651
|
Farid AS, Fath EM, Mido S, Nonaka N, Horii Y. Paraoxonase-1 activity is related to Trichinella spiralis-induced hepatitis in rats. Eur J Clin Invest 2017; 47:250-261. [PMID: 28103386 DOI: 10.1111/eci.12731] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 01/16/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Little is known about the potential adverse effects of a chronic zoonotic nematode Trichinella spiralis infection on hepatic inflammation and its relationship to paraoxonase (PON)-1 and butyrylcholinesterase (BuChE) activities. Therefore, we aimed to examine the effects of T. spiralis infection on hepatic synthesis of PON1. METHODS Wistar rats were infected with 2500 first-stage larvae (L1) of T. spiralis, and serum PON1 and BuChE activities were evaluated. Hepatic expression levels of PON1, BuChE and various cytokines and chemokines [interleukin (IL)-1, IL-4, IL-6, IL-10, tumour necrosis factor (TNF)-α, monocyte chemoattractant protein (MCP)-1, macrophage inflammatory protein (MIP)-1α, and transforming growth factor (TGF)-β1] were evaluated for up to 9 weeks post-infection (p.i.). The effect of these changes on the degree of hepatic apoptosis was also investigated. RESULTS Trichinella spiralis infection in rats induced significant decreases in serum PON1 activities from day 2 until week 7 p.i. and BuChE activity starting from day 4 until 2 weeks p.i. Moreover, T. spiralis infection increased serum pro-inflammatory cytokines IL-1, IL-6 and TNF-α as well as chemokines MCP-1, MIP-1α and TGF-β1 during the enteral phase of the parasite life cycle. The anti-inflammatory cytokines IL-4 and IL-10 showed significant increases during the enteral phase for the former and the muscle phase for the latter. These were associated with hepatic inflammation and apoptosis. These events typically decreased hepatic PON1 and BuChE mRNA expression. CONCLUSIONS Immune responses mounted against T. spiralis infection in rats were associated with hepatic inflammation and a subsequent decrease in serum PON1 and BuChE activities.
Collapse
Affiliation(s)
- Ayman Samir Farid
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Qalyubia, Egypt
| | - Eman Mohamed Fath
- Laboratory of Veterinary Parasitic Diseases, Interdisciplinary Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki, Japan
| | - Shogo Mido
- Laboratory of Veterinary Parasitic Diseases, Interdisciplinary Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki, Japan
| | - Nariaki Nonaka
- Laboratory of Veterinary Parasitic Diseases, Interdisciplinary Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki, Japan.,Center for Animal Disease Control, University of Miyazaki, Miyazaki, Japan
| | - Yoichiro Horii
- Laboratory of Veterinary Parasitic Diseases, Interdisciplinary Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki, Japan.,Center for Animal Disease Control, University of Miyazaki, Miyazaki, Japan
| |
Collapse
|
652
|
Schisandra Lignan Extract Protects against Carbon Tetrachloride-Induced Liver Injury in Mice by Inhibiting Oxidative Stress and Regulating the NF- κB and JNK Signaling Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:5140297. [PMID: 28246539 PMCID: PMC5299172 DOI: 10.1155/2017/5140297] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 12/13/2016] [Accepted: 12/27/2016] [Indexed: 12/25/2022]
Abstract
Schisandra chinensis (S. chinensis) is a traditional Chinese herbal medicine widely used for the treatment of liver disease, whose main active components are lignans. However, the action mechanisms of the lignans in S. chinensis remain unclear. This study aimed to investigate the protective effect and related molecular mechanism of Schisandra lignan extract (SLE) against carbon tetrachloride- (CCl4-) induced acute liver injury in mice. Different doses of SLE at 50, 100, and 200 mg/kg were administered daily by gavage for 5 days before CCl4 treatment. The results showed that SLE significantly decreased the activities of serum ALT/AST and reduced liver pathologic changes induced by CCl4. Pretreatment with SLE not only decreased the content of MDA but increased SOD, GSH, and GSH-Px activities in the liver, suggesting that SLE attenuated CCl4-induced oxidative stress. The expression levels of inflammatory cytokines TNF-a, IL-1β, and IL-6 were decreased after oral administration of SLE, probably because lignans inhibited the NF-κB activity. Additionally, SLE also inhibited hepatocyte apoptosis by suppressing JNK activation and regulating Bcl-2/Bax signaling pathways. In conclusion, these results suggested that SLE prevented CCl4-induced liver injury through a combination of antioxidative stress, anti-inflammation, and antihepatocyte apoptosis and alleviated inflammation and apoptosis by regulating the NF-κB, JNK, and Bcl-2/Bax signaling pathways.
Collapse
|
653
|
Yang L, Miura K, Zhang B, Matsushita H, Yang YM, Liang S, Song J, Roh YS, Seki E. TRIF Differentially Regulates Hepatic Steatosis and Inflammation/Fibrosis in Mice. Cell Mol Gastroenterol Hepatol 2017; 3:469-483. [PMID: 28462384 PMCID: PMC5403956 DOI: 10.1016/j.jcmgh.2016.12.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 12/25/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Toll-like receptor 4 (TLR4) signaling is activated through 2 adaptor proteins: MyD88 and TIR-domain containing adaptor-inducing interferon-β (TRIF). TLR4 and MyD88 are crucial in nonalcoholic steatohepatitis (NASH) and fibrosis. However, the role of TRIF in TLR4-mediated NASH and fibrosis has been elusive. This study investigated the differential roles of TRIF in hepatic steatosis and inflammation/fibrosis. METHODS A choline-deficient amino acid defined (CDAA) diet was used for the mouse NASH model. On this diet, the mice develop hepatic steatosis, inflammation, and fibrosis. TLR4 wild-type and TLR4-/- bone marrow chimeric mice and TRIF-/- mice were fed CDAA or a control diet for 22 weeks. Hepatic steatosis, inflammation, and fibrosis were examined. RESULTS In the CDAA diet-induced NASH, the mice with wild-type bone marrow had higher alanine aminotransferase and hepatic tumor necrosis factor levels than the mice with TLR4-/- bone marrow. The nonalcoholic fatty liver disease activity score showed that both wild-type and TLR4-/- bone marrow chimeras had reduced hepatic steatosis, and that both types of chimeras had similar levels of inflammation and hepatocyte ballooning to whole-body wild-type mice. Notably, wild-type recipients showed more liver fibrosis than TLR4-/- recipients. Although TRIF-/- mice showed reduced hepatic steatosis, these mice showed more liver injury, inflammation, and fibrosis than wild-type mice. TRIF-/- stellate cells and hepatocytes produced more C-X-C motif chemokine ligand 1 (CXCL1) and C-C motif chemokine ligand than wild-type cells in response to lipopolysaccharide. Consistently, TRIF-/- mice showed increased CXCL1 and CCL3 expression along with neutrophil and macrophage infiltration, which promotes liver inflammation and injury. CONCLUSIONS In TLR4-mediated NASH, different liver cells have distinct roles in hepatic steatosis, inflammation, and fibrosis. TRIF promotes hepatic steatosis but it inhibits injury, inflammation, and fibrosis.
Collapse
Key Words
- ALT, alanine aminotransferase
- BM, bone marrow
- BMT, bone marrow transplantation
- CDAA, choline-deficient amino acid defined
- DGAT2, diacylglycerol acyltransferase 2
- HFD, high-fat diet
- HSC, hepatic stellate cell
- Hepatocyte Apoptosis
- IL, interleukin
- LDH, lactate dehydrogenase
- LPS
- LPS, lipopolysaccharide
- NAFLD, nonalcoholic fatty liver disease
- NASH, nonalcoholic steatohepatitis
- Neutrophils
- PCR, polymerase chain reaction
- TLR4
- TLR4, Toll-like receptor 4
- TNF, tumor necrosis factor
- α-SMA, α-smooth muscle actin
Collapse
Affiliation(s)
- Ling Yang
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, La Jolla, California,Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kouichi Miura
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, La Jolla, California,Department of Gastroenterology, Akita University Graduate School of Medicine, Akita, Japan
| | - Bi Zhang
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, La Jolla, California
| | - Hiroshi Matsushita
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, La Jolla, California,Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Yoon Mee Yang
- Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Shuang Liang
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, La Jolla, California
| | - Jingyi Song
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, La Jolla, California
| | - Yoon Seok Roh
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, La Jolla, California,Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California,Department of Pharmacy, Chungbuk National University College of Pharmacy, Chungbuk, South Korea
| | - Ekihiro Seki
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, La Jolla, California,Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California,Department of Medicine, University of California Los Angeles, David Geffen School of Medicine, Los Angeles, California,Correspondence Address correspondence to: Ekihiro Seki, MD, PhD, Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Davis Research Building, Suite 2099, Los Angeles, California 90048. fax: (310) 423-0157.Division of GastroenterologyDepartment of MedicineCedars-Sinai Medical Center8700 Beverly BoulevardDavis Research BuildingSuite 2099Los AngelesCalifornia 90048
| |
Collapse
|
654
|
The Role of Tissue Macrophage-Mediated Inflammation on NAFLD Pathogenesis and Its Clinical Implications. Mediators Inflamm 2017; 2017:8162421. [PMID: 28115795 PMCID: PMC5237469 DOI: 10.1155/2017/8162421] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 11/22/2016] [Accepted: 12/04/2016] [Indexed: 02/06/2023] Open
Abstract
The obese phenotype is characterized by a state of chronic low-grade systemic inflammation that contributes to the development of comorbidities, including nonalcoholic fatty liver disease (NAFLD). In fact, NAFLD is often associated with adipocyte enlargement and consequent macrophage recruitment and inflammation. Macrophage polarization is often associated with the proinflammatory state in adipose tissue. In particular, an increase of M1 macrophages number or of M1/M2 ratio triggers the production and secretion of various proinflammatory signals (i.e., adipocytokines). Next, these inflammatory factors may reach the liver leading to local M1/M2 macrophage polarization and consequent onset of the histological damage characteristic of NAFLD. Thus, the role of macrophage polarization and inflammatory signals appears to be central for pathogenesis and progression of NAFLD, even if the heterogeneity of macrophages and molecular mechanisms that govern their phenotype switch remain incompletely understood. In this review, we discuss the role of adipose and liver tissue macrophage-mediated inflammation in experimental and human NAFLD. This focus is relevant because it may help researchers that approach clinical and experimental studies on this disease advancing the knowledge of mechanisms that could be targeted in order to revert NAFLD-related fibrosis.
Collapse
|
655
|
Jiang X, Shen T, Tang X, Yang W, Guo H, Ling W. Cyanidin-3-O-β-glucoside combined with its metabolite protocatechuic acid attenuated the activation of mice hepatic stellate cells. Food Funct 2017; 8:2945-2957. [DOI: 10.1039/c7fo00265c] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cyanidin-3-O-β-glucoside with the aid of its metabolite protocatechuic acid attenuated the activation of mice hepatic stellate cells.
Collapse
Affiliation(s)
- Xinwei Jiang
- Department of Food Science and Engineering
- Institute of Science and Technology
- Jinan University
- Guangzhou 510632
- People's Republic of China
| | - Tianran Shen
- Department of Nutrition
- School of Public Health
- Sun Yat-Sen University
- Guangzhou 510080
- People's Republic of China
| | - Xilan Tang
- Department of Nutrition
- School of Public Health
- Sun Yat-Sen University
- Guangzhou 510080
- People's Republic of China
| | - Wenqi Yang
- Department of Nutrition
- School of Public Health
- Sun Yat-Sen University
- Guangzhou 510080
- People's Republic of China
| | - Honghui Guo
- Department of Nutrition
- Henry Fok School of Food Science and Engineering
- Shaoguan University
- Shaoguan 512005
- People's Republic of China
| | - Wenhua Ling
- Department of Nutrition
- School of Public Health
- Sun Yat-Sen University
- Guangzhou 510080
- People's Republic of China
| |
Collapse
|
656
|
Longato L, Andreola F, Davies SS, Roberts JL, Fusai G, Pinzani M, Moore K, Rombouts K. Reactive gamma-ketoaldehydes as novel activators of hepatic stellate cells in vitro. Free Radic Biol Med 2017; 102:162-173. [PMID: 27890721 DOI: 10.1016/j.freeradbiomed.2016.11.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 11/14/2016] [Accepted: 11/21/2016] [Indexed: 11/28/2022]
Abstract
AIMS Products of lipid oxidation, such as 4-hydroxynonenal (4-HNE), are key activators of hepatic stellate cells (HSC) to a pro-fibrogenic phenotype. Isolevuglandins (IsoLG) are a family of acyclic γ-ketoaldehydes formed through oxidation of arachidonic acid or as by-products of the cyclooxygenase pathway. IsoLGs are highly reactive aldehydes which are efficient at forming protein adducts and cross-links at concentrations 100-fold lower than 4-hydroxynonenal. Since the contribution of IsoLGs to liver injury has not been studied, we synthesized 15-E2-IsoLG and used it to investigate whether IsoLG could induce activation of HSC. RESULTS Primary human HSC were exposed to 15-E2-IsoLG for up to 48h. Exposure to 5μM 15-E2-IsoLG in HSCs promoted cytotoxicity and apoptosis. At non-cytotoxic doses (50 pM-500nM) 15-E2-IsoLG promoted HSC activation, indicated by increased expression of α-SMA, sustained activation of ERK and JNK signaling pathways, and increased mRNA and/or protein expression of cytokines and chemokines, which was blocked by inhibitors of JNK and NF-kB. In addition, IsoLG promoted formation of reactive oxygen species, and induced an early activation of ER stress, followed by autophagy. Inhibition of autophagy partially reduced the pro-inflammatory effects of IsoLG, suggesting that it might serve as a cytoprotective response. INNOVATION This study is the first to describe the biological effects of IsoLG in primary HSC, the main drivers of hepatic fibrosis. CONCLUSIONS IsoLGs represent a newly identified class of activators of HSC in vitro, which are biologically active at concentrations as low as 500 pM, and are particularly effective at promoting a pro-inflammatory response and autophagy.
Collapse
Affiliation(s)
- Lisa Longato
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, University College London, Royal Free, London, UK
| | - Fausto Andreola
- Liver Failure Group, Institute for Liver & Digestive Health, University College of London, Royal Free, London, UK
| | - Sean S Davies
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Jackson L Roberts
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Giuseppe Fusai
- Division of Surgery, University College London, Royal Free, London, UK
| | - Massimo Pinzani
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, University College London, Royal Free, London, UK
| | - Kevin Moore
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, University College London, Royal Free, London, UK
| | - Krista Rombouts
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, University College London, Royal Free, London, UK.
| |
Collapse
|
657
|
Xu W, Zeng S, Li M, Fan Z, Zhou B. Aggf1 attenuates hepatic inflammation and activation of hepatic stellate cells by repressing Ccl2 transcription. J Biomed Res 2017; 31:428-436. [PMID: 28958996 PMCID: PMC5706435 DOI: 10.7555/jbr.30.20160046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Liver injury represents a continuum of pathophysiological processes involving a complex interplay between hepatocytes, macrophages, and hepatic stellate cells. The mechanism whereby these intercellular interactions contribute to liver injury and fibrosis is not completely understood. We report here that angiogenic factor with G patch and FHA domains 1 (Aggf1) was downregulated in the livers of cirrhotic patients compared to healthy controls and in primary hepatocytes in response to carbon tetrachloride (CCl4) stimulation. Overexpression of Aggf1 attenuated macrophage chemotaxis. Aggf1 interacted with NF-κB to block its binding to theCcl2 gene promoter and repressed Ccl2 transcription in hepatocytes. Macrophages cultured in the conditioned media collected from Aggf1-overexpressing hepatocytes antagonized HSC activation. Taken together, our data illustrate a novel role for Aggf1 in regulating hepatic inflammation and provide insights on the development of interventional strategies against cirrhosis.
Collapse
Affiliation(s)
- Wenping Xu
- Department of Nursing, Jiangsu Jiankang Vocational University, Nanjing, Jiangsu 210029, China
| | - Sheng Zeng
- Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Min Li
- Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Zhiwen Fan
- Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Bisheng Zhou
- Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
658
|
Li S, Hong M, Tan HY, Wang N, Feng Y. Insights into the Role and Interdependence of Oxidative Stress and Inflammation in Liver Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:4234061. [PMID: 28070230 PMCID: PMC5192343 DOI: 10.1155/2016/4234061] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 11/02/2016] [Indexed: 02/06/2023]
Abstract
The crucial roles of oxidative stress and inflammation in the development of hepatic diseases have been unraveled and emphasized for decades. From steatosis to fibrosis, cirrhosis and liver cancer, hepatic oxidative stress, and inflammation are sustained and participated in this pathological progressive process. Notably, increasing evidences showed that oxidative stress and inflammation are tightly related, which are regarded as essential partners that present simultaneously and interact with each other in various pathological conditions, creating a vicious cycle to aggravate the hepatic diseases. Clarifying the interaction of oxidative stress and inflammation is of great importance to provide new directions and targets for developing therapeutic intervention. Herein, this review is concerned with the regulation and interdependence of oxidative stress and inflammation in a variety of liver diseases. In addition to classical mediators and signaling, particular emphasis is placed upon immune suppression, a potential linkage of oxidative stress and inflammation, to provide new inspiration for the treatment of liver diseases. Furthermore, since antioxidation and anti-inflammation have been extensively attempted as the strategies for treatment of liver diseases, the application of herbal medicines and their derived compounds that protect liver from injury via regulating oxidative stress and inflammation collectively were reviewed and discussed.
Collapse
Affiliation(s)
- Sha Li
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Ming Hong
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Hor-Yue Tan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
659
|
Role and mechanism of the AMPK pathway in waterborne Zn exposure influencing the hepatic energy metabolism of Synechogobius hasta. Sci Rep 2016; 6:38716. [PMID: 27934965 PMCID: PMC5146659 DOI: 10.1038/srep38716] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 11/15/2016] [Indexed: 12/18/2022] Open
Abstract
Previous studies have investigated the physiological responses in the liver of Synechogobius hasta exposed to waterborne zinc (Zn). However, at present, very little is known about the underlying molecular mechanisms of these responses. In this study, RNA sequencing (RNA-seq) was performed to analyse the differences in the hepatic transcriptomes between control and Zn-exposed S. hasta. A total of 36,339 unigenes and 1,615 bp of unigene N50 were detected. These genes were further annotated to the Nonredundant protein (NR), Nonredundant nucleotide (Nt), Swiss-Prot, Kyoto Encyclopedia of Genes and Genomes (KEGG), Clusters of Orthologous Groups (COG) and Gene Ontology (GO) databases. After 60 days of Zn exposure, 708 and 237 genes were significantly up- and down-regulated, respectively. Many differentially expressed genes (DEGs) involved in energy metabolic pathways were identified, and their expression profiles suggested increased catabolic processes and reduced biosynthetic processes. These changes indicated that waterborne Zn exposure increased the energy production and requirement, which was related to the activation of the AMPK signalling pathway. Furthermore, using the primary hepatocytes of S. hasta, we identified the role of the AMPK signalling pathway in Zn-influenced energy metabolism.
Collapse
|
660
|
Affo S, Yu LX, Schwabe RF. The Role of Cancer-Associated Fibroblasts and Fibrosis in Liver Cancer. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2016; 12:153-186. [PMID: 27959632 DOI: 10.1146/annurev-pathol-052016-100322] [Citation(s) in RCA: 498] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Liver cancer is the second leading cause of cancer mortality worldwide, causing more than 700,000 deaths annually. Because of the wide landscape of genomic alterations and limited therapeutic success of targeting tumor cells, a recent focus has been on better understanding and possibly targeting the microenvironment in which liver tumors develop. A unique feature of liver cancer is its close association with liver fibrosis. More than 80% of hepatocellular carcinomas (HCCs) develop in fibrotic or cirrhotic livers, suggesting an important role of liver fibrosis in the premalignant environment (PME) of the liver. Cholangiocarcinoma (CCA), in contrast, is characterized by a strong desmoplasia that typically occurs in response to the tumor, suggesting a key role of cancer-associated fibroblasts (CAFs) and fibrosis in its tumor microenvironment (TME). Here, we discuss the functional contributions of myofibroblasts, CAFs, and fibrosis to the development of HCC and CCA in the hepatic PME and TME, focusing on myofibroblast- and extracellular matrix-associated growth factors, fibrosis-associated immunosuppressive pathways, as well as mechanosensitive signaling cascades that are activated by increased tissue stiffness. Better understanding of the role of myofibroblasts in HCC and CCA development and progression may provide the basis to target these cells for tumor prevention or therapy.
Collapse
Affiliation(s)
- Silvia Affo
- Department of Medicine, Columbia University, New York, NY 10032;
| | - Le-Xing Yu
- Department of Medicine, Columbia University, New York, NY 10032;
| | - Robert F Schwabe
- Department of Medicine, Columbia University, New York, NY 10032;
| |
Collapse
|
661
|
CXCL12 expression and PD-L1 expression serve as prognostic biomarkers in HCC and are induced by hypoxia. Virchows Arch 2016; 470:185-196. [PMID: 27913861 DOI: 10.1007/s00428-016-2051-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/02/2016] [Accepted: 11/21/2016] [Indexed: 02/08/2023]
Abstract
Anti-PD-1 treatment increases anti-tumour immune responses in animal models of hepatocellular carcinoma (HCC). Sorafenib, the mainstay of treatment of HCC patients, however, leads to tumour hypoxia and thereby abrogates the efficacy of anti-PD-1 treatment. This served as a rationale to implement CXCR4 inhibition as adjunct to sorafenib and anti-PD-1 treatment in murine HCC models. We studied the relationship between tumour hypoxia, PD-L1 and CXCL12 expression in human HCC, aiming to test the rationale for triple therapy combining sorafenib, PD-1 immune checkpoint inhibitors and CXCR4 inhibitors. Expression of CXCL12, PD-L1 and of surrogate markers for tumour hypoxia was evaluated at messenger RNA (mRNA) level in a cohort of HCC patients from The Cancer Genome Atlas and immunohistochemically in an independent cohort from the University Hospital of Bonn. Retrospective survival analyses were conducted. CXCL12 mRNA level significantly correlated with markers indicating tumour hypoxia in HCC (HIF1-α ρ = 0.104, p = 0.047). PD-L1 expression was significantly increased in tumours with a high number of tumour-infiltrating lymphocytes (ρ = 0.533, p < 0.001). In Cox proportional hazard analyses, high PD-L1 expression and loss of nuclear CXCL12 expression showed significant prognostic value in terms of overall survival (hazard ratio (HR) = 3.35 [95%CI 1.33-8.46], p = 0.011 for PD-L1; HR = 2.64 [95%CI 1.18-5.88], p = 0.018 for CXCL12, respectively). This study supports the rationale to combine CXCR4 inhibitors and PD-1 immune checkpoint inhibitors in patients with HCC, as sorafenib-induced tumour hypoxia leads to upregulation of PD-L1 and CXCL12.
Collapse
|
662
|
FXR Agonists: From Bench to Bedside, a Guide for Clinicians. Dig Dis Sci 2016; 61:3395-3404. [PMID: 27734248 DOI: 10.1007/s10620-016-4334-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 09/28/2016] [Indexed: 12/15/2022]
|
663
|
Hepatic SATB1 induces paracrine activation of hepatic stellate cells and is upregulated by HBx. Sci Rep 2016; 6:37717. [PMID: 27883059 PMCID: PMC5121621 DOI: 10.1038/srep37717] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 10/28/2016] [Indexed: 12/23/2022] Open
Abstract
Chronic hepatitis B virus (HBV) infection is a major cause of chronic liver diseases, but its involvement in hepatic fibrogenesis remains unclear. Special AT-rich binding protein 1 (SATB1) has been implicated in reprogramming chromatin organization and transcription profiles in many cancers and non-cancer-related conditions. We found that hepatic SATB1 expression was significantly up-regulated in fibrotic tissues from chronic hepatitis B virus (HBV)-infected patients and HBV transgenic (HBV-Tg) mouse model. Knockdown of SATB1 in the liver significantly alleviated CCl4-induced fibrosis in HBV-Tg mouse model. Moreover, we suggested HBV encoded x protein (HBx) induced SATB1 expression through activation of JNK and ERK pathways. Enforced expression of SATB1 in hepatocytes promoted the activation and proliferation of hepatic stellate cells (HSCs) by secretion of connective tissue growth factor (CTGF), Interleukin-6 (IL-6) and platelet derived growth factor-A (PDGF-AA). Our findings demonstrated that HBx upregulated hepatic SATB1 which exerted pro-fibrotic effects by paracrine activation of stellate cells in HBV-related fibrosis.
Collapse
|
664
|
Shao J, Zeng S, Zhou B, Xu H, Bian Y, Xu Y. Angiogenic factor with G patch and FHA domains 1 (Aggf1) promotes hepatic steatosis in mice. Biochem Biophys Res Commun 2016; 482:134-140. [PMID: 27865839 DOI: 10.1016/j.bbrc.2016.10.071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 10/19/2016] [Indexed: 11/18/2022]
Abstract
Increased uptake of nutrients coupled with reduced activity leads to the development of a host of metabolic disorders in humans. In the present study we examined the role of angiogenic factor with G patch and FHA domains 1 (Aggf1) in the pathogenesis of steatosis, characterized by accumulation of lipids in the liver and consequently hepatic insulin resistance. We report here that Aggf1 expression was up-regulated in the liver in both genetically predisposed and diet-induced mouse model of steatosis. Aggf1 expression was also stimulated by free fatty acids in primary hepatocytes. Over-expression of Aggf1 in mice promoted steatosis. On the contrary, Aggf1 depletion ameliorated steatosis in mice. Mechanistically, Aggf1 activated the expression of gluconeogenesis gene and skewed the insulin signaling pathway to induce insulin resistance. Taken together, our data suggest that Aggf1 plays a role in steatosis in vivo and as such may be a new target in the development of therapeutics solutions against steatosis.
Collapse
Affiliation(s)
- Jing Shao
- College of Basic Medical Sciences, Nanjing University of Chinese Medicine, Nanjing, China
| | - Sheng Zeng
- Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - Bisheng Zhou
- Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, China.
| | - Huihui Xu
- Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - Yaoyao Bian
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yong Xu
- Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
665
|
Tornai T, Vitalis Z, Sipeki N, Dinya T, Tornai D, Antal-Szalmas P, Karanyi Z, Tornai I, Papp M. Macrophage activation marker, soluble CD163, is an independent predictor of short-term mortality in patients with cirrhosis and bacterial infection. Liver Int 2016; 36:1628-1638. [PMID: 27031405 DOI: 10.1111/liv.13133] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 03/29/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Innate immune system dysfunction is common in advanced cirrhosis, with a central role of the monocyte/macrophage system. Monocytes and macrophages express the scavenger receptor CD163, which is regulated by inflammatory mediators. Cleavage of the receptor leads to the formation of soluble (s)CD163 that represents an anti-inflammatory response. We aimed to study the clinical importance of sCD163 in cirrhosis. METHODS Sera of 378 patients were assayed for sCD163 by ELISA [193 outpatients and 185 patients with acute decompensation (AD)]. A 5-year follow-up observational study was conducted to assess the possible association between sCD163 level and poor disease outcomes. RESULTS sCD163 level was associated with disease severity, but not with the presence of varices or prior variceal bleeding. In outpatients, sCD163 level did not predict the development of disease-specific complications or the long-term mortality. In patients with AD episode, sCD163 level was significantly higher compared to outpatients but only in the presence of bacterial infection (INF) (AD-INF:4586, AD-NON-INF:3792 and outpatients: 3538 ng/ml, P < 0.015 and P = 0.001, respectively). sCD163 level gradually increased according to severity of infection. During bacterial infections, high sCD163 level (>7000 ng/ml) was associated with increased mortality rate (42% vs. 17%, P < 0.001) and was identified as an independent predictor of 28-day mortality (hazard ratio:2.96, 95% confidence intervals:1.27-6.95) in multivariate Cox-regression model comprising aetiology, co-morbidity, model for end-stage liver disease score and leucocyte count as covariates. CONCLUSIONS High sCD163 level is useful to identify patients with high-risk of death during an AD episode complicated by bacterial infection. This finding serves as an additional hint towards the significance of anti-inflammatory response during bacterial infection.
Collapse
Affiliation(s)
- Tamas Tornai
- Division of Gastroenterology, Department of Internal Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsuzsanna Vitalis
- Division of Gastroenterology, Department of Internal Medicine, University of Debrecen, Debrecen, Hungary
| | - Nora Sipeki
- Division of Gastroenterology, Department of Internal Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamas Dinya
- Institute of Surgery, University of Debrecen, Debrecen, Hungary
| | - David Tornai
- Department of Laboratory Medicine, University of Debrecen, Debrecen, Hungary
| | - Peter Antal-Szalmas
- Department of Laboratory Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Karanyi
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Istvan Tornai
- Division of Gastroenterology, Department of Internal Medicine, University of Debrecen, Debrecen, Hungary
| | - Maria Papp
- Division of Gastroenterology, Department of Internal Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
666
|
Montano-Loza AJ, Thandassery RB, Czaja AJ. Targeting Hepatic Fibrosis in Autoimmune Hepatitis. Dig Dis Sci 2016; 61:3118-3139. [PMID: 27435327 DOI: 10.1007/s10620-016-4254-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/11/2016] [Indexed: 02/06/2023]
Abstract
Hepatic fibrosis develops or progresses in 25 % of patients with autoimmune hepatitis despite corticosteroid therapy. Current management regimens lack reliable noninvasive methods to assess changes in hepatic fibrosis and interventions that disrupt fibrotic pathways. The goals of this review are to indicate promising noninvasive methods to monitor hepatic fibrosis in autoimmune hepatitis and identify anti-fibrotic interventions that warrant evaluation. Laboratory methods can differentiate cirrhosis from non-cirrhosis, but their accuracy in distinguishing changes in histological stage is uncertain. Radiological methods include transient elastography, acoustic radiation force impulse imaging, and magnetic resonance elastography. Methods based on ultrasonography are comparable in detecting advanced fibrosis and cirrhosis, but their performances may be compromised by hepatic inflammation and obesity. Magnetic resonance elastography has excellent performance parameters for all histological stages in diverse liver diseases, is uninfluenced by inflammatory activity or body habitus, has been superior to other radiological methods in nonalcoholic fatty liver disease, and may emerge as the preferred instrument to evaluate fibrosis in autoimmune hepatitis. Promising anti-fibrotic interventions are site- and organelle-specific agents, especially inhibitors of nicotinamide adenine dinucleotide phosphate oxidases, transforming growth factor beta, inducible nitric oxide synthase, lysyl oxidases, and C-C chemokine receptors types 2 and 5. Autoimmune hepatitis has a pro-fibrotic propensity, and noninvasive radiological methods, especially magnetic resonance elastography, and site- and organelle-specific interventions, especially selective antioxidants and inhibitors of collagen cross-linkage, may emerge to strengthen current management strategies.
Collapse
Affiliation(s)
- Aldo J Montano-Loza
- Division of Gastroenterology and Liver Unit, University of Alberta Hospital, Edmonton, AB, Canada
| | - Ragesh B Thandassery
- Division of Gastroenterology and Liver Unit, University of Alberta Hospital, Edmonton, AB, Canada
| | - Albert J Czaja
- Professor Emeritus of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, 200 First Street S.W., Rochester, MN, 55905, USA.
| |
Collapse
|
667
|
Wen SL, Feng S, Tang SH, Gao JH, Zhang LH, Tong H, Yan ZP, Fang DZ. Collapsed Reticular Network and its Possible Mechanism during the Initiation and/or Progression of Hepatic Fibrosis. Sci Rep 2016; 6:35426. [PMID: 27739503 PMCID: PMC5064391 DOI: 10.1038/srep35426] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/29/2016] [Indexed: 02/05/2023] Open
Abstract
Among the researches on hepatic fibrosis, great attention was paid to both hepatocytes and extracellular matrix (ECM). However, little focus was drawn on reticular fibrous network, which is important for demarcation and support of hepatocytes. The aim of this study was to investigate the change pattern of reticular fibers in hepatic fibrosis/cirrhosis and its underlying mechanism. In this study, thioacetamide (TAA) and bile duct ligation (BDL) were utilized to induce rat hepatic fibrosis respectively, and Human liver cirrhotic microassay was analyzed with IHC to confirm the results in animal experiment and to detect the metalloproteinases (MMPs) expressions. As a result, the reticular fibers decreased markedly after 1 week in TAA and 1 day in BDL treated rats. Multiple representative regulators of MMPs and MMPs increased significantly in their expressions and activities. Further more, in human liver cirrhotic microassay, MMPs expressions also showed similar patterns as that of animal experiment. In Conclusions: Degradation or collapse of reticular fibers in hepatic sinusoid can be considered as a pathological feature during the initiation and/or progression of hepatic fibrosis. Moreover, such degradation is associated with and probably caused by the over/dysregulated expression of MMPs.
Collapse
Affiliation(s)
- Shi-Lei Wen
- Department of Human Anatomy, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, PR China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Shi Feng
- Department of Human Anatomy, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Shi-Hang Tang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Jin-Hang Gao
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
- Department of Peptides Related to Human Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Lin-hao Zhang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
- West China School of Medicine, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Huan Tong
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Zhao-Ping Yan
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Ding Zhi Fang
- Department of Biochemistry and Molecular Biology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, PR China
| |
Collapse
|
668
|
Torok NJ. Dysregulation of redox pathways in liver fibrosis. Am J Physiol Gastrointest Liver Physiol 2016; 311:G667-G674. [PMID: 27562057 PMCID: PMC5142204 DOI: 10.1152/ajpgi.00050.2016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 08/18/2016] [Indexed: 02/06/2023]
Abstract
Reactive oxygen species are implicated in physiological signaling and cell fate decisions. In chronic liver diseases persistent and increased production of oxidative radicals drives a fibrogenic response that is a common feature of disease progression. Despite our understanding the biology of the main prooxidant enzymes, their targets, and antioxidant mechanisms in the liver, there is still lack of knowledge concerning their precise role in the pathogenesis of fibrosis. This review will examine the role of physiological redox signaling in the liver, provide an overview on recent advances in prooxidant and antioxidant pathways that are dysregulated during fibrosis, and highlight possible novel treatment targets.
Collapse
Affiliation(s)
- Natalie J. Torok
- UC Davis Medical Center, Sacramento, California; and Northern California VA System, Mather, California
| |
Collapse
|
669
|
Caligiuri A, Gentilini A, Marra F. Molecular Pathogenesis of NASH. Int J Mol Sci 2016; 17:ijms17091575. [PMID: 27657051 PMCID: PMC5037841 DOI: 10.3390/ijms17091575] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/05/2016] [Accepted: 09/07/2016] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is the main cause of chronic liver disease in the Western world and a major health problem, owing to its close association with obesity, diabetes, and the metabolic syndrome. NASH progression results from numerous events originating within the liver, as well as from signals derived from the adipose tissue and the gastrointestinal tract. In a fraction of NASH patients, disease may progress, eventually leading to advanced fibrosis, cirrhosis and hepatocellular carcinoma. Understanding the mechanisms leading to NASH and its evolution to cirrhosis is critical to identifying effective approaches for the treatment of this condition. In this review, we focus on some of the most recent data reported on the pathogenesis of NASH and its fibrogenic progression, highlighting potential targets for treatment or identification of biomarkers of disease progression.
Collapse
Affiliation(s)
- Alessandra Caligiuri
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Firenze 50121, Italy.
| | - Alessandra Gentilini
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Firenze 50121, Italy.
| | - Fabio Marra
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Firenze 50121, Italy.
| |
Collapse
|
670
|
Zhang Y, Tang J, Tian Z, van Velkinburgh JC, Song J, Wu Y, Ni B. Innate Lymphoid Cells: A Promising New Regulator in Fibrotic Diseases. Int Rev Immunol 2016. [PMID: 26222510 DOI: 10.3109/08830185.2015.1068304] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fibrosis is a consequence of chronic inflammation and the persistent accumulation of extracellular matrix, for which the cycle of tissue injury and repair becomes a predominant feature. Both the innate and adaptive immune systems play key roles in the progress of fibrosis. The recently identified subsets of innate lymphoid cells (ILCs), which are mainly localize to epithelial surfaces, have been characterized as regulators of chronic inflammation and tissue remodeling, representing a functional bridge between the innate and adaptive immunity. Moreover, recent research has implicated ILCs as potential contributing factors to several kinds of fibrosis diseases, such as hepatic fibrosis and pulmonary fibrosis. Here, we will summarize and discuss the key roles of ILCs and their related factors in fibrotic diseases and their potential for translation to the clinic.
Collapse
Affiliation(s)
- Yi Zhang
- a Institute of Immunology, PLA, Third Military Medical University , Chongqing , PR China
| | - Jun Tang
- b Department of Dermatology , 105th Hospital of PLA , Hefei , PR China
| | - Zhiqiang Tian
- a Institute of Immunology, PLA, Third Military Medical University , Chongqing , PR China
| | | | - Jianxun Song
- d Department of Microbiology and Immunology , The Pennsylvania State University College of Medicine , Hershey , PA , USA
| | - Yuzhang Wu
- a Institute of Immunology, PLA, Third Military Medical University , Chongqing , PR China
| | - Bing Ni
- a Institute of Immunology, PLA, Third Military Medical University , Chongqing , PR China
| |
Collapse
|
671
|
Buhler S, Giostra E, Gbame C, de Rham C, Mullhaupt B, Dufour JF, Majno P, Negro F, Bochud PY, Villard J. A significant effect of the killer cell immunoglobulin-like receptor ligand human leucocyte antigen-C on fibrosis progression in chronic C hepatitis with or without liver transplantation. Liver Int 2016; 36:1331-9. [PMID: 26717049 DOI: 10.1111/liv.13057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/18/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS The interaction of killer cell immunoglobulin-like receptors with their human leucocyte antigen ligands drives the activation and inhibition of natural killer cells. Natural killer cells could be implicated in the development of liver fibrosis in chronic hepatitis C. METHODS We analysed 206 non-transplanted and 53 liver transplanted patients, selected according to their Metavir fibrosis stage. Several variables such as the number of activator killer cell immunoglobulin-like receptors or the human leucocyte antigen ligands were considered in multinomial and logistic regression models. Possible confounding variables were also investigated. RESULTS The killer cell immunoglobulin-like receptors were not significant predictors of the fibrosis stage. Conversely, a significant reduction of the human leucocyte antigen-C1C2 genotype was observed in the most advanced fibrosis stage group (F4) in both cohorts. Furthermore, the progression rate of fibrosis was almost 10 times faster in the subgroup of patients after liver transplantation, and human leucocyte antigen-C1C2 was significantly reduced in this cohort compared with non-transplanted patients. CONCLUSION This study suggests a possible role of killer cell immunoglobulin-like receptors and their ligands in the development of liver damage. The absence of C1 and C2 ligands heterozygosity could lead to less inhibition of natural killer cells and a quicker progression to a high level of fibrosis in patients infected with hepatitis C virus, especially following liver transplantation.
Collapse
Affiliation(s)
- Stéphane Buhler
- Transplantation Immunology Unit, Service of Nephrology, Department of Internal Medicine Specialties and Genetic and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland.,Laboratory of Anthropology, Genetics and Peopling History, Department of Genetics and Evolution - Anthropology Unit, University of Geneva, Geneva, Switzerland
| | - Emiliano Giostra
- Service of Transplantation, Geneva University Hospitals, Geneva, Switzerland
| | - Corinne Gbame
- Transplantation Immunology Unit, Service of Nephrology, Department of Internal Medicine Specialties and Genetic and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Casimir de Rham
- Transplantation Immunology Unit, Service of Nephrology, Department of Internal Medicine Specialties and Genetic and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Beat Mullhaupt
- Division of Gastroenterology and Hepatology, Zurich University Hospital, Zurich, Switzerland
| | - Jean-François Dufour
- University Clinic of Visceral Surgery and Medicine Inselspital, Bern, Switzerland
| | - Pietro Majno
- Service of Transplantation, Geneva University Hospitals, Geneva, Switzerland
| | - Francesco Negro
- Service of Gastroenterology and Hepatology, Geneva University Hospitals, Geneva, Switzerland.,Service of Clinical Pathology, Geneva University Hospitals, Geneva, Switzerland
| | | | - Jean Villard
- Transplantation Immunology Unit, Service of Nephrology, Department of Internal Medicine Specialties and Genetic and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | | |
Collapse
|
672
|
Ding C, Li Y, Guo F, Jiang Y, Ying W, Li D, Yang D, Xia X, Liu W, Zhao Y, He Y, Li X, Sun W, Liu Q, Song L, Zhen B, Zhang P, Qian X, Qin J, He F. A Cell-type-resolved Liver Proteome. Mol Cell Proteomics 2016; 15:3190-3202. [PMID: 27562671 PMCID: PMC5054343 DOI: 10.1074/mcp.m116.060145] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Indexed: 01/16/2023] Open
Abstract
Parenchymatous organs consist of multiple cell types, primarily defined as parenchymal cells (PCs) and nonparenchymal cells (NPCs). The cellular characteristics of these organs are not well understood. Proteomic studies facilitate the resolution of the molecular details of different cell types in organs. These studies have significantly extended our knowledge about organogenesis and organ cellular composition. Here, we present an atlas of the cell-type-resolved liver proteome. In-depth proteomics identified 6000 to 8000 gene products (GPs) for each cell type and a total of 10,075 GPs for four cell types. This data set revealed features of the cellular composition of the liver: (1) hepatocytes (PCs) express the least GPs, have a unique but highly homogenous proteome pattern, and execute fundamental liver functions; (2) the division of labor among PCs and NPCs follows a model in which PCs make the main components of pathways, but NPCs trigger the pathways; and (3) crosstalk among NPCs and PCs maintains the PC phenotype. This study presents the liver proteome at cell resolution, serving as a research model for dissecting the cell type constitution and organ features at the molecular level.
Collapse
Affiliation(s)
- Chen Ding
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China; **State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Yanyan Li
- ¶School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Feifei Guo
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China
| | - Ying Jiang
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China
| | - Wantao Ying
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China
| | - Dong Li
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China
| | - Dong Yang
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China
| | - Xia Xia
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China
| | - Wanlin Liu
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China
| | - Yan Zhao
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China
| | - Yangzhige He
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China; ¶School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xianyu Li
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China
| | - Wei Sun
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China
| | - Qiongming Liu
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China
| | - Lei Song
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China
| | - Bei Zhen
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China
| | - Pumin Zhang
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China
| | - Xiaohong Qian
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China;
| | - Jun Qin
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China; ‖Alkek Center for Molecular Discovery, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030; **State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Fuchu He
- From the ‡State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing 100039, China; §National Center for Protein Sciences (The PHOENIX center, Beijing), Beijing 102206, China; **State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai 200433, China
| |
Collapse
|
673
|
Pritchard MT, McCracken JM. Identifying Novel Targets for Treatment of Liver Fibrosis: What Can We Learn from Injured Tissues which Heal Without a Scar? Curr Drug Targets 2016; 16:1332-46. [PMID: 26302807 DOI: 10.2174/1389450116666150825111439] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 08/08/2015] [Indexed: 02/07/2023]
Abstract
The liver is unique in that it is able to regenerate. This regeneration occurs without formation of a scar in the case of non-iterative hepatic injury. However, when the liver is exposed to chronic liver injury, the purely regenerative process fails and excessive extracellular matrix proteins are deposited in place of normal liver parenchyma. While much has been discovered in the past three decades, insights into fibrotic mechanisms have not yet lead to effective therapies; liver transplant remains the only cure for advanced liver disease. In an effort to broaden the collection of possible therapeutic targets, this review will compare and contrast the liver wound healing response to that found in two types of wound healing: scarless wound healing of fetal skin and oral mucosa and scar-forming wound healing found in adult skin. This review will examine wound healing in the liver and the skin in relation to the role of humoral and cellular factors, as well as the extracellular matrix, in this process. While several therapeutic targets are similar between fibrotic liver and adult skin wound healing, others are unique and represent novel areas for hepatic anti-fibrotic research. In particular, investigations into the role of hyaluronan in liver fibrosis and fibrosis resolution are warranted.
Collapse
Affiliation(s)
- Michele T Pritchard
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66161, USA.
| | | |
Collapse
|
674
|
Seo W, Eun HS, Kim SY, Yi HS, Lee YS, Park SH, Jang MJ, Jo E, Kim SC, Han YM, Park KG, Jeong WI. Exosome-mediated activation of toll-like receptor 3 in stellate cells stimulates interleukin-17 production by γδ T cells in liver fibrosis. Hepatology 2016; 64:616-31. [PMID: 27178735 DOI: 10.1002/hep.28644] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 04/10/2016] [Accepted: 04/30/2016] [Indexed: 12/11/2022]
Abstract
UNLABELLED During liver injury, hepatocytes secrete exosomes that include diverse types of self-RNAs. Recently, self-noncoding RNA has been recognized as an activator of Toll-like receptor 3 (TLR3). However, the roles of hepatic exosomes and TLR3 in liver fibrosis are not yet fully understood. Following acute liver injury and early-stage liver fibrosis induced by a single or 2-week injection of carbon tetrachloride (CCl4 ), increased interleukin (IL)-17A production was detected primarily in hepatic γδ T cells in wild-type (WT) mice. However, liver fibrosis and IL-17A production by γδ T cells were both significantly attenuated in TLR3 knockout (KO) mice compared with WT mice. More interestingly, IL-17A-producing γδ T cells were in close contact with activated hepatic stellate cells (HSCs), suggesting a role for HSCs in IL-17A production by γδ T cells. In vitro treatments with exosomes derived from CCl4 -treated hepatocytes significantly increased the expression of IL-17A, IL-1β, and IL-23 in WT HSCs but not in TLR3 KO HSCs. Furthermore, IL-17A production by γδ T cells was substantially increased upon coculturing with exosome-treated WT HSCs or conditioned medium from TLR3-activated WT HSCs. However, similar increases were not detected when γδ T cells were cocultured with exosome-treated HSCs from IL-17A KO or TLR3 KO mice. Using reciprocal bone marrow transplantation between WT and TLR3 KO mice, we found that TLR3 deficiency in HSCs contributed to decreased IL-17A production by γδ T cells, as well as liver fibrosis. CONCLUSION In liver injury, the exosome-mediated activation of TLR3 in HSCs exacerbates liver fibrosis by enhancing IL-17A production by γδ T cells, which might be associated with HSC stimulation by unknown self-TLR3 ligands from damaged hepatocytes. Therefore, TLR3 might be a novel therapeutic target for liver fibrosis. (Hepatology 2016;64:616-631).
Collapse
Affiliation(s)
- Wonhyo Seo
- Laboratory of Liver Research, Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon, Republic of Korea
| | - Hyuk Soo Eun
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - So Yeon Kim
- Laboratory of Liver Research, Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon, Republic of Korea
| | - Hyon-Seung Yi
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Young-Sun Lee
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seol-Hee Park
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
- Department of Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Mi-Jin Jang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Eunjung Jo
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Sun Chang Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
- Intelligent Synthetic Biology Center, Daejeon, Republic of Korea
| | - Yong-Mahn Han
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Keun-Gyu Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Won-Il Jeong
- Laboratory of Liver Research, Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon, Republic of Korea
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
675
|
Inzaugarat ME, Wree A, Feldstein AE. Hepatocyte mitochondrial DNA released in microparticles and toll-like receptor 9 activation: A link between lipotoxicity and inflammation during nonalcoholic steatohepatitis. Hepatology 2016; 64:669-71. [PMID: 27239763 PMCID: PMC4956555 DOI: 10.1002/hep.28666] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
| | - Alexander Wree
- Department of Internal Medicine III, University Hospital,
RWTH-Aachen, Germany,Department of Pediatric Gastroenterology, University of California
San Diego (UCSD), and Rady Children’s Hospital, San Diego, California,
United States
| | - Ariel E. Feldstein
- Department of Pediatric Gastroenterology, University of California
San Diego (UCSD), and Rady Children’s Hospital, San Diego, California,
United States,Address for correspondence: Dr. Ariel E. Feldstein, Professor of
Pediatrics, Chief, Division of Pediatric Gastroenterology, Hepatology, and
Nutrition UCSD, 3020 Children’s Way, MC 5030, San Diego, CA 92103-8450,
Tel: (858) 966-8907,
| |
Collapse
|
676
|
Mehal W, To U. New approaches for fibrosis regression in alcoholic cirrhosis. Hepatol Int 2016; 10:773-8. [PMID: 27460408 DOI: 10.1007/s12072-016-9752-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 06/21/2016] [Indexed: 01/18/2023]
Abstract
Liver fibrosis is a dynamic process of fibrinogenesis and fibrinolysis. It is sequelae of recurrent injury and inflammation to the liver. Only recently has there been significant progress in understanding the pathophysiology behind liver fibrosis. This has allowed for the development of identifiable targets for potential therapies. In this article we will discuss the underlying general cellular mechanisms that play a key role in the pathway of fibrinogenesis and fibrinolysis and then focus on the mechanisms that are key in alcohol-induced liver fibrosis. Challenges in formulating potential fibrosis therapies as well as current potential targets for liver fibrosis will be reviewed as well.
Collapse
Affiliation(s)
- Wajahat Mehal
- Section of Digestive Diseases, Department of Veterans Affairs Connecticut Healthcare, West Haven, CT, 06516, USA. .,Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, CT, 06520, USA.
| | - Uyen To
- Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, CT, 06520, USA
| |
Collapse
|
677
|
Lee HS, Shin HS, Choi J, Bae SJ, Wee HJ, Son T, Seo JH, Park JH, Kim SW, Kim KW. AMP-activated protein kinase activator, HL156A reduces thioacetamide-induced liver fibrosis in mice and inhibits the activation of cultured hepatic stellate cells and macrophages. Int J Oncol 2016; 49:1407-14. [DOI: 10.3892/ijo.2016.3627] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/04/2016] [Indexed: 11/05/2022] Open
|
678
|
MicroRNA-146a-5p Negatively Regulates Pro-Inflammatory Cytokine Secretion and Cell Activation in Lipopolysaccharide Stimulated Human Hepatic Stellate Cells through Inhibition of Toll-Like Receptor 4 Signaling Pathways. Int J Mol Sci 2016; 17:ijms17071076. [PMID: 27399683 PMCID: PMC4964452 DOI: 10.3390/ijms17071076] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 06/20/2016] [Accepted: 06/28/2016] [Indexed: 12/13/2022] Open
Abstract
Lipopolysaccharide (LPS)/toll-like receptor 4 (TLR4) signaling pathway is demonstrated to be involved in the hepatic fibrosis. MicroRNA (miR)-146a-5p is a key regulator of the innate immune response. The functional significance of miR-146a-5p during the LPS/TLR4 mediated hepatic fibrosis process remains unclear. In this study, we found that TLR4 and α-smooth muscle actin (α-SMA) were up-regulated and miR-146a-5p was down-regulated in human hepatic stellate cell (HSC) line LX2 after LPS stimulation. Overexpression of miR-146a-5p inhibited LPS induced pro-inflammatory cytokines secretion through down-regulating the expression levels of TLR-4, IL-1 receptor-associated kinase 1 (IRAK1), TNF receptor associated factor-6 (TRAF6) and phosphorylation of nuclear factor-kappa B (NF-κB). Knockdown of IRAK1 and TRAF6 also suppressed pro-inflammatory cytokine production by inhibiting NF-κB phosphorylation. In addition, miR-146a-5p mimic blocked LPS induced TRAF6 dependent c-Jun N-terminal kinase (JNK) and Smad2 activation as well as α-SMA production. Taken together, these results suggest that miR-146a-5p suppresses pro-inflammatory cytokine secretion and cell activation of HSC through inhibition of TLR4/NF-κB and TLR4/TRAF6/JNK pathway.
Collapse
|
679
|
Pentoxifylline inhibits liver fibrosis via hedgehog signaling pathway. ACTA ACUST UNITED AC 2016; 36:372-376. [PMID: 27376806 DOI: 10.1007/s11596-016-1594-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 04/22/2016] [Indexed: 12/29/2022]
Abstract
Infection of schistosomiasis japonica may eventually lead to liver fibrosis, and no effective antifibrotic therapies are available but liver transplantation. Hedgehog (HH) signaling pathway has been involved in the process and is a promising target for treating liver fibrosis. This study aimed to explore the effects of pentoxifylline (PTX) on liver fibrosis induced by schistosoma japonicum infection by inhibiting the HH signaling pathway. Phorbol12-myristate13-acetate (PMA) was used to induce human acute mononuclear leukemia cells THP-1 to differentiate into macrophages. The THP-1-derived macrophages were stimulated by soluble egg antigen (SEA), and the culture supernatants were collected for detection of activation of macrophages. Cell Counting Kit-8 (CCK-8) was used to detect the cytotoxicity of the culture supernatant and PTX on the LX-2 cells. The LX-2 cells were administered with activated culture supernatant from macrophages and(or) PTX to detect the transforming growth factor-β gene expression. The mRNA expression of shh and gli-1, key parts in HH signaling pathway, was detected. The mRNA expression of shh and gli-1 was increased in LX-2 cells treated with activated macrophages-derived culture supernatant, suggesting HH signaling pathway may play a key role in the activation process of hepatic stellate cells (HSCs). The expression of these genes decreased in LX-2 cells co-cultured with both activated macrophages-derived culture supernatant and PTX, indicating PTX could suppress the activation process of HSCs. In conclusion, these data provide evidence that PTX prevents liver fibrogenesis in vitro by the suppression of HH signaling pathway.
Collapse
|
680
|
Elkrief L, Rautou PE, Sarin S, Valla D, Paradis V, Moreau R. Diabetes mellitus in patients with cirrhosis: clinical implications and management. Liver Int 2016; 36:936-48. [PMID: 26972930 DOI: 10.1111/liv.13115] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 03/07/2016] [Indexed: 12/12/2022]
Abstract
Disorders of glucose metabolism, namely glucose intolerance and diabetes, are frequent in patients with chronic liver diseases. In patients with cirrhosis, diabetes can be either a classical type 2 diabetes mellitus or the so-called hepatogenous diabetes, i.e. a consequence of liver insufficiency and portal hypertension. This review article provides an overview of the possible pathophysiological mechanisms explaining diabetes in patients with cirrhosis. Cirrhosis is associated with portosystemic shunts as well as reduced hepatic mass, which can both impair insulin clearance by the liver, contributing to peripheral insulin resistance through insulin receptors down-regulation. Moreover, cirrhosis is associated with increased levels of advanced-glycation-end products and hypoxia-inducible-factors, which may play a role in the development of diabetes. This review also focuses on the clinical implications of diabetes in patients with cirrhosis. First, diabetes is an independent factor for poor prognosis in patients with cirrhosis. Specifically, diabetes is associated with the occurrence of major complications of cirrhosis, including ascites and renal dysfunction, hepatic encephalopathy and bacterial infections. Diabetes is also associated with an increased risk of hepatocellular carcinoma in patients with chronic liver diseases. Last, the management of patients with concurrent diabetes and liver disease is also addressed. Recent findings suggest a beneficial impact of metformin in patients with chronic liver diseases. Insulin is often required in patients with advanced cirrhosis. However, the favourable impact of controlling diabetes in patients with cirrhosis has not been demonstrated yet.
Collapse
Affiliation(s)
- Laure Elkrief
- Service de Gastroentérologie et Hépatologie, Hôpitaux Universitaires de Genève, Genève, Suisse
| | - Pierre-Emmanuel Rautou
- DHU UNITY, Service d'Hépatologie, Hôpital Beaujon, APHP, Clichy, France.,Université Paris-Diderot, Sorbonne Paris Cité, Paris, France.,Inserm U970, Paris Research Cardiovascular Center, Paris, France
| | - Shiv Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Dominique Valla
- DHU UNITY, Service d'Hépatologie, Hôpital Beaujon, APHP, Clichy, France.,Université Paris-Diderot, Sorbonne Paris Cité, Paris, France.,Inserm U1149, Centre de Recherche sur l'Inflammation CRI, Clichy, France
| | - Valérie Paradis
- Université Paris-Diderot, Sorbonne Paris Cité, Paris, France.,Inserm U1149, Centre de Recherche sur l'Inflammation CRI, Clichy, France.,DHU UNITY, Pathology Department, Hôpital Beaujon, APHP, Clichy, France
| | - Richard Moreau
- DHU UNITY, Service d'Hépatologie, Hôpital Beaujon, APHP, Clichy, France.,Université Paris-Diderot, Sorbonne Paris Cité, Paris, France.,Inserm U1149, Centre de Recherche sur l'Inflammation CRI, Clichy, France
| |
Collapse
|
681
|
Oh Y, Park O, Swierczewska M, Hamilton JP, Park JS, Kim TH, Lim SM, Eom H, Jo DG, Lee CE, Kechrid R, Mastorakos P, Zhang C, Hahn SK, Jeon OC, Byun Y, Kim K, Hanes J, Lee KC, Pomper MG, Gao B, Lee S. Systemic PEGylated TRAIL treatment ameliorates liver cirrhosis in rats by eliminating activated hepatic stellate cells. Hepatology 2016; 64:209-23. [PMID: 26710118 PMCID: PMC4917440 DOI: 10.1002/hep.28432] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 11/20/2015] [Accepted: 12/23/2015] [Indexed: 12/20/2022]
Abstract
UNLABELLED Liver fibrosis is a common outcome of chronic liver disease that leads to liver cirrhosis and hepatocellular carcinoma. No US Food and Drug Administration-approved targeted antifibrotic therapy exists. Activated hepatic stellate cells (aHSCs) are the major cell types responsible for liver fibrosis; therefore, eradication of aHSCs, while preserving quiescent HSCs and other normal cells, is a logical strategy to stop and/or reverse liver fibrogenesis/fibrosis. However, there are no effective approaches to specifically deplete aHSCs during fibrosis without systemic toxicity. aHSCs are associated with elevated expression of death receptors and become sensitive to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced cell death. Treatment with recombinant TRAIL could be a potential strategy to ameliorate liver fibrosis; however, the therapeutic application of recombinant TRAIL is halted due to its very short half-life. To overcome this problem, we previously generated PEGylated TRAIL (TRAILPEG ) that has a much longer half-life in rodents than native-type TRAIL. In this study, we demonstrate that intravenous TRAILPEG has a markedly extended half-life over native-type TRAIL in nonhuman primates and has no toxicity in primary human hepatocytes. Intravenous injection of TRAILPEG directly induces apoptosis of aHSCs in vivo and ameliorates carbon tetrachloride-induced fibrosis/cirrhosis in rats by simultaneously down-regulating multiple key fibrotic markers that are associated with aHSCs. CONCLUSION TRAIL-based therapies could serve as new therapeutics for liver fibrosis/cirrhosis and possibly other fibrotic diseases. (Hepatology 2016;64:209-223).
Collapse
Affiliation(s)
- Yumin Oh
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,The Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ogyi Park
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,The Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Magdalena Swierczewska
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,The Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - James P. Hamilton
- Divison of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jong-Sung Park
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,The Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tae Hyung Kim
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,The Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sung-Mook Lim
- College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Hana Eom
- College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Dong Gyu Jo
- College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Choong-Eun Lee
- Department of Biological Science, Sungkyunkwan University, Suwon, Korea
| | - Raouf Kechrid
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Panagiotis Mastorakos
- The Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Clark Zhang
- The Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sei Kwang Hahn
- Department of Materials Science and Engineering, Pohang University of Science and Technology, Pohang, Korea
| | - Ok-Cheol Jeon
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Youngro Byun
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Kwangmeyung Kim
- Center for Theragnosis, Korea Institute of Science and Technology, Seoul, Korea
| | - Justin Hanes
- The Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kang Choon Lee
- College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Martin G. Pomper
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA,Correspondence authors: Bin Gao, M.D., Ph.D., Laboratory of Liver Diseases, NIAAA/NIH, 5625 Fishers Lane, Bethesda, MD 20892. Tel: 301-443-3998; and Seulki Lee, Ph.D. The Russell H. Morgan Department of Radiology and Radiological Sciences, The Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University, School of Medicine, Baltimore, MD 21231. Tel: 443-287-4892;
| | - Seulki Lee
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,The Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,Correspondence authors: Bin Gao, M.D., Ph.D., Laboratory of Liver Diseases, NIAAA/NIH, 5625 Fishers Lane, Bethesda, MD 20892. Tel: 301-443-3998; and Seulki Lee, Ph.D. The Russell H. Morgan Department of Radiology and Radiological Sciences, The Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University, School of Medicine, Baltimore, MD 21231. Tel: 443-287-4892;
| |
Collapse
|
682
|
Geng XX, Huang RG, Lin JM, Jiang N, Yang XX. Transient elastography in clinical detection of liver cirrhosis: A systematic review and meta-analysis. Saudi J Gastroenterol 2016; 22:294-303. [PMID: 27488324 PMCID: PMC4991200 DOI: 10.4103/1319-3767.187603] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/27/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND/AIMS Transient elastography is a noninvasive method for measuring liver fibrosis. This meta-analysis assesses the diagnostic performance of transient elastography of detecting liver cirrhosis in patients with liver disease. PATIENTS AND METHODS We searched MEDLINE, Cochrane, EMBASE databases until Jan 31, 2015, using the following search terms: elastography and liver cirrhosis. Included studies assessed patients with a diagnosis of liver cirrhosis, with an index test of transient elastography, and with the reference standard being a histopathological exam by liver biopsy. Sensitivity analysis and assessment of risk of bias and publication bias were performed. RESULTS Fifty-seven studies were included in the meta-analysis with a total of 10,504 patients. The pooled estimate for the sensitivity of transient elastography for detecting liver fibrosis was 81% and the specificity was 88%. The imputed diagnostic odds ratio (DOR) was 26.08 and the area under the receiver-operating characteristic (AUROC) curve was 0.931. CONCLUSION Our findings indicate that transient elastography shows good sensitivity, specificity and a high accuracy for detecting liver cirrhosis. Transient elastography can be used as an additional method for the clinical diagnosis of liver fibrosis and cirrhosis.
Collapse
Affiliation(s)
- Xiao-Xia Geng
- Department of Infectious Diseases, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu 610072, Sichuan Province, China
| | - Ren-Gang Huang
- Department of Infectious Diseases, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu 610072, Sichuan Province, China
| | - Jian-Mei Lin
- Department of Infectious Diseases, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu 610072, Sichuan Province, China
| | - Nan Jiang
- Department of Infectious Diseases, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu 610072, Sichuan Province, China
| | - Xing-Xiang Yang
- Department of Infectious Diseases, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu 610072, Sichuan Province, China
| |
Collapse
|
683
|
Yang JH, Kim SC, Kim KM, Jang CH, Cho SS, Kim SJ, Ku SK, Cho IJ, Ki SH. Isorhamnetin attenuates liver fibrosis by inhibiting TGF-β/Smad signaling and relieving oxidative stress. Eur J Pharmacol 2016; 783:92-102. [DOI: 10.1016/j.ejphar.2016.04.042] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 04/18/2016] [Accepted: 04/20/2016] [Indexed: 12/28/2022]
|
684
|
Shi L, Qin E, Zhou J, Zhao J, Nie W, Jiang T, Chen W, Wu D, Huang L, Liu L, Lv L, Zhao M, Zhang Z, Wang F. HIV and HCV Co-Culture Promotes Profibrogenic Gene Expression through an Epimorphin-Mediated ERK Signaling Pathway in Hepatic Stellate Cells. PLoS One 2016; 11:e0158386. [PMID: 27362846 PMCID: PMC4928874 DOI: 10.1371/journal.pone.0158386] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 06/15/2016] [Indexed: 01/06/2023] Open
Abstract
Accelerated fibrosis in patients co-infected with hepatitis C virus (HCV) and human immunodeficiency virus (HIV) has been a major cause of mortality in the highly active anti-retroviral therapy (HAART) era. However, the role of co-infection in accelerating the progression of liver fibrosis, particularly with regard to the effects of co-infection on hepatic stellate cells (HSCs), remains unclear. We hypothesized that HIV and HCV induce liver fibrosis synergistically by altering the regulation of epimorphin production, and thereby indirectly alter HSC function. Here, we examined the effects of epimorphin on HSC proliferation and invasion, and the changes in fibrogenesis-related gene activity in HSCs (LX2) in the presence of inactivated CXCR4-tropic HIV and HCV (JFH1). The combination of HIV and HCV significantly increased epimorphin expression, which increased the proliferation and invasion capabilities of HSCs. Epimorphin also induced the expression of profibrogenic tissue inhibitor of metalloproteinase 1 (TIMP1) in an extracellular signal-regulated kinase (ERK)-dependent manner. These data indicated that the effects of HIV/HCV co-infection on hepatic fibrosis might be mediated in part by EPM. Strategies to limit the expression of EPM might represent a novel therapeutic approach to prevent the progression of hepatic fibrosis during HIV/HCV co-infection.
Collapse
Affiliation(s)
- Lei Shi
- Medical School of Chinese PLA, Beijing, China
- Treatment and Research Center for Infectious Diseases, Beijing 302 Hospital, Beijing, China
| | - Enqiang Qin
- Treatment and Research Center for Infectious Diseases, Beijing 302 Hospital, Beijing, China
| | - Junnian Zhou
- Beijing Institute of Transfusion Medicine, Beijing, China
| | - Juanjuan Zhao
- Research Center for Clinical and Translational Medicine, Beijing 302 Hospital, Beijing, China
| | - Weimin Nie
- Treatment and Research Center for Infectious Diseases, Beijing 302 Hospital, Beijing, China
| | - Tianjun Jiang
- Treatment and Research Center for Infectious Diseases, Beijing 302 Hospital, Beijing, China
| | - Weiwei Chen
- Treatment and Research Center for Infectious Diseases, Beijing 302 Hospital, Beijing, China
| | - Dan Wu
- Treatment and Research Center for Infectious Diseases, Beijing 302 Hospital, Beijing, China
| | - Lei Huang
- Treatment and Research Center for Infectious Diseases, Beijing 302 Hospital, Beijing, China
| | - Liying Liu
- Tumor Radiotherapy Center, Beijing 302 Hospital, Beijing, China
| | - Liping Lv
- Beijing Institute of Transfusion Medicine, Beijing, China
| | - Min Zhao
- Treatment and Research Center for Infectious Diseases, Beijing 302 Hospital, Beijing, China
| | - Zheng Zhang
- Medical School of Chinese PLA, Beijing, China
- Research Center for Clinical and Translational Medicine, Beijing 302 Hospital, Beijing, China
- * E-mail: (FW); (ZZ)
| | - Fusheng Wang
- Medical School of Chinese PLA, Beijing, China
- Treatment and Research Center for Infectious Diseases, Beijing 302 Hospital, Beijing, China
- * E-mail: (FW); (ZZ)
| |
Collapse
|
685
|
Transcriptional repression of SIRT1 by protein inhibitor of activated STAT 4 (PIAS4) in hepatic stellate cells contributes to liver fibrosis. Sci Rep 2016; 6:28432. [PMID: 27323886 PMCID: PMC4914937 DOI: 10.1038/srep28432] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 06/06/2016] [Indexed: 02/06/2023] Open
Abstract
Interstitial fibrosis represents a key pathological process in non-alcoholic steatohepatitis (NASH). In the liver, fibrogenesis is primarily mediated by activated hepatic stellate cells (HSCs) transitioning from a quiescent state in response to a host of stimuli. The molecular mechanism underlying HSC activation is not completely understood. Here we report that there was a simultaneous up-regulation of PIAS4 expression and down-regulation of SIRT1 expression accompanying increased hepatic fibrogenesis in an MCD-diet induced mouse model of NASH. In cultured primary mouse HSCs, stimulation with high glucose activated PIAS4 while at the same time repressed SIRT1. Over-expression of PIAS4 directly repressed SIRT1 promoter activity. In contrast, depletion of PIAS4 restored SIRT1 expression in HSCs treated with high glucose. Estrogen, a known NASH-protective hormone, antagonized HSC activation by targeting PIAS4. Lentivirus-mediated delivery of short hairpin RNA (shRNA) targeting PIAS4 in mice ameliorated MCD diet induced liver fibrosis by normalizing SIRT1 expression in vivo. PIAS4 promoted HSC activation in a SIRT1-dependent manner in vitro. Mechanistically, PIAS4 mediated SIRT1 repression led to SMAD3 hyperacetylation and enhanced SMAD3 binding to fibrogenic gene promoters. Taken together, our data suggest SIRT1 trans-repression by PIAS4 plays an important role in HSC activation and liver fibrosis.
Collapse
|
686
|
Nwosu ZC, Alborzinia H, Wölfl S, Dooley S, Liu Y. Evolving Insights on Metabolism, Autophagy, and Epigenetics in Liver Myofibroblasts. Front Physiol 2016; 7:191. [PMID: 27313533 PMCID: PMC4887492 DOI: 10.3389/fphys.2016.00191] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 05/12/2016] [Indexed: 12/14/2022] Open
Abstract
Liver myofibroblasts (MFB) are crucial mediators of extracellular matrix (ECM) deposition in liver fibrosis. They arise mainly from hepatic stellate cells (HSCs) upon a process termed “activation.” To a lesser extent, and depending on the cause of liver damage, portal fibroblasts, mesothelial cells, and fibrocytes may also contribute to the MFB population. Targeting MFB to reduce liver fibrosis is currently an area of intense research. Unfortunately, a clog in the wheel of antifibrotic therapies is the fact that although MFB are known to mediate scar formation, and participate in liver inflammatory response, many of their molecular portraits are currently unknown. In this review, we discuss recent understanding of MFB in health and diseases, focusing specifically on three evolving research fields: metabolism, autophagy, and epigenetics. We have emphasized on therapeutic prospects where applicable and mentioned techniques for use in MFB studies. Subsequently, we highlighted uncharted territories in MFB research to help direct future efforts aimed at bridging gaps in current knowledge.
Collapse
Affiliation(s)
- Zeribe C Nwosu
- Molecular Hepatology Section, Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg Mannheim, Germany
| | - Hamed Alborzinia
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg Heidelberg, Germany
| | - Stefan Wölfl
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg Heidelberg, Germany
| | - Steven Dooley
- Molecular Hepatology Section, Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg Mannheim, Germany
| | - Yan Liu
- Molecular Hepatology Section, Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg Mannheim, Germany
| |
Collapse
|
687
|
Chen QL, Luo Z, Huang C, Pan YX, Wu K. De novo characterization of the liver transcriptome of javelin goby Synechogobius hasta and analysis of its transcriptomic profile following waterborne copper exposure. FISH PHYSIOLOGY AND BIOCHEMISTRY 2016; 42:979-994. [PMID: 26719065 DOI: 10.1007/s10695-015-0190-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 12/25/2015] [Indexed: 06/05/2023]
Abstract
Previous studies have investigated the physiological responses to chronic copper (Cu) exposure in the liver of Synechogobius hasta; however, little information is available on the underlying molecular mechanisms. In an effort to better understand the mechanisms of Cu toxicity and to illuminate global gene expression patterns modulated by Cu exposure, we obtained the liver transcriptome information of S. hasta by RNA sequencing (RNA-seq) technology and also investigated the differential expression of genes following waterborne Cu exposure. Using the Illumina sequencing platform, as many as 60,217 unigenes were generated, with 815 bp of average length and 1298 bp of unigene N50 after filtering and assembly. For functional annotation analysis, 34,860, 31,526, 31,576, 25,808, 11,542, and 21,721 unigenes were annotated to the NR, NT, Swiss-Prot, KEGG, COG, and GO databases, respectively, and total annotation unigenes were 37,764. After 30 days of exposure to 55 μg Cu/l, a total of 292 and 1076 genes were significantly up- and down-regulated, respectively. By KEGG analysis, 660 had a specific pathway annotation. Subsequent bioinformatics analysis revealed that the differentially expressed genes were mainly related to lipid metabolism, immune system, apoptosis, and signal transduction, suggesting that these signaling pathways may be regulated by Cu exposure. The present study provides comprehensive sequence information for subsequent gene expression studies regarding S. hasta, and the transcriptome profiling after Cu exposure is also expected to improve our understanding of the molecular toxicology of Cu.
Collapse
Affiliation(s)
- Qi-Liang Chen
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Fishery College, Huazhong Agricultural University, Wuhan, 430070, China
- Freshwater Aquaculture Collaborative Innovative Centre of Hubei Province, Wuhan, 430070, China
| | - Zhi Luo
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Fishery College, Huazhong Agricultural University, Wuhan, 430070, China.
- Freshwater Aquaculture Collaborative Innovative Centre of Hubei Province, Wuhan, 430070, China.
- Department of Animal Sciences, Cornell University, Morrison Hall, Ithaca, NY, 14850, USA.
| | - Chao Huang
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Fishery College, Huazhong Agricultural University, Wuhan, 430070, China
- Freshwater Aquaculture Collaborative Innovative Centre of Hubei Province, Wuhan, 430070, China
| | - Ya-Xiong Pan
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Fishery College, Huazhong Agricultural University, Wuhan, 430070, China
- Freshwater Aquaculture Collaborative Innovative Centre of Hubei Province, Wuhan, 430070, China
| | - Kun Wu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of China, Fishery College, Huazhong Agricultural University, Wuhan, 430070, China
- Freshwater Aquaculture Collaborative Innovative Centre of Hubei Province, Wuhan, 430070, China
| |
Collapse
|
688
|
Zhou B, Zeng S, Li L, Fan Z, Tian W, Li M, Xu H, Wu X, Fang M, Xu Y. Angiogenic factor with G patch and FHA domains 1 (Aggf1) regulates liver fibrosis by modulating TGF-β signaling. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1203-13. [DOI: 10.1016/j.bbadis.2016.02.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 01/26/2016] [Accepted: 02/01/2016] [Indexed: 11/26/2022]
|
689
|
HLA-B18 as a risk factor of short-term progression to severe liver fibrosis in HIV/HCV co-infected patients with absent or minimal fibrosis: implications for timing of therapy. THE PHARMACOGENOMICS JOURNAL 2016; 17:551-555. [PMID: 27241060 DOI: 10.1038/tpj.2016.42] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 04/19/2016] [Accepted: 05/02/2016] [Indexed: 02/06/2023]
Abstract
Our aim was to analyze the influence of HLA-B haplotypes on liver fibrosis progression in HIV/hepatitis C virus (HCV) co-infected patients. Retrospective longitudinal study including HIV/HCV, non-cirrhotic and HCV treatment-naïve patients. The main outcome variable was liver fibrosis progression of at least one stage. One hundred and four patients constituted the study population (F0-F1: 62 (59.6%); F2: 22 (21.2%); F3: 20 (19.2%)). During a median follow-up of 54.5 months (IQR: 26.2-77), 45 patients (43.3%) showed an increase in the stage of liver fibrosis (time to event: 29 (IQR: 14-49.5) months). HLA-B18pos patients more frequently had a higher and faster fibrosis progression rate (73.3%; 24 (IQR: 8-29) months) than HLA-B18neg patients (38.2%; 34.5 (IQR: 14.7-51.2) months). This association was also observed in the development of F3-F4 fibrosis among F0-F2 patients (HLA-B18pos: 69.2%; 18 (6.5-37) months vs HLA-B18neg: 28.2%; 37 (IQR: 19-52) months). These results could impact the timing of HCV therapy in F0-F2 patients.
Collapse
|
690
|
Shang H, Wang Z, Song Y. Liver progenitor cells-mediated liver regeneration in liver cirrhosis. Hepatol Int 2016; 10:440-447. [PMID: 26742763 DOI: 10.1007/s12072-015-9693-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 12/07/2015] [Indexed: 02/08/2023]
Abstract
Cirrhosis is defined as the histological development of regenerative nodules surrounded by fibrous bands in response to chronic liver injury. In cirrhotic liver where hepatocytes proliferation is compromised, liver progenitor cells (LPCs) are activated and then differentiated into hepatocytes and cholangiocytes, leading to the generation of regenerative nodules and functional restoration. Here, we summarize and discuss recent findings on the mechanisms underlying LPCs-mediated regeneration in liver cirrhosis. Firstly, we provide recent research on the mechanism underlying LPCs activation in severe or chronic liver injury. Secondly, we present new and exciting data on exploring the origin of LPCs, which reveal that the hepatocytes give rise to duct-like progenitors that then differentiate back into hepatocytes in chronic liver injury or liver cirrhosis. Finally, we highlight recent findings from the literature exploring the role of LPCs niche in directing the behavior and fate of LPCs. This remarkable insight into the cellular and molecular mechanisms of LPCs-mediated regeneration in liver cirrhosis will provide a basis for translating this knowledge into clinical application.
Collapse
Affiliation(s)
- Haitao Shang
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Zhijun Wang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Yuhu Song
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
| |
Collapse
|
691
|
Robinson MW, Harmon C, O'Farrelly C. Liver immunology and its role in inflammation and homeostasis. Cell Mol Immunol 2016; 13:267-76. [PMID: 27063467 PMCID: PMC4856809 DOI: 10.1038/cmi.2016.3] [Citation(s) in RCA: 759] [Impact Index Per Article: 84.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/08/2016] [Accepted: 01/09/2016] [Indexed: 12/12/2022] Open
Abstract
The human liver is usually perceived as a non-immunological organ engaged primarily in metabolic, nutrient storage and detoxification activities. However, we now know that the healthy liver is also a site of complex immunological activity mediated by a diverse immune cell repertoire as well as non-hematopoietic cell populations. In the non-diseased liver, metabolic and tissue remodeling functions require elements of inflammation. This inflammation, in combination with regular exposure to dietary and microbial products, creates the potential for excessive immune activation. In this complex microenvironment, the hepatic immune system tolerates harmless molecules while at the same time remaining alert to possible infectious agents, malignant cells or tissue damage. Upon appropriate immune activation to challenge by pathogens or tissue damage, mechanisms to resolve inflammation are essential to maintain liver homeostasis. Failure to clear 'dangerous' stimuli or regulate appropriately activated immune mechanisms leads to pathological inflammation and disrupted tissue homeostasis characterized by the progressive development of fibrosis, cirrhosis and eventual liver failure. Hepatic inflammatory mechanisms therefore have a spectrum of roles in the healthy adult liver; they are essential to maintain tissue and organ homeostasis and, when dysregulated, are key drivers of the liver pathology associated with chronic infection, autoimmunity and malignancy. In this review, we explore the changing perception of inflammation and inflammatory mediators in normal liver homeostasis and propose targeting of liver-specific immune regulation pathways as a therapeutic approach to treat liver disease.
Collapse
Affiliation(s)
- Mark W Robinson
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin D2, Ireland
| | - Cathal Harmon
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin D2, Ireland
| | - Cliona O'Farrelly
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin D2, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin D2, Ireland
| |
Collapse
|
692
|
Arrese M, Cabrera D, Kalergis AM, Feldstein AE. Innate Immunity and Inflammation in NAFLD/NASH. Dig Dis Sci 2016; 61:1294-303. [PMID: 26841783 PMCID: PMC4948286 DOI: 10.1007/s10620-016-4049-x] [Citation(s) in RCA: 380] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 01/19/2016] [Indexed: 02/06/2023]
Abstract
Inflammation and hepatocyte injury and death are the hallmarks of nonalcoholic steatohepatitis (NASH), the progressive form of nonalcoholic fatty liver disease (NAFLD), which is a currently burgeoning public health problem. Innate immune activation is a key factor in triggering and amplifying hepatic inflammation in NAFLD/NASH. Thus, identification of the underlying mechanisms by which immune cells in the liver recognize cell damage signals or the presence of pathogens or pathogen-derived factors that activate them is relevant from a therapeutic perspective. In this review, we present new insights into the factors promoting the inflammatory response in NASH including sterile cell death processes resulting from lipotoxicity in hepatocytes as well as into the altered gut-liver axis function, which involves translocation of bacterial products into portal circulation as a result of gut leakiness. We further delineate the key immune cell types involved and how they recognize both damage-associated molecular patterns or pathogen-associated molecular patterns through binding of surface-expressed pattern recognition receptors, which initiate signaling cascades leading to injury amplification. The relevance of modulating these inflammatory signaling pathways as potential novel therapeutic strategies for the treatment of NASH is summarized.
Collapse
Affiliation(s)
- Marco Arrese
- Departmento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniel Cabrera
- Departmento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Ciencias Químicas y Biológicas, Facultad de Salud, Universidad Bernardo O Higgins, Santiago, Chile
| | - Alexis M Kalergis
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ariel E Feldstein
- Department of Pediatrics, University of California San Diego (UCSD), San Diego, CA, USA.
- Rady Children's Hospital, San Diego, CA, USA.
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, UCSD, 3020 Children's Way, MC 5030, San Diego, CA, 92103-8450, USA.
| |
Collapse
|
693
|
Fujita T, Narumiya S. Roles of hepatic stellate cells in liver inflammation: a new perspective. Inflamm Regen 2016; 36:1. [PMID: 29259674 PMCID: PMC5721720 DOI: 10.1186/s41232-016-0005-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 04/02/2016] [Indexed: 12/16/2022] Open
Abstract
Connected with the intestinal tract through the portal circulation, liver sinusoids function as the first line of defense against extrahepatic stimuli such as bacterial products and other toxic substances. Hepatic stellate cells (HSCs) are pericytes residing in the perisinusoidal space, between sinusoidal endothelial cells and hepatocytes, store vitamin A, and regulate sinusoidal circulation. Following chronic hepatitis, HSCs actively produce extracellular matrices and cause liver fibrosis. In spite of their close position to the liver sinusoids, however, whether HSCs contribute to liver inflammation has remained elusive. Evidence now accumulates to suggest that HSCs actively take part in the regulation of various forms of liver inflammation. Upon inflammatory stimuli from the sinusoids, HSCs produce various inflammatory molecules and interact with other liver cells, thereby recruiting and then activating infiltrating leukocytes and ultimately causing hepatocyte death. On the other hand, HSCs also exert hepatoprotective effects through inhibition of cytokine and chemokine production or induction of immunosuppressive cell population. HSCs therefore integrate cytokine-mediated inflammatory responses in the sinusoids and relay them to the liver parenchyma, either amplifying liver inflammation or suppressing parenchymal damage through immunoregulatory signaling depending on the context.
Collapse
Affiliation(s)
- Tomoko Fujita
- Center for Innovation in Immunoregulatory Technology and Therapeutics, Faculty of Medicine, Kyoto University, Yoshida Konoecho, Sakyo-ku, Kyoto 606-8501 Japan
| | - Shuh Narumiya
- Center for Innovation in Immunoregulatory Technology and Therapeutics, Faculty of Medicine, Kyoto University, Yoshida Konoecho, Sakyo-ku, Kyoto 606-8501 Japan
| |
Collapse
|
694
|
Tabibian JH, Varghese C, LaRusso NF, O'Hara SP. The enteric microbiome in hepatobiliary health and disease. Liver Int 2016; 36:480-487. [PMID: 26561779 PMCID: PMC4825184 DOI: 10.1111/liv.13009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 11/02/2015] [Indexed: 12/15/2022]
Abstract
Increasing evidence points to the contribution of the intestinal microbiome as a potentially key determinant in the initiation and/or progression of hepatobiliary disease. While current understanding of this dynamic is incomplete, exciting insights are continually being made and more are expected given the developments in molecular and high-throughput omics techniques. In this brief review, we provide a practical and updated synopsis of the interaction of the intestinal microbiome with the liver and its downstream impact on the initiation, progression and complications of hepatobiliary disease.
Collapse
Affiliation(s)
- James H. Tabibian
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA, USA
| | - Cyril Varghese
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Nicholas F. LaRusso
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Steven P. O'Hara
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
695
|
Serum Amyloid A Induces Inflammation, Proliferation and Cell Death in Activated Hepatic Stellate Cells. PLoS One 2016; 11:e0150893. [PMID: 26937641 PMCID: PMC4777566 DOI: 10.1371/journal.pone.0150893] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 02/19/2016] [Indexed: 02/06/2023] Open
Abstract
Serum amyloid A (SAA) is an evolutionary highly conserved acute phase protein that is predominantly secreted by hepatocytes. However, its role in liver injury and fibrogenesis has not been elucidated so far. In this study, we determined the effects of SAA on hepatic stellate cells (HSCs), the main fibrogenic cell type of the liver. Serum amyloid A potently activated IκB kinase, c-Jun N-terminal kinase (JNK), Erk and Akt and enhanced NF-κB-dependent luciferase activity in primary human and rat HSCs. Serum amyloid A induced the transcription of MCP-1, RANTES and MMP9 in an NF-κB- and JNK-dependent manner. Blockade of NF-κB revealed cytotoxic effects of SAA in primary HSCs with signs of apoptosis such as caspase 3 and PARP cleavage and Annexin V staining. Serum amyloid A induced HSC proliferation, which depended on JNK, Erk and Akt activity. In primary hepatocytes, SAA also activated MAP kinases, but did not induce relevant cell death after NF-κB inhibition. In two models of hepatic fibrogenesis, CCl4 treatment and bile duct ligation, hepatic mRNA levels of SAA1 and SAA3 were strongly increased. In conclusion, SAA may modulate fibrogenic responses in the liver in a positive and negative fashion by inducing inflammation, proliferation and cell death in HSCs.
Collapse
|
696
|
Nishikawa H, Enomoto H, Iwata Y, Kishino K, Shimono Y, Hasegawa K, Nakano C, Takata R, Nishimura T, Yoh K, Ishii A, Aizawa N, Sakai Y, Ikeda N, Takashima T, Iijima H, Nishiguchi S. B-Cell Activating Factor Belonging to the Tumor Necrosis Factor Family and Interferon-γ-Inducible Protein-10 in Autoimmune Hepatitis. Medicine (Baltimore) 2016; 95:e3194. [PMID: 27015216 PMCID: PMC4998411 DOI: 10.1097/md.0000000000003194] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/02/2016] [Accepted: 03/03/2016] [Indexed: 12/12/2022] Open
Abstract
The aims of the present study were to examine the relationship between serum B-cell activating factor belonging to the tumor necrosis factor family (BAFF) levels and serum interferon-γ-inducible protein-10 (IP-10) levels in patients with autoimmune hepatitis (AIH).A total of 80 corticosteroid therapy naive AIH patients were analyzed in this analysis. First, we examined the relationship between pretreatment serum BAFF and IP-10 levels and liver histological findings. Next, we investigated the relationship of pretreatment serum BAFF and IP-10 levels and aspartate aminotransferase value (AST), alanine aminotransferase value, and serum Immunoglobulin G (IgG) level as serum liver inflammation markers.Our study included 14 men and 66 women with the median (range) age of 64 (21-83) years. The serum BAFF levels ranged from 122.5 to 7696.0 pg/mL (median value, 1417.8 pg/mL), whereas the serum IP-10 levels ranged from 142.0 to 4198.7 pg/mL (median value, 640.1 pg/mL). The serum BAFF levels were significantly stratified in each 2 liver inflammation stage. Similarly, the serum IP-10 levels were significantly stratified in each 2 liver inflammation stage. Among 3 serum inflammation markers, AST value had the highest rs value in terms of the relationship with BAFF level (rs = 0.511, P < 0.001) and IP-10 level (rs = 0.626, P < 0.001). In addition, the serum BAFF level significantly correlated with serum IP-10 level (rs = 0.561, P < 0.001). In patients without advanced fibrosis (F3 or more), the serum BAFF level significantly correlated with serum IP-10 level (rs = 0.658, P < 0.001), whereas in patients with advanced fibrosis, the serum BAFF level significantly correlated with serum IP-10 level (rs = 0.542, P < 0.001).In conclusion, both BAFF and IP-10 are useful for predicting the degree of liver inflammation activity in AIH. BAFF and IP-10 may have the common clinical implication for liver inflammation activity for AIH patients.
Collapse
Affiliation(s)
- Hiroki Nishikawa
- From the Division of Hepatobiliary and Pancreatic disease, Department of Internal Medicine, Hyogo College of Medicine, Hyogo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
697
|
Zhong Z, Umemura A, Sanchez-Lopez E, Liang S, Shalapour S, Wong J, He F, Boassa D, Perkins G, Ali SR, McGeough MD, Ellisman MH, Seki E, Gustafsson AB, Hoffman HM, Diaz-Meco MT, Moscat J, Karin M. NF-κB Restricts Inflammasome Activation via Elimination of Damaged Mitochondria. Cell 2016; 164:896-910. [PMID: 26919428 PMCID: PMC4769378 DOI: 10.1016/j.cell.2015.12.057] [Citation(s) in RCA: 915] [Impact Index Per Article: 101.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 11/12/2015] [Accepted: 12/29/2015] [Indexed: 12/11/2022]
Abstract
Nuclear factor κB (NF-κB), a key activator of inflammation, primes the NLRP3-inflammasome for activation by inducing pro-IL-1β and NLRP3 expression. NF-κB, however, also prevents excessive inflammation and restrains NLRP3-inflammasome activation through a poorly defined mechanism. We now show that NF-κB exerts its anti-inflammatory activity by inducing delayed accumulation of the autophagy receptor p62/SQSTM1. External NLRP3-activating stimuli trigger a form of mitochondrial (mt) damage that is caspase-1- and NLRP3-independent and causes release of direct NLRP3-inflammasome activators, including mtDNA and mtROS. Damaged mitochondria undergo Parkin-dependent ubiquitin conjugation and are specifically recognized by p62, which induces their mitophagic clearance. Macrophage-specific p62 ablation causes pronounced accumulation of damaged mitochondria and excessive IL-1β-dependent inflammation, enhancing macrophage death. Therefore, the "NF-κB-p62-mitophagy" pathway is a macrophage-intrinsic regulatory loop through which NF-κB restrains its own inflammation-promoting activity and orchestrates a self-limiting host response that maintains homeostasis and favors tissue repair.
Collapse
Affiliation(s)
- Zhenyu Zhong
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Atsushi Umemura
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Elsa Sanchez-Lopez
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Shuang Liang
- Division of Gastroenterology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Shabnam Shalapour
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Jerry Wong
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Feng He
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Daniela Boassa
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Guy Perkins
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Syed Raza Ali
- Department of Pediatrics, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Matthew D McGeough
- Department of Pediatrics, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Ekihiro Seki
- Division of Gastroenterology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Asa B Gustafsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Hal M Hoffman
- Department of Pediatrics, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Maria T Diaz-Meco
- Sanford-Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jorge Moscat
- Sanford-Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
698
|
Ouyang Y, Guo J, Lin C, Lin J, Cao Y, Zhang Y, Wu Y, Chen S, Wang J, Chen L, Friedman SL. Transcriptomic analysis of the effects of Toll-like receptor 4 and its ligands on the gene expression network of hepatic stellate cells. FIBROGENESIS & TISSUE REPAIR 2016; 9:2. [PMID: 26900402 PMCID: PMC4759739 DOI: 10.1186/s13069-016-0039-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/05/2016] [Indexed: 01/28/2023]
Abstract
Background Intact Toll-like receptor 4 (TLR4) has been identified in hepatic stellate cells (HSCs), the primary fibrogenic cell type in liver. Here, we investigated the impact of TLR4 signaling on the gene expression network of HSCs by comparing the transcriptomic changes between wild-type (JS1) and TLR4 knockout (JS2) murine HSCs in response to two TLR4 ligands, lipopolysacchride (LPS), or high-mobility group box 1 (HMGB1). Results Whole mouse genome microarray was performed for gene expression analysis. Gene interaction and co-expression networks were built on the basis of ontology and pathway analysis by Kyoto Encyclopedia of Genes and Genomes (KEGG). Gene expression profiles are markedly different between Wild type (JS1) and TLR4 knockout (JS2) HSCs under basal conditions or following stimulation with LPS or HMGB1. The differentially expressed genes between TLR4 intact and null HSCs were enriched in signaling pathways including p53, mTOR, NOD-like receptor, Jak-STAT, chemokine, focal adhesion with some shared downstream kinases, and transcriptional factors. Venn analysis revealed that TLR4-dependent, LPS-responsive genes were clustered into pathways including Toll-like receptor and PI3K-Akt, whereas TLR4-dependent, HMGB1-responsive genes were clustered into pathways including metabolism and phagosome signaling. Genes differentially expressed that were categorized to be TLR4-dependent and both LPS- and HMGB1-responsive were enriched in cell cycle, ubiquitin mediated proteolysis, and mitogen-activated protein kinase (MAPK) signaling pathways. Conclusions TLR4 mediates complex gene expression alterations in HSCs. The affected pathways regulate a wide spectrum of HSC functions, including inflammation, fibrogenesis, and chemotaxis, as well as cell growth and metabolism. There are common and divergent regulatory signaling downstream of LPS and HMGB1 stimulation via TLR4 on HSCs. These findings emphasize the complex cascades downstream of TLR4 in HSCs that could influence their cellular biology and function. Electronic supplementary material The online version of this article (doi:10.1186/s13069-016-0039-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yangyang Ouyang
- Division of Digestive Diseases, Department of Internal Medicine, Zhong Shan Hospital, Shanghai Medical College, Fu Dan University, 180 Feng Lin Road, Shanghai, 200032 China
| | - Jinsheng Guo
- Division of Digestive Diseases, Department of Internal Medicine, Zhong Shan Hospital, Shanghai Medical College, Fu Dan University, 180 Feng Lin Road, Shanghai, 200032 China
| | - Chenzhao Lin
- Institutes of Biomedical Sciences, Fu Dan University, Shanghai, 200032 China
| | - Jie Lin
- Key Laboratory of Systems Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200032 China
| | - Yirong Cao
- Division of Digestive Diseases, Department of Internal Medicine, Zhong Shan Hospital, Shanghai Medical College, Fu Dan University, 180 Feng Lin Road, Shanghai, 200032 China
| | - Yuanqin Zhang
- Division of Digestive Diseases, Department of Internal Medicine, Zhong Shan Hospital, Shanghai Medical College, Fu Dan University, 180 Feng Lin Road, Shanghai, 200032 China
| | - Yujin Wu
- Division of Digestive Diseases, Department of Internal Medicine, Zhong Shan Hospital, Shanghai Medical College, Fu Dan University, 180 Feng Lin Road, Shanghai, 200032 China
| | - Shiyao Chen
- Division of Digestive Diseases, Department of Internal Medicine, Zhong Shan Hospital, Shanghai Medical College, Fu Dan University, 180 Feng Lin Road, Shanghai, 200032 China
| | - Jiyao Wang
- Division of Digestive Diseases, Department of Internal Medicine, Zhong Shan Hospital, Shanghai Medical College, Fu Dan University, 180 Feng Lin Road, Shanghai, 200032 China
| | - Luonan Chen
- Key Laboratory of Systems Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200032 China
| | - Scott L Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave., Room 11-70C, New York, 10029-6574 NY USA
| |
Collapse
|
699
|
Fujimoto Y, Urashima T, Shimura D, Ito R, Kawachi S, Kajimura I, Akaike T, Kusakari Y, Fujiwara M, Ogawa K, Goda N, Ida H, Minamisawa S. Low Cardiac Output Leads Hepatic Fibrosis in Right Heart Failure Model Rats. PLoS One 2016; 11:e0148666. [PMID: 26863419 PMCID: PMC4749189 DOI: 10.1371/journal.pone.0148666] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 01/21/2016] [Indexed: 12/11/2022] Open
Abstract
Background Hepatic fibrosis progresses with right heart failure, and becomes cardiac cirrhosis in a severe case. Although its causal factor still remains unclear. Here we evaluated the progression of hepatic fibrosis using a pulmonary artery banding (PAB)-induced right heart failure model and investigated whether cardiac output (CO) is responsible for the progression of hepatic fibrosis. Methods and Results Five-week-old Sprague-Dawley rats divided into the PAB and sham-operated control groups. After 4 weeks from operation, we measured CO by echocardiography, and hepatic fibrosis ratio by pathological examination using a color analyzer. In the PAB group, CO was significantly lower by 48% than that in the control group (78.2±27.6 and 150.1±31.2 ml/min, P<0.01). Hepatic fibrosis ratio and serum hyaluronic acid, an index of hepatic fibrosis, were significantly increased in the PAB group than those in the control group (7.8±1.7 and 1.0±0.2%, P<0.01, 76.2±27.5 and 32.7±7.5 ng/ml, P<0.01). Notably, the degree of hepatic fibrosis significantly correlated a decrease in CO. Immunohistological analysis revealed that hepatic stellate cells were markedly activated in hypoxic areas, and HIF-1α positive hepatic cells were increased in the PAB group. Furthermore, by real-time PCR analyses, transcripts of profibrotic and fibrotic factors (TGF-β1, CTGF, procollargen I, procollargen III, MMP 2, MMP 9, TIMP 1, TIMP 2) were significantly increased in the PAB group. In addition, western blot analyses revealed that the protein level of HIF-1α was significantly increased in the PAB group than that in the control group (2.31±0.84 and 1.0±0.18 arbitrary units, P<0.05). Conclusions Our study demonstrated that low CO and tissue hypoxia were responsible for hepatic fibrosis in right failure heart model rats.
Collapse
Affiliation(s)
- Yoshitaka Fujimoto
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Takashi Urashima
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
- * E-mail:
| | - Daisuke Shimura
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| | - Reiji Ito
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Sadataka Kawachi
- Division of Cardiology, Saitama Children’s Medical Center, Saitama, Japan
| | - Ichige Kajimura
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| | - Toru Akaike
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoichiro Kusakari
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| | - Masako Fujiwara
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Kiyoshi Ogawa
- Division of Cardiology, Saitama Children’s Medical Center, Saitama, Japan
| | - Nobuhito Goda
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan
| | - Hiroyuki Ida
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Susumu Minamisawa
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
700
|
Chen Q, Chen L, Wu X, Zhang F, Jin H, Lu C, Shao J, Kong D, Wu L, Zheng S. Dihydroartemisinin prevents liver fibrosis in bile duct ligated rats by inducing hepatic stellate cell apoptosis through modulating the PI3K/Akt pathway. IUBMB Life 2016; 68:220-31. [DOI: 10.1002/iub.1478] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 01/05/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Qin Chen
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Lianyun Chen
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Xiafei Wu
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Feng Zhang
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
- National First-Class Key Discipline for Traditional Chinese Medicine of Nanjing University of Chinese Medicine; Nanjing China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Material Medical; Nanjing University of Chinese Medicine; Nanjing China
| | - Huanhuan Jin
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Chunfeng Lu
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Jiangjuan Shao
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Desong Kong
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
- Department of Science; Technology and Education, the Third Affiliated Hospital of Nanjing University of Chinese Medicine; Nanjing China
| | - Li Wu
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Shizhong Zheng
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
- National First-Class Key Discipline for Traditional Chinese Medicine of Nanjing University of Chinese Medicine; Nanjing China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Material Medical; Nanjing University of Chinese Medicine; Nanjing China
| |
Collapse
|