1
|
Zhang G, Cai Y, Liang J, Jing Z, Wei W, Lv L, Dang X, Song Q. The decrease in zinc-finger E-box-binding homeobox-1 could accelerate steroid-induced osteonecrosis of the femoral head by repressing type-H vessel formation via Wnt/β-catenin pathway. Animal Model Exp Med 2024; 7:802-815. [PMID: 39686556 DOI: 10.1002/ame2.12507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 10/08/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Zinc-finger E-box-binding homeobox-1 (ZEB1) is predominantly found in type-H vessels. However, the roles of ZEB1 and type-H vessels in steroid-induced osteonecrosis of the femoral head (SONFH) are unclear. METHODS Human femoral heads were collected to detect the expression of ZEB1 and the levels of type-H vessels. Then, the SONFH model was developed by injecting C57BL/6 mice with lipopolysaccharide and methylprednisolone. Micro-computed tomography, angiography, double calcein labeling, immunofluorescence, immunohistochemistry, quantitative real-time polymerase chain reaction, and Western blotting were performed to detect the expression of ZEB1, the Wnt/β-catenin pathway, type-H vessels, and the extent to which ZEB1 mediates angiogenesis and osteogenesis. Human umbilical vein endothelial cells were also used to explore the relationship between ZEB1 and the Wnt/β-catenin pathway. RESULTS We found that ZEB1 expression and the formation of type-H vessels decreased in SONFH patients and in a mouse model. The number of vascular endothelial growth factors in the femoral heads also decreased. Moreover, the bone mineral density, trabecular number, mineral apposition rate, and expression of genes related to osteogenesis decreased. After ZEB1 knockdown, angiogenesis and osteogenesis decreased. However, the numbers of type-H vessels and the extent of angiogenesis and osteogenesis improved after activation of the Wnt/β-catenin pathway. CONCLUSIONS The ZEB1 expression decreased in SONFH, causing a decrease in type-H vessel, and it mediated angiogenesis and osteogenesis by regulating the Wnt/β-catenin pathway, ultimately accelerating the process of SONFH.
Collapse
Affiliation(s)
- Guangyang Zhang
- Orthopedic Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuanqing Cai
- Orthopedic Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jialin Liang
- Orthopedic Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhaopu Jing
- Orthopedic Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wang Wei
- Orthopedic Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Leifeng Lv
- Orthopedic Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoqian Dang
- Orthopedic Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qichun Song
- Orthopedic Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
2
|
Gao R, Mao J. Noncoding RNA-Mediated Epigenetic Regulation in Hepatic Stellate Cells of Liver Fibrosis. Noncoding RNA 2024; 10:44. [PMID: 39195573 DOI: 10.3390/ncrna10040044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/09/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
Liver fibrosis is a significant contributor to liver-related disease mortality on a global scale. Despite this, there remains a dearth of effective therapeutic interventions capable of reversing this condition. Consequently, it is imperative that we gain a comprehensive understanding of the underlying mechanisms driving liver fibrosis. In this regard, the activation of hepatic stellate cells (HSCs) is recognized as a pivotal factor in the development and progression of liver fibrosis. The role of noncoding RNAs (ncRNAs) in epigenetic regulation of HSCs transdifferentiation into myofibroblasts has been established, providing new insights into gene expression changes during HSCs activation. NcRNAs play a crucial role in mediating the epigenetics of HSCs, serving as novel regulators in the pathogenesis of liver fibrosis. As research on epigenetics expands, the connection between ncRNAs involved in HSCs activation and epigenetic mechanisms becomes more evident. These changes in gene regulation have attracted considerable attention from researchers in the field. Furthermore, epigenetics has contributed valuable insights to drug discovery and the identification of therapeutic targets for individuals suffering from liver fibrosis and cirrhosis. As such, this review offers a thorough discussion on the role of ncRNAs in the HSCs activation of liver fibrosis.
Collapse
Affiliation(s)
- Ruoyu Gao
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Jingwei Mao
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| |
Collapse
|
3
|
Xu AL, Han L, Yan J, Liu D, Wang W. Effects of Mesenchymal Stem Cells-Derived Extracellular Vesicles on Inhibition of Hepatic Fibrosis by Delivering miR-200a. Tissue Eng Regen Med 2024; 21:609-624. [PMID: 38568409 PMCID: PMC11087440 DOI: 10.1007/s13770-024-00631-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 02/01/2024] [Accepted: 02/13/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Hepatic fibrosis (HF) is a common pathological feature of chronic hepatic diseases. We aimed to illuminate the significance of amniotic mesenchymal stem cells (AMSCs)-derived extracellular vesicles (AMSCs-EVs) in HF. METHODS Human AMSCs-EVs were isolated and identified. HF mice were constructed and treated with EVs. The fibrosis was observed by staining experiments and Western blot (WB) assay. Alanine aminotransferase (ALT), aspartate aminotransferase (AST), total bilirubin (TBIL), and hepatic hydroxyproline (Hyp) were detected to confirm liver function. For the in vitro experiments, human hepatic stellate cells were induced with transforming growth factor-β and treated with EVs. To measure the degree of HF, the expression of alpha-smooth muscle actin (α-SMA) and Collagen I was detected by WB assay, and cell proliferation was detected by cell counting kit 8 assay. The levels of miR-200a, Zinc finger E-box binding homeobox 1 (ZEB1), and phosphoinositide-3-kinase regulatory subunit 3 (PIK3R3) were detected by WB and real-time quantitative polymerase chain reaction. The binding of ZEB1 to PIK3R3 and miR-200a to ZEB1 was analyzed by chromatin immunoprecipitation and dual luciferase assays to validate their relationships. RESULTS Human AMSCs and AMSCs-EVs were obtained. Serum ALT, AST, TBIL, and hepatic Hyp were increased, implying the fibrosis degree was aggravated in HF mice, which was decreased again after EV treatment. EVs inhibited HF degree by reducing α-SMA and Collagen I and promoting cell proliferation. AMSCs-EVs delivered miR-200a into hepatocytes, which up-regulated miR-200a expression, inhibited ZEB1 expression, and reduced its enrichment on the PIK3R3 promoter, therefore inhibiting PIK3R3 expression and alleviating HF. Overexpression of ZEB1 or PIK3R3 attenuated the anti-fibrotic effect of AMSCs-EVs. CONCLUSION Human AMSCs-derived EVs mediated miR-200a delivery and inhibition of intracellular ZEB1/PIK3R3 axis to exert anti-fibrosis effects.
Collapse
Affiliation(s)
- Ai-Lei Xu
- Department of Gastroenterology, Hunan Aerospace Hospital, 189 Fenglin 3rd Road, Yuelu District, Changsha, 410205, Hunan, China
| | - Long Han
- Department of Gastroenterology, Hunan Aerospace Hospital, 189 Fenglin 3rd Road, Yuelu District, Changsha, 410205, Hunan, China
| | - Jun Yan
- Department of Gastroenterology, Hunan Aerospace Hospital, 189 Fenglin 3rd Road, Yuelu District, Changsha, 410205, Hunan, China
| | - Dan Liu
- Department of Gastroenterology, Hunan Aerospace Hospital, 189 Fenglin 3rd Road, Yuelu District, Changsha, 410205, Hunan, China
| | - Wei Wang
- Department of Gastroenterology, Hunan Aerospace Hospital, 189 Fenglin 3rd Road, Yuelu District, Changsha, 410205, Hunan, China.
| |
Collapse
|
4
|
Ciofoaia V, Chen W, Tarek BW, Gay M, Shivapurkar N, Smith JP. The Role of a Cholecystokinin Receptor Antagonist in the Management of Chronic Pancreatitis: A Phase 1 Trial. Pharmaceutics 2024; 16:611. [PMID: 38794273 PMCID: PMC11125239 DOI: 10.3390/pharmaceutics16050611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Chronic pancreatitis (CP) is a rare but debilitating condition with an 8-fold increased risk of developing pancreatic cancer. In addition to the symptoms that come from the loss of endocrine and exocrine function in CP, the management of chronic pain is problematic. We previously showed that the CCK-receptor antagonist called proglumide could decrease inflammation, acinar-ductal metaplasia, and fibrosis in murine models of CP. We hypothesized that proglumide would be safe and diminish pain caused by CP. A Phase 1 open-labeled safety study was performed in subjects with clinical and radiographic evidence of CP with moderate to severe pain. After a 4-week observation period, the subjects were treated with proglumide in 400 mg capsules three times daily (1200 mg per day) by mouth for 12 weeks, and then subjects returned for a safety visit 4 weeks after the discontinuation of the study medication. The results of three pain surveys (Numeric Rating Scale, COMPAT-SF, and NIH PROMIS) showed that the patients had significantly less pain after 12 weeks of proglumide compared to the pre-treatment observation phase. Of the eight subjects in this study, two experienced nausea and diarrhea with proglumide. These side effects resolved in one subject with doses reduced to 800 mg per day. No abnormalities were noted in the blood chemistries. A blood microRNA blood biomarker panel that corresponded to pancreatic inflammation and fibrosis showed significant improvement. We conclude that proglumide is safe and well tolerated in most subjects with CP at a dose of 1200 mg per day. Furthermore, proglumide therapy may have a beneficial effect by decreasing pain associated with CP.
Collapse
Affiliation(s)
- Victor Ciofoaia
- Departments of Gastroenterology and Medicine, MedStar Washington Hospital Center, Washington, DC 20010, USA; (V.C.); (B.W.T.)
| | - Wenqiang Chen
- Department of Medicine, Georgetown University, Washington, DC 20007, USA; (W.C.); (M.G.); (N.S.)
| | - Bakain W. Tarek
- Departments of Gastroenterology and Medicine, MedStar Washington Hospital Center, Washington, DC 20010, USA; (V.C.); (B.W.T.)
| | - Martha Gay
- Department of Medicine, Georgetown University, Washington, DC 20007, USA; (W.C.); (M.G.); (N.S.)
| | - Narayan Shivapurkar
- Department of Medicine, Georgetown University, Washington, DC 20007, USA; (W.C.); (M.G.); (N.S.)
| | - Jill P. Smith
- Department of Medicine, Georgetown University, Washington, DC 20007, USA; (W.C.); (M.G.); (N.S.)
| |
Collapse
|
5
|
Chen J, He F, Peng H, Guo J. The underlying mechanism and targeted therapy strategy of miRNAs cross-regulating EMT process through multiple signaling pathways in hepatocellular carcinoma. Front Mol Biosci 2024; 11:1378386. [PMID: 38584703 PMCID: PMC10995332 DOI: 10.3389/fmolb.2024.1378386] [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: 01/29/2024] [Accepted: 03/04/2024] [Indexed: 04/09/2024] Open
Abstract
The consistent notion holds that hepatocellular carcinoma (HCC) initiation, progression, and clinical treatment failure treatment failure are affected by the accumulation of various genetic and epigenetic alterations. MicroRNAs (miRNAs) play an irreplaceable role in a variety of physiological and pathological states. meanwhile, epithelial-mesenchymal transition (EMT) is a crucial biological process that controls the development of HCC. miRNAs regulate the intermediation state of EMTor mesenchymal-epithelial transition (MTE)thereby regulating HCC progression. Notably, miRNAs regulate key HCC-related molecular pathways, including the Wnt/β-catenin pathway, PTEN/PI3K/AKT pathway, TGF-β pathway, and RAS/MAPK pathway. Therefore, we comprehensively reviewed how miRNAs produce EMT effects by multiple signaling pathways and their potential significance in the pathogenesis and treatment response of HCC. emphasizing their molecular pathways and progression in HCC initiation. Additionally, we also pay attention to regulatory mechanisms that are partially independent of signaling pathways. Finally, we summarize and propose miRNA-targeted therapy and diagnosis and defense strategies forHCC. The identification of the mechanism leading to the activation of EMT programs during HCC disease processes also provides a new protocol for the plasticity of distinct cellular phenotypes and possible therapeutic interventions. Consequently, we summarize the latest progress in this direction, with a promising path for further insight into this fast-moving field.
Collapse
Affiliation(s)
- Juan Chen
- Department of Pathology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Fuguo He
- Department of Pathology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Peng
- Department of Gastroenterology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Jinjun Guo
- Department of Gastroenterology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Li H, Liu T, Yang Y, Cho WC, Flynn RJ, Harandi MF, Song H, Luo X, Zheng Y. Interplays of liver fibrosis-associated microRNAs: Molecular mechanisms and implications in diagnosis and therapy. Genes Dis 2023; 10:1457-1469. [PMID: 37397560 PMCID: PMC10311052 DOI: 10.1016/j.gendis.2022.08.013] [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: 06/15/2022] [Revised: 08/09/2022] [Accepted: 08/20/2022] [Indexed: 11/22/2022] Open
Abstract
microRNAs (miRNAs) are a class of non-coding functional small RNA composed of 21-23 nucleotides, having multiple associations with liver fibrosis. Fibrosis-associated miRNAs are roughly classified into pro-fibrosis or anti-fibrosis types. The former is capable of activating hepatic stellate cells (HSCs) by modulating pro-fibrotic signaling pathways, mainly including TGF-β/SMAD, WNT/β-catenin, and Hedgehog; the latter is responsible for maintenance of the quiescent phenotype of normal HSCs, phenotypic reversion of activated HSCs (aHSCs), inhibition of HSCs proliferation and suppression of the extracellular matrix-associated gene expression. Moreover, several miRNAs are involved in regulation of liver fibrosis via alternative mechanisms, such as interacting between hepatocytes and other liver cells via exosomes and increasing autophagy of aHSCs. Thus, understanding the role of these miRNAs may provide new avenues for the development of novel interventions against hepatic fibrosis.
Collapse
Affiliation(s)
- Hong Li
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Tingli Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Yongchun Yang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang 311300, China
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR 999077, China
| | - Robin J. Flynn
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool L3 5RF, UK
- Graduate Studies Office, Department of Research, Innovation and Graduate Studies, Waterford Institute of Technology, X91 K0EK, Ireland
| | - Majid Fasihi Harandi
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman 7616914115, Iran
| | - Houhui Song
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang 311300, China
| | - Xuenong Luo
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Yadong Zheng
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang 311300, China
| |
Collapse
|
7
|
Zheng Y, Xie L, Yang D, Luo K, Li X. Small-molecule natural plants for reversing liver fibrosis based on modulation of hepatic stellate cells activation: An update. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 113:154721. [PMID: 36870824 DOI: 10.1016/j.phymed.2023.154721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/07/2023] [Accepted: 02/17/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Liver fibrosis (LF) is a trauma repair process carried out by the liver in response to various acute and chronic liver injuries. Its primary pathological characteristics are excessive proliferation and improper dismissal of the extracellular matrix, and if left untreated, it will progress into cirrhosis, liver cancer, and other diseases. Hepatic stellate cells (HSCs) activation is intimately associated to the onset of LF, and it is anticipated that addressing HSCs proliferation can reverse LF. Plant-based small-molecule medications have anti-LF properties, and their mechanisms of action involve suppression of extracellular matrix abnormally accumulating as well as anti-inflammation and anti-oxidative stress. New targeting HSC agents will therefore be needed to provide a potential curative response. PURPOSE The most recent HSC routes and small molecule natural plants that target HSC described domestically and internationally in recent years were examined in this review. METHODS The data was looked up using resources including ScienceDirect, CNKI, Web of Science, and PubMed. Keyword searches for information on hepatic stellate cells included "liver fibrosis", "natural plant", "hepatic stellate cells", "adverse reaction", "toxicity", etc. RESULTS: We discovered that plant monomers can target and control various pathways to prevent the activation and proliferation of HSC and promote the apoptosis of HSC in order to achieve the anti-LF effect in this work by compiling the plant monomers that influence many common pathways of HSC in recent years. It demonstrates the wide-ranging potential of plant monomers targeting different routes to combat LF, with a view to supplying new concepts and new strategies for natural plant therapy of LF as well as research and development of novel pharmaceuticals. The investigation of kaempferol, physalin B, and other plant monomers additionally motivated researchers to focus on the structure-activity link between the main chemicals and LF. CONCLUSION The creation of novel pharmaceuticals can benefit greatly from the use of natural components. They are often harmless for people, non-target creatures, and the environment because they are found in nature, and they can be employed as the starting chemicals for the creation of novel medications. Natural plants are valuable resources for creating new medications with fresh action targets because they feature original and distinctive action mechanisms.
Collapse
Affiliation(s)
- Yu Zheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Long Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Dejun Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Kaipei Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
8
|
Li QY, Gong T, Huang YK, Kang L, Warner CA, Xie H, Chen LM, Duan XQ. Role of noncoding RNAs in liver fibrosis. World J Gastroenterol 2023; 29:1446-1459. [PMID: 36998425 PMCID: PMC10044853 DOI: 10.3748/wjg.v29.i9.1446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/27/2022] [Accepted: 02/27/2023] [Indexed: 03/07/2023] Open
Abstract
Liver fibrosis is a wound-healing response following chronic liver injury caused by hepatitis virus infection, obesity, or excessive alcohol. It is a dynamic and reversible process characterized by the activation of hepatic stellate cells and excess accumulation of extracellular matrix. Advanced fibrosis could lead to cirrhosis and even liver cancer, which has become a significant health burden worldwide. Many studies have revealed that noncoding RNAs (ncRNAs), including microRNAs, long noncoding RNAs and circular RNAs, are involved in the pathogenesis and development of liver fibrosis by regulating signaling pathways including transforming growth factor-β pathway, phosphatidylinositol 3-kinase/protein kinase B pathway, and Wnt/β-catenin pathway. NcRNAs in serum or exosomes have been reported to tentatively applied in the diagnosis and staging of liver fibrosis and combined with elastography to improve the accuracy of diagnosis. NcRNAs mimics, ncRNAs in mesenchymal stem cell-derived exosomes, and lipid nanoparticles-encapsulated ncRNAs have become promising therapeutic approaches for the treatment of liver fibrosis. In this review, we update the latest knowledge on ncRNAs in the pathogenesis and progression of liver fibrosis, and discuss the potentials and challenges to use these ncRNAs for diagnosis, staging and treatment of liver fibrosis. All these will help us to develop a comprehensive understanding of the role of ncRNAs in liver fibrosis.
Collapse
Affiliation(s)
- Qing-Yuan Li
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Tao Gong
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Yi-Ke Huang
- Center for Transfusion-transmitted Infectious Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu 610052, Sichuan Province, China
| | - Lan Kang
- Center for Transfusion-transmitted Infectious Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu 610052, Sichuan Province, China
| | - Charlotte A Warner
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - He Xie
- Department of Clinical Laboratory, The Hospital of Xidian Group, Xi’an 710077, Shaanxi Province, China
| | - Li-Min Chen
- Center for Transfusion-transmitted Infectious Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu 610052, Sichuan Province, China
- Department of Clinical Laboratory, The Hospital of Xidian Group, Xi’an 710077, Shaanxi Province, China
| | - Xiao-Qiong Duan
- Center for Transfusion-transmitted Infectious Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu 610052, Sichuan Province, China
| |
Collapse
|
9
|
Mesenchymal stem cell-derived exosomes and non-coding RNAs: Regulatory and therapeutic role in liver diseases. Biomed Pharmacother 2023; 157:114040. [PMID: 36423545 DOI: 10.1016/j.biopha.2022.114040] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/16/2022] [Accepted: 11/19/2022] [Indexed: 11/22/2022] Open
Abstract
Liver disease has become a major health problem worldwide due to its high morbidity and mortality. In recent years, a large body of literature has shown that mesenchymal stem cell-derived exosomes (MSC-Exo) are able to play similar physiological roles as mesenchymal stem cells (MSCs). More importantly, there is no immune rejection caused by transplanted cells and the risk of tumor formation, which has become a new strategy for the treatment of various liver diseases. Moreover, accumulating evidence suggests that non-coding RNAs (ncRNAs) are the main effectors by which they exert hepatoprotective effects. Therefore, by searching the databases of Web of Science, PubMed, ScienceDirect, Google Scholar and CNKI, this review comprehensively reviewed the therapeutic effects of MSC-Exo and ncRNAs in liver diseases, including liver injury, liver fibrosis, and hepatocellular carcinoma. According to the data, the therapeutic effects of MSC-Exo and ncRNAs on liver diseases are closely related to a variety of molecular mechanisms, including inhibition of inflammatory response, alleviation of liver oxidative stress, inhibition of apoptosis of hepatocytes and endothelial cells, promotion of angiogenesis, blocking the cell cycle of hepatocellular carcinoma, and inhibition of activation and proliferation of hepatic stellate cells. These important findings will provide a direction and basis for us to explore the potential of MSC-Exo and ncRNAs in the clinical treatment of liver diseases in the future.
Collapse
|
10
|
Rabiee A, Gay MD, Shivapurkar N, Cao H, Nadella S, Smith CI, Lewis JH, Bansal S, Cheema A, Kwagyan J, Smith JP. Safety and Dosing Study of a Cholecystokinin Receptor Antagonist in Non-alcoholic Steatohepatitis. Clin Pharmacol Ther 2022; 112:1271-1279. [PMID: 36087237 PMCID: PMC9691615 DOI: 10.1002/cpt.2745] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 09/06/2022] [Indexed: 01/31/2023]
Abstract
High saturated fat diets have been shown to raise blood levels of cholecystokinin (CCK) and induce nonalcoholic steatohepatitis (NASH). CCK receptors are expressed on stellate cells and are responsible for hepatic fibrosis when activated. The purpose of this study was to test the safety and dose of a CCK receptor antagonist, proglumide, in human participants with NASH. An open-label single ascending dose study was conducted in 18 participants with clinical NASH based upon steatosis by liver ultrasound, elevated hepatic transaminases, and a component of the metabolic syndrome. Three separate cohorts (N = 6 each) were treated with oral proglumide for 12 weeks in a sequential ascending fashion with 800 (Cohort 1), 1,200 (Cohort 2), and 1,600 (Cohort 3) mg/day, respectively. Blood hematology, chemistries, proglumide levels, a biomarker panel for fibrosis, and symptom surveys were determined at baseline and every 4 weeks. Abdominal ultrasounds and transient elastography utilizing FibroScan were obtained at baseline and at Week 12. Proglumide was well tolerated at all doses without any serious adverse events. There was no change in body weight from baseline to Week 12. For Cohorts 1, 2, and 3, the median percent change in alanine aminotransferase was 8.42, -5.05, and -22.23 and median percent change in fibrosis score by FibroScan was 8.13, -5.44, and -28.87 (kPa), respectively. Hepatic steatosis as measured by controlled attenuation parameter score significantly decreased with proglumide, (P < 0.05). Blood microRNA biomarkers and serum 4-hydroxyproline were consistent with decreased fibrosis at Week 12 compared with baseline. These findings suggest proglumide exhibits anti-inflammatory and anti-fibrotic properties and this compound is well tolerated in participants with NASH.
Collapse
Affiliation(s)
- Atoosa Rabiee
- Department of MedicineWashington DC Veterans Affairs Medical CenterWashingtonDCUSA
| | - Martha D. Gay
- Department of MedicineGeorgetown University Medical CenterWashingtonDCUSA
| | | | - Hong Cao
- Department of MedicineGeorgetown University Medical CenterWashingtonDCUSA
| | - Sandeep Nadella
- Departments of Gastroenterology and Transplant SurgeryMedStar Georgetown University HospitalWashingtonDCUSA
| | - Coleman I. Smith
- Departments of Gastroenterology and Transplant SurgeryMedStar Georgetown University HospitalWashingtonDCUSA
| | - James H. Lewis
- Departments of Gastroenterology and Transplant SurgeryMedStar Georgetown University HospitalWashingtonDCUSA
| | - Sunil Bansal
- Department of MedicineGeorgetown University Medical CenterWashingtonDCUSA
| | - Amrita Cheema
- Department of MedicineGeorgetown University Medical CenterWashingtonDCUSA
| | - John Kwagyan
- Department of StatisticsHoward UniversityWashingtonDCUSA
| | - Jill P. Smith
- Department of MedicineWashington DC Veterans Affairs Medical CenterWashingtonDCUSA
- Department of MedicineGeorgetown University Medical CenterWashingtonDCUSA
| |
Collapse
|
11
|
ZEB1: Catalyst of immune escape during tumor metastasis. Biomed Pharmacother 2022; 153:113490. [DOI: 10.1016/j.biopha.2022.113490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/23/2022] [Accepted: 07/27/2022] [Indexed: 11/20/2022] Open
|
12
|
Shen J, Ma X. miR‑374a‑5p alleviates sepsis‑induced acute lung injury by targeting ZEB1 via the p38 MAPK pathway. Exp Ther Med 2022; 24:564. [PMID: 35978929 PMCID: PMC9366279 DOI: 10.3892/etm.2022.11501] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 04/19/2022] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the effects of microRNA (miR)-374a-5p on sepsis-induced acute lung injury (ALI) and the associated mechanism. Lipopolysaccharide (LPS)-induced human pulmonary microvascular endothelial cells (HPMVECs) were used to construct the cellular model of sepsis. A luciferase reporter assay was performed to confirm the association between miR-374a-5p and zinc finger E-box binding homeobox 1 (ZEB1). Reverse transcription-quantitative polymerase chain reaction and western blot analysis were performed to assess the relative expression of miR-374a-5p, ZEB1 and apoptosis-related proteins. Cell viability and apoptosis were determined by Cell Counting Kit-8 assay and flow cytometry, respectively. Enzyme-linked immunosorbent assays were used to evaluate inflammatory cytokines. The results revealed that miR-374a-5p was downregulated in sepsis patients and LPS-treated HPMVECs. Upregulation of miR-374a-5p alleviated LPS-triggered cell injury in HPMVECs, as evidenced by restoration of cell viability, and inhibition of apoptosis and the production of proinflammatory cytokines. In addition, ZEB1 was revealed to be a downstream target of miR-374a-5p, and overexpression of ZEB1 could reverse the anti-apoptotic and anti-inflammatory effects of miR-374a-5p on an LPS-induced sepsis cell model. Moreover, miR-374a-5p-induced protective effects involved the p38 MAPK signaling pathway. Collectively, miR-374a-5p exerted a protective role in sepsis-induced ALI by regulating the ZEB1-mediated p38 MAPK signaling pathway, providing a potential target for the diagnosis and treatment of sepsis.
Collapse
Affiliation(s)
- Jia Shen
- Department of Intensive Care Unit, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750002, P.R. China
| | - Xiaojun Ma
- Department of Orthopedics, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| |
Collapse
|
13
|
Elevated LINC01232 is associated with poor prognosis and HBV infection in hepatocellular carcinoma patients and contributes to tumor progression in vitro. Clin Res Hepatol Gastroenterol 2022; 46:101813. [PMID: 34583064 DOI: 10.1016/j.clinre.2021.101813] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/09/2021] [Accepted: 09/19/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) had high prevalence and poor prognosis, and hepatitis B virus (HBV) infection is a major risk factor. The aim of this study is to analyze the role of long intergenic noncoding RNA 01232 (LINC01232) in the prognosis and progression of HCC, and explore the relationship between LINC01232 and HBV infection. METHODS LINC01232 expression and its prognostic value were firstly analyzed using TCGA database. Quantitative real-time PCR was used to evaluate the expression of LINC01232 in HCC patients and cell lines. Kaplan-Meier curves were used to analyze the relationship between LINC01232 expression and HCC overall survival prognosis. Function-loss in vitro experiments were performed to demonstrate the role of LINC01232 in HCC progression. A luciferase reporter assay and Pearson correlation were used to confirm the relationship between LINC01232 and microRNA (miR)-708-5p in HCC. RESULTS The expression of LINC01232 was upregulated in HCC tissues and cell lines, and high LINC01232 was associated with worse overall survival in HCC. LINC01232 reduction inhibited HCC cells proliferation, migration and invasion. LINC01232 expression was significantly correlated with HBV infection and liver cirrhosis, and showed potential to distinguish HBV-infected HCC patients. miR-708-5p, as a HBV-related miRNA, was a potential target of LINC01232, and was negatively correlated with LINC01232 in HCC. CONCLUSION Our findings found that highly expressed LINC01232 may be a biomarker to indicate survival prognosis in HCC patients, especially in HBV-infected cases. In addition, LINC01232 plays as an oncogene in HCC progression, and its function may exert by sponging miR-708-5p.
Collapse
|
14
|
Abstract
Hepatic fibrosis is a reversible wound healing process following liver injury. Although this process is necessary for maintaining liver integrity, severe excessive extracellular matrix accumulation (ECM) could lead to permanent scar formation and destroy the liver structure. The activation of hepatic stellate cells (HSCs) is a key event in hepatic fibrosis. Previous studies show that most antifibrotic therapies focus on the apoptosis of HSCs and the prevention of HSC activation. Noncoding RNAs (ncRNAs) play a substantial role in HSC activation and are likely to be biomarkers or therapeutic targets for the treatment of hepatic fibrosis. This review summarizes and discusses the previously reported ncRNAs, including the microRNAs, long noncoding RNAs, and circular RNAs, highlighting their regulatory roles and interactions in the signaling pathways that regulate HSC activation in hepatic fibrosis.
Collapse
|
15
|
Li LY, Yang JF, Rong F, Luo ZP, Hu S, Fang H, Wu Y, Yao R, Kong WH, Feng XW, Chen BJ, Li J, Xu T. ZEB1 serves an oncogenic role in the tumourigenesis of HCC by promoting cell proliferation, migration, and inhibiting apoptosis via Wnt/β-catenin signaling pathway. Acta Pharmacol Sin 2021; 42:1676-1689. [PMID: 33514855 PMCID: PMC8463676 DOI: 10.1038/s41401-020-00575-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023]
Abstract
Zinc finger E-box-binding homeobox 1 (ZEB1), a functional protein of zinc finger family, was aberrant expressed in many kinds of liver disease including hepatic fibrosis and Hepatitis C virus. Bioinformatics results showed that ZEB1 was abnormally expressed in HCC tissues. However, to date, the potential regulatory role and molecular mechanisms of ZEB1 are still unclear in the occurrence and development of HCC. This study demonstrated that the expression level of ZEB1 was significantly elevated both in liver tissues of HCC patients and cell lines (HepG2 and SMMC-7721 cells). Moreover, ZEB1 could promote the proliferation, migration, and invasion of HCC cells. On the downstream regulation mechanism, ZEB1 could activate the Wnt/β-catenin signaling pathway by upregulating the protein expression levels of β-catenin, c-Myc, and cyclin D1. Novel studies showed that miR-708 particularly targeted ZEB1 3'-UTR regions and inhibited the HCC cell proliferation, migration, and invasion. Furthermore, results of nude mice experiments of HCC model indicated that miR-708 could inhibit tumor growth and xenograft metastasis model was established to validate that miR-708 could inhibit HCC cell metastasis through tail-vein injection in vivo. Together, the study suggested that ZEB1 modulated by miR-708 might be a potential therapeutic target for HCC therapy.
Collapse
Affiliation(s)
- Liang-Yun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China
| | - Jun-Fa Yang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
| | - Fan Rong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China
- Lujiang County People's Hospital of Anhui Province, Hefei, 231500, China
| | - Zhi-Pan Luo
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China
- The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Shuang Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China
| | - Hui Fang
- Department of Pharmocology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, 310015, China
| | - Ying Wu
- The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Rui Yao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
| | - Wei-Hao Kong
- The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Xiao-Wen Feng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China
| | - Bang-Jie Chen
- First Clinical Medical College of Anhui Medical University, Hefei, 230032, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
16
|
Malchiodi ZX, Cao H, Gay MD, Safronenka A, Bansal S, Tucker RD, Weinberg BA, Cheema A, Shivapurkar N, Smith JP. Cholecystokinin Receptor Antagonist Improves Efficacy of Chemotherapy in Murine Models of Pancreatic Cancer by Altering the Tumor Microenvironment. Cancers (Basel) 2021; 13:4949. [PMID: 34638432 PMCID: PMC8508339 DOI: 10.3390/cancers13194949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/22/2021] [Accepted: 09/25/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer is resistant to chemotherapy in part due to the dense desmoplastic fibrosis surrounding the tumor, the immunosuppressive cells in the tumor microenvironment (TME), and the early rate of metastases. In this study, we examined the effects of a CCK receptor antagonist, proglumide, alone and in combination with gemcitabine in murine models of pancreatic cancer. Tumor growth rate, metastases, and survival were assessed in mice bearing syngeneic murine or human pancreatic tumors treated with PBS (control), gemcitabine, proglumide, or the combination of gemcitabine and proglumide. Excised tumors were evaluated histologically for fibrosis, immune cells, molecular markers, and uptake of chemotherapy by mass spectroscopy. Peripheral blood was analyzed with a microRNAs biomarker panel associated with fibrosis and oncogenesis. Differentially expressed genes between tumors of mice treated with gemcitabine monotherapy and combination therapy were compared by RNAseq. When given in combination the two compounds exhibited inhibitory effects by decreasing tumor growth rate by 70%, metastases, and prolonging survival. Proglumide monotherapy altered the TME by decreasing fibrosis, increasing intratumoral CD8+ T-cells, and decreasing arginase-positive cells, thus rendering the tumor sensitive to chemotherapy. Proglumide altered the expression of genes involved in fibrosis, epithelial-mesenchymal transition, and invasion. CCK-receptor antagonism with proglumide renders pancreatic cancer susceptible to chemotherapy.
Collapse
Affiliation(s)
- Zoe X. Malchiodi
- Department of Oncology, Georgetown University, Washington, DC 20057, USA; (Z.X.M.); (S.B.); (A.C.)
| | - Hong Cao
- Department of Medicine, Georgetown University, Washington, DC 20057, USA; (H.C.); (M.D.G.); (A.S.); (B.A.W.)
| | - Martha D. Gay
- Department of Medicine, Georgetown University, Washington, DC 20057, USA; (H.C.); (M.D.G.); (A.S.); (B.A.W.)
| | - Anita Safronenka
- Department of Medicine, Georgetown University, Washington, DC 20057, USA; (H.C.); (M.D.G.); (A.S.); (B.A.W.)
| | - Sunil Bansal
- Department of Oncology, Georgetown University, Washington, DC 20057, USA; (Z.X.M.); (S.B.); (A.C.)
| | - Robin D. Tucker
- Department of Pathology, Georgetown University, Washington, DC 20057, USA;
| | - Benjamin A. Weinberg
- Department of Medicine, Georgetown University, Washington, DC 20057, USA; (H.C.); (M.D.G.); (A.S.); (B.A.W.)
| | - Amrita Cheema
- Department of Oncology, Georgetown University, Washington, DC 20057, USA; (Z.X.M.); (S.B.); (A.C.)
| | - Narayan Shivapurkar
- Department of Medicine, Georgetown University, Washington, DC 20057, USA; (H.C.); (M.D.G.); (A.S.); (B.A.W.)
| | - Jill P. Smith
- Department of Oncology, Georgetown University, Washington, DC 20057, USA; (Z.X.M.); (S.B.); (A.C.)
- Department of Medicine, Georgetown University, Washington, DC 20057, USA; (H.C.); (M.D.G.); (A.S.); (B.A.W.)
| |
Collapse
|
17
|
Yang J, Xu C, Wu M, Wu Y, Jia X, Zhou C, Zhang X, Ge S, Li Z, Zhang L. MicroRNA-124 inhibits hepatic stellate cells inflammatory cytokines secretion by targeting IQGAP1 through NF-κB pathway. Int Immunopharmacol 2021; 95:107520. [PMID: 33743313 DOI: 10.1016/j.intimp.2021.107520] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/17/2021] [Accepted: 02/17/2021] [Indexed: 01/01/2023]
Abstract
Liver fibrosis is a health concern that leads to organ failure mediated via production of inflammatory cytokines and fibrotic biomarkers. To date, there was no direct approved antifibrotic therapy, and current treatment was mainly the removal of the causative factor. Recent studies demonstrated that aberrant expression of miR-124 was involved in the progression of various liver diseases including hepatocellular carcinoma (HCC). However, whether miR-124 could function as a transcriptional regulator in the inflammatory cytokines secretion of liver fibrosis remains unclear. In this study, we demonstrated that the expression of miR-124 was downregulated in liver fibrosis tissues and TNF-α-induced LX-2 cells, concomitant with the upregulated expression of IQGAP1, suggesting that miR-124 and IQGAP1 might be associated with the development of inflammation in liver fibrosis. Therefore, we demonstrated that the overexpression of miR-124 and knockdown of IQGAP1 could lead to the downregulation of TNF-α, IL-1β and IL-6. While knockdown of miR-124 or overexpression of IQGAP1 showed reversed results. Moreover, dual luciferase reporter assays demonstrated that miR-124 specifically targeted the 3'-UTR of IQGAP1, and thus inhibited the expression of IQGAP1. Mechanistically, we found that the expression changes of miR-124 and IQGAP1 could be involved in inhibition or activation of NF-κB signaling pathway in response to TNF-α. In conclusion, these results indicated that miR-124 plays a crucial role in TNF-α-induced LX-2 cells via regulating NF-κB signaling pathway.
Collapse
Affiliation(s)
- Junfa Yang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China; School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Changqing Xu
- The Third People's Hospital of Hefei (Hefei Third Clinical College of Anhui Medical University), Hefei, Anhui Province, China
| | - Maomao Wu
- Department of Pharmacy, Anhui Chest Hospital, Hefei, Anhui Province, China
| | - Ying Wu
- The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Xiaodi Jia
- Fujian Normal University, Fuzhou 350007, China
| | - Chang Zhou
- School of Basic Medical Science, Anhui Medical University, Hefei 230032, China
| | - Xianzheng Zhang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Shenglin Ge
- The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China.
| | - Zeng Li
- School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| | - Lingling Zhang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China.
| |
Collapse
|
18
|
Ezhilarasan D. MicroRNA interplay between hepatic stellate cell quiescence and activation. Eur J Pharmacol 2020; 885:173507. [PMID: 32858048 DOI: 10.1016/j.ejphar.2020.173507] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/20/2020] [Accepted: 08/23/2020] [Indexed: 02/08/2023]
Abstract
Hepatic stellate cells (HSCs) activation play a significant role in the progression of hepatic fibrosis. During chronic liver diseases, hepatocytes are damaged severely and secrete several pro-inflammatory markers and profibrogenic cytokines via modulation of a variety of signaling pathways that are responsible for the activation of HSCs. The microRNAs (miRNA or miR) have the potential to modulate fibrogenic signaling pathways in HSCs. A variety of miRNAs are identified as profibrogenic and are capable of activating HSCs by modulating fibrosis-associated signaling pathways such as transforming growth factor-β/Smad, Wnt/β-catenin, Hedgehog, Snail and Notch in the injured liver. On the other hand, HSCs also have certain antifibrotic miRNAs and these include miR-16, miR-19b, miR-29, miR-30, miR-101, miR-122, miR-133a, miR-144, miR-146a, miR-150-5p, miR-155, miR-195, miR-200a, miR-214, miR-335, miR-370, miR-454, miR-483, etc. are responsible for maintenance of the quiescent phenotype of normal HSCs, apoptosis induction and phenotypic reversion of activated HSCs, inhibition of HSCs proliferation, suppression of the extracellular matrix-associated gene expressions, etc. Thus, understanding of HSCs specific miRNAs regulation may provide new ideas for the targeted therapy of hepatic fibrosis at molecular level in the near future. Therefore, this review focusses on the modulation of miRNAs profile during the HSCs activation in the fibrotic liver.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Biomedical Research Unit and Laboratory Animal Centre, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), No.162, PH Road, Chennai, Tamil Nadu, 600 077, India.
| |
Collapse
|