51
|
Mukherjee S, Kumar R, Tsakem Lenou E, Basrur V, Kontoyiannis DL, Ioakeimidis F, Mosialos G, Theiss AL, Flavell RA, Venuprasad K. Deubiquitination of NLRP6 inflammasome by Cyld critically regulates intestinal inflammation. Nat Immunol 2020; 21:626-635. [PMID: 32424362 DOI: 10.1038/s41590-020-0681-x] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 04/06/2020] [Indexed: 02/08/2023]
Abstract
The inflammasome NLRP6 plays a crucial role in regulating inflammation and host defense against microorganisms in the intestine. However, the molecular mechanisms by which NLRP6 function is inhibited to prevent excessive inflammation remain unclear. Here, we demonstrate that the deubiquitinase Cyld prevents excessive interleukin 18 (IL-18) production in the colonic mucosa by deubiquitinating NLRP6. We show that deubiquitination inhibited the NLRP6-ASC inflammasome complex and regulated the maturation of IL-18. Cyld deficiency in mice resulted in elevated levels of active IL-18 and severe colonic inflammation following Citrobacter rodentium infection. Further, in patients with ulcerative colitis, the concentration of active IL-18 was inversely correlated with CYLD expression. Thus, we have identified a novel regulatory mechanism that inhibits the NLRP6-IL-18 pathway in intestinal inflammation.
Collapse
Affiliation(s)
- Sandip Mukherjee
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.,Department of Immunology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ritesh Kumar
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.,Department of Immunology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Elviche Tsakem Lenou
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.,Department of Immunology, UT Southwestern Medical Center, Dallas, TX, USA
| | | | - Dimitris L Kontoyiannis
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece.,Division of Immunology, Biomedical Sciences Research Center Alexander Fleming, Athens, Greece
| | - Fotis Ioakeimidis
- Division of Immunology, Biomedical Sciences Research Center Alexander Fleming, Athens, Greece
| | - George Mosialos
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Arianne L Theiss
- Division of Gastroenterology and Hepatology, School of Medicine at the Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - Richard A Flavell
- Department of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - K Venuprasad
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA. .,Department of Immunology, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
52
|
Yang G, Jang JH, Kim SW, Han SH, Ma KH, Jang JK, Kang HC, Cho YY, Lee HS, Lee JY. Sweroside Prevents Non-Alcoholic Steatohepatitis by Suppressing Activation of the NLRP3 Inflammasome. Int J Mol Sci 2020; 21:ijms21082790. [PMID: 32316419 PMCID: PMC7216241 DOI: 10.3390/ijms21082790] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH), a type of non-alcoholic fatty liver disease, is characterized as steatosis and inflammation in the liver. NLRP3 inflammasome activation is associated with NASH pathology. We hypothesized that suppressing the NLRP3 inflammasome could be effective in preventing NASH. We searched substances that could inhibit the activation of the NLRP3 inflammasome and identified sweroside as an NLRP3 inhibitor. We investigated whether sweroside can be applied to prevent the pathological symptoms associated with NASH in a methionine–choline-deficient (MCD) diet-induced NASH mouse model. The activation of the NLRP3 inflammasome was determined by detecting the production of caspase-1 and IL-1β from pro-caspase-1 and pro-IL-1β in primary mouse macrophages and mouse liver. In a NASH model, mice were fed an MCD diet for two weeks with daily intraperitoneal injections of sweroside. Sweroside effectively inhibited NLRP3 inflammasome activation in primary macrophages as shown by a decrease in IL-1β and caspase-1 production. In a MCD diet-induced NASH mouse model, intraperitoneal injection of sweroside significantly reduced serum aspartate transaminase and alanine transaminase levels, hepatic immune cell infiltration, hepatic triglyceride accumulation, and liver fibrosis. The improvement of NASH symptoms by sweroside was accompanied with its inhibitory effects on the hepatic NLRP3 inflammasome as hepatic IL-1β and caspase-1 were decreased. Furthermore, sweroside blocked de novo synthesis of mitochondrial DNA in the liver, contributing to suppression of the NLRP3 inflammasome. These results suggest that targeting the NLRP3 inflammasome with sweroside could be beneficially employed to improve NASH symptoms.
Collapse
Affiliation(s)
- Gabsik Yang
- BK21plus Team, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Korea; (G.Y.); (J.H.J.); (S.W.K.); (H.C.K.); (Y.-Y.C.); (H.S.L.)
- Department of Pharmacology, College of Korean Medicine, Woosuk University, Jeonbuk 55338, Korea
| | - Joo Hyeon Jang
- BK21plus Team, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Korea; (G.Y.); (J.H.J.); (S.W.K.); (H.C.K.); (Y.-Y.C.); (H.S.L.)
| | - Sung Wook Kim
- BK21plus Team, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Korea; (G.Y.); (J.H.J.); (S.W.K.); (H.C.K.); (Y.-Y.C.); (H.S.L.)
| | - Sin-Hee Han
- Department of Herbal Crop Research, National Institute of Horticultural & Herbal Science, Rural Development Administration, Eumsung 27709, Korea; (S.-H.H.); (K.-H.M.); (J.-K.J.)
| | - Kyung-Ho Ma
- Department of Herbal Crop Research, National Institute of Horticultural & Herbal Science, Rural Development Administration, Eumsung 27709, Korea; (S.-H.H.); (K.-H.M.); (J.-K.J.)
| | - Jae-Ki Jang
- Department of Herbal Crop Research, National Institute of Horticultural & Herbal Science, Rural Development Administration, Eumsung 27709, Korea; (S.-H.H.); (K.-H.M.); (J.-K.J.)
| | - Han Chang Kang
- BK21plus Team, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Korea; (G.Y.); (J.H.J.); (S.W.K.); (H.C.K.); (Y.-Y.C.); (H.S.L.)
| | - Yong-Yeon Cho
- BK21plus Team, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Korea; (G.Y.); (J.H.J.); (S.W.K.); (H.C.K.); (Y.-Y.C.); (H.S.L.)
| | - Hye Suk Lee
- BK21plus Team, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Korea; (G.Y.); (J.H.J.); (S.W.K.); (H.C.K.); (Y.-Y.C.); (H.S.L.)
| | - Joo Young Lee
- BK21plus Team, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Korea; (G.Y.); (J.H.J.); (S.W.K.); (H.C.K.); (Y.-Y.C.); (H.S.L.)
- Correspondence: ; Tel.: +82-2-2164-4095; Fax: +82-2-2164-4059
| |
Collapse
|
53
|
Antecedent Dietary Glutamine Supplementation Benefits Modulation of Liver Pyroptosis in Mice with Polymicrobial Sepsis. Nutrients 2020; 12:nu12041086. [PMID: 32295272 PMCID: PMC7230693 DOI: 10.3390/nu12041086] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 12/13/2022] Open
Abstract
The liver is the main organ responsible for bacterial and endotoxin clearance. Pyroptosis is a form of proinflammatory programmed cell death activated by caspase-1/11 and gasdermin D (GadD). Pyroptosis protects the host against bacterial infection; however, overactive pyroptosis can lead to organ injury. Glutamine (GLN) is a specific amino acid with anti-inflammatory and immunomodulatory properties. This study investigated the effects of GLN pretreatment on liver pyroptosis in a mouse model of polymicrobial sepsis. Mice were assigned to sham, sepsis control (Sepsis-C), and sepsis GLN (Sepsis-G) groups. The sham and Sepsis-C groups were fed the AIN-93G diet. The Sepsis-G group was provided with identical diet components except that part of the casein was replaced by GLN. After feeding the respective diets for 2 weeks, a cecal ligation and puncture (CLP) procedure was performed in the sepsis groups. An antibiotic was administered after CLP. Mice were sacrificed at either 24 or 72 h after CLP. The results showed that sepsis resulted in upregulated liver caspase-1/11 expression. Compared to the Sepsis-C group, the Sepsis-G group had higher liver caspase-11 and NLRP3 gene expressions at 24 h and lower active caspase-1/11 and cleaved GadD protein levels at 72 h after sepsis. Additionally, liver inflammatory cytokine gene expressions had decreased by 72 h post-CLP. The findings suggest that prophylactic administration of GLN initially upregulated liver pyroptosis to eradicate pathogens, yet the process of pyroptosis was suppressed in the late phase of sepsis. This may have beneficially attenuated liver inflammation and injury in an antibiotic-treated septic condition.
Collapse
|
54
|
Mai W, Xu Y, Xu J, Zhao D, Ye L, Yu G, Wang Z, Lu Q, Lin J, Yang T, Gu C, Liu S, Zhong Y, Yang H. Berberine Inhibits Nod-Like Receptor Family Pyrin Domain Containing 3 Inflammasome Activation and Pyroptosis in Nonalcoholic Steatohepatitis via the ROS/TXNIP Axis. Front Pharmacol 2020; 11:185. [PMID: 32194416 PMCID: PMC7063468 DOI: 10.3389/fphar.2020.00185] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 02/10/2020] [Indexed: 12/11/2022] Open
Abstract
Berberine (BBR), an isoquinoline alkaloid originating from herbal plants, has been deemed beneficial for non-alcoholic fatty liver disease. Increasing evidence has demonstrated that Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome activation and the subsequent pyroptosis contribute to the progression of non-alcoholic steatohepatitis (NASH). However, whether BBR impacts NLRP3 inflammasome activation and pyroptosis in NASH and the potential mechanism remains unclear. In the current study, we found that BBR significantly decreased lipid accumulation, ameliorated reactive oxygen species (ROS) and lipid peroxides, Tumor necrosis factor alpha (TNF-α) expression, and phosphorylation of Nuclear factor kappa B (NF-κB) p65 both in vivo and in vitro. In particular, BBR significantly inhibited NLRP3 expression, caspase-1 activity, and the pyroptosis executor, GSDMD-N, expression. In addition, BBR displayed similar inhibitory effects on NLRP3 inflammasome and pyroptosis with a decrease in ROS levels and TXNIP expression as N-acetyl-cysteine, a ROS scavenger, did. Whereas, the inhibitory effect of BBR on ROS, TXNIP expression, NLRP3 inflammasome activation and pyroptosis could be reversed by H2O2 in AML12 cells. This study demonstrates that BBR's inhibitory effect on NLRP3 inflammasome activation and pyroptosis may be mediated by ROS/TXNIP axis in vitro for the first time. Our findings suggest BBR is a potential candidate for the treatment of NASH.
Collapse
Affiliation(s)
- Weijian Mai
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yangzhi Xu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiahui Xu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dan Zhao
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liangying Ye
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ganxiang Yu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhilei Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qianting Lu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiaen Lin
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tao Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chengxin Gu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shiming Liu
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yun Zhong
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hui Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
55
|
Dong Z, Zhuang Q, Ning M, Wu S, Lu L, Wan X. Palmitic acid stimulates NLRP3 inflammasome activation through TLR4-NF-κB signal pathway in hepatic stellate cells. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:168. [PMID: 32309315 PMCID: PMC7154441 DOI: 10.21037/atm.2020.02.21] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background The NLRP3 inflammasome activation plays an important role in the development of NASH and fibrogenesis. However, the mechanisms involved in NLRP3 activation in hepatic stellate cells (HSCs) have been unclear. The aim of this study was to investigate the mechanism of NLRP3 activation in HSCs and the role of NLPR3 inflammasome activation in HSCs on the development of nonalcoholic steatohepatitis (NASH) to fibrosis. Methods Primary HSCs isolated from SD rats were incubated with palmitic acid and/or LPS, respectively. For in vivo animal experiment, 4-week-old SD rats were fed with high fat diet (HF-diet) for 12 weeks, SD rats were sacrificed at 0, 4, 8 and 12 w. In another group of animal experiment, 4-week-old SD rats were fed with HF-diet and a NLRP3 inhibitor (intraperitoneal injection of NLRP3 inhibitor glybenclamide 5 mg/kg, injected every 3 days) for 12 weeks. Liver tissue and serum were harvested. RT-PCR, WB, ELISA, immunofluorescence and immunohistochemistry were performed to assess the NLRP3 inflammasome activation and signal molecules. Results Palmitic acid stimulated NLPR3 inflammasome activation and fibrotic phenotype change in primary HSCs, LPS sensitizes the response of HSCs to palmitic acid. TLR4-NF-κB signal pathway was involved in NLRP3 inflammasome activation in palmitic acid-exposed HSCs and HF diet-induced NASH. It is evident that administration of NLRP3 inhibitor reduced the development of NASH to liver fibrosis in the NASH rat model. Conclusions Palmitic acid stimulates NLRP3 inflammasome activation through the TLR4-NF-κB signal pathway in HSCs. NLRP3 inflammasome activation in HSCs exacerbates the development of NASH to liver fibrosis.
Collapse
Affiliation(s)
- Zhixia Dong
- Digestive Endoscopic Center, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Qian Zhuang
- Digestive Endoscopic Center, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Min Ning
- Digestive Endoscopic Center, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Shan Wu
- Digestive Endoscopic Center, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Lungen Lu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Xinjian Wan
- Digestive Endoscopic Center, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
56
|
Tanwar S, Rhodes F, Srivastava A, Trembling PM, Rosenberg WM. Inflammation and fibrosis in chronic liver diseases including non-alcoholic fatty liver disease and hepatitis C. World J Gastroenterol 2020; 26:109-133. [PMID: 31969775 PMCID: PMC6962431 DOI: 10.3748/wjg.v26.i2.109] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/18/2019] [Accepted: 01/01/2020] [Indexed: 02/06/2023] Open
Abstract
At present chronic liver disease (CLD), the third commonest cause of premature death in the United Kingdom is detected late, when interventions are ineffective, resulting in considerable morbidity and mortality. Injury to the liver, the largest solid organ in the body, leads to a cascade of inflammatory events. Chronic inflammation leads to the activation of hepatic stellate cells that undergo trans-differentiation to become myofibroblasts, the main extra-cellular matrix producing cells in the liver; over time increased extra-cellular matrix production results in the formation of liver fibrosis. Although fibrogenesis may be viewed as having evolved as a “wound healing” process that preserves tissue integrity, sustained chronic fibrosis can become pathogenic culminating in CLD, cirrhosis and its associated complications. As the reference standard for detecting liver fibrosis, liver biopsy, is invasive and has an associated morbidity, the diagnostic assessment of CLD by non-invasive testing is attractive. Accordingly, in this review the mechanisms by which liver inflammation and fibrosis develop in chronic liver diseases are explored to identify appropriate and meaningful diagnostic targets for clinical practice. Due to differing disease prevalence and treatment efficacy, disease specific diagnostic targets are required to optimally manage individual CLDs such as non-alcoholic fatty liver disease and chronic hepatitis C infection. To facilitate this, a review of the pathogenesis of both conditions is also conducted. Finally, the evidence for hepatic fibrosis regression and the mechanisms by which this occurs are discussed, including the current use of antifibrotic therapy.
Collapse
Affiliation(s)
- Sudeep Tanwar
- UCL Institute for Liver and Digestive Health, Division of Medicine, University College London, Royal Free Campus, Hampstead, London NW3 2PF United Kingdom
- Department of Gastroenterology, Whipps Cross University Hospital, Barts Health NHS Trust, Leytonstone, London E11 1NR, United Kingdom
| | - Freya Rhodes
- UCL Institute for Liver and Digestive Health, Division of Medicine, University College London, Royal Free Campus, Hampstead, London NW3 2PF United Kingdom
| | - Ankur Srivastava
- UCL Institute for Liver and Digestive Health, Division of Medicine, University College London, Royal Free Campus, Hampstead, London NW3 2PF United Kingdom
| | - Paul M Trembling
- UCL Institute for Liver and Digestive Health, Division of Medicine, University College London, Royal Free Campus, Hampstead, London NW3 2PF United Kingdom
| | - William M Rosenberg
- UCL Institute for Liver and Digestive Health, Division of Medicine, University College London, Royal Free Campus, Hampstead, London NW3 2PF United Kingdom
| |
Collapse
|
57
|
Oates JR, McKell MC, Moreno-Fernandez ME, Damen MSMA, Deepe GS, Qualls JE, Divanovic S. Macrophage Function in the Pathogenesis of Non-alcoholic Fatty Liver Disease: The Mac Attack. Front Immunol 2019; 10:2893. [PMID: 31921154 PMCID: PMC6922022 DOI: 10.3389/fimmu.2019.02893] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 11/26/2019] [Indexed: 12/15/2022] Open
Abstract
Obesity is a prevalent predisposing factor to non-alcoholic fatty liver disease (NAFLD), the most common chronic liver disease in the developed world. NAFLD spectrum of disease involves progression from steatosis (NAFL), to steatohepatitis (NASH), cirrhosis and hepatocellular carcinoma (HCC). Despite clinical and public health significance, current FDA approved therapies for NAFLD are lacking in part due to insufficient understanding of pathogenic mechanisms driving disease progression. The etiology of NAFLD is multifactorial. The induction of both systemic and tissue inflammation consequential of skewed immune cell metabolic state, polarization, tissue recruitment, and activation are central to NAFLD progression. Here, we review the current understanding of the above stated cellular and molecular processes that govern macrophage contribution to NAFLD pathogenesis and how adipose tissue and liver crosstalk modulates macrophage function. Notably, the manipulation of such events may lead to the development of new therapies for NAFLD.
Collapse
Affiliation(s)
- Jarren R Oates
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Melanie C McKell
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Maria E Moreno-Fernandez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Michelle S M A Damen
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - George S Deepe
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Joseph E Qualls
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
58
|
Wu J, Lin S, Wan B, Velani B, Zhu Y. Pyroptosis in Liver Disease: New Insights into Disease Mechanisms. Aging Dis 2019; 10:1094-1108. [PMID: 31595205 PMCID: PMC6764727 DOI: 10.14336/ad.2019.0116] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/16/2019] [Indexed: 12/12/2022] Open
Abstract
There has been increasing interest in pyroptosis as a novel form of pro-inflammatory programmed cell death. The mechanism of pyroptosis is significantly different from other forms of cell death in its morphological and biochemical features. Pyroptosis is characterized by the activation of two different types of caspase enzymes-caspase-1 and caspase-4/5/11, and by the occurrence of a proinflammatory cytokine cascade and an immune response. Pyroptosis participates in the immune defense mechanisms against intracellular bacterial infections. On the other hand, excessive inflammasome activation can induce sterile inflammation and eventually cause some diseases, such as acute or chronic hepatitis and liver fibrosis. The mechanism and biological significance of this novel form of cell death in different liver diseases will be evaluated in this review. Specifically, we will focus on the role of pyroptosis in alcoholic and non-alcoholic fatty liver disease, as well as in liver failure. Finally, the therapeutic implications of pyroptosis in liver diseases will be discussed.
Collapse
Affiliation(s)
- Jiali Wu
- Liver research center of the First Affiliated Hospital of Fujian Medical University, Fujian 350005, China
| | - Su Lin
- Liver research center of the First Affiliated Hospital of Fujian Medical University, Fujian 350005, China
| | - Bo Wan
- Faculty of Life Sciences and Medicine, King’s College London, London SE1 1UL, United Kingdom
| | - Bharat Velani
- Basildon and Thurrock University Hospitals NHS Foundation Trust, Nethermayne, Basildon, Essex SS16 5NL, United Kingdom
| | - Yueyong Zhu
- Liver research center of the First Affiliated Hospital of Fujian Medical University, Fujian 350005, China
| |
Collapse
|
59
|
Lv Y, Gao X, Luo Y, Fan W, Shen T, Ding C, Yao M, Song S, Yan L. Apigenin ameliorates HFD-induced NAFLD through regulation of the XO/NLRP3 pathways. J Nutr Biochem 2019; 71:110-121. [PMID: 31325892 DOI: 10.1016/j.jnutbio.2019.05.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/21/2019] [Accepted: 05/21/2019] [Indexed: 12/25/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common cause of liver-related morbidity and mortality disease in the world. However, no effective pharmacological treatment for NAFLD has been found. In this study, we used a high fat diet (HFD)-induced NAFLD model to investigate hepatoprotective effect of apigenin (API) against NAFLD and further explored its potential mechanism. Our results demonstrated that gavage administration of API could mitigate HFD-induced liver injury, enhance insulin sensitivity and markedly reduce lipid accumulation in HFD-fed mice livers. In addition, histological analysis showed that hepatic steatosis and macrophages recruitment in the API treatment group were recovered compared with mice fed with HFD alone. Importantly, API could reverse the HFD-induced activation of the NLRP3 inflammasome, further reduced inflammatory cytokines IL-1β and IL-18 release, accompanied with the inhibition of xanthine oxidase (XO) activity and the reduction of uric acid and reactive oxygen species (ROS) production. The pharmacological role of API was further confirmed using free fatty acid (FFA) induced cell NAFLD model. Taking together, our results demonstrated that API could protect against HFD-induced NAFLD by ameliorating hepatic lipid accumulation and inflammation. These protective effects may be partially attributed to the regulation of XO by API, which further modulated NLRP3 inflammasome activation and inflammatory cytokines IL-1β and IL-18 release. Therefore API is a potential therapeutic agent for the prevention of NAFLD.
Collapse
Affiliation(s)
- Yanan Lv
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Xiaona Gao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Yan Luo
- Administration for Market Regulation of GuangDong Province Key Laboratory of Supervision for Edible Agricultural Products, Shenzhen Centre of Inspection and Testing for Agricultural Products, Shenzhen 518000, GuangDong Province, China
| | - Wentao Fan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Tongtong Shen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Chenchen Ding
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Ming Yao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Suquan Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China.
| | - Liping Yan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China.
| |
Collapse
|
60
|
Mirea AM, Toonen EJM, van den Munckhof I, Munsterman ID, Tjwa ETTL, Jaeger M, Oosting M, Schraa K, Rutten JHW, van der Graaf M, Riksen NP, de Graaf J, Netea MG, Tack CJ, Chavakis T, Joosten LAB. Increased proteinase 3 and neutrophil elastase plasma concentrations are associated with non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes. Mol Med 2019; 25:16. [PMID: 31046673 PMCID: PMC6498541 DOI: 10.1186/s10020-019-0084-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 04/17/2019] [Indexed: 02/07/2023] Open
Abstract
Introduction Non-alcoholic fatty liver disease (NAFLD) is becoming a major health problem worldwide. Inflammation plays an important role in disease pathogenesis and recent studies have shown a potential role for the neutrophil serine proteases (NSPs) proteinase-3 (PR3) and neutrophil elastase (NE) in NAFLD as well as an imbalance between NSPs and their natural inhibitor alpha-1 antitrypsin (AAT). The aim of this study was to investigate whether PR3 and NE plasma concentrations are associated with NAFLD and/or type 2 diabetes. Methods To explore this hypothesis we used several cohorts: a cohort of 271 obese individuals with liver steatosis, a cohort of 41 patients with biopsy-proven NAFLD, a cohort of 401 obese type 2 diabetes patients and a cohort of 205 lean healthy controls; and measured PR3 and NE plasma concentrations. In addition, we measured AAT plasma concentrations in order to investigate if the ratios between NSPs and their natural inhibitor were altered in NAFLD and type 2 diabetes when compared to healthy controls. Results Our data shows an increase in PR3 and NE concentrations and a decrease in AAT concentrations in obese patients when compared to controls. Moreover, PR3 plasma concentrations are increased in patients with liver steatosis. Furthermore, PR3 and NE concentrations in the liver are associated with the advanced stages of NAFLD characterized by NASH and/ or liver fibrosis. Additionally, PR3 and NE concentrations were up-regulated in patients with type 2 diabetes when compared to lean and obese controls. Conclusion We conclude that circulating levels of NSPs associate with obesity-related metabolic disorders. Further research is needed to clearly establish the role of these proteases and investigate whether they could be used as non-invasive markers for NAFLD and/or type 2 diabetes. Electronic supplementary material The online version of this article (10.1186/s10020-019-0084-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andreea-Manuela Mirea
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, The Netherlands.,Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, 400349, Cluj-Napoca, Romania
| | - Erik J M Toonen
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, The Netherlands.,R&D Department, Hycult Biotechnology, Uden, The Netherlands
| | - Inge van den Munckhof
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Isabelle D Munsterman
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Eric T T L Tjwa
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martin Jaeger
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Marije Oosting
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Kiki Schraa
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Joost H W Rutten
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Marinette van der Graaf
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Niels P Riksen
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Jacqueline de Graaf
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, The Netherlands.,Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115, Bonn, Germany
| | - Cees J Tack
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Carl-Gustav-Carus, TU Dresden, Dresden, Germany.,Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany; and German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, The Netherlands. .,Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, 400349, Cluj-Napoca, Romania.
| |
Collapse
|
61
|
Ioannou GN, Landis CS, Jin GY, Haigh WG, Farrell GC, Kuver R, Lee SP, Savard C. Cholesterol Crystals in Hepatocyte Lipid Droplets Are Strongly Associated With Human Nonalcoholic Steatohepatitis. Hepatol Commun 2019; 3:776-791. [PMID: 31168512 PMCID: PMC6545865 DOI: 10.1002/hep4.1348] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 03/04/2019] [Indexed: 12/19/2022] Open
Abstract
It is unclear what drives the development of fibrosing nonalcoholic steatohepatitis (NASH). We aimed to determine whether cholesterol crystallization within hepatocyte lipid droplets (LDs) distinguishes patients with fibrosing NASH from patients with isolated hepatic steatosis and to study pathways leading to cholesterol accumulation in hepatocyte LDs. Patients with fibrosing NASH (n = 16) were compared to patients with isolated steatosis (n = 14). Almost all patients with fibrosing NASH had free cholesterol staining by filipin (16/16) and cholesterol crystals (15/16) in hepatocyte LDs, mostly in association with the LD membrane, compared to only 3/14 with cholesterol crystals and 3/14 with faint filipin staining in patients with isolated steatosis (P < 0.05). We were unable to identify significant differences in the expression of genes in liver tissue related to cholesterol homeostasis or LD proteins between patients with fibrosing NASH and isolated steatosis. Human hepatoma cell line (HepG2) cells were supplemented with low-density lipoprotein (LDL)-cholesterol and oleic acid to develop large LDs, similar to those observed in patients with NASH. Fluorescent markers were used to track the uptake and intracellular trafficking of LDL-cholesterol. LDL-cholesterol was taken up by HepG2 cells and transported through the endosomal-lysosomal compartment directly to LDs, suggesting direct contact sites between late endosomes and LDs. Exposure of HepG2 cells to LDL-cholesterol resulted in a high concentration of cholesterol and cholesterol crystallization in LDs. Conclusion: Excess cholesterol is stored in the liver primarily within hepatocyte LDs where it can crystallize. Our findings are best explained by direct transport of cholesterol from late endosomes/lysosomes to LDs in hepatocytes. We found a strong association between the presence of LD cholesterol crystals and the development of fibrosing NASH in humans, suggesting a causal relationship.
Collapse
Affiliation(s)
- George N Ioannou
- Division of Gastroenterology, Department of Medicine Veterans Affairs Puget Sound Health Care System Seattle WA.,Division of Gastroenterology, Department of Medicine University of Washington Seattle WA.,Research and Development Veterans Affairs Puget Sound Health Care System Seattle WA
| | - Charles S Landis
- Division of Gastroenterology, Department of Medicine University of Washington Seattle WA
| | - Ga-Young Jin
- Research and Development Veterans Affairs Puget Sound Health Care System Seattle WA
| | - W Geoffrey Haigh
- Research and Development Veterans Affairs Puget Sound Health Care System Seattle WA
| | - Geoffrey C Farrell
- Liver Research Group Australian National University Medical School at the Canberra Hospital Garran Australia
| | - Rahul Kuver
- Division of Gastroenterology, Department of Medicine University of Washington Seattle WA
| | - Sum P Lee
- Division of Gastroenterology, Department of Medicine University of Washington Seattle WA
| | - Christopher Savard
- Division of Gastroenterology, Department of Medicine Veterans Affairs Puget Sound Health Care System Seattle WA.,Division of Gastroenterology, Department of Medicine University of Washington Seattle WA.,Research and Development Veterans Affairs Puget Sound Health Care System Seattle WA
| |
Collapse
|
62
|
Li JJ, Zhang P, Fan B, Guo XL, Zheng ZS. The efficacy of saxagliptin in T2DM patients with non-alcoholic fatty liver disease: preliminary data. ACTA ACUST UNITED AC 2019; 65:33-37. [PMID: 30758417 DOI: 10.1590/1806-9282.65.1.33] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 08/27/2018] [Indexed: 01/14/2023]
Abstract
OBJECTIVE To investigate the clinical efficacy and the possible mechanisms of saxagliptin in the treatment of type 2 diabetes mellitus (T2DM) combined with non-alcoholic fatty liver disease (NAFLD). METHODS A total of 95 T2DM and NAFLD patients were randomly divided into group A (saxagliptin group), group B (glimepiride group), and group C (glimepiride combined with polyene phosphatidylcholine group). RESULTS After intervention treatment for 24 w, body mass index (BMI), waist-to-hip ratio (WHR), glycated haemoglobin (HbA1c), fasting plasma glucose (FPG), fasting insulin (FINS), homeostatic model assessment of insulin resistance (HOMA-IR), interleukin-6 (IL-6), triglyceride (TG), total cholesterol (TC), alanine aminotransferase (ALT), aspartate aminotransferase (AST), γ-glutamyltransferase (γ-GT), and quantitative detection of liver steatosis of study subjects were observed, the action of liver steatosis in subjects of groups A and C were significantly different from those of group B; however, there were no differences between groups A and C. The FINS, HOMA-IR, and IL-6 of subjects in group A was lower than those in groups B and C; however, there were no significant differences between the latter two groups. CONCLUSION For T2DM combined with NAFLD patients, the saxagliptin treatment could not only effectively control blood glucose but also attenuate insulin resistance and inflammatory injury of the liver to improve fatty liver further.
Collapse
Affiliation(s)
- Juan-Juan Li
- Health Management Center, Qilu Hospital of Shandong University (Qingdao), Shandong Province, China
| | - Ping Zhang
- Department of Ultrasonography, Qilu Hospital of Shandong University (Qingdao), Shandong Province, China
| | - Bing Fan
- Health Management Center, Qilu Hospital of Shandong University (Qingdao), Shandong Province, China
| | - Xiu-Li Guo
- Department of Ultrasonography, Qilu Hospital of Shandong University (Qingdao), Shandong Province, China
| | - Zhe-Shu Zheng
- Department of Ultrasonography, Qilu Hospital of Shandong University (Qingdao), Shandong Province, China
| |
Collapse
|
63
|
The role of macrophages in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Nat Rev Gastroenterol Hepatol 2019; 16:145-159. [PMID: 30482910 DOI: 10.1038/s41575-018-0082-x] [Citation(s) in RCA: 647] [Impact Index Per Article: 107.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) and its inflammatory and often progressive subtype nonalcoholic steatohepatitis (NASH) are becoming the leading cause of liver-related morbidity and mortality worldwide, and a primary indication for liver transplantation. The pathophysiology of NASH is multifactorial and not yet completely understood; however, innate immunity is a major contributing factor in which liver-resident macrophages (Kupffer cells) and recruited macrophages play a central part in disease progression. In this Review, we assess the evidence for macrophage involvement in the development of steatosis, inflammation and fibrosis in NASH. In this process, not only the polarization of liver macrophages towards a pro-inflammatory phenotype is important, but adipose tissue macrophages, especially in the visceral compartment, also contribute to disease severity and insulin resistance. Macrophage activation is mediated by factors such as endotoxins and translocated bacteria owing to increased intestinal permeability, factors released from damaged or lipoapoptotic hepatocytes, as well as alterations in gut microbiota and defined nutritional components, including certain free fatty acids, cholesterol and their metabolites. Reflecting the important role of macrophages in NASH, we also review studies investigating drugs that target macrophage recruitment to the liver, macrophage polarization and their inflammatory effects as potential treatment options for patients with NASH.
Collapse
|
64
|
Inzaugarat ME, Johnson CD, Holtmann TM, McGeough MD, Trautwein C, Papouchado BG, Schwabe R, Hoffman HM, Wree A, Feldstein AE. NLR Family Pyrin Domain-Containing 3 Inflammasome Activation in Hepatic Stellate Cells Induces Liver Fibrosis in Mice. Hepatology 2019; 69:845-859. [PMID: 30180270 PMCID: PMC6351190 DOI: 10.1002/hep.30252] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 08/14/2018] [Indexed: 12/25/2022]
Abstract
The NLR family pyrin domain-containing 3 (NLRP3) inflammasome plays an important role in liver fibrosis (LF) development. However, the mechanisms involved in NLRP3-induced fibrosis are unclear. Our aim was to test the hypothesis that the NLRP3 inflammasome in hepatic stellate cells (HSCs) can directly regulate their activation and contribute to LF. Primary HSCs isolated from wild-type (WT), Nlrp3-/- , or Nlrp3L351PneoR knock-in crossed to inducible (estrogen receptor Cre-CreT) mice were incubated with lipopolysaccharide (LPS) and adenosine triphosphate (ATP), or 4OH-tamoxifen, respectively. HSC-specific Nlrp3L351P knock-in mice were generated by crossing transgenic mice expressing lecithin retinol acyltransferase (Lrat)-driven Cre and maintained on standard rodent chow for 6 months. Mice were then sacrificed; liver tissue and serum were harvested. Nlrp3 inflammasome activation along with HSC phenotype and fibrosis were assessed by RT-PCR, western blotting, fluorescence-activated cell sorting (FACS), enzyme-linked immunosorbent assay, immunofluorescence (IF), and immunohistochemistry (IHC). Stimulated WT HSCs displayed increased levels of NLRP3 inflammasome-induced reactive oxygen species (ROS) production and cathepsin B activity, accompanied by an up-regulation of mRNA and protein levels of fibrotic makers, an effect abrogated in Nlrp3-/- HSCs. Nlrp3L351P CreT HSCs also showed elevated mRNA and protein expression of fibrotic markers 24 hours after inflammasome activation induced with 4-hydroxytamoxifen (4OHT). Protein and mRNA expression levels of fibrotic markers were also found to be increased in isolated HSCs and whole liver tissue from Nlrp3L351P Lrat Cre mice compared to WT. Liver sections from 24-week-old NlrpL351P Lrat Cre mice showed fibrotic changes with increased alpha smooth muscle actin (αSMA) and desmin-positive cells and collagen deposition, independent of inflammatory infiltrates; these changes were also observed after LPS challenge in 8-week-old NlrpL351P Lrat Cre mice. Conclusion: Our results highlight a direct role for the NLRP3 inflammasome in the activation of HSCs directly triggering LF.
Collapse
Affiliation(s)
| | - Casey D. Johnson
- Department of Pediatrics, University of California San Diego, La Jolla, CA
| | | | | | - Christian Trautwein
- Department of Internal Medicine III, RWTH University Hospital Aachen, Germany
| | | | - Robert Schwabe
- Department of Medicine, Columbia University, College of Physicians and Surgeons, New York, USA
| | - Hal M. Hoffman
- Department of Pediatrics, University of California San Diego, La Jolla, CA
| | - Alexander Wree
- Department of Internal Medicine III, RWTH University Hospital Aachen, Germany,,Department of Pediatrics, University of California San Diego, La Jolla, CA
| | - Ariel E. Feldstein
- Department of Pediatrics, University of California San Diego, La Jolla, CA
| |
Collapse
|
65
|
Li J, Xue J, Wang D, Dai X, Sun Q, Xiao T, Wu L, Xia H, Mostofa G, Chen X, Wei Y, Chen F, Quamruzzaman Q, Zhang A, Liu Q. Regulation of gasdermin D by miR-379-5p is involved in arsenite-induced activation of hepatic stellate cells and in fibrosis via secretion of IL-1β from human hepatic cells. Metallomics 2019; 11:483-495. [DOI: 10.1039/c8mt00321a] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Arsenic is an environmental toxicant and human carcinogen.
Collapse
|
66
|
Schwenger KJP, Bolzon CM, Li C, Allard JP. Non-alcoholic fatty liver disease and obesity: the role of the gut bacteria. Eur J Nutr 2018; 58:1771-1784. [PMID: 30306296 DOI: 10.1007/s00394-018-1844-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/05/2018] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty-liver disease (NAFLD) is now considered one of the leading causes of liver disease worldwide and is associated with metabolic syndrome and obesity. There are several factors contributing to the disease state. Recent research suggests that the intestinal microbiota (IM) and bacterial products may play a role through several mechanisms which include increased energy uptake, intestinal permeability and chronic inflammation. In addition to diet and exercise, treatment options targeting the IM are being investigated and include the use of pre-, pro- and synbiotics as well as the possibility of fecal microbial transfers. This literature review explores the relationship between NAFLD and the IM as well as highlight new IM treatment options that may become available in the near future.
Collapse
Affiliation(s)
- Katherine J P Schwenger
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Toronto General Hospital, University Health Network, 585 University Avenue, 9-973, Toronto, ON, M5G 2C4, Canada
| | - Colin M Bolzon
- Toronto General Hospital, University Health Network, 585 University Avenue, 9-973, Toronto, ON, M5G 2C4, Canada
| | - Carrie Li
- Toronto General Hospital, University Health Network, 585 University Avenue, 9-973, Toronto, ON, M5G 2C4, Canada
| | - Johane P Allard
- Toronto General Hospital, University Health Network, 585 University Avenue, 9-973, Toronto, ON, M5G 2C4, Canada. .,Department of Medicine, University of Toronto, Toronto, Canada. .,Department of Nutritional Sciences, University of Toronto, Toronto, Canada.
| |
Collapse
|
67
|
NLRP3 Inflammasome and IL-33: Novel Players in Sterile Liver Inflammation. Int J Mol Sci 2018; 19:ijms19092732. [PMID: 30213101 PMCID: PMC6163521 DOI: 10.3390/ijms19092732] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 09/09/2018] [Accepted: 09/10/2018] [Indexed: 12/11/2022] Open
Abstract
In sterile liver inflammation, danger signals are released in response to tissue injury to alert the immune system; e.g., by activation of the NLRP3 inflammasome. Recently, IL-33 has been identified as a novel type of danger signal or “alarmin”, which is released from damaged and necrotic cells. IL-33 is a pleiotropic cytokine that targets a broad range of immune cells and exhibits pro- and anti-inflammatory properties dependent on the disease. This review summarizes the immunomodulatory roles of the NLRP3 inflammasome and IL-33 in sterile liver inflammation and highlights potential therapeutic strategies targeting these pathways in liver disease.
Collapse
|
68
|
Han M, Zhang T, Gu W, Yang X, Zhao R, Yu J. 2,3,5,4'-tetrahydroxy-stilbene-2-O-β-D-glucoside attenuates methionine and choline-deficient diet-induced non-alcoholic fatty liver disease. Exp Ther Med 2018; 16:1087-1094. [PMID: 30116360 PMCID: PMC6090268 DOI: 10.3892/etm.2018.6300] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 01/09/2018] [Indexed: 12/20/2022] Open
Abstract
Previous studies have suggested that 2,3,5,4′-tetrahydroxy-stilbene-2-O-β-D-glucoside (TSG) prevents progression of non-alcoholic fatty liver disease (NAFLD) induced by high-fat diet. The present study aimed to evaluate whether TSG could reverse NAFLD induced by a methionine and choline-deficient (MCD) diet and identify the possible mechanism of action. C57BL6/J mice were fed a MCD diet and were treated with TSG, fenofibrate, and resveratrol for 9 weeks. Regulatory effects of several cytokines and enzymes, including Nod-like receptor protein 3, apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain (ASC), caspase-1, interleukin (IL)-18, IL-1β, and gut microbiota balance were investigated. TSG significantly reduced NAFLD biochemical indexes, including total cholesterol, triglyceride, low density lipoprotein cholesterol, very low density lipoprotein cholesterol, aspartate aminotransferase and free fatty acid. Middle dosage (TSG.M, 35 mg/kg) of TSG reduced the expression of ASC and caspase-1. Furthermore, TSG displayed gut microbiota regulatory effects on MCD-induced NAFLD mice. The results of the present study suggested that TSG prevented the occurrence and development of MCD diet-induced NAFLD. The data further indicated that TSG may serve as a promising lead compound that may aid with intervention in NAFLD therapy.
Collapse
Affiliation(s)
- Mingnuan Han
- College of Pharmaceutical Science, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Ting Zhang
- College of Pharmaceutical Science, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Wen Gu
- College of Pharmaceutical Science, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Xingxin Yang
- College of Pharmaceutical Science, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Ronghua Zhao
- College of Pharmaceutical Science, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Jie Yu
- College of Pharmaceutical Science, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
69
|
Mirea AM, Tack CJ, Chavakis T, Joosten LAB, Toonen EJM. IL-1 Family Cytokine Pathways Underlying NAFLD: Towards New Treatment Strategies. Trends Mol Med 2018; 24:458-471. [PMID: 29665983 PMCID: PMC5939989 DOI: 10.1016/j.molmed.2018.03.005] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/11/2018] [Accepted: 03/12/2018] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease worldwide. Pathways responsible for the activation of IL-1 family cytokines are key in the development of NAFLD but underlying mechanisms are not fully understood. Many studies have focused on the inflammasome-caspase-1 pathway and have shown that this pathway is an important inducer of inflammation in NAFLD. However, this pathway is not solely responsible for the activation of proinflammatory cytokines. Also, neutrophil serine proteases (NSPs) are capable of activating cytokines and recent studies reported that these proteases also contribute to NAFLD. These studies provided, for the first time, evidence that this inflammasome-independent pathway is involved in NAFLD. In our opinion, these new insights open up new approaches for therapeutic intervention.
Collapse
Affiliation(s)
- Andreea-Manuela Mirea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cees J Tack
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Erik J M Toonen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; R&D Department, Hycult Biotech, Uden, The Netherlands.
| |
Collapse
|
70
|
Xu B, Jiang M, Chu Y, Wang W, Chen D, Li X, Zhang Z, Zhang D, Fan D, Nie Y, Shao F, Wu K, Liang J. Gasdermin D plays a key role as a pyroptosis executor of non-alcoholic steatohepatitis in humans and mice. J Hepatol 2018; 68:773-782. [PMID: 29273476 DOI: 10.1016/j.jhep.2017.11.040] [Citation(s) in RCA: 317] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 11/22/2017] [Accepted: 11/24/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Gasdermin D (GSDMD)-executed programmed necrosis is involved in inflammation and controls interleukin (IL)-1β release. However, the role of GSDMD in non-alcoholic steatohepatitis (NASH) remains unclear. We investigated the role of GSDMD in the pathogenesis of steatohepatitis. METHODS Human liver tissues from patients with non-alcoholic fatty liver disease (NAFLD) and control individuals were obtained to evaluate GSDMD expression. Gsdmd knockout (Gsdmd-/-) mice, obese db/db mice and their wild-type (WT) littermates were fed with methionine-choline deficient (MCD) or control diet to induce steatohepatitis. The Gsdmd-/- and WT mice were also used in a high-fat diet (HFD)-induced NAFLD model. In addition, Alb-Cre mice were administered an adeno-associated virus (AAV) vector that expressed the gasdermin-N domain (AAV9-FLEX-GSDMD-N) and were fed with either MCD or control diet for 10 days. RESULTS GSDMD and its pyroptosis-inducing fragment GSDMD-N were upregulated in liver tissues of human NAFLD/NASH. Importantly, hepatic GSDMD-N protein levels were significantly higher in human NASH and correlated with the NAFLD activity score and fibrosis. GSDMD-N remained a potential biomarker for the diagnosis of NASH. MCD-fed Gsdmd-/- mice exhibit decreased severity of steatosis and inflammation compared with WT littermates. GSDMD was associated with the secretion of pro-inflammatory cytokines (IL-1β, TNF-α, and MCP-1 [CCL2]) and persistent activation of the NF-ĸB signaling pathway. Gsdmd-/- mice showed lower steatosis, mainly because of reduced expression of the lipogenic gene Srebp1c (Srebf1) and upregulated expression of lipolytic genes, including Pparα, Aco [Klk15], Lcad [Acadl], Cyp4a10 and Cyp4a14. Alb-Cre mice administered with AAV9-FLEX-GSDMD-N showed significantly aggravated steatohepatitis when fed with MCD diet. CONCLUSION As an executor of pyroptosis, GSDMD plays a key role in the pathogenesis of steatohepatitis, by controlling cytokine secretion, NF-ĸB activation, and lipogenesis. LAY SUMMARY Non-alcoholic fatty liver disease has become one of the most feared chronic liver diseases, because it is the most rapidly growing indication for adult liver transplantation and a major cause of hepatocellular carcinoma. However, the mechanisms involved in the transformation of simple steatosis to steatohepatitis remain unclear. Herein, we show that gasdermin D driven pyroptosis is prominent in patients with non-alcoholic steatohepatitis (NASH), and gasdermin-N domain remains a potential biomarker for the diagnosis of NASH. Gasdermin D plays a key role in the pathogenesis of NASH by regulating lipogenesis, the inflammatory response, and the NF-ĸB signaling pathway, revealing potential treatment targets for NASH in humans.
Collapse
Affiliation(s)
- Bing Xu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China
| | - Mingzuo Jiang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China
| | - Yi Chu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China
| | - Weijie Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China
| | - Di Chen
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China
| | - Xiaowei Li
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China
| | - Zhao Zhang
- Lintong Aerial Medical Evaluation and Training Center of Air Force, PLA, Xi'an, Shaanxi, MI 710032, China
| | - Di Zhang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China
| | - Feng Shao
- National Institute of Biological Sciences, Number 7 Science Park Road, Zhongguancun Life Science Park, Beijing, MI 102206, China
| | - Kaichun Wu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China.
| | - Jie Liang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China.
| |
Collapse
|
71
|
Chen Y, Yousaf MN, Mehal WZ. Role of sterile inflammation in fatty liver diseases. LIVER RESEARCH 2018. [DOI: 10.1016/j.livres.2018.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
72
|
Wree A, McGeough MD, Inzaugarat ME, Eguchi A, Schuster S, Johnson CD, Peña CA, Geisler LJ, Papouchado BG, Hoffman HM, Feldstein AE. NLRP3 inflammasome driven liver injury and fibrosis: Roles of IL-17 and TNF in mice. Hepatology 2018; 67:736-749. [PMID: 28902427 PMCID: PMC5849484 DOI: 10.1002/hep.29523] [Citation(s) in RCA: 215] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 07/01/2017] [Accepted: 09/08/2017] [Indexed: 12/11/2022]
Abstract
The NLRP3 inflammasome, a caspase-1 activation platform, plays a key role in the modulation of liver inflammation and fibrosis. Here, we tested the hypothesis that interleukin 17 (IL-17) and tumor necrosis factor (TNF) are key cytokines involved in amplifying and perpetuating the liver damage and fibrosis resulting from NLRP3 activation. To address this hypothesis, gain-of-function Nlrp3A350V knock-in mice were bred onto il17a and Tnf knockout backgrounds allowing for constitutive Nlrp3 activation in myeloid derived cells in mice deficient in IL-17 or TNF. Livers of Nlrp3A350V knock-in mice exhibited severe liver inflammatory changes characterized by infiltration with neutrophils, increased expression of chemokine (C-X-C motif) ligand (CXCL) 1 and CXCL2 chemokines, activated inflammatory macrophages, and elevated levels of IL-17 and TNF. Mutants with ablation of il17a signal showed fewer neutrophils when compared to intact Nlrp3A350V mutants, but still significant inflammatory changes when compared to the nonmutant il17a knockout littermates. The severe inflammatory changes associated with mutant Nlrp3 were almost completely rescued by Tnf knockout in association with a marked decrease in circulating IL-1β levels. Intact Nlrp3A350V mutants showed changes in liver fibrosis, as evidenced by morphometric quantitation of Sirius Red staining and increased mRNA levels of profibrotic genes, including connective tissue growth factor and tissue inhibitor of matrix metalloproteinase 1. Il17a lacking mutants exhibited amelioration of the aforementioned fibrosis, whereas Tnf-deficient mutants showed no signs of fibrosis when compared to littermate controls. Conclusion: Our study uncovers key roles for TNF and, to a lesser extent, IL-17 as mediators of liver inflammation and fibrosis induced by constitutive NLRP3 inflammasome activation in myeloid-derived cells. These findings may lead to therapeutic strategies aimed at halting the progression of liver injury and fibrogenesis in various liver pathogeneses driven by NLRP3 activation. (Hepatology 2018;67:736-749).
Collapse
Affiliation(s)
- Alexander Wree
- Department of Pediatrics, University of California – San Diego, La Jolla
- Department of Internal Medicine III, RWTH University Hospital Aachen, Germany
| | | | | | - Akiko Eguchi
- Department of Pediatrics, University of California – San Diego, La Jolla
| | - Susanne Schuster
- Department of Pediatrics, University of California – San Diego, La Jolla
| | - Casey D. Johnson
- Department of Pediatrics, University of California – San Diego, La Jolla
| | - Carla A. Peña
- Department of Pediatrics, University of California – San Diego, La Jolla
| | - Lukas J. Geisler
- Department of Internal Medicine III, RWTH University Hospital Aachen, Germany
| | | | - Hal M. Hoffman
- Department of Pediatrics, University of California – San Diego, La Jolla
| | - Ariel E. Feldstein
- Department of Pediatrics, University of California – San Diego, La Jolla
| |
Collapse
|
73
|
Kanak MA, Shindo Y, SaiKumar P, Naziruddin B. Role of Inflammasomes in the Development of Gastrointestinal Diseases. EXPERIENTIA SUPPLEMENTUM (2012) 2018; 108:235-268. [PMID: 30536174 DOI: 10.1007/978-3-319-89390-7_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Many diseases of the gastrointestinal tract have been attributed to chronic inflammation, and a few have identified the role of inflammasomes in their pathogenesis. Inflammasomes are a group of protein complexes comprising of several intracellular proteins that link the sensing of microbial products and metabolic stress to the proteolytic activation of the proinflammatory cytokines. Recent studies have implicated activation of several families of NOD-like receptors (NLRs) which are major components of inflammasomes in the development and exacerbation of many diseases of human systems. In this chapter, we discuss the role of inflammasomes in some of the most prevalent diseases of the gastrointestinal tract and highlight potential targets for treatment.
Collapse
Affiliation(s)
- Mazhar A Kanak
- Division of Transplantation, Department of Surgery, Virginia Commonwealth University Medical Center, Richmond, VA, USA
| | - Yoshitaro Shindo
- Division of Transplantation, Department of Surgery, Virginia Commonwealth University Medical Center, Richmond, VA, USA
| | | | - Bashoo Naziruddin
- Sammons Cancer Center, Baylor Simmons Transplant Institute, Dallas, TX, USA.
| |
Collapse
|
74
|
Machado MV, Diehl AM. Pathogenesis of Nonalcoholic Fatty Liver Disease. ZAKIM AND BOYER'S HEPATOLOGY 2018:369-390.e14. [DOI: 10.1016/b978-0-323-37591-7.00025-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
75
|
Pappachan JM, Babu S, Krishnan B, Ravindran NC. Non-alcoholic Fatty Liver Disease: A Clinical Update. J Clin Transl Hepatol 2017; 5:384-393. [PMID: 29226105 PMCID: PMC5719196 DOI: 10.14218/jcth.2017.00013] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/31/2017] [Accepted: 06/24/2017] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is currently the most common chronic liver disease in developed countries because of the obesity epidemic. The disease increases liver-related morbidity and mortality, and often increases the risk for other comorbidities, such as type 2 diabetes and cardiovascular disease. Insulin resistance related to metabolic syndrome is the main pathogenic trigger that, in association with adverse genetic, humoral, hormonal and lifestyle factors, precipitates development of NAFLD. Biochemical markers and radiological imaging, along with liver biopsy in selected cases, help in diagnosis and prognostication. Intense lifestyle changes aiming at weight loss are the main therapeutic intervention to manage cases. Insulin sensitizers, antioxidants, lipid lowering agents, incretin-based drugs, weight loss medications, bariatric surgery and liver transplantation may be necessary for management in some cases along with lifestyle measures. This review summarizes the latest evidence on the epidemiology, natural history, pathogenesis, diagnosis and management of NAFLD.
Collapse
Affiliation(s)
- Joseph M Pappachan
- Department of Endocrinology, Diabetes & Metabolism, Royal Lancaster Infirmary, University Hospitals of Morecambe Bay NHS Trust, Lancaster, UK
| | - Shithu Babu
- Department of Medicine, Dorset County Hospital, Dorchester, UK
| | - Babu Krishnan
- Department of Gastroenterology & Hepatology, Royal Bournemouth Hospital, Bournemouth, UK
| | - Nishal C Ravindran
- Department of Gastroenterology & Hepatology, Hinchingbrooke Hospital, Hinchingbrooke, Huntingdon, UK
| |
Collapse
|
76
|
Wu X, Dong L, Lin X, Li J. Relevance of the NLRP3 Inflammasome in the Pathogenesis of Chronic Liver Disease. Front Immunol 2017; 8:1728. [PMID: 29312290 PMCID: PMC5732938 DOI: 10.3389/fimmu.2017.01728] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 11/22/2017] [Indexed: 12/28/2022] Open
Abstract
Inflammation is a common characteristic of chronic liver disease (CLD). Inflammasomes are multiprotein complexes that can sense and recognize various exogenous and endogenous danger signals, eventually activating interleukin (IL)-1β and IL-18. The sensor component of the inflammasome system is a nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs). The NLRs family pyrin domain containing 3 (NLRP3) inflammasome has been involved in the initiation and progression of CLD. However, the molecular mechanisms by which it triggers liver inflammation and damage remain unclear. Here, we focus on recent advances on the potential role of NLRP3 inflammasome activation in the progression of CLD, including viral hepatitis, non-alcoholic steatohepatitis and alcoholic liver disease, and in particular, its ability to alleviate liver inflammation in animal models. Additionally, we also discuss various pharmacological inhibitors identifying the NLRP3 inflammasome signaling cascade as novel therapeutic targets in the treatment of CLD. In summary, this review summarizes the relevance of the NLRP3 inflammasome in the initiation and progression of CLD, and provides critical targets to suppress the development of CLD in clinical management.
Collapse
Affiliation(s)
- Xiaoqin Wu
- Department of Cardiology, First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Pediatrics, Division of Hematology/Oncology, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, United States.,School of Pharmacy, Institute for Liver Diseases of Anhui Medical University, ILDAMU, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Lei Dong
- Department of Pediatrics, Division of Hematology/Oncology, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, United States.,School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Xianhe Lin
- Department of Cardiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jun Li
- School of Pharmacy, Institute for Liver Diseases of Anhui Medical University, ILDAMU, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| |
Collapse
|
77
|
Mitsuyoshi H, Yasui K, Hara T, Taketani H, Ishiba H, Okajima A, Seko Y, Umemura A, Nishikawa T, Yamaguchi K, Moriguchi M, Minami M, Itoh Y. Hepatic nucleotide binding oligomerization domain-like receptors pyrin domain-containing 3 inflammasomes are associated with the histologic severity of non-alcoholic fatty liver disease. Hepatol Res 2017; 47:1459-1468. [PMID: 28245087 DOI: 10.1111/hepr.12883] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 02/22/2017] [Accepted: 02/25/2017] [Indexed: 12/13/2022]
Abstract
AIM To examine the role of nucleotide binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasomes in the development of non-alcoholic fatty liver disease (NAFLD). METHODS Levels of mRNAs encoding NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain, procaspase-1, interleukin (IL)-1β, and IL-18 were quantified by real-time polymerase chain reaction in 91 liver samples and 37 blood samples from biopsy-proven patients with NAFLD. Adiponutrin (also called PNPLA3) polymorphisms (rs738409, C > G) were determined in 74 samples by genotyping assays. Serum IL-1β and IL-18 levels were measured by enzyme-linked immunosorbent assay and liver tissue caspase-1 expression by immunostaining. RESULTS Hepatic NLRP3, procaspase-1, IL-1β, and IL-18 mRNA levels were significantly higher in NAFLD patients than in controls and were significantly associated with adiponutrin G alleles. Blood procaspase-1 mRNA was significantly higher in NAFLD patients than in healthy controls. Hepatic procaspase-1 and IL-1β mRNA levels correlated significantly with lobular inflammation, hepatocyte ballooning, and NAFLD activity score. Serum IL-18 levels were significantly higher in NAFLD patients than in controls, while IL-1β levels were non-significantly higher. Serum IL-1β and IL-18 concentrations correlated significantly with steatosis, NAFLD activity score, and transaminase levels. Serum IL-1β levels were significantly associated with adiponutrin G alleles. Scattered caspase-1-positive cells were present in portal tracts and inflammatory foci and around ballooning hepatocytes. Immunofluorescence staining showed that caspase-1 colocalized with the macrophage marker CD68. CONCLUSIONS The NLRP3 inflammasomes are primed in the liver, influenced by adiponutrin genotypes, and activated in Kupffer cells and/or macrophages in NAFLD, leading to histological progression through IL-1β and IL-18 production.
Collapse
Affiliation(s)
- Hironori Mitsuyoshi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kohichiroh Yasui
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tasuku Hara
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroyoshi Taketani
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroshi Ishiba
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akira Okajima
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuya Seko
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Atsushi Umemura
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Taichiro Nishikawa
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kanji Yamaguchi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Michihisa Moriguchi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masahito Minami
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshito Itoh
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
78
|
Greuter T, Malhi H, Gores GJ, Shah VH. Therapeutic opportunities for alcoholic steatohepatitis and nonalcoholic steatohepatitis: exploiting similarities and differences in pathogenesis. JCI Insight 2017; 2:95354. [PMID: 28878132 DOI: 10.1172/jci.insight.95354] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Alcoholic steatohepatitis (ASH) and nonalcoholic steatohepatitis (NASH) are among the most frequent causes of chronic liver disease in the United States. Although the two entities are triggered by different etiologies - chronic alcohol consumption (ASH) and obesity-associated lipotoxicity (NASH) - they share overlapping histological and clinical features owing to common pathogenic mechanisms. These pathogenic processes include altered hepatocyte lipid metabolism, organelle dysfunction (i.e., ER stress), hepatocyte apoptosis, innate immune system activation, and hepatic stellate cell activation. Nonetheless, there are several disease-specific molecular signaling pathways, such as differential pathway activation downstream of TLR4 (MyD88-dependence in NASH versus MyD88-independence in ASH), inflammasome activation and IL-1β signaling in ASH, insulin resistance and lipotoxicity in NASH, and dysregulation of different microRNAs, which clearly highlight that ASH and NASH are two distinct biological entities. Both pathogenic similarities and differences have therapeutic implications. In this Review, we discuss these pathogenic mechanisms and their therapeutic implications for each disease, focusing on both shared and distinct targets.
Collapse
Affiliation(s)
- Thomas Greuter
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland.,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Harmeet Malhi
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Gregory J Gores
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Vijay H Shah
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
79
|
NLRP3 inflammasome activation results in liver inflammation and fibrosis in mice infected with Schistosoma japonicum in a Syk-dependent manner. Sci Rep 2017; 7:8120. [PMID: 28808303 PMCID: PMC5556086 DOI: 10.1038/s41598-017-08689-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 07/06/2017] [Indexed: 02/06/2023] Open
Abstract
Granulomatous and fibrosing inflammation in response to soluble egg antigen (SEA) from Schistosoma japonicum (S. japonicum) is the main pathological process of S. japonicum infection. Inflammasome activation has recently been implicated in the pathogenesis of liver disease. However, the role of inflammasome activation in schistosomiasis-associated liver fibrosis (SSLF) has not been extensively studied. In this study, it is demonstrated that the NLRP3 inflammasome is markedly activated in mouse HSCs both in vivo and in vitro during S. japonicum infection. Furthermore, it is demonstrated that inhibition of NLRP3 inflammasome significantly alleviates the liver inflammation and collagen deposition that are induced by infection with S. japonicum. The mechanism of SEA-induced NLRP3 inflammasome activation is studied in isolated, cultured mouse HSCs and it is shown that SEA-induced NLRP3 inflammasome activation in HSCs is dependent upon the activities of spleen tyrosine kinase (Syk), an enzyme usually associated with a pathogen recognition receptor for fungal pathogens. Moreover, it is demonstrated that Dectin-1 and JNK signaling are also involved in SEA-induced NLRP3 inflammasome activation in HSCs. These data shed new light on the mechanisms of NLRP3 inflammasome activation during an infection with S. japonicum, and further characterize its role in schistosomiasis-associated liver fibrosis (SSLF).
Collapse
|
80
|
Yang ZY, Liu F, Liu PH, Guo WJ, Xiong GY, Pan H, Wei L. Obeticholic acid improves hepatic steatosis and inflammation by inhibiting NLRP3 inflammasome activation. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:8119-8129. [PMID: 31966664 PMCID: PMC6965461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 05/28/2017] [Indexed: 06/10/2023]
Abstract
BACKGROUND AND AIM Several pre-clinical and clinical researches have proved that obeticholic acid (OCA)has a potential therapeutic effect on non-alcoholic steatohepatitis (NASH). Our aim was to investigate whether the therapeutic effect of OCA on NASH was attributed to its inhibition effect on cytosolic sensor NLR family pyrin domain containing 3 (NLRP3) inflammasome activation. METHODS We used mice model of methionine-choline-deficient (MCD) diet induced NASH. At different fibrosis stages, the expressions of NLRP3, caspase-1 and IL-1β were analyzed by means of immunohistochemistry and western blot respectively. After daily gavage of 0.4 mg of OCA or vehicle for 24 days, we evaluated the direct effect of OCA on NLRP3 inflammasome activation by analyzing the expressions of NLRP3 and IL-1β. Additionally, liver function and liver histology of mice were assessed. The expressions of NLRP3 and IL-1β above and the expressions of fibrosis-related genes were analyzed by quantitative real-time polymerase chain reaction (PCR). RESULTS NLRP3 inflammasome activation could be observed in liver fibrosis, and we found that the expressions of NLRP3, caspase-1 and IL-1β gradually increased to peak at stage 2-3 but decreased significantly at stage 4 of liver fibrosis in MCD mice model. We also found that short-term OCA treatment could significantly down-regulate the expressions of NLRP3 and IL-1β and therefore improved NASH-associated steatosis and inflammation. CONCLUSIONS NLRP3 inflammasome could be activated and might have an essential role in NASH progression, and short-term OCA treatment could have a potential therapeutic effect on NASH-associated steatosis and inflammation by inhibiting NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Zhi-Yu Yang
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver DiseasesBeijing 100044, China
| | - Feng Liu
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver DiseasesBeijing 100044, China
| | - Pei-Hao Liu
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver DiseasesBeijing 100044, China
| | - Wan-Jun Guo
- Medical Research Department, Kawin Technology Co. LtdBeijing 102600, China
| | - Guo-Yu Xiong
- Medical Research Department, Kawin Technology Co. LtdBeijing 102600, China
| | - Hai Pan
- Medical Research Department, Kawin Technology Co. LtdBeijing 102600, China
| | - Lai Wei
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver DiseasesBeijing 100044, China
| |
Collapse
|
81
|
Mridha AR, Wree A, Robertson AA, Yeh MM, Johnson CD, Van Rooyen DM, Haczeyni F, Teoh NCH, Savard C, Ioannou GN, Masters SL, Schroder K, Cooper MA, Feldstein AE, Farrell GC. NLRP3 inflammasome blockade reduces liver inflammation and fibrosis in experimental NASH in mice. J Hepatol 2017; 66:1037-1046. [PMID: 28167322 PMCID: PMC6536116 DOI: 10.1016/j.jhep.2017.01.022] [Citation(s) in RCA: 807] [Impact Index Per Article: 100.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 01/04/2017] [Accepted: 01/06/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS NOD-like receptor protein 3 (NLRP3) inflammasome activation occurs in Non-alcoholic fatty liver disease (NAFLD). We used the first small molecule NLRP3 inhibitor, MCC950, to test whether inflammasome blockade alters inflammatory recruitment and liver fibrosis in two murine models of steatohepatitis. METHODS We fed foz/foz and wild-type mice an atherogenic diet for 16weeks, gavaged MCC950 or vehicle until 24weeks, then determined NAFLD phenotype. In mice fed an methionine/choline deficient (MCD) diet, we gavaged MCC950 or vehicle for 6weeks and determined the effects on liver fibrosis. RESULTS In vehicle-treated foz/foz mice, hepatic expression of NLRP3, pro-IL-1β, active caspase-1 and IL-1β increased at 24weeks, in association with cholesterol crystal formation and NASH pathology; plasma IL-1β, IL-6, MCP-1, ALT/AST all increased. MCC950 treatment normalized hepatic caspase 1 and IL-1β expression, plasma IL-1β, MCP-1 and IL-6, lowered ALT/AST, and reduced the severity of liver inflammation including designation as NASH pathology, and liver fibrosis. In vitro, cholesterol crystals activated Kupffer cells and macrophages to release IL-1β; MCC950 abolished this, and the associated neutrophil migration. MCD diet-fed mice developed fibrotic steatohepatitis; MCC950 suppressed the increase in hepatic caspase 1 and IL-1β, lowered numbers of macrophages and neutrophils in the liver, and improved liver fibrosis. CONCLUSION MCC950, an NLRP3 selective inhibitor, improved NAFLD pathology and fibrosis in obese diabetic mice. This is potentially attributable to the blockade of cholesterol crystal-mediated NLRP3 activation in myeloid cells. MCC950 reduced liver fibrosis in MCD-fed mice. Targeting NLRP3 is a logical direction in pharmacotherapy of NASH. LAY SUMMARY Fatty liver disease caused by being overweight with diabetes and a high risk of heart attack, termed non-alcoholic steatohepatitis (NASH), is the most common serious liver disease with no current treatment. There could be several causes of inflammation in NASH, but activation of a protein scaffold within cells termed the inflammasome (NLRP3) has been suggested to play a role. Here we show that cholesterol crystals could be one pathway to activate the inflammasome in NASH. We used a drug called MCC950, which has already been shown to block NLRP3 activation, in an attempt to reduce liver injury in NASH. This drug partly reversed liver inflammation, particularly in obese diabetic mice that most closely resembles the human context of NASH. In addition, such dampening of liver inflammation in NASH achieved with MCC950 partly reversed liver scarring, the process that links NASH to the development of cirrhosis.
Collapse
Affiliation(s)
- Auvro R. Mridha
- Liver Research Group, ANU Medical School, Australian National University at The Canberra Hospital, Garran, ACT, Australia
| | - Alexander Wree
- Department of Internal Medicine III, RWTH-Aachen University Hospital, Aachen, Germany;,Department of Pediatrics, University of California – San Diego, La Jolla, San Diego, CA, United States
| | - Avril A.B. Robertson
- Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD, Australia
| | - Matthew M. Yeh
- Department of Pathology, University of Washington, Seattle, WA, United States
| | - Casey D. Johnson
- Department of Pediatrics, University of California – San Diego, La Jolla, San Diego, CA, United States
| | - Derrick M. Van Rooyen
- Liver Research Group, ANU Medical School, Australian National University at The Canberra Hospital, Garran, ACT, Australia
| | - Fahrettin Haczeyni
- Liver Research Group, ANU Medical School, Australian National University at The Canberra Hospital, Garran, ACT, Australia
| | - Narci C.-H. Teoh
- Liver Research Group, ANU Medical School, Australian National University at The Canberra Hospital, Garran, ACT, Australia
| | - Christopher Savard
- Department of Gastroenterology and Hepatology, Veterans Affairs Puget Sound Health Care System and University of Washington, Seattle, WA, United States
| | - George N. Ioannou
- Department of Gastroenterology and Hepatology, Veterans Affairs Puget Sound Health Care System and University of Washington, Seattle, WA, United States
| | - Seth L. Masters
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
| | - Kate Schroder
- Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD, Australia
| | - Matthew A. Cooper
- Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD, Australia
| | - Ariel E. Feldstein
- Department of Pediatrics, University of California – San Diego, La Jolla, San Diego, CA, United States
| | - Geoffrey C. Farrell
- Liver Research Group, ANU Medical School, Australian National University at The Canberra Hospital, Garran, ACT, Australia;,Corresponding author. Address: Gastroenterology and Hepatology Unit, The Canberra Hospital, PO Box 111, Woden, ACT 2605, Australia. Fax: +61 2 62443235. (G.C. Farrell)
| |
Collapse
|
82
|
Wang X, Gao Y, Song J, Tang C, Wang M, Que L, Liu L, Zhu G, Chen Q, Yao Y, Xu Y, Li J, Li Y. The TIR/BB-loop mimetic AS-1 prevents non-alcoholic steatohepatitis and hepatic insulin resistance by inhibiting NLRP3-ASC inflammasome activation. Br J Pharmacol 2017; 174:1841-1856. [PMID: 28306139 DOI: 10.1111/bph.13786] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 03/09/2017] [Accepted: 03/13/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Non-alcoholic steatohepatitis (NASH) is characterized by excessive intracellular lipid accumulation, inflammation and hepatic insulin resistance. As the incidence of NASH is increasing worldwide, there is an urgent need to find novel interventional approaches. The pro-inflammatory cytokine IL-1β, generated and released from Kupffer cells, is considered to initiate the development of NASH. AS-1, a synthetic low-molecule mimetic of myeloid differentiation primary response gene 88 (MyD88), disrupts the interaction between the IL-1 receptor and MyD88. Here, we investigated whether AS-1 could attenuate the pathogenesis of NASH with an emphasis on hepatic insulin resistance. EXPERIMENTAL APPROACH Eight-week-old db/db mice were fed a control diet or a methionine- and choline-deficient (MCD) diet. AS-1 (50 mg·kg-1 ) or vehicle was administered i.p. KEY RESULTS AS-1 administration significantly ameliorated NASH as evidenced by alanine aminotransferase levels and CD68 levels in livers of MCD-fed mice. AS-1 inhibited the MCD diet-induced activation of caspase 1 and the NLRP3-ASC inflammasome, and also reduced the enhanced levels of ROS, malondialdehyde, 3-nitrotyrosine, NADPH oxidase complex and CYP reductase-associated cytochrome p450 2E1 (CYP2E1) expression in the liver. In addition, AS-1 decreased ROS, inflammasome activation and IL-1β production in free fatty acid-LPS-treated Kupffer cells. Finally, pretreatment with AS-1 significantly ameliorated gluconeogenesis and insulin desensitization induced by IL-1β, probably by blocking the interaction between MyD88 and the IL-1 receptor. CONCLUSIONS AND IMPLICATIONS Our results indicate that AS-1 can ameliorate NASH and hepatic insulin resistance and could be considered as a potential strategy for the prevention and treatment of NASH.
Collapse
Affiliation(s)
- Xiaolu Wang
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yun Gao
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu, China.,The Affiliated Kunshan Hospital of Jiangsu University, The First People's Hospital of Kunshan, Suzhou, Jiangsu, China
| | - Juan Song
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chao Tang
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Man Wang
- Department of Ophthalmology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Linli Que
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Li Liu
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guoqing Zhu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qi Chen
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yong Yao
- Department of Ophthalmology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Yong Xu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiantao Li
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuehua Li
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
83
|
Cao Z, Fang Y, Lu Y, Tan D, Du C, Li Y, Ma Q, Yu J, Chen M, Zhou C, Pei L, Zhang L, Ran H, He M, Yu Z, Zhou Z. Melatonin alleviates cadmium-induced liver injury by inhibiting the TXNIP-NLRP3 inflammasome. J Pineal Res 2017; 62. [PMID: 28099758 DOI: 10.1111/jpi.12389] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 01/12/2017] [Indexed: 02/06/2023]
Abstract
Cadmium (Cd) is a persistent environmental and occupational contaminant that accumulates in the liver and induces oxidative stress and inflammation. Melatonin possesses potent hepatoprotective properties against the development and progression of acute and chronic liver injury. Nevertheless, the molecular mechanism underlying the protective effects of melatonin against Cd-induced hepatotoxicity remains obscure. In this study, we aimed to investigate the effects of melatonin on Cd-induced liver inflammation and hepatocyte death. Male C57BL/6 mice were intraperitoneally injected with melatonin (10 mg/kg) once a day for 3 days before exposure to CdCl2 (2.0 mg/kg). We found that Cd induced hepatocellular damage and inflammatory infiltration as well as increased serum ALT/AST enzymes. In addition, we showed that Cd triggered an inflammatory cell death, which is mediated by the NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome. Moreover, melatonin treatment significantly alleviated Cd-induced liver injury by decreasing serum ALT/AST levels, suppressing pro-inflammatory cytokine production, inhibiting NLRP3 inflammasome activation, ameliorating oxidative stress, and attenuating hepatocyte death. Most importantly, melatonin markedly abrogated Cd-induced TXNIP overexpression and decreased the interaction between TXNIP and NLRP3 in vivo and in vitro. However, treatment with siRNA targeting TXNIP blocked the protective effects of melatonin in Cd-treated primary hepatocytes. Collectively, our results suggest that melatonin confers protection against Cd-induced liver inflammation and hepatocyte death via inhibition of the TXNIP-NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Zhengwang Cao
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Yiliang Fang
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Yonghui Lu
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Dunxian Tan
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Changhong Du
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, Third Military Medical University, Chongqing, China
| | - Yuming Li
- Institute of Hepatobiliary Surgery, XinQiao Hospital, Third Military Medical University, Chongqing, China
| | - Qinlong Ma
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Junmei Yu
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Mengyan Chen
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Chao Zhou
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Liping Pei
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Lei Zhang
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Haiying Ran
- Biomedical Analysis Center, Third Military Medical University, Chongqing, China
| | - Mindi He
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Zhengping Yu
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Zhou Zhou
- Department of Occupational Health, Third Military Medical University, Chongqing, China
- Department of Occupational Health, School of Medicine, Guangxi University, Nanning, China
| |
Collapse
|
84
|
Lana JP, Martins LB, Oliveira MCD, Menezes-Garcia Z, Yamada LTP, Vieira LQ, Teixeira MM, Ferreira AVM. TNF and IL-18 cytokines may regulate liver fat storage under homeostasis conditions. Appl Physiol Nutr Metab 2017; 41:1295-1302. [PMID: 27863204 DOI: 10.1139/apnm-2016-0265] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The inflammation induced by obesogenic diets is associated with deposition of fat in the liver. On the other hand, anti-inflammatory and immunosuppressive therapies may impact in body fat storage and in liver lipid dynamics. It is important to study specific inflammatory mediators in this context, since their role on hepatic damage is not fully clarified. This study aimed to evaluate the role of interleukin (IL)-18 and tumor necrosis factor (TNF) receptor in liver dysfunction induced by diet. Male C57BL/6 wild-type (WT), IL-18, and TNF receptor 1 knockout mice (IL-18-/- and TNFR1-/-) were divided according to the experimental diets: chow diet or a high-refined carbohydrate-containing diet. Alanine aminotransferase was quantified by colorimetric analysis. Total fat content in the liver was determined by Folch methods. Levels of TNF, IL-6, IL-4, and IL-13 in liver samples were measured by ELISA assay. IL-18 and TNFR knockout mice fed with chow diet showed higher liver triglycerides deposition than WT mice fed with the same diet (WT: 131.9 ± 24.5; IL-18-/-: 239.4 ± 38.12*; TNF-/-: 179.6 ± 50.45*; *P < 0.01). Furthermore, these animals also showed a worse liver histopathological score and lower levels of TNF, IL-6, IL-4, and IL-13 in the liver. Interestingly, treatment with a high-carbohydrate diet did not exacerbate liver damage in IL-18-/- and TNFR1-/- mice. Our data suggest that IL-18 and TNF may be involved on hepatic homeostasis mainly in a context of a healthy diet.
Collapse
Affiliation(s)
- Jaqueline Pereira Lana
- a Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,b Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Laís Bhering Martins
- a Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,b Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marina Chaves de Oliveira
- a Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,b Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Zélia Menezes-Garcia
- b Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Leda Quercia Vieira
- b Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mauro Martins Teixeira
- b Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Adaliene Versiani Matos Ferreira
- a Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,b Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
85
|
Burgy O, Wettstein G, Bellaye PS, Decologne N, Racoeur C, Goirand F, Beltramo G, Hernandez JF, Kenani A, Camus P, Bettaieb A, Garrido C, Bonniaud P. Deglycosylated bleomycin has the antitumor activity of bleomycin without pulmonary toxicity. Sci Transl Med 2016; 8:326ra20. [PMID: 26888428 DOI: 10.1126/scitranslmed.aad7785] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Bleomycin (BLM) is a potent anticancer drug used to treat different malignancies, mainly lymphomas, germ cell tumors, and melanomas. Unfortunately, BLM has major, dose-dependent, pulmonary toxicity that affects 20% of treated individuals. The most severe form of BLM-induced pulmonary toxicity is lung fibrosis. Deglyco-BLM is a molecule derived from BLM in which the sugar residue d-mannosyl-l-glucose disaccharide has been deleted. The objective of this study was to assess the anticancer activity and lung toxicity of deglyco-BLM. We compared the antitumor activity and pulmonary toxicity of intraperitoneally administrated deglyco-BLM and BLM in three rodent models. Pulmonary toxicity was examined in depth after intratracheal administration of both chemotherapeutic agents. The effect of both drugs was further studied in epithelial alveolar cells in vitro. We demonstrated in rodent cancer models, including a human Hodgkin's lymphoma xenograft and a syngeneic melanoma model, that intraperitoneal deglyco-BLM is as effective as BLM in inducing tumor regression. Whereas the antitumor effect of BLM was accompanied by a loss of body weight and the development of pulmonary toxicity, deglyco-BLM did not affect body weight and did not engender lung injury. Both molecules induced lung epithelial cell apoptosis after intratracheal administration, but deglyco-BLM lost the ability to induce caspase-1 activation and the production of ROS (reactive oxygen species), transforming growth factor-β1, and other profibrotic and inflammatory cytokines in the lungs of mice and in vitro. Deglyco-BLM should be considered for clinical testing as a less toxic alternative to BLM in cancer therapy.
Collapse
Affiliation(s)
- Olivier Burgy
- INSERM, LNC UMR 866, Laboratoire d'Excellence LipSTIC, Dijon 21079, France. Equipe "Heat Shock Proteins" Labellisée par la Ligue Nationale contre le Cancer, Dijon 21079, France. Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France
| | - Guillaume Wettstein
- INSERM, LNC UMR 866, Laboratoire d'Excellence LipSTIC, Dijon 21079, France. Equipe "Heat Shock Proteins" Labellisée par la Ligue Nationale contre le Cancer, Dijon 21079, France. Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France
| | - Pierre S Bellaye
- INSERM, LNC UMR 866, Laboratoire d'Excellence LipSTIC, Dijon 21079, France. Equipe "Heat Shock Proteins" Labellisée par la Ligue Nationale contre le Cancer, Dijon 21079, France. Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France
| | - Nathalie Decologne
- Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France. EPHE, Tumor Immunology and Immunotherapy Laboratory, Dijon 21079, France
| | - Cindy Racoeur
- Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France. EPHE, Tumor Immunology and Immunotherapy Laboratory, Dijon 21079, France
| | - Françoise Goirand
- INSERM, LNC UMR 866, Laboratoire d'Excellence LipSTIC, Dijon 21079, France. Equipe "Heat Shock Proteins" Labellisée par la Ligue Nationale contre le Cancer, Dijon 21079, France. Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France
| | - Guillaume Beltramo
- INSERM, LNC UMR 866, Laboratoire d'Excellence LipSTIC, Dijon 21079, France. Equipe "Heat Shock Proteins" Labellisée par la Ligue Nationale contre le Cancer, Dijon 21079, France. Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France. Service de Pneumologie et Soins Intensifs Respiratoires, Centre Hospitalier Universitaire (CHU), Dijon 21079, France
| | - Jean-François Hernandez
- Institut des Biomolécules Max Mousseron, Faculty of Pharmacy, University of Montpellier, Montpellier 34093, France
| | - Abderraouf Kenani
- Department of Biochemistry, University of Monastir, Monastir 5000, Tunisia
| | - Philippe Camus
- INSERM, LNC UMR 866, Laboratoire d'Excellence LipSTIC, Dijon 21079, France. Equipe "Heat Shock Proteins" Labellisée par la Ligue Nationale contre le Cancer, Dijon 21079, France. Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France. Service de Pneumologie et Soins Intensifs Respiratoires, Centre Hospitalier Universitaire (CHU), Dijon 21079, France
| | - Ali Bettaieb
- Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France. EPHE, Tumor Immunology and Immunotherapy Laboratory, Dijon 21079, France
| | - Carmen Garrido
- INSERM, LNC UMR 866, Laboratoire d'Excellence LipSTIC, Dijon 21079, France. Equipe "Heat Shock Proteins" Labellisée par la Ligue Nationale contre le Cancer, Dijon 21079, France. Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France. Anticancer Centre Georges François Leclerc, CGFL, Dijon 21079, France
| | - Philippe Bonniaud
- INSERM, LNC UMR 866, Laboratoire d'Excellence LipSTIC, Dijon 21079, France. Equipe "Heat Shock Proteins" Labellisée par la Ligue Nationale contre le Cancer, Dijon 21079, France. Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France. Service de Pneumologie et Soins Intensifs Respiratoires, Centre Hospitalier Universitaire (CHU), Dijon 21079, France.
| |
Collapse
|
86
|
Hepatocyte TAZ/WWTR1 Promotes Inflammation and Fibrosis in Nonalcoholic Steatohepatitis. Cell Metab 2016; 24:848-862. [PMID: 28068223 PMCID: PMC5226184 DOI: 10.1016/j.cmet.2016.09.016] [Citation(s) in RCA: 315] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 08/06/2016] [Accepted: 09/24/2016] [Indexed: 12/22/2022]
Abstract
Nonalcoholic steatohepatitis (NASH) is a leading cause of liver disease worldwide. However, the molecular basis of how benign steatosis progresses to NASH is incompletely understood, which has limited the identification of therapeutic targets. Here we show that the transcription regulator TAZ (WWTR1) is markedly higher in hepatocytes in human and murine NASH liver than in normal or steatotic liver. Most importantly, silencing of hepatocyte TAZ in murine models of NASH prevented or reversed hepatic inflammation, hepatocyte death, and fibrosis, but not steatosis. Moreover, hepatocyte-targeted expression of TAZ in a model of steatosis promoted NASH features, including fibrosis. In vitro and in vivo mechanistic studies revealed that a key mechanism linking hepatocyte TAZ to NASH fibrosis is TAZ/TEA domain (TEAD)-mediated induction of Indian hedgehog (Ihh), a secretory factor that activates fibrogenic genes in hepatic stellate cells. In summary, TAZ represents a previously unrecognized factor that contributes to the critical process of steatosis-to-NASH progression.
Collapse
|
87
|
Magee N, Zou A, Zhang Y. Pathogenesis of Nonalcoholic Steatohepatitis: Interactions between Liver Parenchymal and Nonparenchymal Cells. BIOMED RESEARCH INTERNATIONAL 2016; 2016:5170402. [PMID: 27822476 PMCID: PMC5086374 DOI: 10.1155/2016/5170402] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/22/2016] [Indexed: 12/14/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common type of chronic liver disease in the Western countries, affecting up to 25% of the general population and becoming a major health concern in both adults and children. NAFLD encompasses the entire spectrum of fatty liver disease in individuals without significant alcohol consumption, ranging from nonalcoholic fatty liver (NAFL) to nonalcoholic steatohepatitis (NASH) and cirrhosis. NASH is a manifestation of the metabolic syndrome and hepatic disorders with the presence of steatosis, hepatocyte injury (ballooning), inflammation, and, in some patients, progressive fibrosis leading to cirrhosis. The pathogenesis of NASH is a complex process and implicates cell interactions between liver parenchymal and nonparenchymal cells as well as crosstalk between various immune cell populations in liver. Lipotoxicity appears to be the central driver of hepatic cellular injury via oxidative stress and endoplasmic reticulum (ER) stress. This review focuses on the contributions of hepatocytes and nonparenchymal cells to NASH, assessing their potential applications to the development of novel therapeutic agents. Currently, there are limited pharmacological treatments for NASH; therefore, an increased understanding of NASH pathogenesis is pertinent to improve disease interventions in the future.
Collapse
Affiliation(s)
- Nancy Magee
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - An Zou
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Yuxia Zhang
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
88
|
Zhong G. Chlamydial Plasmid-Dependent Pathogenicity. Trends Microbiol 2016; 25:141-152. [PMID: 27712952 DOI: 10.1016/j.tim.2016.09.006] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/11/2016] [Accepted: 09/19/2016] [Indexed: 10/20/2022]
Abstract
Most Chlamydia species carry a 7.5kb plasmid encoding eight open reading frames conventionally called plasmid glycoproteins 1-8 or pGP1-8. Although the plasmid is not critical for chlamydial growth in vitro, its role in chlamydial pathogenesis is clearly demonstrated in the genital tracts of mice infected with Chlamydia muridarum, a model for investigating the human pathogen Chlamydia trachomatis. Plasmid-free C. trachomatis is also attenuated in both the mouse genital tract and nonhuman primate ocular tissue. Deficiency in pGP3 alone, which is regulated by pGP4, largely reproduced the in vivo but not in vitro phenotypes of the plasmid-free organisms, suggesting that pGP3 is a key in vivo virulence factor. The positive and negative regulations of some chromosomal genes by pGP4 and pGP5, respectively, may allow the plasmid to promote chlamydial adaptation to varied animal tissue environments. The focus of this review is to summarize the progress on the pathogenic functions of the plasmid-encoded open reading frames, which may motivate further investigation of the molecular mechanisms of chlamydial pathogenicity and development of medical utility of the chlamydial plasmid system.
Collapse
Affiliation(s)
- Guangming Zhong
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| |
Collapse
|
89
|
Tilg H, Moschen AR, Szabo G. Interleukin-1 and inflammasomes in alcoholic liver disease/acute alcoholic hepatitis and nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. Hepatology 2016; 64:955-65. [PMID: 26773297 DOI: 10.1002/hep.28456] [Citation(s) in RCA: 246] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 01/09/2016] [Indexed: 12/12/2022]
Abstract
UNLABELLED Both alcoholic liver disease (ALD) and nonalcoholic fatty liver disease are characterized by massive lipid accumulation in the liver accompanied by inflammation, fibrosis, cirrhosis, and hepatocellular carcinoma in a substantial subgroup of patients. At several stages in these diseases, mediators of the immune system, such as cytokines or inflammasomes, are crucially involved. In ALD, chronic ethanol exposure sensitizes Kupffer cells to activation by lipopolysaccharides through Toll-like receptors, e.g., Toll-like receptor 4. This sensitization enhances the production of various proinflammatory cytokines such as interleukin-1 (IL-1) and tumor necrosis factor-alpha, thereby contributing to hepatocyte dysfunction, necrosis, and apoptosis and the generation of extracellular matrix proteins leading to fibrosis/cirrhosis. Indeed, neutralization of IL-1 by IL-1 receptor antagonist has recently been shown to potently prevent liver injury in murine models of ALD. As IL-1 is clearly linked to key clinical symptoms of acute alcoholic hepatitis such as fever, neutrophilia, and wasting, interfering with the IL-1 pathway might be an attractive treatment strategy in the future. An important role for IL-1-type cytokines and certain inflammasomes has also been demonstrated in murine models of nonalcoholic fatty liver disease. IL-1-type cytokines can regulate hepatic steatosis; the NLR family pyrin domain containing 3 inflammasome is critically involved in metabolic dysregulation. CONCLUSION IL-1 cytokine family members and various inflammasomes mediate different aspects of both ALD and nonalcoholic fatty liver disease. (Hepatology 2016;64:955-965).
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Alexander R Moschen
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Gyongyi Szabo
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USAMA
| |
Collapse
|
90
|
Bashiardes S, Shapiro H, Rozin S, Shibolet O, Elinav E. Non-alcoholic fatty liver and the gut microbiota. Mol Metab 2016; 5:782-94. [PMID: 27617201 PMCID: PMC5004228 DOI: 10.1016/j.molmet.2016.06.003] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 06/04/2016] [Accepted: 06/08/2016] [Indexed: 02/06/2023] Open
Abstract
Background Non-alcoholic fatty liver (NAFLD) is a common, multi-factorial, and poorly understood liver disease whose incidence is globally rising. NAFLD is generally asymptomatic and associated with other manifestations of the metabolic syndrome. Yet, up to 25% of NAFLD patients develop a progressive inflammatory liver disease termed non-alcoholic steatohepatitis (NASH) that may progress towards cirrhosis, hepatocellular carcinoma, and the need for liver transplantation. In recent years, several lines of evidence suggest that the gut microbiome represents a significant environmental factor contributing to NAFLD development and its progression into NASH. Suggested microbiome-associated mechanisms contributing to NAFLD and NASH include dysbiosis-induced deregulation of the gut endothelial barrier function, which facilitates systemic bacterial translocation, and intestinal and hepatic inflammation. Furthermore, increased microbiome-modulated metabolites such as lipopolysaccharides, short chain fatty acids (SCFAs), bile acids, and ethanol, may affect liver pathology through multiple direct and indirect mechanisms. Scope of review Herein, we discuss the associations, mechanisms, and clinical implications of the microbiome's contribution to NAFLD and NASH. Understanding these contributions to the development of fatty liver pathogenesis and its clinical course may serve as a basis for development of therapeutic microbiome-targeting approaches for treatment and prevention of NAFLD and NASH. Major conclusions Intestinal host–microbiome interactions play diverse roles in the pathogenesis and progression of NAFLD and NASH. Elucidation of the mechanisms driving these microbial effects on the pathogenesis of NAFLD and NASH may enable to identify new diagnostic and therapeutic targets of these common metabolic liver diseases. This article is part of a special issue on microbiota.
Collapse
Affiliation(s)
- Stavros Bashiardes
- Immunology Department, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Hagit Shapiro
- Immunology Department, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Shachar Rozin
- Immunology Department, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Oren Shibolet
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel; Department of Gastroenterology, Tel Aviv Medical Center, Tel Aviv 6423906, Israel
| | - Eran Elinav
- Immunology Department, Weizmann Institute of Science, 76100 Rehovot, Israel
| |
Collapse
|
91
|
Zhang S, Huang D, Weng J, Huang Y, Liu S, Zhang Q, Li N, Wen M, Zhu G, Lin F, Gu W. Neutralization of Interleukin-17 Attenuates Cholestatic Liver Fibrosis in Mice. Scand J Immunol 2016; 83:102-8. [PMID: 26484852 DOI: 10.1111/sji.12395] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/15/2015] [Indexed: 12/20/2022]
Abstract
Anti-inflammation strategy is one of the proposed therapeutic approaches to hepatic fibrosis. IL-17 is critical in inflammation, but the role of IL-17 in liver fibrosis has not yet been elucidated. In this study, we investigate the role of IL-17 on bile duct ligation-induced liver injury and fibrosis in C57BL/6 mice. Animals were sacrificed at designated times, and serum and liver tissues were collected for analysis of liver function and serum IL-6, IL-1β, tumour necrosis factor-alpha (TNF-α) and transforming growth factor-β (TGF-β) levels. IL-17 blockade with anti-IL-17A mAb significantly improved liver function and decreased hepatocellular necrosis, pro-inflammatory cytokines, neutrophils and macrophages influx. Furthermore, CD3 + and CD8 + lymphocytes, neutrophils and macrophages were found to express IL-17, and neutrophils are the principal IL-17-producing cells after BDL-induced liver injury. These data indicated that IL-17 signal contributes to the pathogenesis of cholestatic liver injury and blocked of IL-17 could potentially benefit patients with cholestatic liver disease.
Collapse
Affiliation(s)
- S Zhang
- Department of Hepatopancreatobiliary Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou Digestive Disease Center, Guangzhou, 510180, China
| | - D Huang
- Department of Hepatopancreatobiliary Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou Digestive Disease Center, Guangzhou, 510180, China
| | - J Weng
- Department of Hepatopancreatobiliary Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou Digestive Disease Center, Guangzhou, 510180, China
| | - Y Huang
- Department of Hepatopancreatobiliary Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou Digestive Disease Center, Guangzhou, 510180, China
| | - S Liu
- Department of Hepatopancreatobiliary Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou Digestive Disease Center, Guangzhou, 510180, China
| | - Q Zhang
- Department of Hepatopancreatobiliary Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou Digestive Disease Center, Guangzhou, 510180, China
| | - N Li
- Department of Hepatopancreatobiliary Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou Digestive Disease Center, Guangzhou, 510180, China
| | - M Wen
- Department of Hepatopancreatobiliary Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou Digestive Disease Center, Guangzhou, 510180, China
| | - G Zhu
- Department of Hepatopancreatobiliary Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou Digestive Disease Center, Guangzhou, 510180, China
| | - F Lin
- Department of Hepatopancreatobiliary Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou Digestive Disease Center, Guangzhou, 510180, China
| | - W Gu
- Department of Hepatopancreatobiliary Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou Digestive Disease Center, Guangzhou, 510180, China
| |
Collapse
|
92
|
Intervention with a caspase-1 inhibitor reduces obesity-associated hyperinsulinemia, non-alcoholic steatohepatitis and hepatic fibrosis in LDLR-/-.Leiden mice. Int J Obes (Lond) 2016; 40:1416-23. [PMID: 27121255 PMCID: PMC5022108 DOI: 10.1038/ijo.2016.74] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 04/07/2016] [Accepted: 04/08/2016] [Indexed: 12/13/2022]
Abstract
Background/Objectives: Non-alcoholic steatohepatitis (NASH) is a serious liver condition, closely associated with obesity and insulin resistance. Recent studies have suggested an important role for inflammasome/caspase-1 in the development of NASH, but the potential therapeutic value of caspase-1 inhibition remains unclear. Therefore, we aimed to investigate the effects of caspase-1 inhibition in the ongoing disease process, to mimic the clinical setting. Subjects/Methods: To investigate effects of caspase-1 inhibition under therapeutic conditions, male LDLR−/−.Leiden mice were fed a high-fat diet (HFD) for 9 weeks to induce a pre-diabetic state before start of treatment. Mice were then continued on HFD for another 12 weeks, without (HFD) or with (HFD-YVAD) treatment with the caspase-1 inhibitor Ac-YVAD-cmk (40 mg kg−1 per day). Results: Nine weeks of HFD feeding resulted in an obese phenotype, with obesity-associated hypertriglyceridemia, hypercholesterolemia, hyperglycemia and hyperinsulinemia. Treatment with Ac-YVAD-cmk did not affect further body weight gain or dyslipidemia, but did attenuate further progression of insulin resistance. Histopathological analysis of livers clearly demonstrated prevention of NASH development in HFD-YVAD mice: livers were less steatotic and neutrophil infiltration was strongly reduced. In addition, caspase-1 inhibition had a profound effect on hepatic fibrosis, as assessed by histological quantification of collagen staining and gene expression analysis of fibrosis-associated genes Col1a1, Acta2 and Tnfa. Conclusions: Intervention with a caspase-1 inhibitor attenuated the development of NASH, liver fibrosis and insulin resistance. Our data support the importance of inflammasome/caspase-1 in the development of NASH and demonstrate that therapeutic intervention in the already ongoing disease process is feasible.
Collapse
|
93
|
Wan X, Xu C, Yu C, Li Y. Role of NLRP3 Inflammasome in the Progression of NAFLD to NASH. Can J Gastroenterol Hepatol 2016; 2016:6489012. [PMID: 27446858 PMCID: PMC4904645 DOI: 10.1155/2016/6489012] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 07/20/2015] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has been recognized as a major public health problem worldwide. Nonalcoholic steatohepatitis (NASH) is an advanced form of NAFLD that may progress to cirrhosis and hepatocellular carcinoma. The pathogenesis of disease progression from NAFLD to NASH has not been fully understood. Immunological mechanisms that have been increasingly recognized in the disease progression include defects in innate immunity, adaptive immunity, Toll-like receptor (TLR) signaling, and gut-liver axis. The NLRP3 inflammasome is an intracellular multiprotein complex involved in the production of mature interleukin 1-beta (IL-1β) and induces metabolic inflammation. NLRP3 inflammasome has been recently demonstrated to play a crucial role in the progression of NASH. This review highlights the recent findings linking NLRP3 inflammasome to the progression of NASH.
Collapse
Affiliation(s)
- Xingyong Wan
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Chengfu Xu
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Chaohui Yu
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Youming Li
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
94
|
Yang G, Lee HE, Lee JY. A pharmacological inhibitor of NLRP3 inflammasome prevents non-alcoholic fatty liver disease in a mouse model induced by high fat diet. Sci Rep 2016; 6:24399. [PMID: 27075683 PMCID: PMC4830938 DOI: 10.1038/srep24399] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/29/2016] [Indexed: 12/17/2022] Open
Abstract
The activation of NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome is closely associated with the development and progression of non-alcoholic fatty liver disease (NAFLD) induced by a high-fat diet. Therefore, we investigated whether oral administration of sulforaphane (SFN) prevented high-fat diet-induced NAFLD in mice by regulation of the NLRP3 inflammasome in the liver. Daily oral administrations of SFN reduced hepatic steatosis scores, serum ALT and AST levels, and hepatic levels of cholesterol, triglycerides, and free fatty acids in mice fed a high-fat diet. These were correlated with the suppression of NLRP3 inflammasome activation in the liver by SFN as evidenced by decrease in mRNA levels of ASC and caspase-1, caspase-1 enzyme activity, and IL-1β levels. SFN inhibited saturated fatty acid-induced activation of the NLRP3 inflammasome in primary mouse hepatocytes, accompanied by the restoration of mitochondrial dysfunction. The suppression of NLRP3 inflammasome by SFN was mediated by the regulation of AMP-activated protein kinase-autophagy axis. Our findings demonstrated that the suppression of NLRP3 inflammasome activation by an orally available small molecule inhibitor leads to the alleviation of the hepatic steatosis symptoms associated with NAFLD induced by a high-fat diet.
Collapse
Affiliation(s)
- Gabsik Yang
- Integrated Research Institute of Pharmaceutical Sciences, College of Pharmacy, The Catholic University of Korea, Bucheon, Republic of Korea, 420-743, South Korea
| | - Hye Eun Lee
- Integrated Research Institute of Pharmaceutical Sciences, College of Pharmacy, The Catholic University of Korea, Bucheon, Republic of Korea, 420-743, South Korea
| | - Joo Young Lee
- Integrated Research Institute of Pharmaceutical Sciences, College of Pharmacy, The Catholic University of Korea, Bucheon, Republic of Korea, 420-743, South Korea
| |
Collapse
|
95
|
Wang P, Koyama Y, Liu X, Xu J, Ma HY, Liang S, Kim IH, Brenner DA, Kisseleva T. Promising Therapy Candidates for Liver Fibrosis. Front Physiol 2016; 7:47. [PMID: 26909046 PMCID: PMC4754444 DOI: 10.3389/fphys.2016.00047] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 02/01/2016] [Indexed: 12/11/2022] Open
Abstract
Liver fibrosis is a wound-healing process in response to repeated and chronic injury to hepatocytes and/or cholangiocytes. Ongoing hepatocyte apoptosis or necrosis lead to increase in ROS production and decrease in antioxidant activity, which recruits inflammatory cells from the blood and activate hepatic stellate cells (HSCs) changing to myofibroblasts. Injury to cholangiocytes also recruits inflammatory cells to the liver and activates portal fibroblasts in the portal area, which release molecules to activate and amplify cholangiocytes. No matter what origin of myofibroblasts, either HSCs or portal fibroblasts, they share similar characteristics, including being positive for α-smooth muscle actin and producing extracellular matrix. Based on the extensive pathogenesis knowledge of liver fibrosis, therapeutic strategies have been designed to target each step of this process, including hepatocyte apoptosis, cholangiocyte proliferation, inflammation, and activation of myofibroblasts to deposit extracellular matrix, yet the current therapies are still in early-phase clinical development. There is an urgent need to translate the molecular mechanism of liver fibrosis to effective and potent reagents or therapies in human.
Collapse
Affiliation(s)
- Ping Wang
- Department of Surgery, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Medicine, University of CaliforniaSan Diego, La Jolla, CA, USA; Liver Research Center, Beijing Friendship Hospital, Capital Medical UniversityBeijing, China
| | - Yukinori Koyama
- Department of Surgery, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Medicine, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Surgery, Graduate School of Medicine, Kyoto UniversityKyoto, Japan
| | - Xiao Liu
- Department of Surgery, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Medicine, University of CaliforniaSan Diego, La Jolla, CA, USA
| | - Jun Xu
- Department of Surgery, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Medicine, University of CaliforniaSan Diego, La Jolla, CA, USA
| | - Hsiao-Yen Ma
- Department of Surgery, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Medicine, University of CaliforniaSan Diego, La Jolla, CA, USA
| | - Shuang Liang
- Department of Surgery, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Medicine, University of CaliforniaSan Diego, La Jolla, CA, USA
| | - In H Kim
- Department of Surgery, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Medicine, University of CaliforniaSan Diego, La Jolla, CA, USA
| | - David A Brenner
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Tatiana Kisseleva
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
96
|
Ioannou GN. The Role of Cholesterol in the Pathogenesis of NASH. Trends Endocrinol Metab 2016; 27:84-95. [PMID: 26703097 DOI: 10.1016/j.tem.2015.11.008] [Citation(s) in RCA: 372] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 11/12/2015] [Accepted: 11/16/2015] [Indexed: 01/01/2023]
Abstract
Lipotoxicity drives the development of progressive hepatic inflammation and fibrosis in a subgroup of patients with nonalcoholic fatty liver disease (NAFLD), causing nonalcoholic steatohepatitis (NASH) and even progression to cirrhosis and hepatocellular carcinoma (HCC). While the underlying molecular mechanisms responsible for the development of inflammation and fibrosis that characterize progressive NASH remain unclear, emerging evidence now suggests that hepatic free cholesterol (FC) is a major lipotoxic molecule critical in the development of experimental and human NASH. In this review, we examine the effects of excess FC in hepatocytes, Kupffer cells (KCs), and hepatic stellate cells (HSCs), and the subcellular mechanisms by which excess FC can induce cellular toxicity or proinflammatory and profibrotic effects in these cells.
Collapse
Affiliation(s)
- George N Ioannou
- Research and Development, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA; Division of Gastroenterology, Department of Medicine, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA; University of Washington, Seattle, WA, USA.
| |
Collapse
|
97
|
Wree A, Mehal WZ, Feldstein AE. Targeting Cell Death and Sterile Inflammation Loop for the Treatment of Nonalcoholic Steatohepatitis. Semin Liver Dis 2016; 36:27-36. [PMID: 26870930 PMCID: PMC4955833 DOI: 10.1055/s-0035-1571272] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease represents a wide spectrum of conditions and is currently the most common form of chronic liver disease affecting both adults and children in the United States and many other parts of the world. Great effort has been focused on the development of novel therapies for those patients with the more advanced forms of the disease, in particular those with nonalcoholic steatohepatitis (NASH) and liver fibrosis that can be associated with significant morbidity and mortality. In this review, the authors focus on the role of cell death and sterile inflammatory pathways as well as the self-perpetuating deleterious cycle they may trigger as novel therapeutic targets for the treatment of fibrotic NASH.
Collapse
Affiliation(s)
- Alexander Wree
- Department of Pediatrics, University of California San Diego (UCSD), and Rady Children’s Hospital, San Diego, California,Department of Internal Medicine III, University Hospital, RWTH-Aachen, Germany
| | - Wajahat Z. Mehal
- Yale University, and West Haven Veterans Medical Center, New Haven, Connecticut
| | - Ariel E. Feldstein
- Department of Pediatrics, University of California San Diego (UCSD), and Rady Children’s Hospital, San Diego, California
| |
Collapse
|
98
|
Zhao YX, Ju J, Wang W, Ye JF. Role of interleukin 17 in fatty liver disease and other liver diseases. Shijie Huaren Xiaohua Zazhi 2015; 23:5790-5796. [DOI: 10.11569/wcjd.v23.i36.5790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fatty liver disease is a lipid metabolism disorder in the liver, with a variety of etiologies which result in the accumulation of fat in the liver. The pathogenesis of fatty liver disease is still unclear and symptomatic treatment is the main method. In recent years, the incidence of fatty liver disease in China is increasing year by year, and the age of onset becomes younger. Recent studies suggest that interleukin 17 (IL-17) is involved not only in the pathogenesis of various diseases, such as autoimmune diseases, inflammatory diseases, and malignant tumors, but also in fatty liver and other liver diseases through inducing insulin resistance, activating some important pathways in the liver, mediating some inflammatory factors and so on. This paper will review the role of IL-17 in fatty liver disease and other liver diseases.
Collapse
|
99
|
Ye JF, Ju J, Wang W, Zhao YX. Effect of Clematis polysaccharide on serum IL-17 and TNF-α levels and hepatic pathological changes in rats with non-alcoholic steatohepatitis. Shijie Huaren Xiaohua Zazhi 2015; 23:4864-4870. [DOI: 10.11569/wcjd.v23.i30.4864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of Clematis polysaccharide on serum levels of interleukin-17 (IL-17) and tumor necrosis factor alpha (TNF-α) and hepatic pathological changes in rats with non-alcoholic steatohepatitis (NASH), and to analyze the correlation between serum levels of IL-17 and TNF-α.
METHODS: A rat model of NASH was established by feeding SD rats a high-fat diet. The levels of fasting IL-17 and TNF-α were tested, and the liver fatty degeneration grade and liver inflammation grade were assessed for NASH model rats, NASH rats treated with Clematis polysaccharide, NASH rats treated with polyene phosphatidylcholine, and normal rats. The differences in fasting IL-17 and TNF-α levels and hepatic pathological changes among all groups were compared, and the correlation between serum levels of IL-17 and TNF-α was analyzed.
RESULTS: Compared with the model group, NASH rats treated with Clematis polysaccharide had milder liver fatty degeneration and liver inflammation (P < 0.05 for both) and lower serum levels of IL-17 and TNF-α (P < 0.01 for both). Compared with the polyene phosphatidylcholine group, NASH rats treated with Clematis polysaccharide had a similar liver fatty degeneration grade and liver inflammation grade, and similar serum IL-17 and TNF-α levels (P > 0.05 for all).
CONCLUSION: Clematis polysaccharide is effective in NASH rats, its therapeutic effect is equivalent to that of polyene phosphatidylcholine, and the underlying mechanism may be associated with reducing serum IL-17 and TNF-α levels, and improving hepatic pathological change.
Collapse
|
100
|
Jeftic I, Jovicic N, Pantic J, Arsenijevic N, Lukic ML, Pejnovic N. Galectin-3 Ablation Enhances Liver Steatosis, but Attenuates Inflammation and IL-33-Dependent Fibrosis in Obesogenic Mouse Model of Nonalcoholic Steatohepatitis. Mol Med 2015; 21:453-65. [PMID: 26018806 DOI: 10.2119/molmed.2014.00178] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 05/21/2015] [Indexed: 12/14/2022] Open
Abstract
The importance of Galectin-3 (Gal-3) in obesity-associated liver pathology is incompletely defined. To dissect the role of Gal-3 in fibrotic nonalcoholic steatohepatitis (NASH), Gal-3-deficient (LGALS3(-/-)) and wild-type (LGALS3(+/+)) C57Bl/6 mice were placed on an obesogenic high fat diet (HFD, 60% kcal fat) or standard chow diet for 12 and 24 wks. Compared to WT mice, HFD-fed LGALS3(-/-) mice developed, in addition to increased visceral adiposity and diabetes, marked liver steatosis, which was accompanied with higher expression of hepatic PPAR-γ, Cd36, Abca-1 and FAS. However, as opposed to LGALS3(-/-) mice, hepatocellular damage, inflammation and fibrosis were more extensive in WT mice which had an elevated number of mature myeloid dendritic cells, proinflammatory CD11b(+)Ly6C(hi) monocytes/macrophages in liver, peripheral blood and bone marrow, and increased hepatic CCL2, F4/80, CD11c, TLR4, CD14, NLRP3 inflammasome, IL-1β and NADPH-oxidase enzymes mRNA expression. Thus, obesity-driven greater steatosis was uncoupled with attenuated fibrotic NASH in Gal-3-deficient mice. HFD-fed WT mice had a higher number of hepatocytes that strongly expressed IL-33 and hepatic CD11b(+)IL-13(+) cells, increased levels of IL-33 and IL-13 and up-regulated IL-33, ST2 and IL-13 mRNA in liver compared with LGALS3(-/-) mice. IL-33 failed to induce ST2 upregulation and IL-13 production by LGALS3(-/-) peritoneal macrophages in vitro. Administration of IL-33 in vivo enhanced liver fibrosis in HFD-fed mice in both genotypes, albeit to a significantly lower extent in LGALS3(-/-) mice, which was associated with less numerous hepatic IL-13-expressing CD11b(+) cells. The present study provides evidence of a novel role for Gal-3 in regulating IL-33-dependent liver fibrosis.
Collapse
Affiliation(s)
- Ilija Jeftic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.,Institute of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nemanja Jovicic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.,Institute of Histology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Jelena Pantic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nebojsa Arsenijevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Miodrag L Lukic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nada Pejnovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.,Institute of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|