1
|
Hu X, Lin H, Qian S, Xu Z, Li Z, Qian S, Yang F, Hou H, Xie Q, Wu W, Hu C, Abou-Elnour A, He Y, Huang Y. A novel experimental mouse model of diabetic nonalcoholic steatohepatitis: A critical role for acid-sensitive Ion Channel 1a. Biomed Pharmacother 2024; 178:117184. [PMID: 39142252 DOI: 10.1016/j.biopha.2024.117184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND A two-way relationship exists between type 2 diabetes (T2DM) and human nonalcoholic steatohepatitis (NASH). Several diabetic NASH models have the disadvantages of long cycles or inconsistent with the actual incidence of human disease, which would be costly and time-consuming to investigate disease pathogenesis and develop drugs. Therefore, there is an urgent need to establish a diabetic NASH mouse model. METHODS The combination between Fructose-palmitate-cholesterol diet (FPC) and Streptozotocin (STZ) (FPC+STZ) was used to construct diabetic NASH mouse model. The in vivo effects of silencing acid-sensitive Ion Channel 1a (ASIC1a) were examined with an adeno-associated virus 9 (AAV9) carrying ASIC1a short hairpin RNA (shRNA) in FPC+STZ model. RESULTS The mice fed with FPC for 12 weeks had insulin resistance, hyperinsulinemia, lipid accumulation, and increased hepatic levels of inflammatory factors. However, it still did not develop remarkable liver fibrosis. Most interestingly, noticeable fibrotic scars were observed in the liver of mice from FPC+STZ group. Furthermore, insulin therapy significantly ameliorated FPC+STZ-induced NASH-related liver fibrosis, indicating that hyperglycemia is of great significance in NASH development and progression. Importantly, ASIC1a was found to be involved in the pathogenesis of diabetic NASH as demonstrated that silencing ASIC1a in HSCs significantly ameliorated FPC+STZ-induced NASH fibrosis. Mechanistically, ASIC1a interacted with Poly Adp-adenosine ribose polymerase (PARP1) to promote HSC activation by inducing autophagy. CONCLUSION A FPC diet combined with an injection of STZ induces a diabetic NASH mouse model in a shorter period. Targeting ASIC1a may provide a novel therapeutic target for the treatment of diabetic NASH.
Collapse
Affiliation(s)
- Xiaojie Hu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Huimin Lin
- Department of Pharmacy, the Second Affiliated Hospital of Anhui Medical University, China
| | - Shengying Qian
- Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Zhou Xu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Zihao Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Shishun Qian
- Anhui Province Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Furong Yang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Hui Hou
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qinxiu Xie
- Department of Infection, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wenyong Wu
- Hospital of The Second People's Hospital of Anhui Province, Hefei, China
| | - Chengmu Hu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Amira Abou-Elnour
- School of International Education, Anhui Medical University, Hefei, China
| | - Yong He
- Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Yan Huang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Medical University, Hefei, China.
| |
Collapse
|
2
|
Ekpor E, Akyirem S, Adade Duodu P. Prevalence of metabolic dysfunction-associated fatty liver disease and its association with glycemic control in persons with type 2 diabetes in Africa: A systematic review and meta-analysis. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0002835. [PMID: 38709759 PMCID: PMC11073701 DOI: 10.1371/journal.pgph.0002835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/11/2024] [Indexed: 05/08/2024]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) and type 2 diabetes (T2D) are interconnected metabolic disorders with significant health implications. However, a comprehensive understanding of the extent of their co-occurrence in Africa is lacking. The aim of this review was to determine the prevalence of MAFLD and its association with glycemic control (HbA1c) in persons with T2D in Africa. A systematic search was conducted on PubMed, Medline, Embase, Scopus, Global Health, and Web of Science from their inception to December 6, 2023. Data on MAFLD prevalence and correlation coefficients regarding its association with glycemic control were pooled through random effect meta-analyses. Potential sources of heterogeneity were investigated using subgroup analysis and meta-regression. A total of 10 studies were included in the meta-analysis of MAFLD prevalence, while 2 were incorporated in the analysis of the association between MAFLD and glycemic control. The pooled prevalence of MAFLD in persons with T2D was 48.1% (95% CI: 36.1-60.3). The subgroup analysis revealed regional variations in MAFLD prevalence, with rates of 44.7% (95% CI: 28.7-62.0) in sub-Saharan Africa and 55.3% (95% CI: 36.2-73.0) in Northern Africa. Additionally, we observed an increasing trend in MAFLD prevalence, recording 55.1% (95% CI: 43.6-66.1) in the recent five years. There was a weak positive correlation between MAFLD and HbA1c (r = 0.33, 95% CI: 0.18-0.47). The findings of this study highlight a high prevalence of MAFLD in persons with T2D in Africa, with a suggested link between MAFLD and suboptimal glycemic control. Therefore, healthcare providers should prioritize the screening and management of MAFLD in individuals with T2D to enhance their metabolic health.
Collapse
Affiliation(s)
- Emmanuel Ekpor
- School of Nursing and Midwifery, University of Ghana, Accra, Ghana
- Christian Health Association of Ghana, Accra, Ghana
| | - Samuel Akyirem
- Yale School of Nursing, Yale University, New Haven, Connecticut, United States of America
| | - Precious Adade Duodu
- Department of Nursing, School of Human and Health Sciences, University of Huddersfield, Huddersfield, England, United Kingdom
| |
Collapse
|
3
|
Cao R, Cao C, Hu X, Du K, Zhang J, Li M, Li B, Lin H, Zhang A, Li Y, Wu L, Huang Y. Kaempferol attenuates carbon tetrachloride (CCl 4)-induced hepatic fibrosis by promoting ASIC1a degradation and suppression of the ASIC1a-mediated ERS. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155125. [PMID: 37820466 DOI: 10.1016/j.phymed.2023.155125] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 09/15/2023] [Accepted: 09/27/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND Kaempferol is a flavonoid derived from the herb, Kaempferia galanga L., in addition to exhibiting a wide range of pharmacological properties, kaempferol is also an anti-inflammatory, anti-lipid metabolizing, and anti-oxidative stress agent. The underlying molecular mechanisms of its effects on vascular endothelial growth factor (VEGF) secretion and activation of hepatic stellate cells (HSCs) are yet unknown. Activated HSCs induces VEGF release and extracellular matrix (ECM) accumulation which are important factors in hepatic fibrosis. PURPOSE Our aim is to explore how kaempferol may affect hepatic fibrosis and the mechanisms behind its effects. METHODS The in vivo model was Sprague-Dawley rats induced with carbon tetrachloride (CCl4). Histological staining was used to observe histological features of the liver. The levels of (alanine aminotransferase) ALT and (aspartate aminotransferase) AST were detected by the corresponding kits. Platelet-derived growth factor (PDGF) was used to stimulate the HSC-T6 rat hepatic stellate cells. The mechanisms underlying this process were investigated using a variety of molecular approaches, including immunofluorescence, RT-qPCR, and western blotting. Moreover, intracellular Ca2+ were observed by laser confocal microscope. RESULTS It was found that kaempferol significantly reduced the expression of ASIC1a, VEGF, α-SMA and Collagen-I proteins in a model of CCl4-induced hepatic fibrosis in rats. In HSC-T6, kaempferol inhibits activation of HSCs by decreasing expression of ASIC1a, eIF2α, p-eIF2α and ATF-4. Laser confocal fluorescence showed that kaempferol inhibited Ca2+ influx and reduced Ca2+ concentration around the endoplasmic reticulum. Molecular docking and cellular thermal shift assay (CETSA) results further indicated that kaempferol interacted with ASIC1a. We found that kaempferol may promote the degradation of ASIC1a and inhibited ASIC1a- mediated upregulation of ERS. CONCLUSION The data from our in vivo experiments demonstrate that kaempferol effectively attenuates hepatic fibrosis. In vitro studies we further propose a novel mechanism of kaempferol against hepatic fibrosis which can interact with ASIC1a and promote ASIC1a degradation while inhibiting the activation and VEGF release of HSCs by suppressing the ASIC1a-eIF2α-ATF-4 signaling pathway.
Collapse
Affiliation(s)
- Rui Cao
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Chun Cao
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Xiaojie Hu
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Kang Du
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Jingrong Zhang
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Mengxue Li
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Bowen Li
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Huimin Lin
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Anqi Zhang
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Yangyang Li
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Li Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China.
| | - Yan Huang
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
4
|
Macias-Ceja DC, Barrachina MD, Ortiz-Masià D. Autophagy in intestinal fibrosis: relevance in inflammatory bowel disease. Front Pharmacol 2023; 14:1170436. [PMID: 37397491 PMCID: PMC10307973 DOI: 10.3389/fphar.2023.1170436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/06/2023] [Indexed: 07/04/2023] Open
Abstract
Chronic inflammation is often associated with fibrotic disorders in which an excessive deposition of extracellular matrix is a hallmark. Long-term fibrosis starts with tissue hypofunction and finally ends in organ failure. Intestinal fibrosis is not an exception, and it is a frequent complication of inflammatory bowel disease (IBD). Several studies have confirmed the link between deregulated autophagy and fibrosis and the presence of common prognostic markers; indeed, both up- and downregulation of autophagy are presumed to be implicated in the progression of fibrosis. A better knowledge of the role of autophagy in fibrosis may lead to it becoming a potential target of antifibrotic therapy. In this review we explore novel advances in the field that highlight the relevance of autophagy in fibrosis, and give special focus to fibrosis in IBD patients.
Collapse
Affiliation(s)
- Dulce C. Macias-Ceja
- Departamento de Farmacología and CIBER, Facultad de Medicina y Odontología, Universitat de Valencia, Valencia, Spain
| | - María D. Barrachina
- Departamento de Farmacología and CIBER, Facultad de Medicina y Odontología, Universitat de Valencia, Valencia, Spain
| | - Dolores Ortiz-Masià
- Departamento de Farmacología and CIBER, Facultad de Medicina y Odontología, Universitat de Valencia, Valencia, Spain
- Departamento de Medicina, Facultad de Medicina y Odontología, Universitat de Valencia, Valencia, Spain
| |
Collapse
|
5
|
Petta S. Glycemic control, body weight reduction, or glucose-lowering therapy in diabetic patients: What is better for the liver? Nutr Metab Cardiovasc Dis 2023; 33:649-650. [PMID: 36710108 DOI: 10.1016/j.numecd.2022.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 12/28/2022] [Indexed: 01/07/2023]
Affiliation(s)
- Salvatore Petta
- Section of Gastroenterology and Hepatology, PROMISE, University of Palermo, Italy.
| |
Collapse
|
6
|
Colosimo S, Tan GD, Petroni ML, Marchesini G, Tomlinson JW. Improved glycaemic control in patients with type 2 diabetes has a beneficial impact on NAFLD, independent of change in BMI or glucose lowering agent. Nutr Metab Cardiovasc Dis 2023; 33:640-648. [PMID: 36710114 DOI: 10.1016/j.numecd.2022.12.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIM The current focus of the treatment of Non-Alcoholic Fatty Liver Disease (NAFLD) is lifestyle intervention with the aim of significant weight loss alongside aggressive cardiovascular risk reduction. NAFLD is tightly associated with type 2 diabetes (T2D) and obesity. In patients with T2D, glucose lowering agents that promote weight loss have shown a beneficial impact on NAFLD. However, it remains unclear as to whether glucose lowering can improve NALFD in patients with T2D, independent of weight loss. METHODS AND RESULTS In a retrospective analysis of data from 637 people with T2D, we examined the longitudinal impact of optimizing glycaemic control with DPP-IV inhibitors, GLP-1RAs and SGLT2 inhibitors on Fatty liver index (FLI) and Fibrosis score 4 (Fib-4) adjusting for changes in BMI and choice of glucose lowering regimen over a 12-month period. Multiple linear regression analysis demonstrated a significant correlation between the change in glycated haemoglobin and change in FLI after adjustment for change in BMI, age, sex, and drug class (R = 0.467, p = 0.031). The greatest reduction in FLI was observed in patients with the largest reduction in glycated haemoglobin (p < 0.0001). The probability of improvements in FLI with optimization of glycaemic control was similar with all 3 glucose lowering agents, despite differences in weight reduction. Similar relationships were observed examining the changes in glycaemic control and Fib-4. CONCLUSIONS Improvements in glucose control that are independent of weight loss are associated with improvement in NAFLD and should form an integral part of the management patients with co-existent NAFLD and T2D.
Collapse
Affiliation(s)
- Santo Colosimo
- School of Nutrition Science, University of Milan, Milan, Italy; Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Garry D Tan
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | | | - Giulio Marchesini
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK.
| |
Collapse
|
7
|
ASIC1a induces mitochondrial apoptotic responses in acute lung injury. Eur J Pharmacol 2022; 934:175296. [PMID: 36162458 DOI: 10.1016/j.ejphar.2022.175296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/19/2022] [Accepted: 09/19/2022] [Indexed: 11/20/2022]
Abstract
AIM This study aimed to investigate the promoting effect of acid-sensing ion channel 1a (ASIC1a) on lipopolysaccharide (LPS)-induced acute lung injury (ALI) and its mechanisms. METHODS In this experiment, the ALI rat model was induced by intratracheal injection of LPS, and the ASIC1a specific blocker psalmotoxin-1 (PcTx-1) was injected into the tail vein before LPS administration once. Western blot, immunofluorescence, immunohistochemistry and real-time PCR methods were used to detect ASIC1a and apoptosis-related proteins expressions in lung tissue and RLE-6TN rat type II alveolar epithelial cells. Confocal Laser Scanning Microscopy was used to detect Ca2+ fluorescence intensity in RLE-6TN cells. RESULTS PcTx-1 pretreatment not only inhibited the pathological changes of LPS-induced ALI in lung tissue, but also inhibited lung dysfunction. PcTx-1 also reduced the increased levels of the apoptosis-related proteins B-cell lymphoma-2-associated X (Bax) and cleaved cysteinyl aspartate specific proteinase 3 (Cleaved caspase-3) and increased the decreased level of B-cell lymphoma-2 (Bcl-2) in the lung tissue of the model group. LPS-induced changes in mitochondrial membrane potential and calcium influx in alveolar epithelial cells were also reversed by PcTx-1. CONCLUSION ASIC1a induces an apoptotic response in ALI through mitochondrial apoptosis.
Collapse
|
8
|
Su SB, Tao L, Liang XL, Chen W. Long noncoding RNA GAS5 inhibits LX-2 cells activation by suppressing NF-κB signalling through regulation of the miR-433-3p/TLR10 axis. Dig Liver Dis 2022; 54:1066-1075. [PMID: 34903500 DOI: 10.1016/j.dld.2021.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/31/2021] [Accepted: 11/02/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Liver fibrosis is a common disease that can lead to hepatic failure. AIMS Our aims were to reveal the role of GAS5 in the regulation of liver fibrosis. METHODS LX-2 human hepatic satellite cells (HSCs) were cultured and activated using TGF-β1 treatment. A CCK-8 assay was performed to assess cell viability. A luciferase assay was employed to monitor the interactions between miR-433-3p and GAS5 or toll-like receptor 10 (TLR10). Western blotting and real-time quantitative PCR (RT-qPCR) were applied to detect the expression levels of α-SMA, Col. I, PCNA-, MMP2-, MMP9-, TLR10-, and NF-κB-related molecules at the protein and RNA levels. RESULTS GAS5 and TLR10 were decreased while miR-433-3p was upregulated in TGF-β1-activated LX-2 cells. Upregulation of GAS5 or downregulation of miR-433-3p suppressed HSC activation, and luciferase assays indicated that miR-433-3p binds with GAS5 and the 3'-UTR of TLR10. MiR-433-3p upregulation and TLR10 downregulation rescued the impacts of GAS5 overexpression or miR-433-3p knockdown on LX-2 cells. Upregulation of GAS5 also suppressed the phosphorylation of NF-κB through the miR-433-3p/TLR10 axis. CONCLUSION LncRNA GAS5 exerts an inhibitory effect on HSC activation by suppressing NF-κB signalling through regulation of the miR-433-3p/TLR10 axis.
Collapse
Affiliation(s)
- Si-Biao Su
- Department of Gastroenterology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, P.R. China.
| | - Lin Tao
- Department of Gastroenterology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, P.R. China
| | - Xiao-Le Liang
- Basic Medical College, Guangxi Medical University, Nanning, 530021, Guangxi Province, P.R. China
| | - Wen Chen
- Department of Teaching Affairs, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, P.R. China
| |
Collapse
|
9
|
Zhang Y, Liang J, Cao N, Gao J, Xie Y, Zhou S, Tang X. ASIC1α up-regulates MMP-2/9 expression to enhance mobility and proliferation of liver cancer cells via the PI3K/AKT/mTOR pathway. BMC Cancer 2022; 22:778. [PMID: 35840921 PMCID: PMC9287982 DOI: 10.1186/s12885-022-09874-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/24/2022] [Indexed: 11/28/2022] Open
Abstract
A major challenge in the treatment of liver cancer is that a large proportion of patients fail to achieve long-term disease control, with death from liver cancer cell migration and invasion. Acid-sensitive ion channel 1α (ASIC1α) is involved in the migration, invasion, and proliferation of liver cancer cells. Therefore, we explored the mechanism of ASIC1α-mediated liver cancer cell migration and invasion. We determined the levels of ASIC1α by western blotting and immunofluorescence in HepG2 and SK-Hep1 cells cultured in various acidic conditions. In addition, wound healing assay, transwell invasion assay, and MTT assay were conducted to assess the migration, invasion, and proliferation abilities of liver cancer cells. Western blotting was conducted to determine the levels of MMP2, MMP9, ASIC1α, p-PI3Kp85, t-PI3Kp85, p-AKT(Ser473), t-AKT, p-mTOR (Ser2448), t-mTOR. We first found that the levels of ASIC1α in the HepG2 and SK-Hep1 cells in acidic conditions (pH 6.5) were significantly increased. Inhibition and knockdown of ASIC1α down-regulated MMP-2/9 expression and inhibited the migration, invasion, and proliferation of HepG2 and SK-Hep1 cells; overexpression of ASIC1α had the opposite effect. We further demonstrated that ASIC1α up-regulates MMP-2/9 via activation of the PI3K/AKT/mTOR pathway, thereby promoting migration, invasion, and proliferation of liver cancer cells. Overexpression of MMP-2/9 and activation of AKT reversed these effects on liver cancer cells caused by inhibition of ASIC1α. We conclude that ASIC1α can regulate migration, invasion, and proliferation of liver cancer cells through the MMP-2/9/PI3K/AKT/mTOR pathway. These observations may provide a new reference for liver cancer chemotherapy.
Collapse
Affiliation(s)
- Yinci Zhang
- Medcial School, Anhui University of Science & Technology, Huainan, 232001, China.,Institute of Environment-Friendly Materials and Occupational Health of Anhui, University of Science and Technology, Wuhu, 241003, China
| | - Jiaojiao Liang
- Medcial School, Anhui University of Science & Technology, Huainan, 232001, China.,Institute of Environment-Friendly Materials and Occupational Health of Anhui, University of Science and Technology, Wuhu, 241003, China
| | - Niandie Cao
- Medcial School, Anhui University of Science & Technology, Huainan, 232001, China.,Institute of Environment-Friendly Materials and Occupational Health of Anhui, University of Science and Technology, Wuhu, 241003, China
| | - Jiafeng Gao
- Medcial School, Anhui University of Science & Technology, Huainan, 232001, China.,Institute of Environment-Friendly Materials and Occupational Health of Anhui, University of Science and Technology, Wuhu, 241003, China
| | - Yinghai Xie
- Medcial School, Anhui University of Science & Technology, Huainan, 232001, China.,First Affiliated Hospital, Anhui University of Science & Technology, Huainan, 232001, China
| | - Shuping Zhou
- Medcial School, Anhui University of Science & Technology, Huainan, 232001, China.,First Affiliated Hospital, Anhui University of Science & Technology, Huainan, 232001, China
| | - Xiaolong Tang
- Medcial School, Anhui University of Science & Technology, Huainan, 232001, China. .,Institute of Environment-Friendly Materials and Occupational Health of Anhui, University of Science and Technology, Wuhu, 241003, China.
| |
Collapse
|
10
|
Luan SH, Yang YQ, Ye MP, Liu H, Rao QF, Kong JL, Wu FR. ASIC1a promotes hepatic stellate cell activation through the exosomal miR-301a-3p/BTG1 pathway. Int J Biol Macromol 2022; 211:128-139. [PMID: 35561854 DOI: 10.1016/j.ijbiomac.2022.05.041] [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: 12/27/2021] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 12/11/2022]
Abstract
Activation of hepatic stellate cells (HSCs) is a key cause of liver fibrosis. However, the mechanisms leading to the activation of HSCs are not fully understood. In the pathological process, acid-sensing ion channel 1a (ASIC1a) is widely involved in the development of inflammatory diseases, suggesting that ASIC1a may play an important role in liver fibrosis. We found that in an acidic environment, ASIC1a leads to HSC-T6 cell activation. Meanwhile, exosomes produced by activated HSC-T6 cells (HSC-EXOs) can be reabsorbed by quiescent HSC-T6 cells to promote their activation. Exosomes mainly carry miRNAs involved in intercellular information exchange. We performed exosome miRNA whole transcriptome sequencing. The results indicated that the acidic environment could alter the miRNA expression profile in the exosomes of HSC-T6 cells. Further studies revealed that ASIC1a promotes the activation of HSCs by regulating miR-301a-3p targeting B-cell translocation gene 1 (BTG1). In conclusion, our study found that ASIC1a may affect HSC activation through the exosomal miR-301a-3p/BTG1 axis, and inhibiting ASIC1a may be a promising treatment strategy for liver fibrosis.
Collapse
Affiliation(s)
- Shao-Hua Luan
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | | | - Man-Ping Ye
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Hui Liu
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Qiu-Fan Rao
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Jin-Ling Kong
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Fan-Rong Wu
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China.
| |
Collapse
|
11
|
Jin J, Yang H, Hu L, Wang Y, Wu W, Hu C, Wu K, Wu Z, Cheng W, Huang Y. Inonotsuoxide B suppresses hepatic stellate cell activation and proliferation via the PI3K/AKT and ERK1/2 pathway. Exp Ther Med 2022; 23:417. [PMID: 35601068 DOI: 10.3892/etm.2022.11344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 08/04/2020] [Indexed: 11/05/2022] Open
Affiliation(s)
- Juan Jin
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230032, P.R. China
| | - Hui Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230032, P.R. China
| | - Lili Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230032, P.R. China
| | - Yinghong Wang
- Department of Pharmacy, Division of Life Sciences and Medicine, Anhui Provincial Cancer Hospital, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui 230001, P.R. China
| | - Wenyong Wu
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, Hefei, Anhui 230041, P.R. China
| | - Chengmu Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230032, P.R. China
| | - Kun Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230032, P.R. China
| | - Zehua Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230032, P.R. China
| | - Wenming Cheng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230032, P.R. China
| | - Yan Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
12
|
Mastoridou EM, Goussia AC, Glantzounis GK, Kanavaros P, Charchanti AV. Autophagy and Exosomes: Cross-Regulated Pathways Playing Major Roles in Hepatic Stellate Cells Activation and Liver Fibrosis. Front Physiol 2022; 12:801340. [PMID: 35185602 PMCID: PMC8850693 DOI: 10.3389/fphys.2021.801340] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic liver injury, regardless of the underlying disease, results in gradual alteration of the physiological hepatic architecture and in excessive production of extracellular matrix, eventually leading to cirrhosis Liver cellular architecture consists of different cell populations, among which hepatic stellate cells (HSCs) have been found to play a major role in the fibrotic process. Under normal conditions, HSCs serve as the main storage site for vitamin A, however, pathological stimuli lead to their transdifferentiation into myofibroblast cells, with autophagy being the key regulator of their activation, through lipophagy of their lipid droplets. Nevertheless, the role of autophagy in liver fibrosis is multifaceted, as increased autophagic levels have been associated with alleviation of the fibrotic process. In addition, it has been found that HSCs receive paracrine stimuli from neighboring cells, such as injured hepatocytes, Kupffer cells, sinusoidal endothelial cells, which promote liver fibrosis. These stimuli have been found to be transmitted via exosomes, which are incorporated by HSCs and can either be degraded through lysosomes or be secreted back into the extracellular space via fusion with the plasma membrane. Furthermore, it has been demonstrated that autophagy and exosomes may be concomitantly or reciprocally regulated, depending on the cellular conditions. Given that increased levels of autophagy are required to activate HSCs, it is important to investigate whether autophagy levels decrease at later stages of hepatic stellate cell activation, leading to increased release of exosomes and further propagation of hepatic fibrosis.
Collapse
Affiliation(s)
- Eleftheria M. Mastoridou
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Anna C. Goussia
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Georgios K. Glantzounis
- Hepato-Pancreatico-Biliary Unit, Department of Surgery, University General Hospital of Ioannina and School of Medicine, University of Ioannina, Ioannina, Greece
| | - Panagiotis Kanavaros
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Antonia V. Charchanti
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
- *Correspondence: Antonia V. Charchanti,
| |
Collapse
|
13
|
Gu Z, Fang L, Ma P. The angiotensin-converting enzyme inhibitor, captopril, suppressed hepatic stellate cell activation via NF-kappaB or wnt3α/β-catenin pathway. Bioengineered 2021; 12:8370-8377. [PMID: 34607529 PMCID: PMC8806896 DOI: 10.1080/21655979.2021.1987091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Activation of hepatic stellate cells (HSC) is associated with hepatic fibrogenesis, which is one of complications of diabetes mellitus. Captopril possesses potent anti-inflammation, oxidative stress and fibrosis effects. However, the specific molecular mechanism of captopril in high glucose (HG)-induced hepatic stellate cells has not been elucidated. Following the treatment of HG or captopril treatment for rat hepatic stellate cells (HSC-T6), cell activities were detected by Cell Counting Kit-8 (CCK8) assay. Reactive oxygen species (ROS) levels were determined by ROS staining. The expression of inflammation-related proteins (Interleukin (IL)-1β, IL-6 and IL-8) and fibrosis-related proteins (fibronectin (FN), collagen I, collagen III, collagen IV, matrix metallopeptidase (MMP-2 and MMP-9) were determined by Western blot. Captopril significantly decreased HSC-T6 cell viability induced by HG in a dose-dependent manner, as well as decreased levels of malondialdehyde (MDA), ROS, pro-inflammatory markers and fibrosis-related proteins, while upregulated superoxide dismutase (SOD) activities. We further found that captopril decreased the ratio of p-IκBα/IκBα and the ratio of p-p65/p65. Intriguing, phorbol myristate acetate (PMA) or LiCl was able to significantly reverse the captopril-induced alteration of oxidative stress-, inflammation- and fibrosis-marker levels. In conclusion, in HG-stimulated HSC-T6 cells, captopril displayed a potent ability to inhibit oxidative stress, inflammation and hepatic fibrogenesis via NF-kappaB or wnt3α/β-catenin. These results demonstrated the mechanism of captopril as well as the role of the NF-kappaB or wnt3α/β-catenin on HSC-T6 activation induced by HG.
Collapse
Affiliation(s)
- Zhaodi Gu
- Internal Medicine Department, Shaoxing Yuecheng People's Hospital, Shaoxing City, Zhejiang Province, China
| | - Linjun Fang
- Internal Medicine Department, Shaoxing Yuecheng People's Hospital, Shaoxing City, Zhejiang Province, China
| | - Peijun Ma
- Internal Medicine Department, Shaoxing Yuecheng People's Hospital, Shaoxing City, Zhejiang Province, China
| |
Collapse
|
14
|
Salinas Castellanos LC, Uchitel OD, Weissmann C. Signaling Pathways in Proton and Non-proton ASIC1a Activation. Front Cell Neurosci 2021; 15:735414. [PMID: 34675777 PMCID: PMC8523820 DOI: 10.3389/fncel.2021.735414] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/09/2021] [Indexed: 11/13/2022] Open
Abstract
Acid-sensing ion channels (ASICs) regulate synaptic activities and play important roles in neurodegenerative diseases as well as pain conditions. Classically, ASICs are described as transiently activated by a reduced pH, followed by desensitization; the activation allows sodium influx, and in the case of ASIC1a-composed channels, also calcium to some degree. Several factors are emerging and extensively analyzed as modulators, activating, inhibiting, and potentiating specific channel subunits. However, the signaling pathways triggered by channel activation are only starting to be revealed.The channel has been recently shown to be activated through a mechanism other than proton-mediated. Indeed, the large extracellular loop of these channels opens the possibility that other non-proton ligands might exist. One such molecule discovered was a toxin present in the Texas coral snake venom. The finding was associated with the activation of the channel at neutral pH via the toxin and causing intense and unremitting pain.By using different pharmacological tools, we analyzed the downstream signaling pathway triggered either by the proton and non-proton activation for human, mouse, and rat ASIC1a-composed channels in in vitro models. We show that for all species analyzed, the non-protonic mode of activation determines the activation of the ERK signaling cascade at a higher level and duration compared to the proton mode.This study adds to the growing evidence of the important role ASIC1a channels play in different physiological and pathological conditions and also hints at a possible pathological mechanism for a sustained effect.
Collapse
Affiliation(s)
| | | | - Carina Weissmann
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE—UBA CONICET), Facultad de Ciencias, Exactas y Naturales de la Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
15
|
Tao J, Lu Z, Su J, Qian X, Zhang Y, Xu Y, Song S, Hang X, Peng X, Chen F. ASIC1a promotes the proliferation of synovial fibroblasts via the ERK/MAPK pathway. J Transl Med 2021; 101:1353-1362. [PMID: 34282280 DOI: 10.1038/s41374-021-00636-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 06/21/2021] [Accepted: 06/21/2021] [Indexed: 12/15/2022] Open
Abstract
Synovial hyperplasia, a profound alteration in the structure of synovial tissue, is the basis for cumulative joint destruction in rheumatoid arthritis (RA). It is generally accepted that controlling synovial hyperplasia can delay the progression of RA. As one of the most intensively studied isoforms of acid-sensing ion channels (ASICs), ASIC1a contributes to various physiopathologic conditions, including RA, due to its unique property of being permeable to Ca2+. However, the role and the regulatory mechanisms of ASIC1a in synovial hyperplasia are poorly understood. Here, rats induced with adjuvant arthritis (AA) and human primary synovial fibroblasts were used in vivo and in vitro to investigate the role of ASIC1a in the proliferation of RA synovial fibroblasts (RASFs). The results show that the expression of ASIC1a was significantly increased in synovial tissues and RASFs obtained from patients with RA as well as in the synovium of rats with AA. Moreover, extracellular acidification improved the ability of RASFs colony formation and increased the expression of proliferation cell nuclear antigen (PCNA) and Ki67, which was abrogated by the specific ASIC1a inhibitor psalmotoxin-1 (PcTX-1) or ASIC1a-short hairpin RNA (ASIC1a-shRNA), suggesting that extracellular acidification promotes the proliferation of RASFs by activating ASIC1a. In addition, the activation of c-Raf and extracellular signal-regulated protein kinases (ERKs) signaling was blocked with PcTX-1 or ASIC1a-shRNA and the proliferation of RASFs was further inhibited by the ERK inhibitor (U0126), indicating that ERK/MAPK signaling contributes to the proliferation process of RASFs promoted by the activation of ASIC1a. These findings gave us an insight into the role of ASIC1a in the proliferation of RASFs, which may provide solid foundation for ASIC1a as a potential target in the treatment of RA.
Collapse
Affiliation(s)
- Jingjing Tao
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Zheng Lu
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Jingwen Su
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Xuewen Qian
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Yihao Zhang
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Yayun Xu
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Sujing Song
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Xiaoyu Hang
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Xiaoqing Peng
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Feihu Chen
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China.
| |
Collapse
|
16
|
Wang Y, Zhang J, Jiang P, Li K, Sun Y, Huang Y. ASIC1a promotes acidic microenvironment-induced HCC cells migration and invasion by inducing autophagy. Eur J Pharmacol 2021; 907:174252. [PMID: 34116040 DOI: 10.1016/j.ejphar.2021.174252] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/07/2021] [Accepted: 06/07/2021] [Indexed: 01/10/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common types of liver cancer with high incidence and metastatic rate. Recent studies have shown that the high metastasis of HCC is closely related to the acidic microenvironment of HCC cells. Acid-sensing ion Channel 1a (ASIC1a) plays an important role in HCC development, which can mediate tumor cell migration and invasion. However, the underlying mechanism of how ASIC1a promotes HCC cell migration and invasion in acidic microenvironments remains unclear, while autophagy may act as a mechanism for tumor cells to adapt to acidic microenvironment. Therefore, this study aims to investigate whether ASIC1a mediates autophagy and its effects on the migration and invasion of HCC cells. Interestingly, our study has shown that ASIC1a and autophagy were increased in HepG2 cells in acidic microenvironment, and both of them can promote HCC cells migration and invasion. Moreover, inhibition of ASIC1a with PcTx1 or ASIC1a ShRNA reduced the autophagy flux. Collectively, ASIC1a can promote acidic microenvironment-induced HepG2 cells migration and invasion by inducing autophagy, which may be correlated with Ca2+ influx.
Collapse
Affiliation(s)
- Yinghong Wang
- Department of Pharmacy, Anhui Provincial Cancer Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China
| | - Jin Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Peng Jiang
- Department of Pharmacy, Anhui Provincial Cancer Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China
| | - Kai Li
- Department of Pharmacy, Anhui Provincial Cancer Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China
| | - Yancai Sun
- Department of Pharmacy, Anhui Provincial Cancer Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China.
| | - Yan Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, 230032, China.
| |
Collapse
|
17
|
Jia X, Zhang A, Li Z, Peng X, Tian X, Gao F. Activation of spinal PDGFRβ in microglia promotes neuronal autophagy via p38 MAPK pathway in morphine-tolerant rats. J Neurochem 2021; 158:373-390. [PMID: 33950542 DOI: 10.1111/jnc.15383] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/29/2021] [Accepted: 05/01/2021] [Indexed: 12/14/2022]
Abstract
The adverse side effects of opioids, especially antinociceptive tolerance, limit their clinical application. A recent study reported that platelet-derived growth factor receptor β (PDGFRβ) blockage selectively inhibited morphine tolerance. Autophagy has been reported to contribute to the cellular and behavioral responses to morphine. However, little is known about the relationship between PDGFRβ and autophagy in the mechanisms of morphine tolerance. In this study, rats were intrathecally administered with morphine twice daily for 7 days to induce antinociceptive tolerance, which was evaluated using a tail-flick latency test. By administration autophagy inhibitor 3-Methyladenine, PDGFRβ inhibitor imatinib, p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580 hydrochloride and minocycline hydrochloride, western blot, immunofluorescence, and transmission electron microscopy techniques were used to elucidate the roles of PDGFRβ, autophagy, and related signaling pathways in morphine tolerance. This study demonstrated for the first time that spinal PDGFRβ in microglia promotes autophagy in gamma-aminobutyric acid (GABA) interneurons through activating p38 MAPK pathway during the development of morphine tolerance, which suggest a potential strategy for preventing the development of morphine tolerance clinically, thereby improving the use of opioids in pain management.
Collapse
Affiliation(s)
- Xiaoqian Jia
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anqi Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Li
- Department of Anesthesiology, Shenzhen Second People's Hospital, Shenzhen, China
| | - Xiaoling Peng
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuebi Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Gao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
18
|
Lucantoni F, Martínez-Cerezuela A, Gruevska A, Moragrega ÁB, Víctor VM, Esplugues JV, Blas-García A, Apostolova N. Understanding the implication of autophagy in the activation of hepatic stellate cells in liver fibrosis: are we there yet? J Pathol 2021; 254:216-228. [PMID: 33834482 DOI: 10.1002/path.5678] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/29/2021] [Accepted: 04/07/2021] [Indexed: 01/18/2023]
Abstract
Liver fibrosis (LF) occurs as a result of persistent liver injury and can be defined as a pathologic, chronic, wound-healing process in which functional parenchyma is progressively replaced by fibrotic tissue. As a phenomenon involved in the majority of chronic liver diseases, and therefore prevalent, it exerts a significant impact on public health. This impact becomes even more patent given the lack of a specific pharmacological therapy, with LF only being ameliorated or prevented through the use of agents that alleviate the underlying causes. Hepatic stellate cells (HSCs) are fundamental mediators of LF, which, activated in response to pro-fibrotic stimuli, transdifferentiate from a quiescent phenotype into myofibroblasts that deposit large amounts of fibrotic tissue and mediate pro-inflammatory effects. In recent years, much effort has been devoted to understanding the mechanisms through which HSCs are activated or inactivated. Using cell culture and/or different animal models, numerous studies have shown that autophagy is enhanced during the fibrogenic process and have provided specific evidence to pinpoint the fundamental role of autophagy in HSC activation. This effect involves - though may not be limited to - the autophagic degradation of lipid droplets. Several hepatoprotective agents have been shown to reverse the autophagic alteration present in LF, but clinical confirmation of these effects is pending. On the other hand, there is evidence that implicates autophagy in several anti-fibrotic mechanisms in HSCs that stimulate HSC cell cycle arrest and cell death or prevent the generation of pro-fibrotic mediators, including excess collagen accumulation. The objective of this review is to offer a comprehensive analysis of published evidence of the role of autophagy in HSC activation and to provide hints for possible therapeutic targets for the treatment and/or prevention of LF related to autophagy. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Federico Lucantoni
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
- FISABIO - Hospital Universitario Doctor Peset, Valencia, Spain
| | | | - Aleksandra Gruevska
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
- FISABIO - Hospital Universitario Doctor Peset, Valencia, Spain
| | - Ángela B Moragrega
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
- FISABIO - Hospital Universitario Doctor Peset, Valencia, Spain
| | - Víctor M Víctor
- FISABIO - Hospital Universitario Doctor Peset, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Valencia, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Juan V Esplugues
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
- FISABIO - Hospital Universitario Doctor Peset, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Valencia, Spain
| | - Ana Blas-García
- FISABIO - Hospital Universitario Doctor Peset, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Valencia, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Nadezda Apostolova
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
- FISABIO - Hospital Universitario Doctor Peset, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Valencia, Spain
| |
Collapse
|
19
|
Pan X, Zhu Y, Wu X, Liu L, Ying R, Wang L, Du N, Zhang J, Jin J, Meng X, Dai F, Huang Y. The interaction of ASIC1a and ERS mediates nerve cell apoptosis induced by insulin deficiency. Eur J Pharmacol 2020; 893:173816. [PMID: 33345857 DOI: 10.1016/j.ejphar.2020.173816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 10/22/2022]
Abstract
Diabetes-related brain complications are the most serious complications of terminal diabetes. The increasing evidence have showed that the predisposing factor is not only hyperglycemia, but also insulin deficiency. In this study, we demonstrated that insulin deficiency was involved in the apoptosis of nerve cells, and it was related to the interaction between acid-sensitive ion channel 1a (ASIC1a) and endoplasmic reticulum stress (ERS). By silencing C/EBP homologous protein (CHOP) and ASIC1a, the pro-apoptotic effect of insulin deficiency on NS20y cells was relieved. Further research found that the binding of CHOP and C/EBPα was increased in the nucleus of cells cultured without insulin, and C/EBPα was competitively inhibited as a negative regulator of ASIC1a, which further increased the ERS and lead to neuronal apoptosis. In summary, ERS and ASIC1a play an important role in neurological damage caused by insulin deficiency. Our finding may lead to new ideas and treatment of diabetes-related brain complications.
Collapse
Affiliation(s)
- Xuesheng Pan
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Yueqin Zhu
- Department of Pharmacy, West Branch of the First Affiliated Hospital of University of Science and Technology of China (Anhui Provincial Cancer Hospital), Hefei, 230031, China
| | - Xian Wu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Lan Liu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China; Department of Pharmacy, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
| | - Ruixue Ying
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Lili Wang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Na Du
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Jin Zhang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Juan Jin
- Department of Pharmacology, School of Basic Medicine, Anhui Medical University. Hefei, 230032, China
| | - Xiaoming Meng
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Fang Dai
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| | - Yan Huang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China.
| |
Collapse
|
20
|
Zhu Y, Pan X, Du N, Li K, Hu Y, Wang L, Zhang J, Liu Y, Zuo L, Meng X, Hu C, Wu X, Jin J, Wu W, Chen X, Wu F, Huang Y. ASIC1a regulates miR‐350/SPRY2 by N
6
‐methyladenosine to promote liver fibrosis. FASEB J 2020; 34:14371-14388. [DOI: 10.1096/fj.202001337r] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/03/2020] [Accepted: 08/11/2020] [Indexed: 01/05/2023]
Affiliation(s)
- Yueqin Zhu
- Anhui Provincial laboratory of inflammatory and immunity disease Anhui Institute of Innovative Drugs School of Pharmacy Anhui Medical University, 230032, China Hefei230032China
| | - Xuesheng Pan
- Anhui Provincial laboratory of inflammatory and immunity disease Anhui Institute of Innovative Drugs School of Pharmacy Anhui Medical University, 230032, China Hefei230032China
| | - Na Du
- Anhui Provincial laboratory of inflammatory and immunity disease Anhui Institute of Innovative Drugs School of Pharmacy Anhui Medical University, 230032, China Hefei230032China
| | - Kuayue Li
- Anhui Provincial laboratory of inflammatory and immunity disease Anhui Institute of Innovative Drugs School of Pharmacy Anhui Medical University, 230032, China Hefei230032China
| | - Yamin Hu
- Anhui Provincial laboratory of inflammatory and immunity disease Anhui Institute of Innovative Drugs School of Pharmacy Anhui Medical University, 230032, China Hefei230032China
| | - Lili Wang
- Anhui Provincial laboratory of inflammatory and immunity disease Anhui Institute of Innovative Drugs School of Pharmacy Anhui Medical University, 230032, China Hefei230032China
| | - Jin Zhang
- Anhui Provincial laboratory of inflammatory and immunity disease Anhui Institute of Innovative Drugs School of Pharmacy Anhui Medical University, 230032, China Hefei230032China
| | - Yanyi Liu
- Anhui Provincial laboratory of inflammatory and immunity disease Anhui Institute of Innovative Drugs School of Pharmacy Anhui Medical University, 230032, China Hefei230032China
| | - Longquan Zuo
- Department of Pharmacy Hospital of Armed Police of Anhui Province Hefei230041China
| | - Xiaoming Meng
- Anhui Provincial laboratory of inflammatory and immunity disease Anhui Institute of Innovative Drugs School of Pharmacy Anhui Medical University, 230032, China Hefei230032China
| | - Chengmu Hu
- Anhui Provincial laboratory of inflammatory and immunity disease Anhui Institute of Innovative Drugs School of Pharmacy Anhui Medical University, 230032, China Hefei230032China
| | - xian Wu
- Anhui Provincial laboratory of inflammatory and immunity disease Anhui Institute of Innovative Drugs School of Pharmacy Anhui Medical University, 230032, China Hefei230032China
| | - Juan Jin
- Department of Pharmacology, School of Basic Medical Sciences Anhui Medical University Hefei230032China
| | - Wenyong Wu
- 4Department of General Surgery First Affiliated Hospital of Anhui Medical University Hefei230022China
| | - Xiangtao Chen
- Anhui Provincial laboratory of inflammatory and immunity disease Anhui Institute of Innovative Drugs School of Pharmacy Anhui Medical University, 230032, China Hefei230032China
| | - Fanrong Wu
- Anhui Provincial laboratory of inflammatory and immunity disease Anhui Institute of Innovative Drugs School of Pharmacy Anhui Medical University, 230032, China Hefei230032China
| | - Yan Huang
- Anhui Provincial laboratory of inflammatory and immunity disease Anhui Institute of Innovative Drugs School of Pharmacy Anhui Medical University, 230032, China Hefei230032China
| |
Collapse
|
21
|
Li Y, Liu R, Wu J, Li X. Self-eating: friend or foe? The emerging role of autophagy in fibrotic diseases. Am J Cancer Res 2020; 10:7993-8017. [PMID: 32724454 PMCID: PMC7381749 DOI: 10.7150/thno.47826] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/16/2020] [Indexed: 01/18/2023] Open
Abstract
Fibrosis occurs in most human organs including the liver, lung, heart and kidney, and is crucial for the progression of most chronic diseases. As an indispensable catabolic process for intracellular quality control and homeostasis, autophagy occurs in most mammalian cells and is implicated in many biological processes including fibrogenesis. Although advances have been made in understanding autophagy process, the potential role of autophagy in fibrotic diseases remains controversial and has recently attracted a great deal of attention. In the current review, we summarize the commonalities of autophagy affecting different types of fibrosis in different organs, including the liver, lung, heart, and kidney as well as in cystic fibrosis, systematically outline the contradictory results and highlight the distinct role of autophagy during the various stages of fibrosis. In summary, the exact role autophagy plays in fibrogenesis depends on specific cell types and different stimuli, and identifying and evaluating the pathogenic contribution of autophagy in fibrogenesis will promote the discovery of novel therapeutic strategies for the clinical management of these fibrotic diseases.
Collapse
|
22
|
Fujii H, Kawada N. The Role of Insulin Resistance and Diabetes in Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2020; 21:ijms21113863. [PMID: 32485838 PMCID: PMC7312931 DOI: 10.3390/ijms21113863] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/22/2020] [Accepted: 05/24/2020] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) consists of the entire spectrum of fatty liver disease in patients without significant alcohol consumption, ranging from nonalcoholic fatty liver (NAFL) to nonalcoholic steatohepatitis (NASH) to cirrhosis, with NASH recently shown as an important cause of hepatocellular carcinoma (HCC). There is a close relationship between insulin resistance (IR) and NAFLD, with a five-fold higher prevalence of NAFLD in patients with type 2 diabetes (T2DM) compared to that in patients without T2DM. IR is involved in the progression of disease conditions such as steatosis and NASH, as well as hepatic fibrosis progression. The mechanisms underlying these processes involve genetic factors, hepatic fat accumulation, alterations in energy metabolism, and inflammatory signals derived from various cell types including immune cells. In NASH-associated fibrosis, the principal cell type responsible for extracellular matrix production is the hepatic stellate cell (HSC). HSC activation by IR involves “direct” and “indirect” pathways. This review will describe the molecular mechanisms of inflammation and hepatic fibrosis in IR, the relationship between T2DM and hepatic fibrosis, and the relationship between T2DM and HCC in patients with NAFLD.
Collapse
Affiliation(s)
- Hideki Fujii
- Department of Premier Preventive Medicine, Graduate School of Medicine, Osaka City University, Osaka 545-8585, Japan;
| | - Norifumi Kawada
- Department of Hepatology, Graduate School of Medicine, Osaka City University, Osaka 545-8585, Japan
- Correspondence: ; Tel.: +81-6-6645-3897
| | | |
Collapse
|
23
|
Wang W, Zhang F, Yan X, Tan Q. Wnt7a regulates high autophagic and inflammatory response of epidermis in high-glucose environment. Burns 2020; 46:121-127. [DOI: 10.1016/j.burns.2019.07.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/23/2019] [Accepted: 07/18/2019] [Indexed: 12/23/2022]
|
24
|
Zhang SL, Ma L, Zhao J, You SP, Ma XT, Ye XY, Liu T. The Phenylethanol Glycoside Liposome Inhibits PDGF-Induced HSC Activation via Regulation of the FAK/PI3K/Akt Signaling Pathway. Molecules 2019; 24:E3282. [PMID: 31505837 PMCID: PMC6766902 DOI: 10.3390/molecules24183282] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/04/2019] [Accepted: 09/06/2019] [Indexed: 01/30/2023] Open
Abstract
Cistanche tubulosa is a traditional Chinese herbal medicine that is widely used to regulate immunity, and phenylethanol glycosides (CPhGs) are among the primary components responsible for this activity. However, the application of CPhGs is negatively affected by their poor absorption and low oral utilization. Targeted drug delivery is an important development direction for pharmaceutics. Previous studies have indicated that CPhGs could block the conduction of the signaling pathways in TGF-β1/smad and inhibit the activation of hepatic stellate cells (HSCs). The aim of this study was to evaluate the anti-hepatic fibrosis effect of CPhG liposomes by inhibiting HSC activation, promoting apoptosis, blocking the cell cycle, suppressing the conduction of signaling pathways in focal adhesion kinase(FAK)/phosphatidylinositol-3-kinase(PI3K)/protein kinase B(Akt), and determining their in vitro hepatoprotective activity. In vitro release studies demonstrated that CPhG liposomes have a sustained release effect compared to drug CPhGs. HSC proliferation was inhibited after treatment with the CPhG liposomes (29.45, 14.72, 7.36 µg/mL), with IC50 values of 42.54 µg/mL in the MTT assay. Different concentrations of the CPhG liposomes could inhibit HSC proliferation, promote apoptosis, and block the cell cycle. The MTT method showed an obvious inhibition of HSC proliferation after CPhG liposome and Recombinant Rat Platelet-derived growth factor-BB(rrPDGF-BB) treatment. The levels of collagen-1, metallopeptidase inhibitor 1 (TIMP-1), α smooth muscle actin (α-SMA), and phosphorylated PI3K/Akt were downregulated, and matrix metalloproteinase-1 (MMP-1) was upregulated, by pretreatment with different concentrations of CPhG liposomes. Moreover, 29.45 μg/mL of CPhG liposomes could decrease the expression of the FAK protein and the phosphorylated PI3K and Akt protein downstream of FAK by overexpression of the FAK gene. This experiment suggests that CPhG liposomes may inhibit the activation of HSCs by inhibiting FAK and then reducing the expression of phosphorylated Akt/PI3K, thereby providing new insights into the application of CPhGs for liver fibrosis.
Collapse
Affiliation(s)
- Shi-Lei Zhang
- Department of Toxicology, School of Public Health, Xinjiang Medical University, Xinjiang Uyghur Autonomous Region, Xinyi Road No.393, Urumqi 830011, China.
| | - Long Ma
- Department of Toxicology, School of Public Health, Xinjiang Medical University, Xinjiang Uyghur Autonomous Region, Xinyi Road No.393, Urumqi 830011, China.
| | - Jun Zhao
- Key Laboratory for Uighur Medicine, Institute of Materia Medica of Xinjiang, Xinjiang Uyghur Autonomous Region, Tianshan District, Xinhua South Road No. 140, Urumqi 830004, China.
| | - Shu-Ping You
- Department of Toxicology, School of Public Health, Xinjiang Medical University, Xinjiang Uyghur Autonomous Region, Xinyi Road No.393, Urumqi 830011, China.
| | - Xiao-Ting Ma
- Department of Toxicology, School of Public Health, Xinjiang Medical University, Xinjiang Uyghur Autonomous Region, Xinyi Road No.393, Urumqi 830011, China.
| | - Xiao-Yan Ye
- Department of Toxicology, School of Public Health, Xinjiang Medical University, Xinjiang Uyghur Autonomous Region, Xinyi Road No.393, Urumqi 830011, China.
| | - Tao Liu
- Department of Toxicology, School of Public Health, Xinjiang Medical University, Xinjiang Uyghur Autonomous Region, Xinyi Road No.393, Urumqi 830011, China.
| |
Collapse
|
25
|
Shan L, Liu Z, Ci L, Shuai C, Lv X, Li J. Research progress on the anti-hepatic fibrosis action and mechanism of natural products. Int Immunopharmacol 2019; 75:105765. [PMID: 31336335 DOI: 10.1016/j.intimp.2019.105765] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 07/15/2019] [Indexed: 12/15/2022]
Abstract
Hepatic fibrosis is the most common pathological feature of most chronic liver diseases, and its continuous deterioration gradually develops into liver cirrhosis and eventually leads to liver cancer. At present, there are many kinds of drugs used to treat liver fibrosis. However, Western drugs tend to only target single genes/proteins and induce many adverse reactions. Most of the mechanisms and active ingredients of traditional Chinese medicine (TCM) are not clear, and there is a lack of unified diagnosis and treatment standards. Natural products, which are characterized by structural diversity, low toxicity, and origination from a wide range of sources, have unique advantages and great potential in anti-liver fibrosis. This article summarizes the work done over the previous decade, on the active ingredients in natural products that are reported to have anti-hepatic fibrosis effects. The effective anti-hepatic fibrosis ingredients identified can be generally divided into flavonoids, saponins, polysaccharides and alkaloids. Mechanisms of anti-liver fibrosis include inhibition of liver inflammation, anti-lipid peroxidation injury, inhibition of the activation and proliferation of hepatic stellate cells (HSCs), modulation of the synthesis and secretion of pro-fibrosis factors, and regulation of the synthesis and degradation of the extracellular matrix (ECM). This review provides suggestions for the development of anti-hepatic fibrosis drugs.
Collapse
Affiliation(s)
- Liang Shan
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Zhenni Liu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Leilei Ci
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Chen Shuai
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xiongwen Lv
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China.
| | - Jun Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| |
Collapse
|
26
|
IGFBPrP1 accelerates autophagy and activation of hepatic stellate cells via mutual regulation between H19 and PI3K/AKT/mTOR pathway. Biomed Pharmacother 2019; 116:109034. [PMID: 31152924 DOI: 10.1016/j.biopha.2019.109034] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/04/2019] [Accepted: 05/22/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Our previous study found that insulin-like growth factor binding protein-associated protein (IGFBPrP1) drives hepatic stellate cells (HSCs) activation, and IGFBPrP1 and transforming growth factor β1 (TGFβ1) likely interact with each other to promote HSCs activation. TGFβ1 reportedly promotes autophagy and contributes to HSCs activation; however, the mechanism between IGFBPrP1 and autophagy in liver fibrogenesis is yet unknown. Moreover, long noncoding RNA (lncRNA) H19 participates in autophagy regulation and plays a crucial function in liver fibrosis. AIMS To define the relationship between IGFBPrP1 and autophagy and the role of H19 in IGFBPrP1-induced hepatic fibrosis. METHODS IGFBPrP1 and autophagy were detected in bile duct ligation (BDL)-induced hepatic fibrosis. Adenovirus-mediated IGFBPrP1 was transfected into mouse liver and JS-1 cells with or without LY294002 or rapamycin to examine the effects of IGFBPrP1 on HSCs activation and autophagy as well as the PI3K/AKT/mTOR pathway. lncRNA H19 in liver fibrosis tissues and JS-1 cells induced by IGFBPrP1 were detected, then autophagy and HSCs activation level were detected in JS-1 cells by IGFBPrP1 with H19 overexpression or knowdown. RESULTS IGFBPrP1 expression and autophagy level were concomitantly increased in liver tissue with BDL-induced hepatic fibrosis. Furthermore, we found that IGFBPrP1 stimulated autophagy and HSCs activation in vivo and in vitro, and PI3K/AKT/mTOR signaling pathway was involved in the regulation of autophagy by IGFBPrP1. In addition, H19 promoted autophagy by interacting with the PI3K/AKT/mTOR pathway in IGFBPrP1-induced HSCs activation. CONCLUSIONS IGFBPrP1 promoted autophagy and contributed to HSCs activation via mutual regulation between H19 and the PI3K/AKT/mTOR pathway.
Collapse
|
27
|
George J, Tsuchishima M, Tsutsumi M. Molecular mechanisms in the pathogenesis of N-nitrosodimethylamine induced hepatic fibrosis. Cell Death Dis 2019; 10:18. [PMID: 30622238 PMCID: PMC6325159 DOI: 10.1038/s41419-018-1272-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 12/03/2018] [Accepted: 12/06/2018] [Indexed: 12/13/2022]
Abstract
Hepatic fibrosis is marked by excessive synthesis and deposition of connective tissue proteins, especially interstitial collagens in the extracellular matrix of the liver. It is a result of an abnormal wound healing in response to chronic liver injury from various causes such as ethanol, viruses, toxins, drugs, or cholestasis. The chronic stimuli involved in the initiation of fibrosis leads to oxidative stress and generation of reactive oxygen species that serve as mediators of molecular events involved in the pathogenesis of hepatic fibrosis. These processes lead to cellular injury and initiate inflammatory responses releasing a variety of cytokines and growth factors that trigger activation and transformation of resting hepatic stellate cells into myofibroblast like cells, which in turn start excessive synthesis of connective tissue proteins, especially collagens. Uncontrolled and extensive fibrosis results in distortion of lobular architecture of the liver leading to nodular formation and cirrhosis. The perpetual injury and regeneration process could also results in genomic aberrations and mutations that lead to the development of hepatocellular carcinoma. This review covers most aspects of the molecular mechanisms involved in the pathogenesis of hepatic fibrosis with special emphasize on N-Nitrosodimethylamine (NDMA; Dimethylnitorsmaine, DMN) as the inducing agent.
Collapse
Affiliation(s)
- Joseph George
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY, 10029, USA.
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan.
| | - Mutsumi Tsuchishima
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| | - Mikihiro Tsutsumi
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| |
Collapse
|