21
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The Potential Role of Elk-3/Egr-1 Signaling Pathway in the Epithelial-Mesenchymal Transition during Liver Fibrosis

      editorial
      Gut and Liver
      Editorial Office of Gut and Liver

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Liver fibrosis is characterized by a dysregulated wound healing process that results in excessive deposition of the extracellular matrix (ECM), mainly collagen type I, and scar formation. Hepatic myofibroblasts are part of the heterogenous cell group, which has a fundamental role for the development of liver fibrosis.1 However, the origin of myofibroblasts in the fibrotic liver has not been fully elucidated and is still under heated debate, despite extensive research.2 Although activated hepatic stellate cells have been considered as the major source, since they are the main ECM-producing cells in the injured liver, previous studies suggested that the epithelial-mesenchymal transition (EMT) is one of the mechanisms that give rise to hepatic myofibroblasts in liver fibrosis.3 During EMT, epithelial cells lose key epithelial features, such as apical-basal polarity, intercellular adhesion complexes, and adherence to a basal basement membrane, while gradually obtaining multiple mesenchymal phenotypes, including spindle-shaped appearance, increased cell motility, invasiveness, and increased production of ECM components.3 EMT is divided into three distinct categories: type 1 occurring in development, type 2 in fibrosis, and type 3 in cancer and metastasis.3 Type 2 EMT has been associated with organ fibrosis and regeneration, occurring in the liver, lung, kidney and intestine. The cells that undergo EMT show a loss of epithelial adhesion protein, E-cadherin, counterbalanced by the aberrant expression of N-cadherin. Fibroblast-specific protein 1 (FSP1; also known as S100A4), α-smooth muscle actin (α-SMA), and collagen 1 are characterized markers of the mesenchymal products generated by the EMT during organ fibrosis development.3 Other biomarkers, including vimentin, desmin, fibronectin, and discoidin domain receptor 2 have been used to demonstrate the epithelial cells undergoing EMT.4 Through the EMT process, epithelial cells eventually lose their epithelial markers (E-cadherin and zonula occludens-1), acquiring a fully fibroblastic mesenchymal phenotype (N-cadherin, vimentin, FSP1, collagen-1, and α-SMA). Transforming growth factor β (TGF-β) is believed to be a potent inducer of EMT and a well-established profibrogenic cytokine in liver fibrosis.5 Hepatocyte EMT was observed in the primary hepatocytes incubated with TGF-β1 and primary hepatocytes from CCl4-induced cirrhotic liver.6,7 In the TGF β signaling pathway, active TGF-β1 ligands initiate the signaling process by binding to TGF-β receptor type I (TβRI) and TβRII serine/threonine kinases.3 TβRI phosphorylates Smad2 and Smad3, which form a complex with Smad4 and translocate to the nucleus. Smad proteins convey signals from TGF-β to the nucleus. Once in the nucleus, the complex of Smads can regulate the transcription of target genes. The activation of several Smad independent pathways have been identified to be a crucial component for EMT induction by TGF-β, including phosphoinositide 3-kinase (PI3K)-Akt, focal adhesion kinase, p38 the mitogen activated protein kinases (MAPK), and ERK. In addition, recent studies have implicated Krüppel-like factor-8, hyaluronan synthase 2, and microRNA miR-203 as critical regulators for EMT.3 Li et al.8 investigated novel molecular mechanisms involved in the EMT of hepatocytes and liver fibrosis. They demonstrated that the expression levels of Elk-3 and early growth response-1 (Egr-1) were significantly increased during the TGF-β1-induced EMT of hepatocytes, in both the CCl4-induced mouse liver fibrotic tissues and in the human liver cirrhotic tissues. E26 transformation-specific (ETS) proteins form one of the largest families of signal-dependent transcriptional regulators, which mediate cellular proliferation, differentiation, and tumorigenesis.9 Elk-3 (Net/Sap-2/Erp) is a member of the ternary complex factors subfamily of ETS proteins, along with Elk-1 and Sap-1. Elk-3 plays an important role in wound healing, angiogenesis, cell migration, and tumorigenesis. Elk-3 is activated by the expression of Ras and phosphorylated by ERK and p38. Egr-1, a zinc finger-containing transcription factor, is immediately expressed in response to a variety of stimuli, such as growth factors and lipopolysaccharide. It has been reported that Egr-1 can regulate genes involved in the wound-healing process and immune response. However, to the best of our knowledge, there have not been any studies investigating the role of Elk-3 and Egr-1 during EMT of hepatocyte in liver fibrosis. Li et al.8 demonstrated that the expression levels of Elk-3 and Egr-1 were significantly increased during TGF-β1-induced EMT of hepatocytes, in both the CCl4-induced mouse liver fibrotic tissues and in the human liver cirrhotic tissues. Furthermore, they investigated the molecular relationships among Elk-3, Egr-1, and the p38 MAPK pathway during EMT in hepatocytes. According to the results, the silencing of Elk-3 and inhibition of the Ras-Elk-3 pathway with an inhibitor suppressed the expression of EMT-related markers. Moreover, Elk-3 expression was regulated by p38 MAPK phosphorylation during EMT. Therefore, the study by Li et al.8 suggested Elk-3/Egr-1 MAPK signaling as a novel molecular pathway in the progression of liver fibrosis via the regulation of hepatocyte EMT. However, since recent studies using “lineage tracing” method reported evidence against EMT in the liver, the EMT theory is one of the most controversial issues in this field of research.10 Taura et al.11 bred triple transgenic mice expressing ROSA26 stop β-galactosidase (β-gal), Albumin Cre, and collagen α (I) green fluorescent protein (GFP), in which the hepatocyte-derived cells are permanently labeled by β-gal and type I collagen-expressing cells are labeled by GFP. The study examined the expression of four different mesenchymal markers (FSP 1, α-SMA, vimentin, and desmin) in the primary hepatocyte from the untreated and CCl4-treated livers and fibrotic liver tissues induced by CCl4 injections. According to the results, type I collagen-producing cells do not originate from the hepatocytes. Hepatocytes in vivo neither acquire the mesenchymal marker expression nor exhibit a morphological change that clearly distinguishes from the normal hepatocytes. Thus, these findings strongly challenge the concept that hepatocytes in vivo acquire a mesenchymal phenotype through EMT to produce ECM in liver fibrosis. However, these contradictory studies against EMT in liver fibrosis also has some methodological concerns, including incomplete efficiency of Cre-mediated recombination, use of only a few mesenchymal markers on immunostaining, and discordance of experimental models from human chronic liver disease. These criticisms cannot completely rule out the possible role of EMT in liver fibrosis. Indeed, many studies have still been investigating the possibility of EMT and related molecular mechanisms in hepatic fibrogenesis. Therefore, regardless of the discourse on EMT, it is necessary for future research on EMT to provide noble insights into the plasticity of cellular phenotypes, molecular signaling pathways, and any possible therapeutic targets in liver fibrosis.

          Related collections

          Most cited references10

          • Record: found
          • Abstract: found
          • Article: not found

          Transforming growth factor-beta1 induces an epithelial-to-mesenchymal transition state in mouse hepatocytes in vitro.

          Liver fibrosis is a progressive pathologic process that involves deposition of excess extracellular matrix leading to distorted architecture and culminating in cirrhosis. The role of transforming growth factor-beta (TGF-beta) as a key molecule in the development and progression of hepatic fibrosis via the activation of hepatic stellate cells, among other fibroblast populations, is without controversy. We hereby show that TGF-beta1 induces an epithelial-to-mesenchymal transition (EMT) state in mature hepatocytes in vitro. EMT state was marked by significant upregulation of alpha(1)(I) collagen mRNA expression and type I collagen deposition. Similar changes were found in a "normal" mouse hepatocyte cell line (AML12), thus confirming that hepatocytes are capable of EMT changes and type I collagen synthesis. We also show that in hepatocytes in the EMT state, TGF-beta1 induces the snail-1 transcription factor and activates the Smad2/3 pathway. Evidence for a central role of the TGF-beta1/Smad pathway is further supported by the inhibition of EMT by Smad4 silencing using small interference RNA technology. In conclusion, TGF-beta1, a known pro-apoptotic cytokine in mature hepatocytes, is capable of mediating phenotypic changes and plasticity in the form of EMT, resulting in collagen deposition. Our findings support a potentially crucial role for EMT in the development and progression of hepatic fibrogenesis.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Hepatocytes do not undergo epithelial-mesenchymal transition in liver fibrosis in mice.

            The origin of fibrogenic cells in liver fibrosis remains controversial. We assessed the emerging concept that hepatocytes contribute to production of extracellular matrix (ECM) in liver fibrosis through epithelial-mesenchymal transition (EMT). We bred triple transgenic mice expressing ROSA26 stop beta-galactosidase (beta-gal), albumin Cre, and collagen alpha1(I) green fluorescent protein (GFP), in which hepatocyte-derived cells are permanently labeled by beta-gal and type I collagen-expressing cells are labeled by GFP. We induced liver fibrosis by repetitive carbon tetrachloride (CCl(4)) injections. Liver sections and isolated cells were evaluated for GFP and beta-gal as well as expression of alpha-smooth muscle actin (alpha-SMA) and fibroblast-specific protein 1 (FSP-1). Upon stimulation with transforming growth factor beta-1, cultured hepatocytes isolated from untreated liver expressed both GFP and beta-gal with a fibroblast-like morphological change but lacked expression of other mesenchymal markers. Cells from CCl(4)-treated livers never showed double-positivity for GFP and beta-gal. All beta-gal-positive cells exhibited abundant cytoplasm, a typical morphology of hepatocytes, and expressed none of the mesenchymal markers including alpha-SMA, FSP-1, desmin, and vimentin. In liver sections of CCl(4)-treated mice, GFP-positive areas were coincident with fibrotic septa and never overlapped X-gal-positive areas. Type I collagen-producing cells do not originate from hepatocytes. Hepatocytes in vivo neither acquire mesenchymal marker expression nor exhibit a morphological change clearly distinguishable from normal hepatocytes. Our results strongly challenge the concept that hepatocytes in vivo acquire a mesenchymal phenotype through EMT to produce the ECM in liver fibrosis.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Epithelial-mesenchymal transition in liver fibrosis.

              Liver fibrosis is the result of a sustained wound healing response to sustained chronic liver injury, which includes viral, alcoholic and autoimmune hepatitis. Hepatic regeneration is the dominant outcome of liver damage. The outcomes of successful repair are the replacement of dead epithelial cells with healthy epithelial cells, and reconstruction of the normal hepatic structure and function. Prevention of the development of epithelial-mesenchymal transition (EMT) may control and even reverse liver fibrosis. EMT is a critical process for an epithelial cell to undergo a conversion to a mesenchymal phenotype, and is believed to be an inflammation-induced response, which may have a central role in liver fibrosis. The origin of fibrogenic cells in liver fibrosis remains controversial. Numerous studies have investigated the origin of all fibrogenic cells within the liver and the mechanism of the signaling pathways that lead to the activation of EMT programs during numerous chronic liver diseases. The present study aimed to summarize the evidence to explain the possible role of EMT in liver fibrosis.
                Bookmark

                Author and article information

                Journal
                Gut Liver
                Gut Liver
                Gut and Liver
                Editorial Office of Gut and Liver
                1976-2283
                2005-1212
                January 2017
                15 January 2017
                : 11
                : 1
                : 11-12
                Affiliations
                Department of Internal Medicine, Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea
                Author notes
                Correspondence to: In Hee Kim, Department of Internal Medicine, Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University Medical School, 20 Geonji-ro, Deokjin-gu, Jeonju 54907, Korea, Tel: +82-63-250-1677, Fax: +82-63-254-1609, E-mail: ihkimmd@ 123456jbnu.ac.kr
                Article
                gnl-11-011
                10.5009/gnl16564
                5221856
                28053299
                e9a2ead8-e5db-4f3f-a464-baeee5763d82
                Copyright © 2017 by The Korean Society of Gastroenterology, the Korean Society of Gastrointestinal Endoscopy, the Korean Society of Neurogastroenterology and Motility, Korean College of Helicobacter and Upper Gastrointestinal Research, Korean Association the Study of Intestinal Diseases, the Korean Association for the Study of the Liver, Korean Pancreatobiliary Association, and Korean Society of Gastrointestinal Cancer.

                This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License ( http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

                History
                Categories
                Editorial

                Gastroenterology & Hepatology
                Gastroenterology & Hepatology

                Comments

                Comment on this article