1
|
Dai J, Li H, Gou L, Tian Y, Cheng C, Yuan F, Xie J, Zhang L, Ji J, Zhang L, Wang X. Mechanistic Study of Purple Sweet Potato Anthocyanins: Multifaceted Anti-Fibrotic Effects and Targeting of PDGFRβ in Liver Fibrosis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:27861-27875. [PMID: 39626114 DOI: 10.1021/acs.jafc.4c05796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
The purple sweet potato anthocyanins (PSPA) are known for their diverse health benefits, yet their hepatoprotective effects and the mechanisms by which they combat liver fibrosis have not been thoroughly investigated. This study aimed to elucidate these effects by employing a carbon tetrachloride (CCl4)-induced mouse model of liver fibrosis. We conducted a comprehensive analysis of the effects of PSPA on liver injury, oxidative stress, inflammation, and fibrosis-related signaling pathways. Our results demonstrate that PSPA can mitigate liver damage in mice, regulate key antioxidant enzymes such as catalase and SOD, and reduce oxidative stress as indicated by lowered MDA levels. PSPA also decrease the expression of inflammatory proteins, including CD3, CD4, CD45, IL-1β, TNF-α, and IL-17A, and reduce the accumulation of fibrotic markers like type I and III collagens and α-SMA. Additionally, PSPA have demonstrated the ability to inhibit key fibrogenic signaling proteins, including TGFβR2, p-Smad2, p-Smad3, p-PDGFRβ, p-AKT, p-ERK1/2, p-JNK1/2, and p-p38. Furthermore, we identified two potent monomers, PSPA-1 and PSPA-2, which directly target the PDGFRβ, a key player in fibrosis. The mechanism of action involves the inhibition of PDGF-B binding to PDGFRβ, thus disrupting the PDGF-B/PDGFRβ signaling pathway. These findings suggest that the hepatoprotective and antifibrotic effects of PSPA are due to their multifunctional bioactivities and the presence of specific active components that can effectively target fibrogenic protein.
Collapse
Affiliation(s)
- Jun Dai
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
- Anhui Engineering Laboratory for Conservation and Sustainable Utilization of Traditional Chinese Medicine Resources, West Anhui University, Lu'an 237012, Anhui, China
| | - Huansong Li
- Department of General Surgery of XuZhou Central Hospital, XuZhou Clinical School of Xuzhou Medical University, XuZhou 221009, Jiangsu, China
| | - Lingshan Gou
- Center for Genetic Medicine, Xuzhou Maternity and Child Health Care Hospital, Xuzhou 221009, Jiangsu, China
| | - Yuanzhi Tian
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Chao Cheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Fukang Yuan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Jun Xie
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Lidan Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Jia Ji
- Department of Cell Biology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221009, Jiangsu, China
| | - Liming Zhang
- Institute of Field Crops, Shandong Academy of Agricultural Sciences, Jinan 250100, Shandong, China
| | - Xingqi Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
- Anhui Engineering Laboratory for Conservation and Sustainable Utilization of Traditional Chinese Medicine Resources, West Anhui University, Lu'an 237012, Anhui, China
| |
Collapse
|
2
|
Ni Q, Lin J, Huang W, Yang L, Li R, Tu T, He G, Gao Y, Sun X, Kong X, Zhu X. Kehuang capsule inhibits MAPK and AKT signaling pathways to mitigate CCl 4-induced acute liver injury. LIVER RESEARCH 2024; 8:269-281. [PMID: 39958915 PMCID: PMC11771260 DOI: 10.1016/j.livres.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/27/2024] [Accepted: 11/28/2024] [Indexed: 02/18/2025]
Abstract
Background and aims Kehuang (KH) capsule is an herbal medical product approved for the treatment of liver diseases, including liver injury, in China. However, the mechanism is still unclear. This study aimed to elucidate the protective effects of KH capsule against carbon tetrachloride (CCl4)-induced acute liver injury (ALI) in a murine model. Methods Mice were randomly divided into control, model (CCl4), CCl4+KH_Low and CCl4+KH_High group. Liver enzyme levels and histological changes were assessed to evaluate liver injury. Oxidative stress markers and inflammatory cell infiltration in liver tissues were measured. Additionally, network pharmacology was employed to explore the potential mechanisms of KH capsule. Results KH capsule significantly reduced serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, as well as the necrotic area in liver tissue. KH capsule also decreased the infiltration of macrophages and neutrophils, thereby inhibiting the expression of interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and interleukin-1 beta (IL-1β). Furthermore, KH capsule decreased liver malondialdehyde (MDA) levels and increased superoxide dismutase (SOD) activity. The number of terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labeling (TUNEL)-positive cells in liver tissue was also reduced. The expression of nuclear factor erythroid 2 related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) proteins was significantly elevated, while the protein expression of cytochrome P450 2E1 (CYP2E1) was significantly reduced. Mass spectrometry identified genistein, galangin, wogonin, skullcapflavone II, and hispidulin as potential active ingredients of KH capsule. Network pharmacology analysis revealed enrichment in the mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT) signaling pathways. Western blot analysis confirmed that KH capsule suppressed AKT, extracellular signal-regulated kinase (ERK), and p38 signaling. Conclusions These findings suggest that KH capsule could exert protective effects against CCl4-induced ALI, with the inhibition of MAPK and PI3K-AKT signaling pathways playing a crucial role in its mechanism of action.
Collapse
Affiliation(s)
- Qinyu Ni
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiacheng Lin
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weifan Huang
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liu Yang
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ran Li
- Kexing Biopharm Co., Ltd, Jinan, Shandong, China
| | - Tianzhi Tu
- Kexing Biopharm Co., Ltd, Jinan, Shandong, China
| | - Guangfu He
- Kexing Biopharm Co., Ltd, Jinan, Shandong, China
| | - Yueqiu Gao
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuehua Sun
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoni Kong
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaojun Zhu
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
3
|
Ramos-Tovar E, Muriel P. NLRP3 inflammasome in hepatic diseases: A pharmacological target. Biochem Pharmacol 2023; 217:115861. [PMID: 37863329 DOI: 10.1016/j.bcp.2023.115861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
The NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome pathway is mainly responsible for the activation and release of a cascade of proinflammatory mediators that contribute to the development of hepatic diseases. During alcoholic liver disease development, the NLRP3 inflammasome pathway contributes to the maturation of caspase-1, interleukin (IL)-1β, and IL-18, which induce a robust inflammatory response, leading to fibrosis by inducing profibrogenic hepatic stellate cell (HSC) activation. Substantial evidence demonstrates that nonalcoholic fatty liver disease (NAFLD) progresses to nonalcoholic steatohepatitis (NASH) via NLRP3 inflammasome activation, ultimately leading to fibrosis and hepatocellular carcinoma (HCC). Activation of the NLRP3 inflammasome in NASH can be attributed to several factors, such as reactive oxygen species (ROS), gut dysbiosis, leaky gut, which allow triggers such as cardiolipin, cholesterol crystals, endoplasmic reticulum stress, and uric acid to reach the liver. Because inflammation triggers HSC activation, the NLRP3 inflammasome pathway performs a central function in fibrogenesis regardless of the etiology. Chronic hepatic activation of the NLRP3 inflammasome can ultimately lead to HCC; however, inflammation also plays a role in decreasing tumor growth. Some data indicate that NLRP3 inflammasome activation plays an important role in autoimmune hepatitis, but the evidence is scarce. Most researchers have reported that NLRP3 inflammasome activation is essential in liver injury induced by a variety of drugs and hepatotropic virus infection; however, few reports indicate that this pathway can play a beneficial role by inducing liver regeneration. Modulation of the NLRP3 inflammasome appears to be a suitable strategy to treat liver diseases.
Collapse
Affiliation(s)
- Erika Ramos-Tovar
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina-IPN, Apartado Postal 11340, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, Ciudad de México, México
| | - Pablo Muriel
- Laboratorio de Hepatología Experimental, Departamento de Farmacología, Cinvestav-IPN, Apartado Postal 14-740, Ciudad de México, México.
| |
Collapse
|
4
|
Ning W, Gao G, Zhou Y, Li WQ, Yang HH, Duan XB, Li X, Gong YB, Li D, Guo R. Calcitonin gene-related peptide ameliorates sepsis-induced intestinal injury by suppressing NLRP3 inflammasome activation. Int Immunopharmacol 2023; 116:109747. [PMID: 36706592 DOI: 10.1016/j.intimp.2023.109747] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/08/2023] [Accepted: 01/14/2023] [Indexed: 01/26/2023]
Abstract
Intestinal damage has long been viewed as the primary cause of sepsis-induced multiple organ dysfunction syndrome (MODS). Previous studies have demonstrated that calcitonin gene-related peptide (CGRP) exhibits anti-inflammatory and protective effects in mice exposed to endotoxin. This study investigated whether CGRP protects against sepsis-induced intestinal damage and its underlying mechanisms. Using a murine caecal ligation and puncture (CLP) model, we observed elevated serum and intestinal CGRP levels in septic mice. CGRP knockout (KO) mice showed more severe intestinal barrier damage, excessive NLRP3 inflammasome activation and higher levels of inflammation. In vitro, we used lipopolysaccharide (LPS) and adenosine triphosphate (ATP) to activate the NLRP3 inflammasome in MODE-K murine intestinal epithelial cells. CGRP inhibited NF-κB pathway activation; prevented ASC assembly and ROS accumulation; significantly decreased NLRP3, Caspase-1 p10, and IL-1β levels and LDH release; and increased cell viability. Treatment with an IL-1β inhibitor or CGRP suppressed p38 MAPK and ERK1/2 pathway activation and increased ZO-1 and Occludin protein levels in LPS+ATP-treated MODE-K cells. Finally, we used the CGRP upstream agonist drug rutaecarpine (RUT) to control endogenous CGRP release in mice, and this drug demonstrated good therapeutic effects on septic intestinal injury. In conclusion, our results suggest that CGRP ameliorates sepsis-induced intestinal damage, providing valuable insights for drug development.
Collapse
Affiliation(s)
- Wei Ning
- Laboratory Department, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Ge Gao
- Laboratory Department, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Wen-Qun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Xiang-Bing Duan
- Laboratory Department, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Xin Li
- Laboratory Department, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Yi-Bo Gong
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Dai Li
- Phase I Clinical Research Center, Xiangya Hospital, Central South University, Changsha 410005, China.
| | - Ren Guo
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| |
Collapse
|
5
|
Zhang C, Zhang Q, Yang D, Qiao Y, Wang B, Yan J, Li Z, Huang Z, Zhou Y, Hu K, Zhang Y. Chitosan degradation products promote healing of burn wounds of rat skin. Front Bioeng Biotechnol 2022; 10:1002437. [PMID: 36304900 PMCID: PMC9592717 DOI: 10.3389/fbioe.2022.1002437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/26/2022] [Indexed: 11/23/2022] Open
Abstract
Burns can impair the barrier function of the skin, and small burns can also cause high mortality. The WHO has described that over 180,000 people die of burns worldwide each year. Thus, the treatment of burn wounds is a major clinical challenge. Chitooligosaccharides (COS) are alkaline amino oligosaccharides with small molecular weights obtained by enzyme or chemical degradation of chitosan. With the characteristics of biocompatibility, water solubility and degradability, it has attracted increasing attention in the fields of biomedicine. In the present study, we used COS to treat deep second-degree burn wounds of rat skin and found that COS was able to promote wound healing. We also revealed that COS could promote fibroblast proliferation. Transcriptome sequencing analysis was performed on COS-treated fibroblasts to identify the underlying mechanisms. The results showed that COS was able to promote wound healing through regulation of the mitogen-activated protein kinase (MAPK) pathway and growth factor Hepatocyte Growth Factor (HGF). Our results provide a potential drug for burn wound therapy and the related molecular mechanism.
Collapse
Affiliation(s)
- Chuwei Zhang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Qingrong Zhang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Third Military Medical University (Army Medical University), Chongqing, China
| | - Dongmei Yang
- Outpatient Treatment Center, Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yating Qiao
- Department of Gastrointestinal Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Bolin Wang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Jun Yan
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Zihan Li
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Zhanghao Huang
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Youlang Zhou
- The Hand Surgery Research Center, Department of Hand Surgery, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Youlang Zhou, ; Kesu Hu, ; Yi Zhang,
| | - Kesu Hu
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Youlang Zhou, ; Kesu Hu, ; Yi Zhang,
| | - Yi Zhang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Youlang Zhou, ; Kesu Hu, ; Yi Zhang,
| |
Collapse
|
6
|
Recent Advancements in Antifibrotic Therapies for Regression of Liver Fibrosis. Cells 2022; 11:cells11091500. [PMID: 35563807 PMCID: PMC9104939 DOI: 10.3390/cells11091500] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 11/18/2022] Open
Abstract
Cirrhosis is a severe form of liver fibrosis that results in the irreversible replacement of liver tissue with scar tissue in the liver. Environmental toxicity, infections, metabolic causes, or other genetic factors including autoimmune hepatitis can lead to chronic liver injury and can result in inflammation and fibrosis. This activates myofibroblasts to secrete ECM proteins, resulting in the formation of fibrous scars on the liver. Fibrosis regression is possible through the removal of pathophysiological causes as well as the elimination of activated myofibroblasts, resulting in the reabsorption of the scar tissue. To date, a wide range of antifibrotic therapies has been tried and tested, with varying degrees of success. These therapies include the use of growth factors, cytokines, miRNAs, monoclonal antibodies, stem-cell-based approaches, and other approaches that target the ECM. The positive results of preclinical and clinical studies raise the prospect of a viable alternative to liver transplantation in the near future. The present review provides a synopsis of recent antifibrotic treatment modalities for the treatment of liver cirrhosis, as well as a brief summary of clinical trials that have been conducted to date.
Collapse
|
7
|
Charan HV, Dwivedi DK, Khan S, Jena G. Mechanisms of NLRP3 inflammasome-mediated hepatic stellate cell activation: therapeutic potential for liver fibrosis. Genes Dis 2022; 10:480-494. [DOI: 10.1016/j.gendis.2021.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 11/09/2021] [Accepted: 12/01/2021] [Indexed: 01/18/2023] Open
|
8
|
SB203580-A Potent p38 MAPK Inhibitor Reduces the Profibrotic Bronchial Fibroblasts Transition Associated with Asthma. Int J Mol Sci 2021; 22:ijms222312790. [PMID: 34884593 PMCID: PMC8657816 DOI: 10.3390/ijms222312790] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/21/2021] [Accepted: 11/23/2021] [Indexed: 01/23/2023] Open
Abstract
Subepithelial fibrosis is a component of the remodeling observed in the bronchial wall of patients diagnosed with asthma. In this process, human bronchial fibroblasts (HBFs) drive the fibroblast-to-myofibroblast transition (FMT) in response to transforming growth factor-β1 (TGF-β1), which activates the canonical Smad-dependent signaling. However, the pleiotropic properties of TGF-β1 also promote the activation of non-canonical signaling pathways which can affect the FMT. In this study we investigated the effect of p38 mitogen-activated protein kinase (MAPK) inhibition by SB203580 on the FMT potential of HBFs derived from asthmatic patients using immunocytofluorescence, real-time PCR and Western blotting methods. Our results demonstrate for the first time the strong effect of p38 MAPK inhibition on the TGF-β1-induced FMT potential throughout the strong attenuation of myofibroblast-related markers: α-smooth muscle actin (α-SMA), collagen I, fibronectin and connexin 43 in HBFs. We suggest the pleiotropic mechanism of SB203580 on FMT impairment in HBF populations by the diminishing of TGF-β/Smad signaling activation and disturbances in the actin cytoskeleton architecture along with the maturation of focal adhesion sites. These observations justify future research on the role of p38 kinase in FMT efficiency and bronchial wall remodeling in asthma.
Collapse
|
9
|
Kaminsky LW, Al-Sadi R, Ma TY. IL-1β and the Intestinal Epithelial Tight Junction Barrier. Front Immunol 2021; 12:767456. [PMID: 34759934 PMCID: PMC8574155 DOI: 10.3389/fimmu.2021.767456] [Citation(s) in RCA: 194] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/11/2021] [Indexed: 02/06/2023] Open
Abstract
The intestinal epithelial tight junction (TJ) barrier controls the paracellular permeation of contents from the intestinal lumen into the intestinal tissue and systemic circulation. A defective intestinal TJ barrier has been implicated as an important pathogenic factor in inflammatory diseases of the gut including Crohn's disease, ulcerative colitis, necrotizing enterocolitis, and celiac disease. Previous studies have shown that pro-inflammatory cytokines, which are produced during intestinal inflammation, including interleukin-1β (IL-1β), tumor necrosis factor-α, and interferon-γ, have important intestinal TJ barrier-modulating actions. Recent studies have shown that the IL-1β-induced increase in intestinal TJ permeability is an important contributing factor of intestinal inflammation. The IL-1β-induced increase in intestinal TJ permeability is mediated by regulatory signaling pathways and activation of nuclear transcription factor nuclear factor-κB, myosin light chain kinase gene activation, and post-transcriptional occludin gene modulation by microRNA and contributes to the intestinal inflammatory process. In this review, the regulatory role of IL-1β on intestinal TJ barrier, the intracellular mechanisms that mediate the IL-1β modulation of intestinal TJ permeability, and the potential therapeutic targeting of the TJ barrier are discussed.
Collapse
Affiliation(s)
- Lauren W Kaminsky
- Section of Allergy, Asthma, and Immunology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Rana Al-Sadi
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Thomas Y Ma
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
10
|
Rodríguez-Antonio I, López-Sánchez GN, Uribe M, Chávez-Tapia NC, Nuño-Lámbarri N. Role of the inflammasome, gasdermin D, and pyroptosis in non-alcoholic fatty liver disease. J Gastroenterol Hepatol 2021; 36:2720-2727. [PMID: 34050551 DOI: 10.1111/jgh.15561] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/17/2021] [Accepted: 05/26/2021] [Indexed: 12/25/2022]
Abstract
Pyroptosis is a type of programmed cell death mediated by a multiprotein complex called the inflammasome through the pro-inflammatory activity of gasdermin D. This study aimed to recognize the final biological product that leads to pore formation in the cell membrane, lysis, pro-inflammatory cytokines release, and the establishment of an immune response. An exhaustive search engine investigation of an elevated immune response can induce a sustained inflammation that directly links this mechanism to non-alcoholic fatty liver disease and its progression to non-alcoholic steatohepatitis. Clinical studies and systematic reviews suggest that gasdermin D is a critical molecule between the immune response and the disease manifestation, which could be considered a therapeutic target for highly prevalent diseases characterized by presenting perpetuated inflammatory processes. Both basic and clinical research show evidence on the expression and regulation of the inflammasome-gasdermin D-pyroptosis trinomial for the progression of non-alcoholic fatty liver disease to non-alcoholic steatohepatitis.
Collapse
Affiliation(s)
| | | | - Misael Uribe
- Obesity and Digestive Diseases Unit, Médica Sur Clinic Foundation, Mexico City, Mexico
| | - Norberto C Chávez-Tapia
- Traslational Research Unit, Médica Sur Clinic Foundation, Mexico City, Mexico.,Obesity and Digestive Diseases Unit, Médica Sur Clinic Foundation, Mexico City, Mexico
| | | |
Collapse
|
11
|
Kochumon S, Al-Sayyar A, Jacob T, Hasan A, Al-Mulla F, Sindhu S, Ahmad R. TNF-α Increases IP-10 Expression in MCF-7 Breast Cancer Cells via Activation of the JNK/c-Jun Pathways. Biomolecules 2021; 11:biom11091355. [PMID: 34572567 PMCID: PMC8464892 DOI: 10.3390/biom11091355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/04/2021] [Accepted: 09/06/2021] [Indexed: 12/28/2022] Open
Abstract
IP-10 (also called CXCL10) plays a significant role in leukocyte homing to inflamed tissues, and increased IP-10 levels are associated with the pathologies of various inflammatory disorders, including type 2 diabetes, atherosclerosis, and cancer. TNF-α is a potent activator of immune cells and induces inflammatory cytokine expression in these cells. However, it is unclear whether TNF-α is able to induce IP-10 expression in MCF-7 breast cancer cells. We therefore determined IP-10 expression in TNF-α-treated MCF-7 cells and investigated the mechanism involved. Our data show that TNF-α induced/upregulated the IP-10 expression at both mRNA and protein levels in MCF-7 cells. Inhibition of JNK (SP600125) significantly suppressed the TNF-α-induced IP-10 in MCF-7 cells, while the inhibition of p38 MAPK (SB203580), MEK1/2 (U0126), and ERK1/2 (PD98059) had no significant effect. Furthermore, TNF-α-induced IP-10 expression was abolished in MCF-7 cells deficient in JNK. Similar results were obtained using MCF-7 cells deficient in c-Jun. Moreover, the JNK kinase inhibitor markedly reduced the TNF-α-induced JNK and c-Jun phosphorylation. The kinase activity of JNK induced by TNF-α stimulation of MCF-7 cells was significantly inhibited by SP600125. Altogether, our novel findings provide the evidence that TNF-α induces IP-10 expression in MCF-7 breast cancer cells via activation of the JNK/c-Jun signaling pathway.
Collapse
Affiliation(s)
- Shihab Kochumon
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (A.A.-S.); (T.J.); (A.H.); (S.S.)
| | - Amnah Al-Sayyar
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (A.A.-S.); (T.J.); (A.H.); (S.S.)
| | - Texy Jacob
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (A.A.-S.); (T.J.); (A.H.); (S.S.)
| | - Amal Hasan
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (A.A.-S.); (T.J.); (A.H.); (S.S.)
| | - Fahd Al-Mulla
- Genetics & Bioinformatics Department, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| | - Sardar Sindhu
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (A.A.-S.); (T.J.); (A.H.); (S.S.)
- Animal and Imaging Core Facility, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Rasheed Ahmad
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (A.A.-S.); (T.J.); (A.H.); (S.S.)
- Correspondence:
| |
Collapse
|
12
|
Du JJ, Sun JC, Li N, Li XQ, Sun WY, Wei W. β-Arrestin2 deficiency attenuates oxidative stress in mouse hepatic fibrosis through modulation of NOX4. Acta Pharmacol Sin 2021; 42:1090-1100. [PMID: 33116250 PMCID: PMC8209231 DOI: 10.1038/s41401-020-00545-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatic fibrosis is a disease characterized by excessive deposition of extracellular matrix (ECM) in the liver. Activation of hepatic stellate cells (HSCs) is responsible for most of ECM production. Oxidative stress and reactive oxygen species (ROS) may be important factors leading to liver fibrosis. NADPH oxidase 4 (NOX4) is the main source of ROS in hepatic fibrosis, but the mechanism by which NOX4 regulates oxidative stress is not fully understood. β-Arrestin2 is a multifunctional scaffold protein that regulates receptor endocytosis, signaling and trafficking. In this study, we investigated whether β-arrestin2 regulated oxidative stress in hepatic fibrosis. Both β-arrestin2 knockout (Arrb2 KO) mice and wild-type mice were intraperitoneally injected with carbon tetrachloride (CCl4) to induce hepatic fibrosis. Arrb2 KO mice showed significantly attenuated liver fibrosis, decreased ROS levels and NOX4 expression, and reduced collagen levels in their livers. In vitro, NOX4 knockdown significantly inhibited ROS production, and decreased expression of alpha-smooth muscle actin in angiotensin II-stimulated human HSC cell line LX-2. Through overexpression or depletion of β-arrestin2 in LX-2 cells, we revealed that decreased β-arrestin2 inhibited ROS levels and NOX4 expression, and reduced collagen production; it also inhibited activation of ERK and JNK signaling pathways. These results demonstrate that β-arrestin2 deficiency protects against liver fibrosis by downregulating ROS production through NOX4. This effect appears to be mediated by ERK and JNK signaling pathways. Thus, targeted inhibition of β-arrestin2 might reduce oxidative stress and inhibit the progression of liver fibrosis.
Collapse
Affiliation(s)
- Jia-Jia Du
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Jia-Chang Sun
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Nan Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Xiu-Qin Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Wu-Yi Sun
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China.
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China.
| |
Collapse
|
13
|
Ribera J, Vilches C, Sanz V, de Miguel I, Portolés I, Córdoba-Jover B, Prat E, Nunes V, Jiménez W, Quidant R, Morales-Ruiz M. Treatment of Hepatic Fibrosis in Mice Based on Targeted Plasmonic Hyperthermia. ACS NANO 2021; 15:7547-7562. [PMID: 33720693 DOI: 10.1021/acsnano.1c00988] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Liver fibrosis is a major health problem with multiple associated complications, which, to date, has no effective treatment. Hepatic stellate cells are the main responsible cells for fibrosis formation; upon their activation, excess accumulation of extracellular matrix and collagen deposits occurs. The mitogen platelet-derived growth factor (PDGF) and its receptor β (PDGFRβ) play a major role in hepatic stellate cells activation and are, therefore, promising targets for antifibrotic therapies. Gold nanorods hold great potential for diseased liver treatments, since their passive hepatic accumulation enhances active targeting strategies, hence increasing therapeutic efficiency. In addition, gold nanorods have photothermal properties that, combined with specific cell delivery, can be exploited to induce localized near-infrared light-mediated thermal ablation. Here, we demonstrate that gold nanorods coated with anti-PDGFRβ specifically target activated hepatic stellate cells in vivo. Additionally, gold nanorods-PDGFRβ-mediated photothermal therapy decreases fibrosis, hepatic inflammation, and hepatocyte injury in the experimental model of CCl4-induced liver fibrosis in mice.
Collapse
Affiliation(s)
- Jordi Ribera
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08008 Barcelona, Spain
| | - Clara Vilches
- Institut de Ciències Fotòniques (ICFO), The Barcelona Institute of Science and Technology, 08860 Castelldefels, Barcelona, Spain
| | - Vanesa Sanz
- Institut de Ciències Fotòniques (ICFO), The Barcelona Institute of Science and Technology, 08860 Castelldefels, Barcelona, Spain
| | - Ignacio de Miguel
- Institut de Ciències Fotòniques (ICFO), The Barcelona Institute of Science and Technology, 08860 Castelldefels, Barcelona, Spain
| | - Irene Portolés
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08008 Barcelona, Spain
| | - Bernat Córdoba-Jover
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08008 Barcelona, Spain
| | - Esther Prat
- Molecular Genetics Laboratory, Genes, Disease and Therapy Programme, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Spain
- Department of Physiology, Health Science and Medicine Faculty, University of Barcelona (UB), 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Virginia Nunes
- Molecular Genetics Laboratory, Genes, Disease and Therapy Programme, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Spain
- Department of Physiology, Health Science and Medicine Faculty, University of Barcelona (UB), 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Wladimiro Jiménez
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08008 Barcelona, Spain
- Department of Biomedicine-Biochemistry Unit, School of Medicine, University of Barcelona (UB), 08008 Barcelona, Spain
| | - Romain Quidant
- Institut de Ciències Fotòniques (ICFO), The Barcelona Institute of Science and Technology, 08860 Castelldefels, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Manuel Morales-Ruiz
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08008 Barcelona, Spain
- Department of Biomedicine-Biochemistry Unit, School of Medicine, University of Barcelona (UB), 08008 Barcelona, Spain
| |
Collapse
|
14
|
Jung J, Lee YH, Fang X, Kim SJ, Kim SH, Kim DH, Song NY, Na HK, Baek JH, Surh YJ. IL-1β induces expression of proinflammatory cytokines and migration of human colon cancer cells through upregulation of SIRT1. Arch Biochem Biophys 2021; 703:108847. [PMID: 33766523 DOI: 10.1016/j.abb.2021.108847] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 10/21/2022]
Abstract
SIRT1 is a mammalian NAD+-dependent deacetylase, which is known to be involved in various physiological events, such as adaptive response to environmental stresses including caloric restriction, as well as in aging and cellular senescence. However, recent studies have revealed overexpression of SIRT1 in many different types of human malignancies, particularly colon cancer. Interleukin-1β (IL-1β) is a proinflammatory cytokine that plays a major role in invasiveness, stemness and progression of colon cancer. However, the interaction between IL-1β and SIRT1 in the tumor development and progression remains elusive. In this study, we found that IL-1β induces SIRT1 protein expression in human colon cancer HCT-116 cells. IL-1β-induced SIRT1 upregulation led to enhanced expression of mRNA transcripts of pro-inflammatory cytokines, IL-6 and IL-8 as well as that of IL-1β. Knockdown of SIRT1 prevented IL-1β-induced phosphorylation and nuclear accumulation of c-Jun. Furthermore, pharmacologic inhibition of SIRT1 abrogated clonogenicity and migrative capability of human colon cancer cells stimulated with IL-1β. In summary, IL-1β-induced SIRT1 upregulation stimulates production of proinflammatory cytokines via a nuclear accumulation of c-Jun, leadng to colon cancer growth and progression.
Collapse
Affiliation(s)
- Jaekyung Jung
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea; Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Yeon-Hwa Lee
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Xizhu Fang
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Su-Jung Kim
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Seong Hoon Kim
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Do-Hee Kim
- Department of Chemistry, College of Convergence and Integrated Science, Kyonggi University, Suwon, Gyeonggi-do, South Korea
| | - Na-Young Song
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, South Korea
| | - Hye-Kyung Na
- Department of Food Science and Biotechnology, College of Knowledge-Based Services Engineering, Sungshin Women's University, Seoul, South Korea
| | - Jeong-Heum Baek
- Division of Colon and Rectal Surgery, Department of Surgery, Gil Medical Center, Gachon University College of Medicine, Incheon, South Korea
| | - Young-Joon Surh
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea; Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea; Cancer Research Institute, Seoul National University, Seoul, South Korea.
| |
Collapse
|
15
|
Sharawy MH, El-Awady MS, Makled MN. Protective effects of paclitaxel on thioacetamide-induced liver fibrosis in a rat model. J Biochem Mol Toxicol 2021; 35:e22745. [PMID: 33749060 DOI: 10.1002/jbt.22745] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 01/13/2021] [Accepted: 02/10/2021] [Indexed: 01/18/2023]
Abstract
Liver fibrosis is a public health burden that is highly associated with morbidity and mortality. Therefore, this study aims to explore the anti-fibrotic effects of low dose of paclitaxel (PTX) against thioacetamide (TAA)-induced liver fibrosis in rats and the possible mechanisms involved. TAA was administered at a dose of 200 mg/kg twice weekly for 6 weeks in rats to induce liver fibrosis similar to that in humans. Liver dysfunction was shown by increased alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, and γ-glutamyl transferase, along with histopathological changes. Liver fibrosis was confirmed by Masson's Trichome staining, increased collagen content, and elevated α-smooth muscle actin (α-SMA) protein expression. In addition, TAA induced liver apoptosis as indicated by the increased terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells in liver tissues. This study demonstrated that the administration of PTX (0.3 mg/kg/i.p.) three times a week for 6 weeks significantly alleviated functional and biochemical changes induced by TAA in addition to improving the liver architecture. PTX attenuated liver fibrosis as reflected by the decreased collagen content and α-SMA protein expression. Additionally, PTX attenuated liver apoptosis as indicated by the decreased TUNEL-positive cells. Moreover, PTX prevented TAA-induced elevation of transforming growth factor-β1 (TGF-β1), platelet-derived growth factor-BB (PDGF-BB), and tissue inhibitor of metalloproteinase 1 (TIMP-1) levels in liver tissues. These findings suggest that the low dose of PTX prevented TAA-induced liver fibrosis in rats, possibly by inhibiting the expression of TGF-β1 and PDGF-BB and subsequently suppressing the apoptosis and the expression of TIMP-1.
Collapse
Affiliation(s)
- Maha H Sharawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Mohammed S El-Awady
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.,Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Al-Madinah Al-Munawwarah, Saudi Arabia
| | - Mirhan N Makled
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
16
|
Allicin Attenuated Advanced Oxidation Protein Product-Induced Oxidative Stress and Mitochondrial Apoptosis in Human Nucleus Pulposus Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6685043. [PMID: 33381267 PMCID: PMC7758128 DOI: 10.1155/2020/6685043] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 12/15/2022]
Abstract
Intervertebral disc degeneration (IDD) is one of the most common chronic degenerative musculoskeletal disorders. Oxidative stress-induced apoptosis of the nucleus pulposus (NP) cells plays a key role during IDD progression. Advanced oxidation protein products (AOPP), novel biomarkers of oxidative stress, have been reported to function in various diseases due to their potential for disrupting the redox balance. The current study is aimed at investigating the function of AOPP in the oxidative stress-induced apoptosis of human NP cells and the alleviative effects of allicin during this process which was known for its antioxidant properties. AOPP were demonstrated to hamper the viability and proliferation of NP cells in a time- and concentration-dependent manner and cause cell apoptosis markedly. High levels of reactive oxygen species (ROS) and lipid peroxidation product malondialdehyde (MDA) were detected in NP cells after AOPP stimulation, which resulted in depolarized mitochondrial transmembrane potential (MTP). Correspondingly, higher levels of AOPP were discovered in the human degenerative intervertebral discs (IVD). It was also found that allicin could protect NP cells against AOPP-mediated oxidative stress and mitochondrial dysfunction via suppressing the p38-MAPK pathway. These results disclosed a significant role of AOPP in the oxidative stress-induced apoptosis of NP cells, which could be involved in the primary pathogenesis of IDD. It was also revealed that allicin could be a promising therapeutic approach against AOPP-mediated oxidative stress during IDD progression.
Collapse
|
17
|
Xu D, Zeng W, Han X, Qian T, Sun J, Qi F, Liu C, Wang Q, Jin H. Honokiol protects against epidural fibrosis by inhibiting fibroblast proliferation and extracellular matrix overproduction in rats post‑laminectomy. Int J Mol Med 2020; 46:2057-2068. [PMID: 33125121 PMCID: PMC7595651 DOI: 10.3892/ijmm.2020.4765] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/28/2020] [Indexed: 12/22/2022] Open
Abstract
Epidural fibrosis (EF)‑induced failed back surgery syndrome (FBSS) in patients post‑laminectomy remains a medical challenge. Although the scarring mechanisms remain unclear, the majority of aetiological studies have reported fibroblast dysfunction. Honokiol, the major bioactive constituent of the magnolia tree, exerts a variety of pharmacological effects, including anti‑proliferative and anti‑fibrotic effects, on various cell types. The present study investigated whether honokiol attenuates EF progression. In vitro, it was found that honokiol inhibited excessive fibroblast proliferation induced by transforming growth factor‑β1 (TGF‑β1) and the synthesis of extracellular matrix (ECM) components, including fibronectin and type I collagen, in a dose‑dependent manner. These effects were attributed to the ability of honokiol to suppress the activity of connective tissue growth factor (CTGF), which is indispensable for the progression of fibrosis. Mechanistically, honokiol attenuated the TGF‑β1‑induced activation of the Smad2/3 and mitogen‑activated protein kinase (MAPK) signalling pathways in fibroblasts. In vivo, honokiol reduced the proliferation of fibroblasts and the synthesis of ECM components, thus ameliorating EF in a rat model post‑laminectomy. Taken together, these preclinical findings suggest that honokiol deserves further consideration as a candidate therapeutic agent for EF.
Collapse
Affiliation(s)
- Daoliang Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Weimin Zeng
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Xuyao Han
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Tianchen Qian
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Jingyu Sun
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Fangzhou Qi
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Chen Liu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Quan Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Haiming Jin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| |
Collapse
|
18
|
Riaz F, Li D. Non-coding RNA Associated Competitive Endogenous RNA Regulatory Network: Novel Therapeutic Approach in Liver Fibrosis. Curr Gene Ther 2020; 19:305-317. [PMID: 31696817 DOI: 10.2174/1566523219666191107113046] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/21/2019] [Accepted: 10/28/2019] [Indexed: 12/19/2022]
Abstract
Liver fibrosis or scarring is the most common pathological feature caused by chronic liver injury, and is widely considered one of the primary causes of morbidity and mortality. It is primarily characterised by hepatic stellate cells (HSC) activation and excessive extracellular matrix (ECM) protein deposition. Overwhelming evidence suggests that the dysregulation of several noncoding RNAs (ncRNAs), mainly long non-coding RNAs (lncRNAs), microRNAs (miRNAs) and circular RNAs (circRNAs) contributes to the activation of HSC and progression of liver fibrosis. These ncRNAs not only bind to their target genes for the development and regression of liver fibrosis but also act as competing endogenous RNAs (ceRNAs) by sponging with miRNAs to form signaling cascades. Among these signaling cascades, lncRNA-miRNA-mRNA and circRNA-miRNA-mRNA are critical modulators for the initiation, progression, and regression of liver fibrosis. Thus, targeting these interacting ncRNA cascades can serve as a novel and potential therapeutic target for inhibition of HSC activation and prevention and regression of liver fibrosis.
Collapse
Affiliation(s)
- Farooq Riaz
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Dongmin Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, China
| |
Collapse
|
19
|
Katsarou A, Moustakas II, Pyrina I, Lembessis P, Koutsilieris M, Chatzigeorgiou A. Metabolic inflammation as an instigator of fibrosis during non-alcoholic fatty liver disease. World J Gastroenterol 2020; 26:1993-2011. [PMID: 32536770 PMCID: PMC7267690 DOI: 10.3748/wjg.v26.i17.1993] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/09/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterized by excessive storage of fatty acids in the form of triglycerides in hepatocytes. It is most prevalent in western countries and includes a wide range of clinical and histopathological findings, namely from simple steatosis to steatohepatitis and fibrosis, which may lead to cirrhosis and hepatocellular cancer. The key event for the transition from steatosis to fibrosis is the activation of quiescent hepatic stellate cells (qHSC) and their differentiation to myofibroblasts. Pattern recognition receptors (PRRs), expressed by a plethora of immune cells, serve as essential components of the innate immune system whose function is to stimulate phagocytosis and mediate inflammation upon binding to them of various molecules released from damaged, apoptotic and necrotic cells. The activation of PRRs on hepatocytes, Kupffer cells, the resident macrophages of the liver, and other immune cells results in the production of proinflammatory cytokines and chemokines, as well as profibrotic factors in the liver microenvironment leading to qHSC activation and subsequent fibrogenesis. Thus, elucidation of the inflammatory pathways associated with the pathogenesis and progression of NAFLD may lead to a better understanding of its pathophysiology and new therapeutic approaches.
Collapse
Affiliation(s)
- Angeliki Katsarou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
- 251 Hellenic Airforce General Hospital, Athens 11525, Greece
| | - Ioannis I Moustakas
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Iryna Pyrina
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden 01307, Germany
| | - Panagiotis Lembessis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden 01307, Germany.
| |
Collapse
|
20
|
Sanches RCO, Souza C, Marinho FV, Mambelli FS, Morais SB, Guimarães ES, Oliveira SC. NLRP6 Plays an Important Role in Early Hepatic Immunopathology Caused by Schistosoma mansoni Infection. Front Immunol 2020; 11:795. [PMID: 32431709 PMCID: PMC7214731 DOI: 10.3389/fimmu.2020.00795] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/07/2020] [Indexed: 12/11/2022] Open
Abstract
Schistosomiasis is a debilitating parasitic disease that affects more than 200 million people worldwide and causes approximately 280,000 deaths per year. Inside the definitive host, eggs released by Schistosoma mansoni lodge in the intestine and especially in the liver where they induce a granulomatous inflammatory process, which can lead to fibrosis. The molecular mechanisms initiating or promoting hepatic granuloma formation remain poorly understood. Inflammasome activation has been described as an important pathway to induce pathology mediated by NLRP3 receptor. Recently, other components of the inflammasome pathway, such as NLRP6, have been related to liver diseases and fibrotic processes. Nevertheless, the contribution of these components in schistosomiasis-associated pathology is still unknown. In the present study, using dendritic cells, we demonstrated that NLRP6 sensor is important for IL-1β production and caspase-1 activation in response to soluble egg antigens (SEA). Furthermore, the lack of NLRP6 has been shown to significantly reduce periovular inflammation, collagen deposition in hepatic granulomas and mRNA levels of α-SMA and IL-13. Livers of Nlrp6–/– mice showed reduced levels of CXCL1/KC, CCL2, CCL3, IL-5, and IL-10 as well as Myeloperoxidase (MPO) and Eosinophilic Peroxidase (EPO) enzymatic activity. Consistently, the frequency of macrophage and neutrophil populations were lower in the liver of NLRP6 knockout mice, after 6 weeks of infection. Finally, it was further demonstrated that the onset of hepatic granuloma and collagen deposition were also compromised in Caspase-1–/–, IL-1R–/– and Gsdmd–/– mice. Our findings suggest that the NLRP6 inflammasome is an important component for schistosomiasis-associated pathology.
Collapse
Affiliation(s)
- Rodrigo C O Sanches
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Cláudia Souza
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fabio Vitarelli Marinho
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fábio Silva Mambelli
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Suellen B Morais
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Erika S Guimarães
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Sergio Costa Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, Salvador, Brazil
| |
Collapse
|
21
|
Elfeky MG, Mantawy EM, Gad AM, Fawzy HM, El-Demerdash E. Mechanistic aspects of antifibrotic effects of honokiol in Con A-induced liver fibrosis in rats: Emphasis on TGF-β/SMAD/MAPK signaling pathways. Life Sci 2019; 240:117096. [PMID: 31760097 DOI: 10.1016/j.lfs.2019.117096] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/11/2019] [Accepted: 11/19/2019] [Indexed: 02/08/2023]
Abstract
Aim Liver fibrosis represents a massive global health burden with limited therapeutic options. Thus, the need for curative options is evident. Thus, this study aimed to assess the potential antifibrotic effect of honokiol in Concanavalin A (Con A) induced immunological model of liver fibrosis as well the possible underlying molecular mechanisms. METHODS Male Sprague-Dawley rats were treated with either Con A (20 mg/kg, IV) and/or honokiol (10 mg/kg, orally) for 4 weeks. Hepatotoxicity indices were as well as histopathological evaluation was done. Hepatic fibrosis was assessed by measuring alpha smooth muscle actin (α-SMA) expression and collagen fibers deposition by Masson's trichrome stain and hydroxyproline content. To elucidate the underlying molecular mechanisms, the effect of honokiol on oxidative stress, inflammatory markers as well as transforming growth factor beta (TGF-β)/SMAD and mitogen-activated protein kinase (MAPK) pathways was assessed. KEY FINDINGS Honokiol effectively reversed the hepatotoxicity indices elevations and abnormal histopathological changes induced by Con A. Besides, honokiol attenuated Con A-induced liver fibrosis by down-regulation of hydroxyproline levels, α-SMA expression together with a marked decrease in collagen fibers deposition. Mechanistically Con A induced oxidative stress, provocation of inflammatory responses and activation of TGF-β/SMAD/MAPK pathways. Contrariwise, honokiol co-treatment significantly restored antioxidant defence mechanisms, down-regulated inflammatory cascades and inhibited TGF-β/SMAD/MAPK signaling pathways. CONCLUSION The results provide an evidence for the promising antifibrotic effect of honokiol that could be partially due to suppressing oxidative stress and inflammatory processes as well as inhibition of TGF-β/SMAD/MAPK signaling pathways.
Collapse
Affiliation(s)
- Maha G Elfeky
- Department of Pharmacology, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| | - Eman M Mantawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Amany M Gad
- Department of Pharmacology, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| | - Hala M Fawzy
- Department of Pharmacology, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
22
|
Down-regulation of RalGTPase-Activating Protein Promotes Colitis-Associated Cancer via NLRP3 Inflammasome Activation. Cell Mol Gastroenterol Hepatol 2019; 9:277-293. [PMID: 31622786 PMCID: PMC6957823 DOI: 10.1016/j.jcmgh.2019.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND & AIMS Ral guanosine triphosphatase-activating protein α2 (RalGAPα2) is the major catalytic subunit of the negative regulators of the small guanosine triphosphatase Ral, a member of the Ras subfamily. Ral regulates tumorigenesis and invasion/metastasis of some cancers; however, the role of Ral in colitis-associated cancer (CAC) has not been investigated. We aimed to elucidate the role of Ral in the mechanism of CAC. METHODS We used wild-type (WT) mice and RalGAPα2 knockout (KO) mice that showed Ral activation, and bone marrow chimeric mice were generated as follows: WT to WT, WT to RalGAPα2 KO, RalGAPα2 KO to WT, and RalGAPα2 KO to RalGAPα2 KO mice. CAC was induced in these mice by intraperitoneal injection of azoxymethane followed by dextran sulfate sodium intake. Intestinal epithelial cells were isolated from colon tissues, and we performed complementary DNA microarray analysis. Cytokine expression in normal colon tissues and CAC was analyzed by quantitative polymerase chain reaction. RESULTS Bone marrow chimeric mice showed that immune cell function between WT mice and RalGAPα2 KO mice was not significantly different in the CAC mechanism. RalGAPα2 KO mice had a significantly larger tumor number and size and a significantly higher proportion of tumors invading the submucosa than WT mice. Higher expression levels of matrix metalloproteinase-9 and matrix metalloproteinase-13 were observed in RalGAPα2 KO mice than in WT mice. The expression levels of interleukin 1β, NLRP3, apoptosis associated speck-like protein containing a CARD, and caspase-1 were apparently increased in the tumors of RalGAPα2 KO mice compared with WT mice. NLRP3 inhibitor reduced the number of invasive tumors. CONCLUSIONS Ral activation participates in the mechanism of CAC development via NLRP3 inflammasome activation.
Collapse
|
23
|
Wu J, Lin S, Wan B, Velani B, Zhu Y. Pyroptosis in Liver Disease: New Insights into Disease Mechanisms. Aging Dis 2019; 10:1094-1108. [PMID: 31595205 PMCID: PMC6764727 DOI: 10.14336/ad.2019.0116] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/16/2019] [Indexed: 12/12/2022] Open
Abstract
There has been increasing interest in pyroptosis as a novel form of pro-inflammatory programmed cell death. The mechanism of pyroptosis is significantly different from other forms of cell death in its morphological and biochemical features. Pyroptosis is characterized by the activation of two different types of caspase enzymes—caspase-1 and caspase-4/5/11, and by the occurrence of a proinflammatory cytokine cascade and an immune response. Pyroptosis participates in the immune defense mechanisms against intracellular bacterial infections. On the other hand, excessive inflammasome activation can induce sterile inflammation and eventually cause some diseases, such as acute or chronic hepatitis and liver fibrosis. The mechanism and biological significance of this novel form of cell death in different liver diseases will be evaluated in this review. Specifically, we will focus on the role of pyroptosis in alcoholic and non-alcoholic fatty liver disease, as well as in liver failure. Finally, the therapeutic implications of pyroptosis in liver diseases will be discussed.
Collapse
Affiliation(s)
- Jiali Wu
- 1Liver research center of the First Affiliated Hospital of Fujian Medical University, Fujian 350005, China
| | - Su Lin
- 1Liver research center of the First Affiliated Hospital of Fujian Medical University, Fujian 350005, China
| | - Bo Wan
- 2Faculty of Life Sciences and Medicine, King's College London, London SE1 1UL, United Kingdom
| | - Bharat Velani
- 3Basildon and Thurrock University Hospitals NHS Foundation Trust, Nethermayne, Basildon, Essex SS16 5NL, United Kingdom
| | - Yueyong Zhu
- 1Liver research center of the First Affiliated Hospital of Fujian Medical University, Fujian 350005, China
| |
Collapse
|
24
|
Ma X, Zheng X, Pan L, Zhang X. NLRP3 inflammasome activation in liver cirrhotic patients. Biochem Biophys Res Commun 2018; 505:40-44. [DOI: 10.1016/j.bbrc.2018.09.055] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 09/09/2018] [Indexed: 02/06/2023]
|
25
|
Song HL, Zhang X, Wang WZ, Liu RH, Zhao K, Liu MY, Gong WM, Ning B. Neuroprotective mechanisms of rutin for spinal cord injury through anti-oxidation and anti-inflammation and inhibition of p38 mitogen activated protein kinase pathway. Neural Regen Res 2018; 13:128-134. [PMID: 29451217 PMCID: PMC5840978 DOI: 10.4103/1673-5374.217349] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Rutin has anti-inflammatory, antioxidant, anti-viral, anti-tumor and immune regulatory effects. However, the neuroprotective effects of rutin in spinal cord injury are unknown. The p38 mitogen activated protein kinase (p38 MAPK) pathway is the most important member of the MAPK family that controls inflammation. We assumed that the mechanism of rutin in the repair of spinal cord injury is associated with the inhibition of p38 MAPK pathway. Allen's method was used to establish a rat model of spinal cord injury. The rat model was intraperitoneally injected with rutin (30 mg/kg) for 3 days. After treatment with rutin, Basso, Beattie and Bresnahan locomotor function scores increased. Water content, tumor necrosis factor alpha, interleukin 1 beta, and interleukin 6 levels, p38 MAPK protein expression and caspase-3 and -9 activities in T8–9 spinal cord decreased. Oxidative stress related markers superoxide dismutase and glutathione peroxidase levels increased in peripheral blood. Rutin exerts neuroprotective effect through anti-oxidation, anti-inflammation, anti-apoptosis and inhibition of p38 MAPK pathway.
Collapse
Affiliation(s)
- Hong-Liang Song
- Department of Spinal Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong Province, China
| | - Xiang Zhang
- Hospital Pharmacy, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong Province, China
| | - Wen-Zhao Wang
- Department of Spinal Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong Province, China
| | - Rong-Han Liu
- Department of Spinal Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong Province, China
| | - Kai Zhao
- Department of Spinal Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong Province, China
| | - Ming-Yuan Liu
- Department of Gynecology and Obstetrics, Jinan Maternity and Child Care Hospital, Jinan, Shandong Province, China
| | - Wei-Ming Gong
- Department of Spinal Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong Province, China
| | - Bin Ning
- Department of Spinal Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
26
|
Ying HZ, Chen Q, Zhang WY, Zhang HH, Ma Y, Zhang SZ, Fang J, Yu CH. PDGF signaling pathway in hepatic fibrosis pathogenesis and therapeutics (Review). Mol Med Rep 2017; 16:7879-7889. [PMID: 28983598 PMCID: PMC5779870 DOI: 10.3892/mmr.2017.7641] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 07/20/2017] [Indexed: 02/06/2023] Open
Abstract
The platelet‑derived growth factor (PDFG) signaling pathway exerts persistent activation in response to a variety of stimuli and facilitates the progression of hepatic fibrosis. Since this pathway modulates a broad spectrum of cellular processes, including cell growth, differentiation, inflammation and carcinogenesis, it has emerged as a therapeutic target for hepatic fibrosis and liver‑associated disorders. The present review exhibits the current knowledge of the role of the PDGF signaling pathway and its pathological profiles in hepatic fibrosis, and assesses the potential of inhibitors which have been investigated in the experimental hepatic fibrosis model, in addition to the clinical challenges associated with these inhibitors.
Collapse
Affiliation(s)
- Hua-Zhong Ying
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Qin Chen
- Department of Clinical Laboratory Medicine, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Wen-You Zhang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Huan-Huan Zhang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Yue Ma
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Song-Zhao Zhang
- Department of Clinical Laboratory Medicine, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Jie Fang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Chen-Huan Yu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| |
Collapse
|
27
|
Wang D, Chen Y, Jiang J, Zhou A, Pan L, Chen Q, Qian Y, Chu M, Chen C. Carvedilol has stronger anti-inflammation and anti-virus effects than metoprolol in murine model with coxsackievirus B3-induced viral myocarditis. Gene 2014; 547:195-201. [PMID: 24905653 DOI: 10.1016/j.gene.2014.06.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 05/16/2014] [Accepted: 06/02/2014] [Indexed: 12/26/2022]
Abstract
AIMS This study aims to compare the effects of carvedilol and metoprolol in alleviating viral myocarditis (VMC) induced by coxsackievirus B3 (CVB3) in mice. METHODS A total of 116 Balb/c mice were included in this study. Ninety-six mice were inoculated intraperitoneally with CVB3 to induce VMC. The CVB3 inoculated mice were evenly divided into myocarditis group (n=32), carvedilol group (n=32) and metoprolol group (n=32). Twenty mice (control group) were inoculated intraperitoneally with normal saline. Hematoxylin and eosin staining and histopathologic scoring were used to investigate the effects of carvedilol and metoprolol on myocardial histopathologic changes on days 3 and 5. In addition, serum cTn-I levels, cytokine levels and virus titers were determined using chemiluminescence immunoassay, enzyme-linked immunosorbent assay and plaque assay, respectively, on days 3 and 5. Finally, the levels of phosphorylated p38MAPK were studied using immunohistochemical staining and Western blotting on day 5. RESULTS Carvedilol had a stronger effect than metoprolol in reducing the pathological scores of VMC induced by CVB3. Both carvedilol and metoprolol reduced the levels of cTn-I, but the effect of carvedilol was stronger. Carvedilol and metoprolol decreased the levels of myocardial pro-inflammatory cytokines and increased the expression of anti-inflammatory cytokine, with the effects of carvedilol being stronger than those of metoprolol. Carvedilol had a stronger effect in reducing myocardial virus concentration compared with metoprolol. Carvedilol was stronger than metoprolol in decreasing the levels of myocardial phosphorylated p38MAPK. CONCLUSIONS In conclusion, carvedilol was more potent than metoprolol in ameliorating myocardial lesions in VMC, probably due to its stronger modulation of the balance between pro- and anti-inflammatory cytokines by inhibiting the activation of p38MAPK pathway through β1- and β2-adrenoreceptors.
Collapse
Affiliation(s)
- Dan Wang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, PR China; Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, PR China
| | - Yiming Chen
- Department of Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, PR China
| | - Jianbin Jiang
- Department of Pediatrics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, PR China
| | - Aihua Zhou
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, PR China
| | - Lulu Pan
- Department of Pediatrics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, PR China
| | - Qi Chen
- Department of Pediatrics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, PR China
| | - Yan Qian
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, PR China
| | - Maoping Chu
- Department of Pediatrics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, PR China.
| | - Chao Chen
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, PR China.
| |
Collapse
|