1
|
Sultana M, Islam MA, Khairnar R, Kumar S. A guide to pathophysiology, signaling pathways, and preclinical models of liver fibrosis. Mol Cell Endocrinol 2025; 598:112448. [PMID: 39755140 DOI: 10.1016/j.mce.2024.112448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/23/2024] [Accepted: 12/28/2024] [Indexed: 01/06/2025]
Abstract
Liver fibrosis is potentially a reversible form of liver disease that evolved from the early stage of liver scarring as a consequence of chronic liver injuries. Recurrent injuries in the liver without any appropriate medication cause the injuries to get intense and deeper, which gradually leads to the progression of irreversible cirrhosis or carcinoma. Unfortunately, there are no approved treatment strategies for reversing hepatic fibrosis, making it one of the significant risk factors for developing advanced liver disorders and liver disease-associated mortality. Consequently, the interpretation of the fundamental mechanisms, etiology, and pathogenesis is crucial for identifying the potential therapeutic target as well as evaluating novel anti-fibrotic therapy. However, despite innumerable research, the functional mechanism and disease characteristics are still obscure. To accelerate the understanding of underlying disease pathophysiology, molecular pathways and disease progression mechanism, it is crucial to mimic human liver disease through the formation of precise disease models. Although various in vitro and in vivo liver fibrotic models have emerged and developed already, a perfect clinical model replicating human liver diseases is yet to be established, which is one of the major challenges in discovering proper therapeutics. This review paper will shed light on pathophysiology, signaling pathways, preclinical models of liver fibrosis, and their limitations.
Collapse
Affiliation(s)
- Mehonaz Sultana
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Md Asrarul Islam
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Rhema Khairnar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Sunil Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| |
Collapse
|
2
|
Li L, Gao PP, Chen TT, Li N, Zhang HJ, Li MQ, Chen YN, Wei W, Wang H, Sun WY. SUMO: A new perspective to decipher fibrosis. Acta Physiol (Oxf) 2024; 240:e14240. [PMID: 39404508 DOI: 10.1111/apha.14240] [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: 04/24/2024] [Revised: 09/13/2024] [Accepted: 09/25/2024] [Indexed: 11/10/2024]
Abstract
Fibrosis is characterized by excessive extracellular matrix (ECM) deposition resulting from dysregulated wound healing and connective tissue repair mechanisms. Excessive accumulation of ECM leads to fibrous tissue formation, impairing organ function and driving the progression of various fibrotic diseases. Recently, the role of small ubiquitin-like modifiers (SUMO) in fibrotic diseases has attracted significant attention. SUMO-mediated SUMOylation, a highly conserved posttranslational modification, participates in a variety of biological processes, including nuclear-cytosolic transport, cell cycle progression, DNA damage repair, and cellular metabolism. Conversely, SUMO-specific proteases cleave the isopeptide bond of SUMO conjugates, thereby regulating the deSUMOylation process. Mounting evidence indicates that SUMOylation and deSUMOylation regulate the functions of several proteins, such as Smad3, NF-κB, and promyelocytic leukemia protein, which are implicated in fibrotic diseases like liver fibrosis, myocardial fibrosis, and pulmonary fibrosis. This review summarizes the role of SUMO in fibrosis-related pathways and explores its pathological relevance in various fibrotic diseases. All evidence suggest that the SUMO pathway is important targets for the development of treatments for fibrotic diseases.
Collapse
Affiliation(s)
- Ling Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
| | - Ping-Ping Gao
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
| | - Ting-Ting Chen
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
| | - Nan Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
| | - Hui-Juan Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
| | - Meng-Qi Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
| | - Ya-Ning Chen
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
| | - Hua Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Wu-Yi Sun
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
| |
Collapse
|
3
|
Song H, Zhang Q, Fang G, Luo X, Wu D, Li H, Zhou K, Zhao X, Xu F, Zhang Y, Huang A. Unraveling the Mechanisms of MicroRNA in Suppressing Hepatitis B Virus Progression: A Comprehensive Review for Designing Treatment Strategies. HEPATITIS MONTHLY 2024; 24. [DOI: 10.5812/hepatmon-144239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 06/22/2024] [Accepted: 07/13/2024] [Indexed: 01/02/2025]
Abstract
: Liver cancer and cirrhosis caused by the Hepatitis B virus (HBV) remain significant global health challenges due to the virus's high prevalence and contagious nature. Hepatitis B virus can be transmitted through various means, leading to mild or severe liver disease. Although an effective prophylactic vaccine is available, it offers limited benefits for those already chronically infected. Current treatments often fail to consistently eliminate the virus and can cause severe adverse effects. In response to these challenges, researchers have begun exploring microRNAs (miRNAs) as novel therapeutic targets. Studying miRNA-virus interactions presents a promising opportunity to identify potential therapeutic targets. By manipulating host miRNAs, researchers aim to enhance antiviral defenses, restore cellular balance, and prevent viral replication. The text concludes by highlighting the potential for personalized medicine in Hepatitis B treatment, guided by individual miRNA profiles. Numerous studies have been conducted to understand how different miRNAs inhibit HBV replication, paving the way for the development of innovative and effective therapeutic strategies.
Collapse
|
4
|
Song X, Liu F, Chen M, Zhu M, Zheng H, Wang W, Chen D, Li M, Chen S. MiR-21 regulates skeletal muscle atrophy and fibrosis by targeting TGF-beta/SMAD7-SMAD2/3 signaling pathway. Heliyon 2024; 10:e33062. [PMID: 39027432 PMCID: PMC11254527 DOI: 10.1016/j.heliyon.2024.e33062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024] Open
Abstract
Long-term denervation-induced atrophy and fibrosis of skeletal muscle due to denervation leads to poor recovery of muscle function. Studies have shown that the transforming growth factor-β1 (TGF-β1)-Smad signaling pathway plays a central role in muscle atrophy and fibrosis. Recent studies demonstrate the role of microRNAs (miRs) in various pathological conditions, including muscle regeneration. miR-21 has been shown to play a dynamic role in inflammatory responses and in accelerating injury responses to fibrosis. We used both RNA sequencing and quantitative RT-PCR strategies to examine the alternations of miRNAs during denervation-induced gastrocnemius muscle atrophy and fibrosis. Our data showed that MiR-21 was upregulated in denervated gastrocnemius muscle tissue, and TGF-β1treatment increased miR-21 expression. Inhibition of miR-21 reduced gastrocnemius muscle fibrosis and significantly downregulated the expression of p-SMAD2/3 and the fibrosis-associated markers TGF-β1, connective tissue growth factor, alpha smooth muscle actin. Masson's trichrome staining revealed that atrophy and fibrosis in gastrocnemius muscle tissue were reduced in the miR-21 inhibition group compared to the control group. We confirmed that SMAD7 is a direct target of miR-21 using a dual luciferase assay. Furthermore, Immunofluorescence and Western blot analyses revealed that miR-21 inhibition reduced SMAD2/3 phosphorylation and nuclear translocation. While SMAD7-siRNA abolished the effect. Consequently, the discovery that miR-21 regulates the atrophy and fibrosis of the gastrocnemius muscle offers a possible therapeutic approach for their management.
Collapse
Affiliation(s)
- Xianmin Song
- From the Department of Otorhinolaryngology & Head and Neck Surgery, Changhai Hospital, Naval Military Medical University (The Second Military Medical University), Shanghai, 200433, China
| | - Fei Liu
- From the Department of Otorhinolaryngology & Head and Neck Surgery, Changhai Hospital, Naval Military Medical University (The Second Military Medical University), Shanghai, 200433, China
| | - Mengjie Chen
- From the Department of Otorhinolaryngology & Head and Neck Surgery, Changhai Hospital, Naval Military Medical University (The Second Military Medical University), Shanghai, 200433, China
| | - Minhui Zhu
- From the Department of Otorhinolaryngology & Head and Neck Surgery, Changhai Hospital, Naval Military Medical University (The Second Military Medical University), Shanghai, 200433, China
| | - Hongliang Zheng
- From the Department of Otorhinolaryngology & Head and Neck Surgery, Changhai Hospital, Naval Military Medical University (The Second Military Medical University), Shanghai, 200433, China
| | - Wei Wang
- From the Department of Otorhinolaryngology & Head and Neck Surgery, Changhai Hospital, Naval Military Medical University (The Second Military Medical University), Shanghai, 200433, China
| | - Donghui Chen
- Department of Otorhinolaryngology, The First Affiliate Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Meng Li
- From the Department of Otorhinolaryngology & Head and Neck Surgery, Changhai Hospital, Naval Military Medical University (The Second Military Medical University), Shanghai, 200433, China
| | - Shicai Chen
- From the Department of Otorhinolaryngology & Head and Neck Surgery, Changhai Hospital, Naval Military Medical University (The Second Military Medical University), Shanghai, 200433, China
| |
Collapse
|
5
|
Xue X, Li Y, Yao Y, Zhang S, Peng C, Li Y. A comprehensive review of miR-21 in liver disease: Big impact of little things. Int Immunopharmacol 2024; 134:112116. [PMID: 38696909 DOI: 10.1016/j.intimp.2024.112116] [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: 02/21/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 05/04/2024]
Abstract
microRNAs (miRNAs), a class of non-coding RNA with 20-24 nucleotides, are defined as the powerful regulators for gene expression. miR-21 is a multifunctional miRNA enriched in the circulatory system and multiple organs, which not only serves as a non-invasive biomarker in disease diagnosis, but also participates in many cellular activities. In various chronic liver diseases, the increase of miR-21 affects glycolipid metabolism, viral infection, inflammatory and immune cell activation, hepatic stellate cells activation and tissue fibrosis, and autophagy. Moreover, miR-21 is also a liaison in the deterioration of chronic liver disease to hepatocellular carcinoma (HCC), and it impacts on cell proliferation, apoptosis, migration, invasion, angiogenesis, immune escape, and epithelial-mesenchymal transformation by regulating target genes expression in different signaling pathways. In current research on miRNA therapy, some natural products can exert the hepatoprotective effects depending on the inhibition of miR-21 expression. In addition, miR-21-based therapeutic also play a role in regulating intracellular miR-21 levels and enhancing the efficacy of chemotherapy drugs. Herein, we systemically summarized the recent progress of miR-21 on biosynthesis, biomarker function, molecular mechanism and miRNA therapy in chronic liver disease and HCC, and looked forward to outputting some information to enable it from bench to bedside.
Collapse
Affiliation(s)
- Xinyan Xue
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yanzhi Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yuxin Yao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shenglin Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
6
|
Zheng J, Wu J, Xie L, Huang Y, Hong J, Chen C. Paclitaxel Aggravating Radiation-Induced Pulmonary Fibrosis Is Associated with the Down-Regulation of the Negative Regulatory Function of Spry2. J Pharmacol Exp Ther 2024; 389:197-207. [PMID: 37918858 DOI: 10.1124/jpet.123.001695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/27/2023] [Accepted: 09/18/2023] [Indexed: 11/04/2023] Open
Abstract
Paclitaxel (PTX) is capable of aggravating radiation-induced pulmonary fibrosis (RIPF), but the mechanism is unknown. Spry2 is a negative regulator of receptor tyrosine kinase-related Ras/Raf/extracellular signal regulated kinase (ERK) pathway. This experiment was aimed at exploring whether the aggravation of RIPF by PTX is related to Spry2. The RIPF model was established with C57BL/6 mice by thoracic irradiation, and PTX was administered concurrently. Western blot was used to detect the expression level of ERK signaling molecules and the distribution of Spry2 in the plasma membrane/cytoplasm. Co-immunoprecipitation (co-IP) and immunofluorescence were used to observe the colocalization of Spry2 with the plasma membrane and tubulin. The results showed that PTX-concurrent radiotherapy could aggravate fibrotic lesions in RIPF, downregulate the content of membrane Spry2, and upregulate the levels of p-c-Raf and p-ERK in lung tissue. It was found that knockdown of Spry2 in fibroblast abolished the upregulation of p-c-Raf and p-ERK by PTX. Both co-IP results and immunofluorescence staining showed that PTX increased the binding of Spry2 to tubulin, and microtubule depolymerizing agents could abolish PTX's inhibition of Spry2 membrane distribution and inhibit PTX's upregulation of Raf/ERK signaling. Both nintedanib and ERK inhibitor were effective in relieving PTX-exacerbated RIPF. Taken together, the mechanism of PTX's aggravating RIPF was related to its ability to enhance Spry2's binding to tubulin, thus attenuating Spry2's negative regulation on Raf/ERK pathway. SIGNIFICANCE STATEMENT: This study revealed that paclitaxel (PTX) concurrent radiation therapy exacerbates radiation-induced pulmonary fibrosis during the treatment of thoracic tumors, which is associated with PTX restraining Spry2 and upregulating the Raf/extracellular signal regulated kinase signaling pathway, and provided drug targets for mitigating this complication.
Collapse
Affiliation(s)
- Jianxing Zheng
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China (J.Z.); Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China (J.W., J.H.); School of Pharmacy (L.X., Y.H., C.C.) and Fujian Key Laboratory of Natural Medicine Pharmacology (C.C.), Fujian Medical University, Fuzhou, China; and Key Laboratory of Radiation Biology of Fujian higher education institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China (J.H.)
| | - Jiandong Wu
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China (J.Z.); Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China (J.W., J.H.); School of Pharmacy (L.X., Y.H., C.C.) and Fujian Key Laboratory of Natural Medicine Pharmacology (C.C.), Fujian Medical University, Fuzhou, China; and Key Laboratory of Radiation Biology of Fujian higher education institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China (J.H.)
| | - Lingfeng Xie
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China (J.Z.); Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China (J.W., J.H.); School of Pharmacy (L.X., Y.H., C.C.) and Fujian Key Laboratory of Natural Medicine Pharmacology (C.C.), Fujian Medical University, Fuzhou, China; and Key Laboratory of Radiation Biology of Fujian higher education institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China (J.H.)
| | - Yihao Huang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China (J.Z.); Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China (J.W., J.H.); School of Pharmacy (L.X., Y.H., C.C.) and Fujian Key Laboratory of Natural Medicine Pharmacology (C.C.), Fujian Medical University, Fuzhou, China; and Key Laboratory of Radiation Biology of Fujian higher education institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China (J.H.)
| | - Jinsheng Hong
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China (J.Z.); Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China (J.W., J.H.); School of Pharmacy (L.X., Y.H., C.C.) and Fujian Key Laboratory of Natural Medicine Pharmacology (C.C.), Fujian Medical University, Fuzhou, China; and Key Laboratory of Radiation Biology of Fujian higher education institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China (J.H.)
| | - Chun Chen
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China (J.Z.); Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China (J.W., J.H.); School of Pharmacy (L.X., Y.H., C.C.) and Fujian Key Laboratory of Natural Medicine Pharmacology (C.C.), Fujian Medical University, Fuzhou, China; and Key Laboratory of Radiation Biology of Fujian higher education institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China (J.H.)
| |
Collapse
|
7
|
Dong BS, Liu FQ, Yang WN, Li XD, Shi MJ, Li MR, Yan XL, Zhang H. Huangqi Decoction, a compound Chinese herbal medicine, inhibits the proliferation and activation of hepatic stellate cells by regulating the long noncoding RNA-C18orf26-1/microRNA-663a/transforming growth factor-β axis. JOURNAL OF INTEGRATIVE MEDICINE 2023; 21:47-61. [PMID: 36456413 DOI: 10.1016/j.joim.2022.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Huangqi Decoction (HQD), a classical traditional Chinese medicine formula, has been used as a valid treatment for alleviating liver fibrosis; however, the underlying molecular mechanism is still unknown. Although our previous studies showed that microRNA-663a (miR-663a) suppresses the proliferation and activation of hepatic stellate cells (HSCs) and the transforming growth factor-β/small mothers against decapentaplegic (TGF-β/Smad) pathway, whether long noncoding RNAs (lncRNAs) are involved in HSC activation via the miR-663a/TGF-β/Smad signaling pathway has not yet reported. The present study aimed to investigate the roles of lncRNA lnc-C18orf26-1 in the activation of HSCs and the mechanism by which HQD inhibits hepatic fibrosis. METHODS The expression levels of lnc-C18orf26-1, miR-663a and related genes were measured by quantitative reverse transcription-polymerase chain reaction. HSCs were transfected with the miR-663a mimic or inhibitor and lnc-C18orf26-1 small interfering RNAs. The water-soluble tetrazolium salt-1 assay was used to assess the proliferation rate of HSCs. Changes in lncRNA expression were evaluated in miR-663a-overexpressing HSCs by using microarray to identify miR-663a-regulated lncRNAs. RNA hybrid was used to predict the potential miR-663a binding sites on lncRNAs. Luciferase reporter assays further confirmed the interaction between miR-663a and the lncRNA. The expression levels of collagen α-2(I) chain (COL1A2), α-smooth muscle actin (α-SMA) and TGF-β/Smad signaling pathway-related proteins were determined using Western blotting. RESULTS Lnc-C18orf26-1 was upregulated in TGF-β1-activated HSCs and competitively bound to miR-663a. Knockdown of lnc-C18orf26-1 inhibited HSC proliferation and activation, downregulated TGF-β1-stimulated α-SMA and COL1A2 expression, and inhibited the TGF-β1/Smad signaling pathway. HQD suppressed the proliferation and activation of HSCs. HQD increased miR-663a expression and decreased lnc-C18orf26-1 expression in HSCs. Further studies showed that HQD inhibited the expression of COL1A2, α-SMA, TGF-β1, TGF-β type I receptor (TGF-βRI) and phosphorylated Smad2 (p-Smad2) in HSCs, and these effects were reversed by miR-663a inhibitor treatment. CONCLUSION Our study identified lnc-C18orf26-1 and miR-663a as promising therapeutic targets for hepatic fibrosis. HQD inhibits HSC proliferation and activation at least partially by regulating the lnc-C18orf26-1/miR-663a/TGF-β1/TGF-βRI/p-Smad2 axis.
Collapse
Affiliation(s)
- Ben-Sheng Dong
- Traditional Chinese Medicine Epigenomics Research Center, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Fu-Qun Liu
- Department of Rheumatology and Immunology, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing 211299, Jiangsu Province, China; Department of Rheumatology and Immunology, Yangzhou University Medical College, Yangzhou 225000, Jiangsu Province, China
| | - Wen-Na Yang
- Department of Rheumatology and Immunology, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing 211299, Jiangsu Province, China; Department of Rheumatology and Immunology, Yangzhou University Medical College, Yangzhou 225000, Jiangsu Province, China
| | - Xiao-Dong Li
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Kita-gun, Kagawa 761-0793, Japan
| | - Miao-Juan Shi
- Traditional Chinese Medicine Epigenomics Research Center, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mao-Rong Li
- Traditional Chinese Medicine Epigenomics Research Center, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiu-Li Yan
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| | - Hui Zhang
- Traditional Chinese Medicine Epigenomics Research Center, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
8
|
Cheng L, Wang T, Gao Z, Wu W, Cao Y, Wang L, Zhang Q. Study on the Protective Effect of Schizandrin B against Acetaminophen-Induced Cytotoxicity in Human Hepatocyte. Biol Pharm Bull 2022; 45:596-604. [DOI: 10.1248/bpb.b21-00965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Ling Cheng
- Medical Intensive Care Unit, The First Affiliated Hospital of Anhui University of Chinese Medicine
| | - Tingting Wang
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine
| | - Zhiling Gao
- Medical Intensive Care Unit, The First Affiliated Hospital of Anhui University of Chinese Medicine
| | - Wenkai Wu
- Department of General Surgery, The First Affiliated Hospital of Anhui University of Chinese Medicine
| | - Yezhi Cao
- Department of General Surgery, The First Affiliated Hospital of Anhui University of Chinese Medicine
| | - Linghu Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui University of Chinese Medicine
| | - Qi Zhang
- Institute of Surgery, Anhui Academy of Chinese Medicine
| |
Collapse
|
9
|
Liu Z, Xu B, Ding Y, Ding X, Yang Z. Guizhi Fuling pill attenuates liver fibrosis in vitro and in vivo via inhibiting TGF-β1/Smad2/3 and activating IFN-γ/Smad7 signaling pathways. Bioengineered 2022; 13:9357-9368. [PMID: 35387552 PMCID: PMC9161976 DOI: 10.1080/21655979.2022.2054224] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Liver fibrosis resulting from chronic liver injuries (CLI) is a common health problem globally. Guizhi Fuling pill (GZFL), a modern preparation from traditional Chinese medicine, exhibited anti-dysmenorrhea, anti-inflammatory, and immune-regulative effects. However, the effect of GZFL on liver fibrosis remains unknown. In this research, LX-2 cells were stimulated with acetaldehyde for mimicking liver fibrosis progression in vitro. In addition, carbon tetrachloride (CCl4)-induced mouse model of liver fibrosis was established as well. The data revealed GZFL obviously suppressed the proliferation and triggered the apoptosis of acetaldehyde-stimulated LX-2 cells. In addition, GZFL prevented acetaldehyde-induced activation of LX-2 cells via downregulation of TGF-β1, p-Smad2, p-Smad3, CUGBP1, and upregulation of p-STAT1 and Smad7. Meanwhile, GZFL significantly alleviated CCl4‑induced liver fibrosis, as evidenced by the decrease of ALT and AST levels. Moreover, GZFL downregulated the expressions of TGF-β1, p-Smad2, p-Smad3, and CUGBP1 in CCl4-treated mice. Furthermore, GZFL remarkably elevated the levels of IFN-γ, p-STAT1, and Smad7 in CCl4-treated mice. To sum up, GZFL was able to inhibit liver fibrosis in vitro and in vivo through suppressing TGF-β1/Smad2/3-CUGBP1 signaling and activating IFN-γ/STAT1/Smad7 signaling. Thus, GZFL might have a potential to act as a therapeutic agent for anti-fibrotic therapy.
Collapse
Affiliation(s)
- Zhongliang Liu
- Department of Oncology, Zhoushan Hospital of Traditional Chinese Medicine (Affiliated to Zhejiang University of Traditional Chinese Medicine), Zhoushan, P.R. China
| | - Baogui Xu
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Yaping Ding
- Department of Nutrition, Zhoushan Hospital of Traditional Chinese Medicine (Affiliated to Zhejiang University of Traditional Chinese Medicine), Zhoushan, P.R. China
| | - Xianjun Ding
- Department of Infectious Diseases, Zhoushan Hospital of Traditional Chinese Medicine (Affiliated to Zhejiang University of Traditional Chinese Medicine), Zhoushan, P.R. China.,Department of Infectious Diseases, Zhoushan Hospital, P.R. China
| | - Zuisu Yang
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| |
Collapse
|
10
|
Chen J, Xu Y, Wu P, Chen X, Weng W, Li D. Transcription Factor FOXO3a Overexpression Inhibits the Progression of Neuroblastoma by Regulating the miR-21/SPRY2/ERK Axis. World Neurosurg 2022; 164:e99-e112. [DOI: 10.1016/j.wneu.2022.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/02/2022] [Indexed: 11/25/2022]
|
11
|
Hassan HM, Cai Q, Liang X, Xin J, Ren K, Jiang J, Shi D, Lu Y, Li T, Shang Y, He L, Chen X, Sun S, Li P, Guo B, Chen J, Yang H, Hu W, Chen X, Li J. Transcriptomics reveals immune-metabolism disorder in acute-on-chronic liver failure in rats. Life Sci Alliance 2021; 5:5/3/e202101189. [PMID: 34853163 PMCID: PMC8645333 DOI: 10.26508/lsa.202101189] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/17/2021] [Accepted: 11/17/2021] [Indexed: 12/03/2022] Open
Abstract
Liver tissue transcriptomics of liver cirrhosis (LC)–based acute-on-chronic liver failure (ACLF) rats reveal immune-metabolism disorder as the core mechanism underlying ACLF development and prognosis. Acute-on-chronic liver failure (ACLF) is clinical syndrome with high mortality rate. This study aimed to perform detailed transcriptomic analysis in liver cirrhosis–based ACLF rats to elucidate ACLF pathogenesis. ACLF was induced by combined porcine serum with D-galactosamine and lipopolysaccharide. Gene expression profile of liver tissues from ACLF rats was generated by transcriptome sequencing to reveal the molecular mechanism. ACLF rats successfully developed with typical characteristics. Total of 2,354/3,576 differentially expressed genes were identified when ACLF was compared to liver cirrhosis and normal control, separately. The functional synergy analysis revealed prominent immune dysregulation at ACLF stage, whereas metabolic disruption was significantly down-regulated. Relative proportions of innate immune–related cells showed significant elevation of monocytes and macrophages, whereas adaptive immune–related cells were reduced. The seven differentially expressed genes underlying the ACLF molecular mechanisms were externally validated, among them THBS1, IL-10, and NR4A3 expressions were confirmed in rats, patient transcriptomics, and liver biopsies, verifying their potential value in the ACLF pathogenesis. This study indicates immune-metabolism disorder in ACLF rats, which may provide clinicians new targets for improving intervention strategies.
Collapse
Affiliation(s)
- Hozeifa M Hassan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qun Cai
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xi Liang
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Jiaojiao Xin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Keke Ren
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Dongyan Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Yingyan Lu
- Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Tan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuxin Shang
- Imperial College London, South Kensington Campus, London, UK
| | - Lulu He
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xi Chen
- Shanghai Pinghe School, Shanghai, China
| | - Suwan Sun
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Beibei Guo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaxian Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wen Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin Chen
- Institute of Pharmaceutical Biotechnology and The First Affiliated Hospital Department of Radiation Oncology, Zhejiang University School of Medicine, Hangzhou, China.,Joint Institute for Genetics and Genome Medicine Between Zhejiang University and University of Toronto, Zhejiang University, Hangzhou, China
| | - Jun Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China .,Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| |
Collapse
|
12
|
Zhou X, Liang Z, Qin S, Ruan X, Jiang H. Serum-derived miR-574-5p-containing exosomes contribute to liver fibrosis by activating hepatic stellate cells. Mol Biol Rep 2021; 49:1945-1954. [PMID: 34843038 PMCID: PMC8863722 DOI: 10.1007/s11033-021-07008-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/23/2021] [Indexed: 12/11/2022]
Abstract
Aim To investigate the association of serum exosomes miR-574-5p with liver fibrosis, and explore the effect and mechanism of serum exosomes on HSC activation. Materials and methods Using serum samples collected from healthy adults and patients with liver cirrhosis, we extracted human serum exosomes via ultra-high-speed centrifugation, and co-cultured them with hepatic stellate cells (HSCs) line LX2. LX-2-mediated intake of human serum exosomes was examined by confocal microscopy. To induce liver fibrosis, we administered 20% CCl4 to mice intraperitoneally and adopted an exoEasy MIDI kit to extract serum exosomes.Liver fibrosis-related molecules were determined via qRT-PCR, Western blot, Masson staining, and Immunohistochemical staining. Results Significantly high miR-574-5p levels were expressed in serum exosomes and were positively correlated with the expression of miR-574-5p, collagen deposition, and α-SMA expression in liver tissues of mice during liver fibrosis. Compared to healthy subjects, serum exosomes from cirrhosis patients were associated with higher expression of miR-574-5p. MiR-574-5p mimic promoted the expression of α-SMA and COL1A1 mRNA and protein in LX-2, whereas miR-574-5p inhibitor exerted no effect. Conclusion This article demonstrates that miR-574-5p expression in serum exosomes is positively correlated with collagen deposition and HSC activation in liver tissues during liver fibrosis.Serum exosomes potentially activate HSC through the transfer of miR-574-5p to HSC during liver fibrosis.
Collapse
Affiliation(s)
- Xia Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.,People's Hospital of Guizhou Province, Guiyang, Guizhou, China
| | - Ziyu Liang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shanyu Qin
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xianxian Ruan
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Haixing Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
13
|
Wang X, Liu T, Huang Y, Dai Y, Lin H. Regulation of transforming growth factor-β signalling by SUMOylation and its role in fibrosis. Open Biol 2021; 11:210043. [PMID: 34753319 PMCID: PMC8580444 DOI: 10.1098/rsob.210043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fibrosis is an abnormal healing process that only repairs the structure of an organ after injury and does not address damaged functions. The pathogenesis of fibrosis is multifactorial and highly complex; numerous signalling pathways are involved in this process, with the transforming growth factor-β (TGF-β) signalling pathway playing a central role. TGF-β regulates the generation of myofibroblasts and the epithelial-mesenchymal transition by regulating transcription and translation of downstream genes and precisely regulating fibrogenesis. The TGF-β signalling pathway can be modulated by various post-translational modifications, of which SUMOylation has been shown to play a key role. In this review, we focus on the function of SUMOylation in canonical and non-canonical TGF-β signalling and its role in fibrosis, providing promising therapeutic strategies for fibrosis.
Collapse
Affiliation(s)
- Xinyi Wang
- First Clinical Medical School, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Ting Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Yifei Huang
- First Clinical Medical School, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Yifeng Dai
- Second Clinical Medical School, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Hui Lin
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| |
Collapse
|
14
|
Khaleel EF. l-Thyroxine induces left ventricular remodeling and fibrosis in rats by upregulating miR-21 in a reactive oxygen-dependent mechanism: a protective role of N-acetylcysteine. Drug Chem Toxicol 2021; 45:2758-2768. [PMID: 34641738 DOI: 10.1080/01480545.2021.1986251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
miR-21 is the most studied pro-fibrotic marker in the majority of mammalian tissues. The precise mechanism by which hyperthyroidism induces left ventricular LV fibrosis and remodeling remains unclear. In this study, we have investigated the role of miR-21 on l-thyroxine (l-Thy)-induced cardiac fibrosis in rats. Adult male Sprague-Dawley rats were divided into four groups as control, l-Thy, l-Thy + miR antagomir (inhibitor), and l-Thy + N-acetylcysteine (NAC/glutathione (GSH) precursor). Administration of l-Thy significantly increased mRNA levels of miR-21 in the LVs of the treated rats. Also, it impaired the LV systolic and diastolic function and increased the production of reactive oxygen species (ROS), the transactivation of NF-κB p65, the expression of NRLP3 inflammasome, and levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in these LVs. Concomitantly, l-Thy increased the ventricular collagen deposition, and stimulated the expression of collagen 1/3, alpha-smooth actin (α-SMA), transforming growth factor-β1, and Smad3/p-Smad3 but suppressed the expression of Smad7. All these effects were reversed by pre-treatment with miR-21 antagomir or co-administration of NAC. In conclusion, l-Thy-induced LV remodeling and fibrosis include a ROS-dependent upregulation of miR-21 which in turns activates NF-κB/NRLP3 inflammasome and suppresses SMad7.
Collapse
Affiliation(s)
- Eman F Khaleel
- Department of Medical Physiology, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia.,Department of Medical Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
15
|
Xu X, Hong P, Wang Z, Tang Z, Li K. MicroRNAs in Transforming Growth Factor-Beta Signaling Pathway Associated With Fibrosis Involving Different Systems of the Human Body. Front Mol Biosci 2021; 8:707461. [PMID: 34381815 PMCID: PMC8350386 DOI: 10.3389/fmolb.2021.707461] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022] Open
Abstract
Fibrosis, a major cause of morbidity and mortality, is a histopathological manifestation of many chronic inflammatory diseases affecting different systems of the human body. Two types of transforming growth factor beta (TGF-β) signaling pathways regulate fibrosis: the canonical TGF-β signaling pathway, represented by SMAD-2 and SMAD-3, and the noncanonical pathway, which functions without SMAD-2/3 participation and currently includes TGF-β/mitogen-activated protein kinases, TGF-β/SMAD-1/5, TGF-β/phosphatidylinositol-3-kinase/Akt, TGF-β/Janus kinase/signal transducer and activator of transcription protein-3, and TGF-β/rho-associated coiled-coil containing kinase signaling pathways. MicroRNA (miRNA), a type of non-coding single-stranded small RNA, comprises approximately 22 nucleotides encoded by endogenous genes, which can regulate physiological and pathological processes in fibrotic diseases, particularly affecting organs such as the liver, the kidney, the lungs, and the heart. The aim of this review is to introduce the characteristics of the canonical and non-canonical TGF-β signaling pathways and to classify miRNAs with regulatory effects on these two pathways based on the influenced organ. Further, we aim to summarize the limitations of the current research of the mechanisms of fibrosis, provide insights into possible future research directions, and propose therapeutic options for fibrosis.
Collapse
Affiliation(s)
- Xiaoyang Xu
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, China
| | - Pengyu Hong
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, China
| | - Zhefu Wang
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, China
| | - Zhangui Tang
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, China
| | - Kun Li
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, China
| |
Collapse
|
16
|
El-Sebaey AM, Abramov PN. Hepatocyte-derived canine familiaris-microRNAs as serum biomarkers of hepatic steatosis or fibrosis as implicated in the pathogenesis of canine cholecystolithiasis. Vet Clin Pathol 2021; 50 Suppl 1:37-46. [PMID: 34031917 DOI: 10.1111/vcp.12942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/01/2020] [Accepted: 09/16/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Hepatic cholesterol accumulation in small breed dogs is a leading risk factor for hepatic fatty changes, gallbladder hypomotility, and cholelith development, which, if not discovered early, could lead to life-threatening choledocholithiasis and acute pancreatitis. OBJECTIVE This study proposed to assess the use of hepatocyte-derived canine familiaris (cfa)-microRNAs (miRNA-122, -34a, and -21) as new diagnostic serum biomarkers of liver steatosis or fibrosis, for which both processes have been implicated in canine cholecystolithiasis. METHODS Forty client-owned dogs diagnosed with cholecystolithiasis and hepatic steatosis (C+HS) or fibrosis (C+HF) based on ultrasonographic, biochemical, and histopathologic findings, and 20 healthy dogs used as controls were included in the study. Serum cfa-miRNA expression was determined using a real-time polymerase chain reaction assay. RESULTS Serum cfa-miRNA-122 and -34a expression was significantly upregulated in the C+HS (P < .001) and C+HF (P < .01) groups compared with the control group and showed a positive correlation with alanine aminotransferase (ALT), aspartate aminotransferase (AST), total bilirubin (TBIL), alkaline phosphatase (ALP), γ-glutamyl transferase (GGT), total cholesterol (TC), and triglycerides (TG) levels in the C+HS group. Cfa-miRNA-122 and -34a expression discriminated the diseased groups from the control group better than traditional serum-derived liver biomarkers, as evidenced by areas under the receiver operating characteristic (AUC-ROC) curve of 0.99 and 0.97 for cfa-miRNA-122 expression in the C+HS and C+HF groups, and 1.0 and 0.96 for cfa-miRNA-34a in the C+HS and C+HF groups, respectively. Cfa-miRNA-21 expression was upregulated only in the C+HF group compared with the C+HS (P < .01) and control (P < .001) groups and showed a positive correlation with serum ALT, AST, TBIL, ALP, and GGT and negative correlation with serum TC and TG levels. Cfa-miRNA-21 expression could also differentiate the C+HF group from the control and C+HS groups with a diagnostic performance superior to that of the conventional serum biochemical variables as evidenced by AUCs of 1.0 and 0.98, respectively. CONCLUSIONS Serum cfa-miRNA-122, -34a, and -21 expression was significantly upregulated in dogs with cholecystolithiasis with hepatic steatosis or fibrosis compared with control dogs. These miRNAs could serve as novel biomarkers for hepatic steatosis or fibrosis, which have been implicated in the pathogenesis of cholecystolithiasis.
Collapse
Affiliation(s)
- Ahmed M El-Sebaey
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt.,Department of Disease Diagnosis, Therapy, Obstetrics, and Animal Reproduction, Moscow State Academy of Veterinary Medicine and Biotechnology - MVA by K. I. Skryabin, Moscow, Russian Federation
| | - Pavel N Abramov
- Department of Disease Diagnosis, Therapy, Obstetrics, and Animal Reproduction, Moscow State Academy of Veterinary Medicine and Biotechnology - MVA by K. I. Skryabin, Moscow, Russian Federation
| |
Collapse
|
17
|
He G, Ding J, Zhang Y, Cai M, Yang J, Cho WC, Zheng Y. microRNA-21: a key modulator in oncogenic viral infections. RNA Biol 2021; 18:809-817. [PMID: 33499700 DOI: 10.1080/15476286.2021.1880756] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Oncogenic viruses are associated with approximately 15% of human cancers. In viral infections, microRNAs play an important role in host-pathogen interactions. miR-21 is a highly conserved non-coding RNA that not only regulates the development of oncogenic viral diseases, but also responds to the regulation of intracellular signal pathways. Oncogenic viruses, including HBV, HCV, HPV, and EBV, co-evolve with their hosts and cause persistent infections. The upregulation of host miR-21 manipulates key cellular pathways to evade host immune responses and then promote viral replication. Thus, a better understanding of the role of miR-21 in viral infections may help us to develop effective genetically-engineered oncolytic virus-based therapies against cancer.
Collapse
Affiliation(s)
- Guitian He
- State Key Laboratory of Veterinary Etiological Biology' and 'Key Laboratory of Veterinary Parasitology of Gansu Province, CAAS, Lanzhou, China
| | - Juntao Ding
- College of Life Science and Technology, College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Yong'e Zhang
- State Key Laboratory of Veterinary Etiological Biology' and 'Key Laboratory of Veterinary Parasitology of Gansu Province, CAAS, Lanzhou, China
| | - Mengting Cai
- State Key Laboratory of Veterinary Etiological Biology' and 'Key Laboratory of Veterinary Parasitology of Gansu Province, CAAS, Lanzhou, China
| | - Jing Yang
- State Key Laboratory of Veterinary Etiological Biology' and 'Key Laboratory of Veterinary Parasitology of Gansu Province, CAAS, Lanzhou, China
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Yadong Zheng
- State Key Laboratory of Veterinary Etiological Biology' and 'Key Laboratory of Veterinary Parasitology of Gansu Province, CAAS, Lanzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou China
| |
Collapse
|
18
|
El-Sebaey AMH, Abramov PN, Borunova SM. Cfa-miRNAs-122 and -21 as modern biomarkers of primary hepatitis in dogs. RUDN JOURNAL OF AGRONOMY AND ANIMAL INDUSTRIES 2020. [DOI: 10.22363/2312-797x-2020-15-3-294-307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Primary hepatitis (PH) is one of the most frequently diagnosed hepatic diseases in dogs. Its popular forms are acute hepatitis (AH) and chronic active hepatitis (CAH) which can progress to hepatic fibrosis and cirrhosis. This investigation aimed to evaluate the use of hepatocyte released Canine familiaris miRNAs(cfamiR)-122 and -21 as serum biological markers for the early and reliable diagnosis of PH and to reveal the onset of hepatic fibrosis. After the ultrasonographic and histological examination, fifteen healthy dogs were involved in the study as control group to compare with other thirty dogs confirmed to have AH or CAH (n = 15). Activity of liver enzymes as well as serum level of globulin and total bilirubin were significantly elevated in AH (P 0.001) and CAH (P 0.01) groups whereas, serum level of total protein, albumin, BUN and A/G ratio were significantly lowered in both PH groups (P 0.01) compared to control. Cfa-miR-122 significantly expressed in AH (P 0.001) and CAH (P 0.01) and exhibited a potential significance in distinguishing these groups from control with an area under the curve (AUC) of 0.98 and 0.96, respectively. Additionally, cfa-miR-122 displayed a potential role in distinguishing AH (P 0.05) from CAH group with an AUC of 0.85. Cfa-miR-21 was only expressed in dogs of CAH group and displayed a potential role in distinguishing this group (P 0.001) from AH and healthy groups with an AUC of 0.99 and 0.88, respectively. Therefore, cfa-miR-122 can be significantly expressed in dogs with two forms of PH whereas, cfa-miR-21 could be potentially enhanced only in chronic form of PH and may act as new non-invasive biomarker for distinguishing AH from CAH.
Collapse
|
19
|
Gao Y, Xi B, Li J, Li Z, Xu J, Zhong M, Xu Q, Lian Y, Wei R, Wang L, Cao H, Jin L, Zhang K, Dong J. Scoparone alleviates hepatic fibrosis by inhibiting the TLR-4/NF-κB pathway. J Cell Physiol 2020; 236:3044-3058. [PMID: 33090488 DOI: 10.1002/jcp.30083] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 01/15/2023]
Abstract
The aim of this study was to investigate the role of scoparone (SCO) in hepatic fibrosis. For this, we conducted in vivo and in vitro experiments. In vivo rats that were divided into six groups, control, carbon tetrachloride, and colchicine, as well as SCO groups, SCO50, SCO100, and SCO200 treated with 50, 100, and 200 mg/kg SCO doses, respectively. Furthermore, SCO was shown to inhibit Toll-like receptor-4 (TLR-4)/nuclear factor kappa-B (NF-κB; TLR-4/NF-κB) signals by inhibiting TLR-4, which in turn downregulates the expression of MyD88, promotes NF-κB inhibitor-α, NF-κB inhibitor-β, and NF-κB inhibitor-ε activation, while inhibiting NF-κB inhibitor-ζ. Subsequently, the decrease of phosphorylation of nuclear factor-κB levels leads to the downregulation of the downstream inflammatory factors' tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and IL-1 beta, thus weakening hepatic fibrosis. Notably, the SCO200 treated group presented the most significant improvement. Hence, we conclude that SCO alleviates hepatic fibrosis by inhibiting TLR-4/NF-κB signals.
Collapse
Affiliation(s)
- Ya Gao
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, China
| | - Boting Xi
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, China
| | - Jiani Li
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, China
| | - Zimeng Li
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, China
| | - Jie Xu
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, China
| | - Mingli Zhong
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, China
| | - Qiongmei Xu
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, China
| | - Yuanyu Lian
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, China
| | - Riming Wei
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, China
| | - Liping Wang
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, China
| | - Houkang Cao
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Ling Jin
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Kefeng Zhang
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, China
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Jianghui Dong
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, China
| |
Collapse
|
20
|
M2 bone marrow-derived macrophage-derived exosomes shuffle microRNA-21 to accelerate immune escape of glioma by modulating PEG3. Cancer Cell Int 2020; 20:93. [PMID: 32231463 PMCID: PMC7099792 DOI: 10.1186/s12935-020-1163-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/04/2020] [Indexed: 02/08/2023] Open
Abstract
Background Growing studies have focused on the role of microRNA-21 (miR-21) in glioma, thus our objective was to discuss the effect of M2 bone marrow-derived macrophage (BMDM)-derived exosomes (BMDM-Exos) shuffle miR-21 on biological functions of glioma cells by regulating paternally expressed gene 3 (PEG3). Methods Seventy-one cases of human glioma tissues and 30 cases of non-tumor normal brain tissues were collected and stored in liquid nitrogen. PEG3 and miR-21 expression in glioma tissues was tested. The fasting venous blood of glioma patients and healthy control was collected and centrifuged, and then the supernatant was stored at - 80 °C refrigerator. The contents of interferon (IFN)-γ and transforming growth factor-β1 (TGF-β1) in serum were tested by ELISA. Glioma cells and normal glial cells were cultured to screen the target cells for further in vitro experiments. BMDM-Exos was obtained by ultra-high speed centrifugation and then was identified. BMDM-Exos was co-cultured with U87 cells to detect the biological functions. The fasting venous blood of glioma patients was extracted and treated with ethylene diamine tetraacetic acid-K2 anti-freezing, and then CD8+T cells were isolated. CD8+T cells were co-cultured with U87 cells to detect the CD8+T proliferation, cell cytotoxic activity, U87 cell activity, as well as IFN-γ and TGF-β1 levels. Moreover, BALB/c-nu/nu mice was taken, and the human-nude mouse glioma orthotopic transplantation model was established with U87 cells, and then mice were grouped to test the trends in tumor growth. The brain of mice (fixed by 10% formaldehyde) was sliced to detect the expression of Ki67 and proliferating cell nuclear antigen (PCNA). The spleen of mice was taken to prepare single-cell suspension, and the percentage of T lymphocytes in spleen to CD8+T cells was detected. Results PEG3 expression was decreased and miR-21 expression was increased in glioma cells and tissues. Depleting miR-21 or restoring PEG3 suppressed growth, migration and invasion as well as accelerated apoptosis of glioma cells, also raised CD8+T proliferation, cell cytotoxic activity, and IFN-γ level as well as decreased U87 cell activity and TGF-β1 level. BMDM-Exos shuttle miR-21 promoted migration, proliferation and invasion as well as suppressed apoptosis of glioma cells by reducing PEG3. Exosomes enhanced the volume of tumor, Ki67 and PCNA expression, reduced the percentage of CD8+T cells in glioma mice. Conclusion BMDM-Exos shuffle miR-21 to facilitate invasion, proliferation and migration as well as inhibit apoptosis of glioma cells via inhibiting PEG3, furthermore, promoting immune escape of glioma cells.
Collapse
|
21
|
M. El-Sebaey A, N. Abramov P, M. Abdelhamid F. Clinical Characteristics, Serum Biochemical Changes, and Expression Profile of Serum Cfa-miRNAs in Dogs Confirmed to Have Congenital Portosystemic Shunts Accompanied by Liver Pathologies. Vet Sci 2020; 7:vetsci7020035. [PMID: 32218339 PMCID: PMC7356535 DOI: 10.3390/vetsci7020035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 03/20/2020] [Accepted: 03/23/2020] [Indexed: 12/24/2022] Open
Abstract
Computed tomography angiography (CTA) and biochemical parameters cannot specify liver pathologies in dogs with congenital portosystemic shunts (CPSS) that are easily determined by invasive histopathology. This study aims to assess the possibility of using circulating serum canine familiaris (cfa) microRNAs (miRNAs) as novel non-invasive serum-based fingerprints for liver injuries associated with various morphologies of extrahepatic and intrahepatic portosystemic shunts (EHPSS and IHPSS). Data were obtained from 12 healthy dogs and 84 dogs confirmed to have EHPSS (splenocaval, splenophrenic, splenoazygos, right gastrocaval (RGC), right gastrocaval with caudal loop (RGC-CL)) and IHPSS (right divisional and left divisional) using CTA. Hepatic pathologies were determined by histopathology. Serum expression of miRNAs was assessed by real-time polymerase chain reaction. Based on the nature of liver injuries in each shunt type, cfa-miR-122 was significantly upregulated in all CPSS groups. Meanwhile, serums cfa-miR-34a and 21 were not significantly expressed in splenophrenic or splenoazygos groups, but they were extensively upregulated in splenocaval, RGC, RGC-CL groups and less frequently in right or left divisional groups. Also, serum cfa-miR126 was significantly upregulated in both IHPSS groups but less significantly expressed in RGC, RGC-CL, and splenocaval groups. Overall, estimated cfa-miRNAs could serve as novel biomarkers to mirror the histopathological and molecular events within the liver in each shunt type.
Collapse
Affiliation(s)
- Ahmed M. El-Sebaey
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt;
- Department of Diagnostics of Diseases, Therapy, Obstetrics and Animal Reproduction, Moscow State Academy of Veterinary Medicine and Biotechnology – MVA named K. I. Skryabin, 109472 Moscow, Russia;
- Correspondence: ; Tel.: +7-966-100-4890
| | - Pavel N. Abramov
- Department of Diagnostics of Diseases, Therapy, Obstetrics and Animal Reproduction, Moscow State Academy of Veterinary Medicine and Biotechnology – MVA named K. I. Skryabin, 109472 Moscow, Russia;
| | - Fatma M. Abdelhamid
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt;
| |
Collapse
|
22
|
POLYMORPHISM OF SMAD7 (RS4939827) AND EIF3H (RS16892766) GENES AS A CRITERIUM OF FIBROSIS PROGRESSION RATE IN PATIENTS WITH CHRONIC HEPATITIS С AND В. WORLD OF MEDICINE AND BIOLOGY 2020. [DOI: 10.26724/2079-8334-2020-4-74-7-10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
23
|
Comparative Pharmacokinetic Study of Taxifolin after Oral Administration of Fructus Polygoni Orientalis Extract in Normal and Fibrotic Rats by UPLC-MS/MS. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:9348075. [PMID: 31976002 PMCID: PMC6955125 DOI: 10.1155/2019/9348075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 01/15/2023]
Abstract
Fructus polygoni orientalis (FPO) is widely used in clinical practice in China, especially in treatment of liver diseases including viral hepatitis, liver fibrosis, and liver cirrhosis. However, its pharmacokinetic (PK) alterations in liver fibrotic rats have rarely been reported. To study whether taxifolin, one of the main flavonoids in FPO can be absorbed into blood after oral administration of FPO extract and to compare the differences in pharmacokinetic parameters of taxifolin to normal and liver fibrotic rats induced by porcine serum (PS), a UPLC-MS/MS method was developed and validated for determination of taxifolin in rat plasma using puerarin as the internal standard (IS). All validation parameters met the acceptance criteria according to regulatory guidelines. The results indicated that after treatment of rats with PS alone for 12 weeks, the liver fibrotic model group was built successfully. The taxifolin can be absorbed into the blood after oral administration of the FPO extract. The C max of taxifolin was 1940 ± 502.2 ng/mL and 2648 ± 208.5 ng/mL (p < 0.05), the AUC0∼t of taxifolin was 4949.7 ± 764.89 h·ng/mL and 6679.9 ± 734.26 h·ng/mL (p < 0.05), the AUC0∼∞ of taxifolin was 5049.4 ± 760.7 and 7095.2 ± 962.3 h·ng/mL (p < 0.05), and the mean residence time (MRT) of taxifolin was 2.46 ± 0.412 h and 3.17 ± 0.039 h (p < 0.05) in the normal and fibrotic model groups, respectively. These results confirmed that the pharmacokinetic parameters of taxifolin are altered in liver fibrosis, manifested as C max, AUC0∼t , AUC0∼∞, and the mean residence time (MRT). It suggested that it is essential to consider the characteristics of pharmacokinetics after oral administration of FPO in liver disease patients.
Collapse
|
24
|
Chen W, Yan X, Yang A, Xu A, Huang T, You H. miRNA-150-5p promotes hepatic stellate cell proliferation and sensitizes hepatocyte apoptosis during liver fibrosis. Epigenomics 2019; 12:53-67. [PMID: 31833387 DOI: 10.2217/epi-2019-0104] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aim: To explore the role of miRNA-150-5p (miR-150-5p) in liver fibrosis. Materials & methods: miRNA expression profiles, CCl4-induced liver fibrosis progression and regression rodent models, quantitative real-time PCR, miR-150-5p mimics and inhibitors, cell proliferation and apoptosis detection, RNA sequencing and bioinformatics analysis were employed. Results: Liver tissue miR-150-5p expression was positively associated with liver fibrosis progression and regression; however, miR-150-5p exhibited a cell-specific expression pattern, namely, it was enhanced in hepatocytes but reduced in hepatic stellate cells (HSCs) during liver fibrosis; miR-150-5p overexpression promoted HSC apoptosis and sensitized hepatocyte apoptosis; miR-150-5p mimic had a larger influence on the transcriptomic stability of HSCs than that of hepatocytes; miR-150-5p mediated activation of interferon signaling pathways might be responsible for HSC apoptosis. Conclusion: miR-150-5p exhibited an opposite regulation and function pattern between HSCs and hepatocytes during liver fibrosis.
Collapse
Affiliation(s)
- Wei Chen
- Experimental & Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China.,Beijing Key Laboratory of Tolerance Induction & Organ Protection in Transplantation, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Xuzhen Yan
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, Beijing, PR China
| | - Aiting Yang
- Experimental & Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China.,Beijing Key Laboratory of Tolerance Induction & Organ Protection in Transplantation, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Anjian Xu
- Experimental & Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China.,Beijing Key Laboratory of Tolerance Induction & Organ Protection in Transplantation, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Tao Huang
- Experimental & Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China.,Beijing Key Laboratory of Tolerance Induction & Organ Protection in Transplantation, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Hong You
- Experimental & Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China.,Beijing Key Laboratory of Tolerance Induction & Organ Protection in Transplantation, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China.,Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, Beijing, PR China
| |
Collapse
|