinois
| | - Christopher Forsyth
- Department Digestive Diseases, Rush University Medical Center, Chicago, Illinois
| | - Ali Keshavarzian
- Department Digestive Diseases, Rush University Medical Center, Chicago, Illinois; Departments of Pharmacology; Molecular Biophysics & Physiology, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
90
|
Wu Q, Chen T, El-Nezami H, Savidge TC. Food ingredients in human health: Ecological and metabolic perspectives implicating gut microbiota function. Trends Food Sci Technol 2020. [DOI: 10.1016/j.tifs.2020.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
91
|
A Role for Gut Microbiome Fermentative Pathways in Fatty Liver Disease Progression. J Clin Med 2020; 9:jcm9051369. [PMID: 32392712 PMCID: PMC7291163 DOI: 10.3390/jcm9051369] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/24/2020] [Accepted: 05/03/2020] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multifactorial disease in which environmental and genetic factors are involved. Although the molecular mechanisms involved in NAFLD onset and progression are not completely understood, the gut microbiome (GM) is thought to play a key role in the process, influencing multiple physiological functions. GM alterations in diversity and composition directly impact disease states with an inflammatory course, such as non-alcoholic steatohepatitis (NASH). However, how the GM influences liver disease susceptibility is largely unknown. Similarly, the impact of strategies targeting the GM for the treatment of NASH remains to be evaluated. This review provides a broad insight into the role of gut microbiota in NASH pathogenesis, as a diagnostic tool, and as a therapeutic target in this liver disease. We highlight the idea that the balance in metabolic fermentations can be key in maintaining liver homeostasis. We propose that an overabundance of alcohol-fermentation pathways in the GM may outcompete healthier, acid-producing members of the microbiota. In this way, GM ecology may precipitate a self-sustaining vicious cycle, boosting liver disease progression.
Collapse
|
92
|
Tributyrin differentially regulates inflammatory markers and modulates goblet cells number along the intestinal tract segments of weaning pigs. Livest Sci 2020. [DOI: 10.1016/j.livsci.2020.103996] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
93
|
Avila MA, Dufour JF, Gerbes AL, Zoulim F, Bataller R, Burra P, Cortez-Pinto H, Gao B, Gilmore I, Mathurin P, Moreno C, Poznyak V, Schnabl B, Szabo G, Thiele M, Thursz MR. Recent advances in alcohol-related liver disease (ALD): summary of a Gut round table meeting. Gut 2020; 69:764-780. [PMID: 31879281 PMCID: PMC7236084 DOI: 10.1136/gutjnl-2019-319720] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 11/28/2019] [Accepted: 12/06/2019] [Indexed: 12/14/2022]
Abstract
Alcohol-related liver disease (ALD), which includes a range of disorders of different severity and is one of the most prevalent types of liver disease worldwide, has recently regained increased attention. Among other reasons, the realisation that any alcohol intake, regardless of type of beverage represents a health risk, and the new therapeutic strategies tested in recently published or undergoing clinical trials spur scientific interest in this area.In April 2019, Gut convened a round table panel of experts during the European Association for the Study of the Liver International Liver Congress in Vienna to discuss critical and up-to-date issues and clinical trial data regarding ALD, its epidemiology, diagnosis, management, pathomechanisms, possible future treatments and prevention. This paper summarises the discussion and its conclusions.
Collapse
Affiliation(s)
- Matias A Avila
- Hepatology, CIBERehd, IdiSNA, CIMA, University of Navarra, Pamplona, Spain
| | - Jean-François Dufour
- Hepatology, Department of Clinical Research and University Clinic for Visceral Surgery and Medicine, Inselspital, University of Bern, Bern, Switzerland
| | - Alexander L Gerbes
- Liver Centre Munich, Department of Medicine II, University Hospital, LMU Munich, Munich, Germany
| | - Fabien Zoulim
- Hepatology Department, INSERM U1052, Hospices Civils de Lyon, Cancer Research Centerl of Lyon, University of Lyon, Lyon, France
| | - Ramon Bataller
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Patrizia Burra
- Multivisceral Transplant Unit, Gastroenterology, Department of Surgery, Oncology and Gastroenterology, Padua University Hospital, Padua, Italy
| | - Helena Cortez-Pinto
- Departamento de Gastroenterologia, CHLN, Laboratorio de Nutriçao, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Ian Gilmore
- Liverpool Centre for Alcohol Research, University of Liverpool, Liverpool, UK
| | - Philippe Mathurin
- Service des Maladies de l'Appareil Digestif, INSERM U795, Hôpital Huriez, Lille, France
| | - Christophe Moreno
- Service de Gastroentérologie, Hépatopancréatologie et Oncologie Digestive, CUB Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Vladimir Poznyak
- Department of Mental Health and Substance Abuse, World Health Organization, Geneve, Switzerland
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Gyongyi Szabo
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Maja Thiele
- Department of Gastroenterology and Hepatology, and Department of Clinical Research, Odense University Hospital and University of Southern Denmark, Odense, Denmark
| | - Mark R Thursz
- Department of Metabolism, Faculty of Medicine, Imperial College, London, UK
| |
Collapse
|
94
|
Neuman MG, Seitz HK, French SW, Malnick S, Tsukamoto H, Cohen LB, Hoffman P, Tabakoff B, Fasullo M, Nagy LE, Tuma PL, Schnabl B, Mueller S, Groebner JL, Barbara FA, Yue J, Nikko A, Alejandro M, Brittany T, Edward V, Harrall K, Saba L, Mihai O. Alcoholic-Hepatitis, Links to Brain and Microbiome: Mechanisms, Clinical and Experimental Research. Biomedicines 2020; 8:63. [PMID: 32197424 PMCID: PMC7148515 DOI: 10.3390/biomedicines8030063] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/02/2020] [Accepted: 03/09/2020] [Indexed: 02/07/2023] Open
Abstract
The following review article presents clinical and experimental features of alcohol-induced liver disease (ALD). Basic aspects of alcohol metabolism leading to the development of liver hepatotoxicity are discussed. ALD includes fatty liver, acute alcoholic hepatitis with or without liver failure, alcoholic steatohepatitis (ASH) leading to fibrosis and cirrhosis, and hepatocellular cancer (HCC). ALD is fully attributable to alcohol consumption. However, only 10-20% of heavy drinkers (persons consuming more than 40 g of ethanol/day) develop clinical ALD. Moreover, there is a link between behaviour and environmental factors that determine the amount of alcohol misuse and their liver disease. The range of clinical presentation varies from reversible alcoholic hepatic steatosis to cirrhosis, hepatic failure, and hepatocellular carcinoma. We aimed to (1) describe the clinico-pathology of ALD, (2) examine the role of immune responses in the development of alcoholic hepatitis (ASH), (3) propose diagnostic markers of ASH, (4) analyze the experimental models of ALD, (5) study the role of alcohol in changing the microbiota, and (6) articulate how findings in the liver and/or intestine influence the brain (and/or vice versa) on ASH; (7) identify pathways in alcohol-induced organ damage and (8) to target new innovative experimental concepts modeling the experimental approaches. The present review includes evidence recognizing the key toxic role of alcohol in ALD severity. Cytochrome p450 CYP2E1 activation may change the severity of ASH. The microbiota is a key element in immune responses, being an inducer of proinflammatory T helper 17 cells and regulatory T cells in the intestine. Alcohol consumption changes the intestinal microbiota and influences liver steatosis and liver inflammation. Knowing how to exploit the microbiome to modulate the immune system might lead to a new form of personalized medicine in ALF and ASH.
Collapse
Affiliation(s)
- Manuela G. Neuman
- In Vitro Drug Safety and Biotechnology, Toronto, ON M5G 1L5, Canada;
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON M5G 1L5, Canada
| | - Helmut Karl Seitz
- Department of Medicine, Centre of Alcohol Research, University of Heidelberg, Salem Medical Centre, 337374 Heidelberg, Germany; (H.K.S.); (S.M.)
| | - Samuel W. French
- Department of Pathology, Harbor-UCLA Medical Center and Los Angeles BioMedical Institute, Torrance, CA Harbor-UCLA Medical Center, Torrance, CA 90509, USA; (S.W.F.); (F.A.B.); (J.Y.); (A.N.); (M.A.); (T.B.); (V.E.)
| | - Stephen Malnick
- Department Internal Medicine C, Kaplan Medical Centre and Hebrew University of Jerusalem, Rehovot 76100, Israel;
| | - Heidekazu Tsukamoto
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089-5311, USA;
- Department of Veterans; Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Lawrence B. Cohen
- Division of Gastroenterology, Sunnybrook Health Sciences Centre, Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON M4N 3M5, Canada;
| | - Paula Hoffman
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045-0511, USA; (P.H.); (B.T.); (K.H.); (L.S.)
| | - Boris Tabakoff
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045-0511, USA; (P.H.); (B.T.); (K.H.); (L.S.)
| | - Michael Fasullo
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12205, USA;
| | - Laura E. Nagy
- Departments of Pathobiology and Gastroenterology, Center for Liver Disease Research, Cleveland Clinic Foundation, Cleveland, OH 44195, USA;
| | - Pamela L. Tuma
- Department of Biology, The Catholic University of America, Washington, DC 20064, USA; (P.L.T.); (J.L.G.)
| | - Bernd Schnabl
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA;
| | - Sebastian Mueller
- Department of Medicine, Centre of Alcohol Research, University of Heidelberg, Salem Medical Centre, 337374 Heidelberg, Germany; (H.K.S.); (S.M.)
| | - Jennifer L. Groebner
- Department of Biology, The Catholic University of America, Washington, DC 20064, USA; (P.L.T.); (J.L.G.)
| | - French A. Barbara
- Department of Pathology, Harbor-UCLA Medical Center and Los Angeles BioMedical Institute, Torrance, CA Harbor-UCLA Medical Center, Torrance, CA 90509, USA; (S.W.F.); (F.A.B.); (J.Y.); (A.N.); (M.A.); (T.B.); (V.E.)
| | - Jia Yue
- Department of Pathology, Harbor-UCLA Medical Center and Los Angeles BioMedical Institute, Torrance, CA Harbor-UCLA Medical Center, Torrance, CA 90509, USA; (S.W.F.); (F.A.B.); (J.Y.); (A.N.); (M.A.); (T.B.); (V.E.)
| | - Afifiyan Nikko
- Department of Pathology, Harbor-UCLA Medical Center and Los Angeles BioMedical Institute, Torrance, CA Harbor-UCLA Medical Center, Torrance, CA 90509, USA; (S.W.F.); (F.A.B.); (J.Y.); (A.N.); (M.A.); (T.B.); (V.E.)
| | - Mendoza Alejandro
- Department of Pathology, Harbor-UCLA Medical Center and Los Angeles BioMedical Institute, Torrance, CA Harbor-UCLA Medical Center, Torrance, CA 90509, USA; (S.W.F.); (F.A.B.); (J.Y.); (A.N.); (M.A.); (T.B.); (V.E.)
| | - Tillman Brittany
- Department of Pathology, Harbor-UCLA Medical Center and Los Angeles BioMedical Institute, Torrance, CA Harbor-UCLA Medical Center, Torrance, CA 90509, USA; (S.W.F.); (F.A.B.); (J.Y.); (A.N.); (M.A.); (T.B.); (V.E.)
| | - Vitocruz Edward
- Department of Pathology, Harbor-UCLA Medical Center and Los Angeles BioMedical Institute, Torrance, CA Harbor-UCLA Medical Center, Torrance, CA 90509, USA; (S.W.F.); (F.A.B.); (J.Y.); (A.N.); (M.A.); (T.B.); (V.E.)
| | - Kylie Harrall
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045-0511, USA; (P.H.); (B.T.); (K.H.); (L.S.)
| | - Laura Saba
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045-0511, USA; (P.H.); (B.T.); (K.H.); (L.S.)
| | - Opris Mihai
- In Vitro Drug Safety and Biotechnology, Toronto, ON M5G 1L5, Canada;
- Department Family Medicine Clinic CAR, 010164 Bucharest, Romania
| |
Collapse
|
95
|
Mandrekar P. Targeting Epigenetic Mechanisms to Alleviate Alcoholic Steatosis. Cell Mol Gastroenterol Hepatol 2020; 9:713-714. [PMID: 32097606 PMCID: PMC7212487 DOI: 10.1016/j.jcmgh.2020.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 01/28/2020] [Accepted: 01/30/2020] [Indexed: 01/01/2023]
Affiliation(s)
- Pranoti Mandrekar
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts.
| |
Collapse
|
96
|
Cresci GAM, Lampe JW, Gibson G. Targeted Approaches for In Situ Gut Microbiome Manipulation. JPEN J Parenter Enteral Nutr 2020; 44:581-588. [PMID: 32027044 PMCID: PMC9291485 DOI: 10.1002/jpen.1779] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/17/2019] [Indexed: 12/16/2022]
Abstract
The 2019 Dudrick Research Symposium, entitled "Targeted Approaches for In Situ Gut Microbiome Manipulation," was held on March 25, 2019, at the American Society for Parenteral and Enteral Nutrition (ASPEN) 2019 Nutrition Science & Practice Conference in Phoenix, AZ. The Dudrick Symposium honors the many pivotal and innovative contributions to the development and advancement of parenteral nutrition (PN) made by Dr Stanley J. Dudrick, physician scientist, academic leader, and a founding member of ASPEN. As the 2018 recipient of the Dudrick award, Dr Gail Cresci organized and chaired the symposium. The symposium addressed the evolving field of nutrition manipulation of the gut microbiome as a means to mitigate disease and support health. Presentations focused on (1) the role of prebiotics as a means to beneficially support gut microbiome composition and function and health; (2) designer synbiotics targeted to support metabolic by-products altered by ethanol exposure and microbial effectors that manipulate host metabolic outcomes; and, lastly, (3) types of intervention designs used to study diet-gut microbiome interactions in humans and a review of findings from recent interventions, which tested the effects of diet on the microbiome and the microbiome's effect on dietary exposures. New molecular techniques and multiomic approaches have improved knowledge of the structure and functional activity of the gut microbiome; however, challenges remain in establishing causal relationships between changes in the gut microbial-community structure and function and health outcomes in humans.
Collapse
Affiliation(s)
- Gail A. M. Cresci
- Department of Pediatric GastroenterologyCleveland Clinic Children's HospitalClevelandOhioUSA
- Department of Inflammation and ImmunityLerner Research InstituteCleveland ClinicClevelandOhioUSA
- Center for Human NutritionDigestive Disease InstituteCleveland ClinicClevelandOhioUSA
| | | | - Glenn Gibson
- Department of Food and Nutritional SciencesThe University of ReadingReadingUK
| |
Collapse
|
97
|
Dietary Synbiotic Supplementation Protects Barrier Integrity of Hepatocytes and Liver Sinusoidal Endothelium in a Mouse Model of Chronic-Binge Ethanol Exposure. Nutrients 2020; 12:nu12020373. [PMID: 32023885 PMCID: PMC7071303 DOI: 10.3390/nu12020373] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 02/07/2023] Open
Abstract
Alcohol overconsumption disrupts the gut microbiota and intestinal barrier, which decreases the production of beneficial microbial metabolic byproducts and allows for translocation of pathogenic bacterial-derived byproducts into the portal-hepatic circulation. As ethanol is known to damage liver sinusoidal endothelial cells (LSEC), here we evaluated dietary supplementation with a previously studied synbiotic on gut microbial composition, and hepatocyte and LSEC integrity in mice exposed to ethanol. We tested a chronic-binge ethanol feeding mouse model in which C57BL/6 female mice were fed ethanol (5% vol/vol) for 10 days and provided a single ethanol gavage (5 g/kg body weight) on day 11, 6 h before euthanasia. An ethanol-treatment group also received oral supplementation daily with a synbiotic; and an ethanol-control group received saline. Control mice were pair-fed and isocalorically substituted maltose dextran for ethanol over the entire exposure period; they received a saline gavage daily. Ethanol exposure decreased gut microbial abundance and diversity. This was linked with diminished expression of adherens junction proteins in hepatocytes and dysregulated expression of receptors for advanced glycation end-products; and this coincided with reduced expression of endothelial barrier proteins. Synbiotic supplementation mitigated these effects. These results demonstrate synbiotic supplementation, as a means to modulate ethanol-induced gut dysbiosis, is effective in attenuating injury to hepatocyte and liver endothelial barrier integrity, highlighting a link between the gut microbiome and early stages of acute liver injury in ethanol-exposed mice.
Collapse
|
98
|
Wang C, Shen Z, Cao S, Zhang Q, Peng Y, Hong Q, Feng J, Hu C. Effects of tributyrin on growth performance, intestinal microflora and barrier function of weaned pigs. Anim Feed Sci Technol 2019. [DOI: 10.1016/j.anifeedsci.2019.114311] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
99
|
Warner DR, Warner JB, Hardesty JE, Song YL, King TN, Kang JX, Chen CY, Xie S, Yuan F, Prodhan MAI, Ma X, Zhang X, Rouchka EC, Maddipati KR, Whitlock J, Li EC, Wang GP, McClain CJ, Kirpich IA. Decreased ω-6:ω-3 PUFA ratio attenuates ethanol-induced alterations in intestinal homeostasis, microbiota, and liver injury. J Lipid Res 2019; 60:2034-2049. [PMID: 31586017 PMCID: PMC6889711 DOI: 10.1194/jlr.ra119000200] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 09/22/2019] [Indexed: 02/07/2023] Open
Abstract
Ethanol (EtOH)-induced alterations in intestinal homeostasis lead to multi-system pathologies, including liver injury. ω-6 PUFAs exert pro-inflammatory activity, while ω-3 PUFAs promote anti-inflammatory activity that is mediated, in part, through specialized pro-resolving mediators [e.g., resolvin D1 (RvD1)]. We tested the hypothesis that a decrease in the ω-6:ω-3 PUFA ratio would attenuate EtOH-mediated alterations in the gut-liver axis. ω-3 FA desaturase-1 (fat-1) mice, which endogenously increase ω-3 PUFA levels, were protected against EtOH-mediated downregulation of intestinal tight junction proteins in organoid cultures and in vivo. EtOH- and lipopolysaccharide-induced expression of INF-γ, Il-6, and Cxcl1 was attenuated in fat-1 and WT RvD1-treated mice. RNA-seq of ileum tissue revealed upregulation of several genes involved in cell proliferation, stem cell renewal, and antimicrobial defense (including Alpi and Leap2) in fat-1 versus WT mice fed EtOH. fat-1 mice were also resistant to EtOH-mediated downregulation of genes important for xenobiotic/bile acid detoxification. Further, gut microbiome and plasma metabolomics revealed several changes in fat-1 versus WT mice that may contribute to a reduced inflammatory response. Finally, these data correlated with a significant reduction in liver injury. Our study suggests that ω-3 PUFA enrichment or treatment with resolvins can attenuate the disruption in intestinal homeostasis caused by EtOH consumption and systemic inflammation with a concomitant reduction in liver injury.
Collapse
Affiliation(s)
- Dennis R Warner
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, KY
| | - Jeffrey B Warner
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, KY
- Department of Medicine Division of Infectious Diseases and Global Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Josiah E Hardesty
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, KY
| | - Ying L Song
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, KY
| | - Taylor N King
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, KY
| | - Jing X Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Chih-Yu Chen
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Shanfu Xie
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Fang Yuan
- Department of Chemistry, University of Louisville, Louisville, KY
| | | | - Xipeng Ma
- Department of Chemistry, University of Louisville, Louisville, KY
| | - Xiang Zhang
- Department of Chemistry, University of Louisville, Louisville, KY
| | - Eric C Rouchka
- Department of Computer Engineering and Computer Science, Speed School of Engineering, University of Louisville, Louisville, KY
| | | | - Joan Whitlock
- Department of Medicine Division of Infectious Diseases and Global Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Eric C Li
- Department of Medicine Division of Infectious Diseases and Global Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Gary P Wang
- Department of Medicine Division of Infectious Diseases and Global Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Craig J McClain
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, KY
- Department of Pharmacology and Toxicology and University of Louisville Alcohol Center, University of Louisville School of Medicine, Louisville, KY
- Robley Rex Veterans Medical Center, Louisville, KY
| | - Irina A Kirpich
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville, Louisville, KY
- Department of Pharmacology and Toxicology and University of Louisville Alcohol Center, University of Louisville School of Medicine, Louisville, KY
| |
Collapse
|
100
|
Furuya S, Argemi J, Uehara T, Katou Y, Fouts DE, Schnabl B, Dubuquoy L, Belorkar A, Vadigepalli R, Kono H, Bataller R, Rusyn I. A Novel Mouse Model of Acute-on-Chronic Cholestatic Alcoholic Liver Disease: A Systems Biology Comparison With Human Alcoholic Hepatitis. Alcohol Clin Exp Res 2019; 44:87-101. [PMID: 31710124 DOI: 10.1111/acer.14234] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 11/05/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Alcohol-related liver disease is the main cause of liver-related mortality worldwide. The development of novel targeted therapies for patients with advanced forms (i.e., alcoholic hepatitis, AH) is hampered by the lack of suitable animal models. Here, we developed a novel mouse model of acute-on-chronic alcohol liver injury with cholestasis and fibrosis and performed an extensive molecular comparative analysis with human AH. METHODS For the mouse model of acute-on-chronic liver injury, we used 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC, 0.05% w/w) diet for 8 weeks to establish cholestatic liver fibrosis. After 1-week washout period, male mice were fed intragastrically for 4 weeks with up to 24 g/kg of ethyl alcohol in a high-fat diet. This animal model was phenotyped using histopathology, clinical chemistry, microbiome, and gene expression approaches. Data were compared to the phenotypes of human alcohol-related liver disease, including AH. RESULTS Mice with cholestatic liver fibrosis and subsequent alcohol exposure (DDC + EtOH) exhibited exacerbated liver fibrosis with a pericellular pattern, increased neutrophil infiltration, and ductular proliferation, all characteristics of human AH. DDC administration had no effect on urine alcohol concentration or liver steatosis. Importantly, DDC- and alcohol-treated mice showed a transcriptomic signature that resembled that of patients with AH. Finally, we show that mice in the DDC + EtOH group had an increased gut barrier dysfunction, mimicking an important pathophysiological mechanism of human AH. CONCLUSIONS We developed a novel mouse model of acute-on-chronic cholestatic alcoholic liver injury that has considerable translational potential and can be used to test novel therapeutic modalities for AH.
Collapse
Affiliation(s)
- Shinji Furuya
- From the , Department of Veterinary Integrative Biosciences (SF, IR), Texas A&M University, College Station, Texas
| | - Josepmaria Argemi
- Center for Liver Diseases, (JA, RB), Pittsburgh Research Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Takeki Uehara
- Laboratory of Veterinary Pathology, (TU, YK), Osaka Prefecture University, Osaka, Japan
| | - Yuuki Katou
- Laboratory of Veterinary Pathology, (TU, YK), Osaka Prefecture University, Osaka, Japan
| | | | - Bernd Schnabl
- Department of Medicine, (BS), University of California San Diego, La Jolla, California
| | - Laurent Dubuquoy
- Unité INSERM 995, (LD), Faculté de Médecine, Hôpital Huriez, Lille Service des Maladies de l'Appareil Digestif, Lille, France
| | - Abha Belorkar
- Department of Pathology, Anatomy and Cell Biology, (AB, RV), Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Rajanikanth Vadigepalli
- Department of Pathology, Anatomy and Cell Biology, (AB, RV), Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Hiroshi Kono
- First Department of Surgery, (HK), University of Yamanashi, Yamanashi Prefecture, Japan
| | - Ramon Bataller
- Center for Liver Diseases, (JA, RB), Pittsburgh Research Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Ivan Rusyn
- From the , Department of Veterinary Integrative Biosciences (SF, IR), Texas A&M University, College Station, Texas
| |
Collapse
|