1
|
Crawford JM, Bioulac-Sage P, Hytiroglou P. Structure, Function and Responses to Injury. MACSWEEN'S PATHOLOGY OF THE LIVER 2024:1-95. [DOI: 10.1016/b978-0-7020-8228-3.00001-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
2
|
Roy AM, Iyer R, Chakraborty S. The extracellular matrix in hepatocellular carcinoma: Mechanisms and therapeutic vulnerability. Cell Rep Med 2023; 4:101170. [PMID: 37652015 PMCID: PMC10518608 DOI: 10.1016/j.xcrm.2023.101170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/04/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023]
Abstract
The tumor microenvironment (TME) is influenced by a "disorganized" extracellular matrix (ECM) that sensitizes cancer cells toward mechanical stress, signaling, and structural alterations. In hepatocellular carcinoma (HCC), lack of knowledge about key ECM proteins driving the TME refractory to targeted therapies poses a barrier to the identification of new therapeutic targets. Herein, we discuss the contributions of various ECM components that impact hepatocytes and their surrounding support network during tumorigenesis. In addition, the underpinnings by which ECM proteins transduce mechanical signals to the liver TME are detailed. Finally, in view of the bidirectional feedback between the ECM, transformed hepatocytes, and immune cells, we highlight the potential role of the ECM disorganization process in shaping responses to immune checkpoint inhibitors and targeted therapies. Our comprehensive characterization of these ECM components may provide a roadmap for innovative therapeutic approaches to restrain HCC.
Collapse
Affiliation(s)
- Arya Mariam Roy
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Renuka Iyer
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Sayan Chakraborty
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Program of Developmental Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263.
| |
Collapse
|
3
|
McQuitty CE, Williams R, Chokshi S, Urbani L. Immunomodulatory Role of the Extracellular Matrix Within the Liver Disease Microenvironment. Front Immunol 2020; 11:574276. [PMID: 33262757 PMCID: PMC7686550 DOI: 10.3389/fimmu.2020.574276] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Chronic liver disease when accompanied by underlying fibrosis, is characterized by an accumulation of extracellular matrix (ECM) proteins and chronic inflammation. Although traditionally considered as a passive and largely architectural structure, the ECM is now being recognized as a source of potent damage-associated molecular pattern (DAMP)s with immune-active peptides and domains. In parallel, the ECM anchors a range of cytokines, chemokines and growth factors, all of which are capable of modulating immune responses. A growing body of evidence shows that ECM proteins themselves are capable of modulating immunity either directly via ligation with immune cell receptors including integrins and TLRs, or indirectly through release of immunoactive molecules such as cytokines which are stored within the ECM structure. Notably, ECM deposition and remodeling during injury and fibrosis can result in release or formation of ECM-DAMPs within the tissue, which can promote local inflammatory immune response and chemotactic immune cell recruitment and inflammation. It is well described that the ECM and immune response are interlinked and mutually participate in driving fibrosis, although their precise interactions in the context of chronic liver disease are poorly understood. This review aims to describe the known pro-/anti-inflammatory and fibrogenic properties of ECM proteins and DAMPs, with particular reference to the immunomodulatory properties of the ECM in the context of chronic liver disease. Finally, we discuss the importance of developing novel biotechnological platforms based on decellularized ECM-scaffolds, which provide opportunities to directly explore liver ECM-immune cell interactions in greater detail.
Collapse
Affiliation(s)
- Claire E. McQuitty
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Roger Williams
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Shilpa Chokshi
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Luca Urbani
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
4
|
Hepatoprotective effect of pyrroloquinoline quinone against alcoholic liver injury through activating Nrf2-mediated antioxidant and inhibiting TLR4-mediated inflammation responses. Process Biochem 2020. [DOI: 10.1016/j.procbio.2020.01.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
5
|
Crawford JM, Bioulac-Sage P, Hytiroglou P. Structure, Function, and Responses to Injury. MACSWEEN'S PATHOLOGY OF THE LIVER 2018:1-87. [DOI: 10.1016/b978-0-7020-6697-9.00001-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
6
|
PrabhuDas MR, Baldwin CL, Bollyky PL, Bowdish DME, Drickamer K, Febbraio M, Herz J, Kobzik L, Krieger M, Loike J, McVicker B, Means TK, Moestrup SK, Post SR, Sawamura T, Silverstein S, Speth RC, Telfer JC, Thiele GM, Wang XY, Wright SD, El Khoury J. A Consensus Definitive Classification of Scavenger Receptors and Their Roles in Health and Disease. THE JOURNAL OF IMMUNOLOGY 2017; 198:3775-3789. [PMID: 28483986 DOI: 10.4049/jimmunol.1700373] [Citation(s) in RCA: 236] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 03/13/2017] [Indexed: 02/07/2023]
Abstract
Scavenger receptors constitute a large family of proteins that are structurally diverse and participate in a wide range of biological functions. These receptors are expressed predominantly by myeloid cells and recognize a diverse variety of ligands including endogenous and modified host-derived molecules and microbial pathogens. There are currently eight classes of scavenger receptors, many of which have multiple names, leading to inconsistencies and confusion in the literature. To address this problem, a workshop was organized by the United States National Institute of Allergy and Infectious Diseases, National Institutes of Health, to help develop a clear definition of scavenger receptors and a standardized nomenclature based on that definition. Fifteen experts in the scavenger receptor field attended the workshop and, after extensive discussion, reached a consensus regarding the definition of scavenger receptors and a proposed scavenger receptor nomenclature. Scavenger receptors were defined as cell surface receptors that typically bind multiple ligands and promote the removal of nonself or altered-self targets. They often function by mechanisms that include endocytosis, phagocytosis, adhesion, and signaling that ultimately lead to the elimination of degraded or harmful substances. Based on this definition, nomenclature and classification of these receptors into 10 classes were proposed. This classification was discussed at three national meetings and input from participants at these meetings was requested. The following manuscript is a consensus statement that combines the recommendations of the initial workshop and incorporates the input received from the participants at the three national meetings.
Collapse
Affiliation(s)
- Mercy R PrabhuDas
- Division of Allergy, Immunology and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852;
| | - Cynthia L Baldwin
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003.,Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, MA 01003
| | - Paul L Bollyky
- Department of Medicine, Stanford University, Stanford, CA 94305
| | - Dawn M E Bowdish
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, M.G. DeGroote Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Kurt Drickamer
- Department of Life Sciences, Imperial College, London SW7 2AZ, United Kingdom
| | - Maria Febbraio
- Department of Dentistry, Katz Group Centre for Pharmacy and Health Research, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390.,Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390.,Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX 75390.,Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 75390.,Peter O'Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Lester Kobzik
- Department of Environmental Health, Harvard School of Public Health, Boston, MA 02115
| | - Monty Krieger
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - John Loike
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032
| | - Benita McVicker
- University of Nebraska Medical Center, Omaha VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105
| | - Terry K Means
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129
| | - Soren K Moestrup
- Department of Biomedicine, University of Aarhus, 8000 Aarhus C, Denmark
| | - Steven R Post
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - Tatsuya Sawamura
- Department of Physiology, Research Institute, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Samuel Silverstein
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032
| | - Robert C Speth
- Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, FL 33328
| | - Janice C Telfer
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003
| | - Geoffrey M Thiele
- Division of Rheumatology, Department of Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68105
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
| | - Samuel D Wright
- Cardiovascular Therapeutics, CSL Behring, King of Prussia, PA 19406; and
| | - Joseph El Khoury
- Infectious Disease Division, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
| |
Collapse
|
7
|
He L, Yuan FH, Chen T, Huang Q, Wang Y, Liu ZG. ER stress-mediated cell damage contributes to the release of EDA + fibronectin from hepatocytes in nonalcoholic fatty liver disease. ACTA ACUST UNITED AC 2017; 37:217-225. [PMID: 28397039 DOI: 10.1007/s11596-017-1718-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/23/2017] [Indexed: 12/12/2022]
Abstract
Fibronectin containing extra domain A (EDA+ FN), a functional glycoprotein participating in several cellular processes, correlates with chronic liver disease. Herein, we aim to investigate the expression and secretion of EDA+ FN from hepatocytes in nonalcoholic fatty liver disease (NAFLD) and the underlying mechanisms. Circulating levels of EDA+ FN were determined by ELISA in clinical samples. Western blotting and flow cytometry were performed on L02 and HepG2 cell lines to analyze whether the levels of EDA+ FN were associated with endoplasmic reticulum (ER) stress-related cell death. Circulating levels of EDA+ FN in NAFLD patients were significantly higher than those in control subjects, and positively related with severity of ultrasonographic steatosis score. In cultured hepatocytes, palmitate up-regulated the expression of EDA+ FN in a dose-dependent manner. Conversely, when the cells were pretreated with 4-phenylbutyrate, a specific inhibitor of ER stress, up-regulation of EDA+ FN could be abrogated. Moreover, silencing CHOP by shRNA enhanced the release of EDA+ FN from hepatocytes following palmitate treatment, which was involved in ER stress-related cell damage. These findings suggest that the up-regulated level of EDA+ FN is associated with liver damage in NAFLD, and ER stress-mediated cell damage contributes to the release of EDA+ FN from hepatocytes.
Collapse
Affiliation(s)
- Lei He
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Fa-Hu Yuan
- School of Medicine, Jianghan University, Wuhan, 430000, China
| | - Ting Chen
- School of Biology and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Qiang Huang
- School of Biology and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Yu Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhi-Guo Liu
- School of Biology and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan, 430023, China.
| |
Collapse
|
8
|
Massey VL, Dolin CE, Poole LG, Hudson SV, Siow DL, Brock GN, Merchant ML, Wilkey DW, Arteel GE. The hepatic "matrisome" responds dynamically to injury: Characterization of transitional changes to the extracellular matrix in mice. Hepatology 2017; 65:969-982. [PMID: 28035785 PMCID: PMC5319876 DOI: 10.1002/hep.28918] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 09/14/2016] [Accepted: 10/16/2016] [Indexed: 12/13/2022]
Abstract
UNLABELLED The extracellular matrix (ECM) consists of diverse components that work bidirectionally with surrounding cells to create a responsive microenvironment. In some contexts (e.g., hepatic fibrosis), changes to the ECM are well recognized and understood. However, it is becoming increasingly accepted that the hepatic ECM proteome (i.e., matrisome) responds dynamically to stress well before fibrosis. The term "transitional tissue remodeling" describes qualitative and quantitative ECM changes in response to injury that do not alter the overall architecture of the organ; these changes in ECM may contribute to early disease initiation and/or progression. The nature and magnitude of these changes to the ECM in liver injury are poorly understood. The goals of this work were to validate analysis of the ECM proteome and compare the impact of 6 weeks of ethanol diet and/or acute lipopolysaccharide (LPS). Liver sections were processed in a series of increasingly rigorous extraction buffers to separate proteins by solubility. Extracted proteins were identified using liquid chromatography/tandem mass spectrometry (LC-MS/MS). Both ethanol and LPS dramatically increased the number of matrisome proteins ∼25%. The enhancement of LPS-induced liver damage by ethanol preexposure was associated with unique protein changes. CONCLUSION An extraction method to enrich the hepatic ECM was characterized. The results demonstrate that the hepatic matrisome responds dynamically to both acute (LPS) and chronic (ethanol) stresses, long before more-dramatic fibrotic changes to the liver occur. The changes to the mastrisome may contribute, at least in part, to the pathological responses to these stresses. It is also interesting that several ECM proteins responded similarly to both stresses, suggesting a common mechanism in both models. Nevertheless, there were responses that were unique to the individual and combined exposures. (Hepatology 2017;65:969-982).
Collapse
Affiliation(s)
- Veronica L. Massey
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY,University of Louisville Alcohol Research Center, Louisville, KY
| | - Christine E. Dolin
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY,University of Louisville Alcohol Research Center, Louisville, KY
| | - Lauren G. Poole
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY,University of Louisville Alcohol Research Center, Louisville, KY
| | - Shanice V. Hudson
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY,University of Louisville Alcohol Research Center, Louisville, KY
| | - Deanna L. Siow
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY,University of Louisville Alcohol Research Center, Louisville, KY
| | - Guy N. Brock
- Department of Bioinformatics and Biostatistics, University of Louisville Health Sciences Center, Louisville, KY
| | - Michael L. Merchant
- University of Louisville Alcohol Research Center, Louisville, KY,Department of Medicine, University of Louisville Health Sciences Center, Louisville, KY
| | - Daniel W. Wilkey
- University of Louisville Alcohol Research Center, Louisville, KY,Department of Medicine, University of Louisville Health Sciences Center, Louisville, KY
| | - Gavin E. Arteel
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY,University of Louisville Alcohol Research Center, Louisville, KY
| |
Collapse
|
9
|
Transitional Remodeling of the Hepatic Extracellular Matrix in Alcohol-Induced Liver Injury. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3162670. [PMID: 27843941 PMCID: PMC5098054 DOI: 10.1155/2016/3162670] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/27/2016] [Indexed: 12/19/2022]
Abstract
Alcohol consumption is a common custom worldwide, and the toxic effects of alcohol on several target organs are well understood. The liver is the primary site of alcohol metabolism and is therefore the major target of alcohol toxicity. Alcoholic liver disease is a spectrum of disease states, ranging from simple steatosis (fat accumulation), to inflammation, and eventually to fibrosis and cirrhosis if untreated. The fibrotic stage of ALD is primarily characterized by robust accumulation of extracellular matrix (ECM) proteins (collagens) which ultimately impairs the function of the organ. The role of the ECM in early stages of ALD is poorly understood, but recent research has demonstrated that a number of changes in the hepatic ECM in prefibrotic ALD not only are present, but may also contribute to disease progression. The purpose of this review is to summarize the established and proposed changes to the hepatic extracellular matrix (ECM) that may contribute to earlier stages of ALD development and to discuss potential mechanisms by which these changes may mediate the progression of the disease.
Collapse
|
10
|
Li S, Wang CX, Liu NZ, Liu P. Anti-inflammatory effects of propofol on lipopolysaccharides-treated rat hepatic Kupffer cells. Cell Biochem Biophys 2015; 71:845-50. [PMID: 25296958 DOI: 10.1007/s12013-014-0272-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This study is set to explore the role of commonly used intravenous anesthetic propofol on the inflammatory response of rat liver Kupffer cells (KCs) induced by lipopolysaccharides (LPS). The isolated KCs were cultured at the density of 1 × 10(5)/ml, divided into five groups randomly after 48 h culture: group C, control group; group L, KCs were treated with 1 μg/ml LPS for 24 h; groups P1, P2, P3, KCs were pretreated with propofol at low (25 μM), medium (50 μM), high (100 μM) concentration for 2 h, respectively, and then were stimulated with 1 μg/ml LPS for 24 h. The expressions of tumor necrosis factor-α (TNF-α) mRNA and interleukin-1β (IL-1β) mRNA of every group were measured by RT-PCR. Nuclear NF-ΚB p65 was determined by Western blot. The concentrations of IL-1β and TNF-α in supernatant were measured by ELISA. Compared with the group C, TNF-α mRNA and IL-1β mRNA in group L were significantly up-regulated and NF-ΚB p65 was significantly up-regulated after LPS treatment (P < 0.05). Meanwhile, TNF-α and IL-1β were also significantly increased (P < 0.05). With propofol the mRNA expressions of aforementioned inflammatory mediators were significantly down-regulated and NF-ΚB p65 was significantly inhibited in group P2 and P3 (P < 0.05), compared with group L. However, low propofol concentration did not exhibit any effect (group P1, P > 0.05). Propofol at medium and high concentration can counteract the LPS-induced inflammatory response in KCs by regulating NF-ΚB p65 protein expression.
Collapse
Affiliation(s)
- Sen Li
- Department of Emergency, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, China
| | | | | | | |
Collapse
|
11
|
The tetrapartite synapse: Extracellular matrix remodeling contributes to corticoaccumbens plasticity underlying drug addiction. Brain Res 2015; 1628:29-39. [PMID: 25838241 DOI: 10.1016/j.brainres.2015.03.027] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 02/10/2015] [Accepted: 03/13/2015] [Indexed: 12/29/2022]
Abstract
Synaptic plasticity has long been known to involve three key elements of neuropil, the presynapse, the postsynapse and adjacent glia. Here we review the role of the extracellular matrix in synaptic plasticity as a necessary component forming the tetrapartite synapse. We describe the role of matrix metalloproteinases as enzymes sculpting extracellular proteins and thereby creating an extracellular signaling domain required for synaptic plasticity. Specifically we focus on the role of the tetrapartite synapse in mediating the effects of addictive drugs at cortico-striatal synapses, and conclude that the extracellular signaling domain and its regulation by matrix metalloproteinases is critical for developing and expressing drug seeking behaviors.
Collapse
|
12
|
Abstract
Kupffer cells are a critical component of the mononuclear phagocytic system and are central to both the hepatic and systemic response to pathogens. Kupffer cells are reemerging as critical mediators of both liver injury and repair. Kupffer cells exhibit a tremendous plasticity; depending on the local metabolic and immune environment, then can express a range of polarized phenotypes, from the proinflammatory M1 phenotype to the alternative/M2 phenotype. Multiple M2 phenotypes can be distinguished, each involved in the resolution of inflammation and wound healing. Here, we have provided an update on recent research that has contributed to the developing delineation of the contribution of Kupffer cells to different types of liver injury, with an emphasis on alcoholic and nonalcoholic liver diseases. These recent advances in our understanding of Kupffer cell function and regulation will likely provide new insights into the potential for therapeutic manipulation of Kupffer cells to promote the resolution of inflammation and enhance wound healing in liver disease.
Collapse
Affiliation(s)
- Laura J Dixon
- Liver Disease Research Center, Case Western Reserve University, Cleveland, Ohio, USA
| | | | | | | | | |
Collapse
|
13
|
Wang Y, Hu Y, Chao C, Yuksel M, Colle I, Flavell RA, Ma Y, Yan H, Wen L. Role of IRAK-M in alcohol induced liver injury. PLoS One 2013; 8:e57085. [PMID: 23437317 PMCID: PMC3578822 DOI: 10.1371/journal.pone.0057085] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 01/17/2013] [Indexed: 12/20/2022] Open
Abstract
Increasing evidence suggests that innate immunity plays an important role in alcohol-induced liver injury and most studies have focused on positive regulation of innate immunity. The main objective of this study was to investigate the negative regulator of innate immunity, IL-1/Toll-like receptor (TLR) signaling pathways and interleukin receptor-associated kinase-M (IRAK-M) in alcoholic liver injury. We established an alcohol-induced liver injury model using wild type and IRAK-M deficient B6 mice and investigated the possible mechanisms. We found that in the absence of IRAK-M, liver damage by alcohol was worse with higher alanine transaminase (ALT), more immune cell infiltration and increased numbers of IFNγ producing cells. We also found enhanced phagocytic activity in CD68+ cells. Moreover, our results revealed altered gut bacteria after alcohol consumption and this was more striking in the absence of IRAK-M. Our study provides evidence that IRAK-M plays an important role in alcohol-induced liver injury and IRAK-M negatively regulates the innate and possibly the adaptive immune response in the liver reacting to acute insult by alcohol. In the absence of IRAK-M, the hosts developed worse liver injury, enhanced gut permeability and altered gut microbiota.
Collapse
Affiliation(s)
- Yipeng Wang
- The Sections of Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Lassaletta AD, Chu LM, Sellke FW. Effects of alcohol on pericardial adhesion formation in hypercholesterolemic swine. J Thorac Cardiovasc Surg 2012; 143:953-9. [PMID: 22244558 DOI: 10.1016/j.jtcvs.2011.12.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 10/24/2011] [Accepted: 12/13/2011] [Indexed: 10/14/2022]
Abstract
OBJECTIVE Reoperative cardiac surgery is complicated in part because of extensive adhesions encountered during the second operation. The purpose of this study was to examine the effects of alcohol with and without resveratrol (red wine vs vodka) on postoperative pericardial adhesion formation in a porcine model of hypercholesterolemia and chronic myocardial ischemia. METHODS Male Yorkshire swine were fed a high-cholesterol diet to simulate conditions of coronary artery disease followed by surgical placement of an ameroid constrictor to induce chronic ischemia. Postoperatively, control pigs continued their high-cholesterol diet alone, whereas the 2 experimental groups had diets supplemented with red wine or vodka. Seven weeks after ameroid placement, all animals underwent reoperative sternotomy. RESULTS Compared with controls, pericardial adhesion grade was markedly reduced in the vodka group, whereas there was no difference in the wine group. Intramyocardial fibrosis was significantly reduced in the vodka group compared with controls. There was no difference in expression of proteins involved in focal adhesion formation between any groups (focal adhesion kinase, integrin alpha-5, integrin beta-1, paxillin, vinculin, protein tyrosine kinase 2, protein kinase C ε, and phosphorylated protein kinase C ε). The wine group exhibited elevated C-reactive protein levels versus the control and vodka groups. CONCLUSIONS Postoperative vodka consumption markedly reduced the formation of pericardial adhesions and intramyocardial fibrosis, whereas red wine had no effect. Analysis of protein expression did not reveal any obvious explanation for this phenomenon, suggesting a post-translational effect of alcohol on fibrous tissue deposition. The difference in adhesion formation in the vodka versus wine groups may be due to increased inflammation in the wine group.
Collapse
Affiliation(s)
- Antonio D Lassaletta
- Department of Surgery, Division of Cardiothoracic Surgery, Warren Alpert School of Medicine, Brown University, Providence, RI, USA
| | | | | |
Collapse
|
15
|
Crawford JM, Burt AD. Anatomy, pathophysiology and basic mechanisms of disease. MACSWEEN'S PATHOLOGY OF THE LIVER 2012:1-77. [DOI: 10.1016/b978-0-7020-3398-8.00001-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
16
|
Bio-informatics based analysis of genes implicated in alcohol mediated liver injury. Gene 2011; 494:130-9. [PMID: 22155707 DOI: 10.1016/j.gene.2011.11.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 10/21/2011] [Accepted: 11/15/2011] [Indexed: 12/18/2022]
Abstract
Alcohol induced liver injury has been studied extensively. Using literature search and bioinformatics tools, the present study characterizes the genes involved in alcohol induced liver injury. The cellular and metabolic processes in which genes involved in alcohol induced liver injury are implicated are also discussed. The genes related to alcohol induced liver injury are also involved in affecting certain molecular functions and metabolism of drugs, besides being associated with diseases. In conclusion, the changes in regulation of genes implicated in alcohol induced liver injury apart from causing alcohol mediated hepatic dysfunction may affect other vital processes in the body.
Collapse
|
17
|
Aziz-Seible RS, Casey CA. Fibronectin: Functional character and role in alcoholic liver disease. World J Gastroenterol 2011; 17:2482-99. [PMID: 21633653 PMCID: PMC3103806 DOI: 10.3748/wjg.v17.i20.2482] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 04/07/2011] [Accepted: 04/14/2011] [Indexed: 02/06/2023] Open
Abstract
Fibronectins are adhesive glycoproteins that can be found in tissue matrices and circulating in various fluids of the body. The variable composition of fibronectin molecules facilitates a diversity of interactions with cell surface receptors that suggest a role for these proteins beyond the structural considerations of the extracellular matrix. These interactions implicate fibronectin in the regulation of mechanisms that also determine cell behavior and activity. The two major forms, plasma fibronectin (pFn) and cellular fibronectin (cFn), exist as balanced amounts under normal physiological conditions. However, during injury and/or disease, tissue and circulating levels of cFn become disproportionately elevated. The accumulating cFn, in addition to being a consequence of prolonged tissue damage, may in fact stimulate cellular events that promote further damage. In this review, we summarize what is known regarding such interactions between fibronectin and cells that may influence the biological response to injury. We elaborate on the effects of cFn in the liver, specifically under a condition of chronic alcohol-induced injury. Studies have revealed that chronic alcohol consumption stimulates excess production of cFn by sinusoidal endothelial cells and hepatic stellate cells while impairing its clearance by other cell types resulting in the build up of this glycoprotein throughout the liver and its consequent increased availability to influence cellular activity that could promote the development of alcoholic liver disease. We describe recent findings by our laboratory that support a plausible role for cFn in the promotion of liver injury under a condition of chronic alcohol abuse and the implications of cFn stimulation on the pathogenesis of alcoholic liver disease. These findings suggest an effect of cFn in regulating cell behavior in the alcohol-injured liver that is worth further characterizing not only to gain a more comprehensive understanding of the role this reactive glycoprotein plays in the progression of injury but also for the insight further studies could provide towards the development of novel therapies for alcoholic liver disease.
Collapse
|