8
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Calcitriol attenuates liver fibrosis through hepatitis C virus nonstructural protein 3-transactivated protein 1-mediated TGF β1/Smad3 and NF-κB signaling pathways

      research-article

      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

          BACKGROUND

          Hepatic fibrosis is a serious condition, and the development of hepatic fibrosis can lead to a series of complications. However, the pathogenesis of hepatic fibrosis remains unclear, and effective therapy options are still lacking. Our group identified hepatitis C virus nonstructural protein 3-transactivated protein 1 (NS3TP1) by suppressive subtractive hybridization and bioinformatics analysis, but its role in diseases including hepatic fibrosis remains undefined. Therefore, additional studies on the function of NS3TP1 in hepatic fibrosis are urgently needed to provide new targets for treatment.

          AIM

          To elucidate the mechanism of NS3TP1 in hepatic fibrosis and the regulatory effects of calcitriol on NS3TP1.

          METHODS

          Twenty-four male C57BL/6 mice were randomized and separated into three groups, comprising the normal, fibrosis, and calcitriol treatment groups, and liver fibrosis was modeled by carbon tetrachloride (CCl 4). To evaluate the level of hepatic fibrosis in every group, serological and pathological examinations of the liver were conducted. TGF-β1 was administered to boost the in vitro cultivation of LX-2 cells. NS3TP1, α-smooth muscle actin (α-SMA), collagen I, and collagen III in every group were examined using a Western blot and real-time quantitative polymerase chain reaction. The activity of the transforming growth factor beta 1 (TGFβ1)/Smad3 and NF-κB signaling pathways in each group of cells transfected with pcDNA-NS3TP1 or siRNA-NS3TP1 was detected. The statistical analysis of the data was performed using the Student’s t test.

          RESULTS

          NS3TP1 promoted the activation, proliferation, and differentiation of hepatic stellate cells (HSCs) and enhanced hepatic fibrosis via the TGFβ1/Smad3 and NF-κB signaling pathways, as evidenced by the presence of α-SMA, collagen I, collagen III, p-smad3, and p-p65 in LX-2 cells, which were upregulated after NS3TP1 overexpression and downregulated after NS3TP1 interference. The proliferation of HSCs was lowered after NS3TP1 interference and elevated after NS3TP1 overexpression, as shown by the luciferase assay. NS3TP1 inhibited the apoptosis of HSCs. Moreover, both Smad3 and p65 could bind to NS3TP1, and p65 increased the promoter activity of NS3TP1, while NS3TP1 increased the promoter activity of TGFβ1 receptor I, as indicated by coimmunoprecipitation and luciferase assay results. Both in vivo and in vitro, treatment with calcitriol dramatically reduced the expression of NS3TP1. Calcitriol therapy-controlled HSCs activation, proliferation, and differentiation and substantially suppressed CCl 4-induced hepatic fibrosis in mice. Furthermore, calcitriol modulated the activities of the above signaling pathways via downregulation of NS3TP1.

          CONCLUSION

          Our results suggest that calcitriol may be employed as an adjuvant therapy for hepatic fibrosis and that NS3TP1 is a unique, prospective therapeutic target in hepatic fibrosis.

          Related collections

          Most cited references37

          • Record: found
          • Abstract: found
          • Article: found

          Liver fibrosis: Pathophysiology, pathogenetic targets and clinical issues

          The progression of chronic liver diseases (CLD), irrespective of etiology, involves chronic parenchymal injury, persistent activation of inflammatory response as well as sustained activation of liver fibrogenesis and wound healing response. Liver fibrogenesis, is a dynamic, highly integrated molecular, cellular and tissue process responsible for driving the excess accumulation of extracellular matrix (ECM) components (i.e., liver fibrosis) sustained by an eterogeneous population of hepatic myofibroblasts (MFs). The process of liver fibrogenesis recognizes a number of common and etiology-independent mechanisms and events but it is also significantly influenced by the specific etiology, as also reflected by peculiar morphological patterns of liver fibrosis development. In this review we will analyze the most relevant established and/or emerging pathophysiological issues underlying CLD progression with a focus on the role of critical hepatic cell populations, mechanisms and signaling pathways involved, as they represent potential therapeutic targets, to finally analyze selected and relevant clinical issues.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Targeting TGF-β signal transduction for fibrosis and cancer therapy

            Transforming growth factor β (TGF-β) has long been identified with its intensive involvement in early embryonic development and organogenesis, immune supervision, tissue repair, and adult homeostasis. The role of TGF-β in fibrosis and cancer is complex and sometimes even contradictory, exhibiting either inhibitory or promoting effects depending on the stage of the disease. Under pathological conditions, overexpressed TGF-β causes epithelial-mesenchymal transition (EMT), extracellular matrix (ECM) deposition, cancer-associated fibroblast (CAF) formation, which leads to fibrotic disease, and cancer. Given the critical role of TGF-β and its downstream molecules in the progression of fibrosis and cancers, therapeutics targeting TGF-β signaling appears to be a promising strategy. However, due to potential systemic cytotoxicity, the development of TGF-β therapeutics has lagged. In this review, we summarized the biological process of TGF-β, with its dual role in fibrosis and tumorigenesis, and the clinical application of TGF-β-targeting therapies.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Animal Models for Fibrotic Liver Diseases: What We Have, What We Need, and What Is under Development

              Liver fibrosis is part of the wound-healing response to liver damage of various origins and represents a major health problem. Although our understanding of the pathogenesis of liver fibrosis has grown considerably over the last 20 years, effective antifibrotic therapies are still lacking. The use of animal models is crucial for determining mechanisms underlying initiation, progression, and resolution of fibrosis and for developing novel therapies. To date, no animal model can recapitulate all the hepatic and extra-hepatic features of liver disease. In this review, we will discuss the current rodent models of liver injuries. We will then focus on the available ways to target specifically particular compounds of fibrogenesis and on the new models of liver diseases like the humanized liver mouse model.
                Bookmark

                Author and article information

                Contributors
                Journal
                World J Gastroenterol
                World J Gastroenterol
                WJG
                World Journal of Gastroenterology
                Baishideng Publishing Group Inc
                1007-9327
                2219-2840
                14 May 2023
                14 May 2023
                : 29
                : 18
                : 2798-2817
                Affiliations
                Department of Infectious Disease Medicine, The First Hospital Affiliated to Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
                China-Japan Friendship Hospital, Department of Infectious Disease China-Japan Friendship Hospital, Beijing 100029, China
                Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
                The Division of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
                Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
                Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
                Institute of Liver Diseases, Beijing Pan-Asia Tongze Institute of Biomedicine Co., Ltd, Beijing 100015, China
                The Division of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
                Department of Rehabilitation Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
                Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
                Beijing Key Laboratory of Emerging Infectious Diseases, Peking University Ditan Teaching Hospital, Beijing 100015, China
                Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
                Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
                Department of Infectious Disease Medicine, The First Hospital Affiliated to Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
                Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
                Department of Infectious Disease Medicine, The First Hospital Affiliated to Xi’an Jiaotong University, Xi'an 710061, Shaanxi Province, China. lsmxjtu@ 123456126.com
                Author notes

                Author contributions: Shi L, Zhou L, Han M, Zhang Y, Zhang Y, Yuan XX, Lu HP, Wang Y, Yang XL, Liu C, Wang J, Liang P, Liu SA, Liu XJ, Cheng J, and Lin SM contributed to the study conception and design; Liu S, Li Z, Ming H, Yu Z, Yang Z, Xue YX, Ping LH, Yun W, Liang YX, Chen L, Jun W, and Jing LX participated in the design and completion of the experiment; Liu S wrote the original draft; Liu S, Jun C, and Mei LS reviewed and edited the manuscript; Pu L and Ai LS finished the project administration; Jun C and Mei LS have the same contribution to the article; all authors approved the final version of the article.

                Supported by the National Key Research and Development Program of China, No. 2017YFC0908104; and National Science and Technology Projects, No. 2017ZX10203201, No. 2017ZX10201201, and No. 2017ZX10202202.

                Corresponding author: Shu-Mei Lin, Doctor, MD, PhD, Doctor, Professor, Teacher, Department of Infectious Disease Medicine, The First Hospital Affiliated to Xi’an Jiaotong University, No. 277 Yanta West Road, Yanta, Xi’an 710061, Shaanxi Province, China. lsmxjtu@ 123456126.com

                Article
                jWJG.v29.i18.pg2798
                10.3748/wjg.v29.i18.2798
                10237113
                f34ef313-2195-4e79-84e6-5cc08387916f
                ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.

                This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial.

                History
                : 13 December 2022
                : 8 March 2023
                : 10 April 2023
                Categories
                Basic Study

                nonstructural protein 3-transactivated protein 1,calcitriol,liver fibrosis,hepatic stellate cells,mouse model,tgfβ1/smad3,nf-κb,signaling pathway

                Comments

                Comment on this article