1
|
Li J, Wu B, Zeng L, Lin Y, Chen Q, Wang H, An L, Zhang J, Chen S, Huang J, Zhan R, Zhang G. Aqueous extract of Amydrium sinense (Engl.) H. Li alleviates hepatic fibrosis by suppressing hepatic stellate cell activation through inhibiting Stat3 signaling. Front Pharmacol 2023; 14:1101703. [PMID: 37383718 PMCID: PMC10293641 DOI: 10.3389/fphar.2023.1101703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 05/30/2023] [Indexed: 06/30/2023] Open
Abstract
Background: The present study aimed to investigate the protective effect of the water extract of Amydrium sinense (Engl.) H. Li (ASWE) against hepatic fibrosis (HF) and clarify the underlying mechanism. Methods: The chemical components of ASWE were analysed by a Q-Orbitrap high-resolution mass spectrometer. In our study, an in vivo hepatic fibrosis mouse model was established via an intraperitoneal injection of olive oil containing 20% CCl4. In vitro experiments were conducted using a hepatic stellate cell line (HSC-T6) and RAW 264.7 cell line. A CCK-8 assay was performed to assess the cell viability of HSC-T6 and RAW264.7 cells treated with ASWE. Immunofluorescence staining was used to examine the intracellular localization of signal transducer and activator of transcription 3 (Stat3). Stat3 was overexpressed to analyse the role of Stat3 in the effect of ASWE on HF. Results: Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses showed that candidate targets of ASWE, associated with protective effects against hepatic fibrosis, were related to inflammation response. ASWE ameliorated CCl4-induced liver pathological damage and reduced the liver index and alanine transaminase (ALT) and aspartate transaminase (AST) levels. ASWE also decreased the serum levels of collagen Ⅰ (Col Ⅰ) and hydroxyproline (Hyp) in CCl4-treated mice. In addition, the expression of fibrosis markers, including α-SMA protein and Acta2, Col1a1, and Col3a1 mRNA, was downregulated by ASWE treatment in vivo. The expression of these fibrosis markers was also decreased by treatment with ASWE in HSC-T6 cells. Moreover, ASWE decreased the expression of inflammatory markers, including the Tnf-α, Il6 and Il1β, in RAW264.7 cells. ASWE decreased the phosphorylation of Stat3 and total Stat3 expression and reduced the mRNA expression of the Stat3 gene in vivo and in vitro. ASWE also inhibited the nuclear shuttling of Stat3. Overexpression of Stat3 weakened the therapeutic effect of ASWE and accelerated the progression of HF. Conclusion: The results show that ASWE protects against CCl4-induced liver injury by suppressing fibrosis, inflammation, HSC activation and the Stat3 signaling pathway, which might lead to a new approach for preventing HF.
Collapse
Affiliation(s)
- Jingyan Li
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Bingmin Wu
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lishan Zeng
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ying Lin
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Qiuhe Chen
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Haixia Wang
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lin An
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jiajun Zhang
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Siyan Chen
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Junying Huang
- College of Life Sciences, Guangzhou University, Guangzhou, Guangdong, China
| | - Ruoting Zhan
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Key Laboratory of Chinese Medicinal Resource from Lingnan, Ministry of Education, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guifang Zhang
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People’s Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Key Laboratory of Chinese Medicinal Resource from Lingnan, Ministry of Education, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
2
|
Zhai H, Zhang J, Shang D, Zhu C, Xiang X. The progress to establish optimal animal models for the study of acute-on-chronic liver failure. Front Med (Lausanne) 2023; 10:1087274. [PMID: 36844207 PMCID: PMC9947362 DOI: 10.3389/fmed.2023.1087274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Acute-on-chronic liver failure (ACLF) defines a complicated and multifaceted syndrome characterized by acute liver dysfunction following an acute insult on the basis of chronic liver diseases. It is usually concurrent with bacterial infection and multi-organ failure resulting in high short-term mortality. Based on the cohort studies in ACLF worldwide, the clinical course of ACLF was demonstrated to comprise three major stages including chronic liver injury, acute hepatic/extrahepatic insult, and systemic inflammatory response caused by over-reactive immune system especially bacterial infection. However, due to the lack of optimal experimental animal models for ACLF, the progress of basic study on ACLF is limping. Though several experimental ACLF models were established, none of them can recapitulate and simulate the whole pathological process of ACLF patients. Recently, we have developed a novel mouse model for ACLF combining chronic liver injury [injection of carbon tetrachloride (CCl4) for 8 weeks], acute hepatic insult (injection of a double dose CCl4), and bacterial infection (intraperitoneal injection of Klebsiella pneumoniae), which could recapitulate the major clinical features of patients with ACLF worsened by bacterial infection.
Collapse
Affiliation(s)
- Hengben Zhai
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Translational Lab of Liver Diseases, Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinming Zhang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Translational Lab of Liver Diseases, Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dabao Shang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Translational Lab of Liver Diseases, Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuanwu Zhu
- Department of Infectious Diseases, The Fifth People’s Hospital of Suzhou, Suzhou, China,Chuanwu Zhu,
| | - Xiaogang Xiang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Translational Lab of Liver Diseases, Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,*Correspondence: Xiaogang Xiang,
| |
Collapse
|
3
|
Qin L, Liu N, Bao CLM, Yang DZ, Ma GX, Yi WH, Xiao GZ, Cao HL. Mesenchymal stem cells in fibrotic diseases-the two sides of the same coin. Acta Pharmacol Sin 2023; 44:268-287. [PMID: 35896695 PMCID: PMC9326421 DOI: 10.1038/s41401-022-00952-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 06/29/2022] [Indexed: 02/06/2023]
Abstract
Fibrosis is caused by extensive deposition of extracellular matrix (ECM) components, which play a crucial role in injury repair. Fibrosis attributes to ~45% of all deaths worldwide. The molecular pathology of different fibrotic diseases varies, and a number of bioactive factors are involved in the pathogenic process. Mesenchymal stem cells (MSCs) are a type of multipotent stem cells that have promising therapeutic effects in the treatment of different diseases. Current updates of fibrotic pathogenesis reveal that residential MSCs may differentiate into myofibroblasts which lead to the fibrosis development. However, preclinical and clinical trials with autologous or allogeneic MSCs infusion demonstrate that MSCs can relieve the fibrotic diseases by modulating inflammation, regenerating damaged tissues, remodeling the ECMs, and modulating the death of stressed cells after implantation. A variety of animal models were developed to study the mechanisms behind different fibrotic tissues and test the preclinical efficacy of MSC therapy in these diseases. Furthermore, MSCs have been used for treating liver cirrhosis and pulmonary fibrosis patients in several clinical trials, leading to satisfactory clinical efficacy without severe adverse events. This review discusses the two opposite roles of residential MSCs and external MSCs in fibrotic diseases, and summarizes the current perspective of therapeutic mechanism of MSCs in fibrosis, through both laboratory study and clinical trials.
Collapse
Affiliation(s)
- Lei Qin
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000 China
| | - Nian Liu
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000 China
| | - Chao-le-meng Bao
- CASTD Regengeek (Shenzhen) Medical Technology Co. Ltd, Shenzhen, 518000 China
| | - Da-zhi Yang
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000 China
| | - Gui-xing Ma
- grid.263817.90000 0004 1773 1790Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055 China
| | - Wei-hong Yi
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000 China
| | - Guo-zhi Xiao
- grid.263817.90000 0004 1773 1790Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055 China
| | - Hui-ling Cao
- grid.263817.90000 0004 1773 1790Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055 China
| |
Collapse
|
4
|
Hepatoprotective Effect of a New FFAR1 Agonist-N-Alkylated Isobornylamine. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28010396. [PMID: 36615590 PMCID: PMC9823450 DOI: 10.3390/molecules28010396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 12/26/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023]
Abstract
Free fatty acid receptor-1 (FFAR1) is one of the possible therapeutic targets in the search for new hepatoprotective drugs. FFAR1 agonists were found to have hypolipidemic, antifibrotic, anti-inflammatory, antiproliferative and antioxidant effects in addition to hypoglycemic action. In this work, we conducted a study of the hepatoprotective effect of the compound QS-528 (previously discovered as an agonist of FFAR1) at doses of 60, 90, 120 and 150 mg/kg on carbon tetrachloride (CCl4)-induced liver injury. At the end of the experiment, a biochemical blood assay demonstrated that the introduction of QS-528 dose-dependently reduces the levels of liver enzymes (AST, ALT and ALKP). Histological and morphometric studies of animals' livers treated with QS-528 at doses of 120 and 150 mg/kg showed a decrease in degenerative/necrotic changes in hepatocytes and an increase in the regenerative activity of the liver. In addition, no toxicity at a single oral dose of 1000 mg/kg and an increase in HepG2 cell viability in vitro were found. Thus, the compound QS-528 was found to exhibit a hepatoprotective effect against CCl4-induced toxic liver damage.
Collapse
|
5
|
Xue L, Li X, Zhu X, Zhang J, Zhou S, Tang W, Chen D, Chen Y, Dai J, Wu M, Wu M, Wang S. Carbon tetrachloride exposure induces ovarian damage through oxidative stress and inflammatory mediated ovarian fibrosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 242:113859. [PMID: 35816842 DOI: 10.1016/j.ecoenv.2022.113859] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/02/2022] [Accepted: 07/03/2022] [Indexed: 06/15/2023]
Abstract
Carbon tetrachloride (CCL4) is widely used as a chemical intermediate and as a feedstock in the production of chlorofluorocarbons. CCL4 is highly toxic in the liver, kidney, testicle, brain and other tissues. However, the effect of CCL4 on ovarian function has not been reported. In this study, we found that the mice treated with CCL4 showed decreased ovarian function with disturbed estrus cycle, decreased serum level of 17β-estradiol and the reduced number of healthy follicles. Ovarian damage was accompanied by oxidative stress and the production of proinflammatory cytokines, especially interleukins. The indicators of oxidative stress, 4-Hydroxynonenal (4-HNE), 8-hydroxy-2´-deoxyguanosine (8-OHdG), 3-Nitrotyrosine (3-NT) and malondialdehyde (MDA), and the levels of proinflammatory cytokines IL-1α, IL-1β, IL-6 and IL-11 were increased, while the antioxidants, including superoxide dismutase (SOD), nuclear factor erythroid2-related factor 2 (NRF2) and heme oxygenase-1 (HO-1), were decreased in the CCL4 group. In the CCL4 treated group, the results of Sirius Red staining, immunohistochemistry and qPCR indicated that proinflammatory cytokines caused further ovarian fibrosis. And CCL4 could also promote ovarian thecal cells to secrete inflammatory cytokines, resulting in fibrosis in vitro. In addition, CCL4 inhibited oocyte development and triggered oocyte apoptosis. In conclusion, CCL4 exposure causes ovarian damage by strong oxidative stress and the high expression of the proinflammatory cytokine mediated ovarian fibrosis.
Collapse
Affiliation(s)
- Liru Xue
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China
| | - Xiang Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China; Department of Obstetrics and Gynecology, Xiehe Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoran Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China
| | - Su Zhou
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China
| | - Weicheng Tang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China
| | - Dan Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China
| | - Yingying Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China.
| | - Mingfu Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China.
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China.
| |
Collapse
|
6
|
Igf2bp2 knockdown improves CCl 4-induced liver fibrosis and TGF-β-activated mouse hepatic stellate cells by regulating Tgfbr1. Int Immunopharmacol 2022; 110:108987. [PMID: 35820364 DOI: 10.1016/j.intimp.2022.108987] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 06/12/2022] [Accepted: 06/17/2022] [Indexed: 01/18/2023]
Abstract
Progressive liver fibrosis is a dynamic process characterized by the net accumulation of extracellular matrix (ECM), which could eventually develop into cirrhosis, leading to malignant transformation. In this study, insulin-like growth factor 2 mRNA binding protein 2 (Igf2bp2) was found to be up-regulated in carbon tetrachloride (CCl4)-induced liver fibrosis and transforming growth factor-beta 1 (TGF-β)-activated hepatic stellate cells (HSCs). Igf2bp2 knockdown in the CCl4-induced hepatic fibrosis mice model significantly improved CCl4-induced liver damage by decreasing necrosis and fibrotic septa, reducing hydroxyproline levels, and down-regulating fibrotic markers levels. In TGF-β-activated HSCs, Igf2bp2 knockdown partially attenuated TGF-β-induced cellular effects by suppressing HSCs viability and DNA synthesis and reducing the ECM-associated factors such as α-SMA, COLLAGEN I, and COLLAGEN III. Integrative network and signaling analysis revealed that the Igf2bp2 could bind to Tgfbr1. Transforming growth factor-beta receptor 1 (Tgfbr1) was found to be significantly up-regulated in the fibrotic liver and activated HSCs, and positively correlated with Igf2bp2. Tgfbr1 knockdown partially eliminated TGF-β-induced fibrotic changes and Igf2bp2 overexpression effects on TGF-β-activated HSCs in vitro. Moreover, Igf2bp2 overexpression promoted the phosphorylation of SMAD2/SMAD3, AKT, and PI3K, whereas Tgfbr1 knockdown exhibited the opposite effect; Tgfbr1 knockdown also partially attenuated the effects of Igf2bp2 overexpression on the phosphorylation of SMAD2/SMAD3, AKT, and PI3K. In closing, Igf2bp2 and Tgfbr1 are up-regulated in CCl4-induced liver fibrosis and TGF-β-activated mHSCs. Igf2bp2 knockdown improved CCl4-induced liver fibrosis and TGF-β-activated HSCs by targeting Tgfbr1, possibly through the PI3K/Akt pathway.
Collapse
|
7
|
Faccioli LA, Dias ML, Paranhos BA, dos Santos Goldenberg RC. Liver cirrhosis: An overview of experimental models in rodents. Life Sci 2022; 301:120615. [DOI: 10.1016/j.lfs.2022.120615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/29/2022] [Accepted: 05/02/2022] [Indexed: 02/07/2023]
|
8
|
Song YR, Jang MH, Jang B, Bae SJ, Bak SB, Lee SM, Yun UJ, Lee JH, Park SM, Jung DH, Sa BS, Song JK, Lee EH, Kim KY, Park KI, Kim YW, Kim SC. Jageum-Jung, the herbal pharmaceuticals, inhibits the hepatic fibrogenesis as mediated with TGF-β1/smad signaling. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-021-00196-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
9
|
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: 12] [Impact Index Per Article: 3.0] [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
|
10
|
Schonfeld M, O’Neil M, Villar MT, Artigues A, Averilla J, Gunewardena S, Weinman SA, Tikhanovich I. A Western diet with alcohol in drinking water recapitulates features of alcohol-associated liver disease in mice. Alcohol Clin Exp Res 2021; 45:1980-1993. [PMID: 34523155 PMCID: PMC9006178 DOI: 10.1111/acer.14700] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Mouse models of alcohol-associated liver disease vary greatly in their ease of implementation and the pathology they produce. Effects range from steatosis and mild inflammation with the Lieber-DeCarli liquid diet to severe inflammation, fibrosis, and pyroptosis seen with the Tsukamoto-French intragastric feeding model. Implementation of all of these models is limited by the labor-intensive nature of the protocols and the specialized skills necessary for successful intragastric feeding. We thus sought to develop a new model to reproduce features of alcohol-induced inflammation and fibrosis with minimal operational requirements. METHODS Over a 16-week period, mice were fed ad libitum with a pelleted high-fat Western diet (WD; 40% calories from fat) and alcohol added to the drinking water. We found the optimal alcohol consumption to be that at which the alcohol concentration was 20% for 4 days and 10% for 3 days per week. Control mice received WD pellets with water alone. RESULTS Alcohol consumption was 18 to 20 g/kg/day in males and 20 to 22 g/kg/day in females. Mice in the alcohol groups developed elevated serum transaminase levels after 12 weeks in males and 10 weeks in females. At 16 weeks, both males and females developed liver inflammation, steatosis, and pericellular fibrosis. Control mice on WD without alcohol had mild steatosis only. Alcohol-fed mice showed reduced HNF4α mRNA and protein expression. HNF4α is a master regulator of hepatocyte differentiation, down-regulation of which is a known driver of hepatocellular failure in alcoholic hepatitis. CONCLUSION A simple-to-administer, 16-week WD alcohol model recapitulates the inflammatory, fibrotic, and gene expression aspects of human alcohol-associated steatohepatitis.
Collapse
Affiliation(s)
- Michael Schonfeld
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, U.S.A
| | - Maura O’Neil
- Department of Pathology, University of Kansas Medical Center, Kansas City, KS 66160, U.S.A
- Liver Center, University of Kansas Medical Center, Kansas City, KS 66160, U.S.A
| | - Maria T Villar
- Department of Biochemistry, University of Kansas Medical Center, Kansas City, KS 66160, U.S.A
| | - Antonio Artigues
- Department of Biochemistry, University of Kansas Medical Center, Kansas City, KS 66160, U.S.A
| | - Janice Averilla
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, U.S.A
| | - Sumedha Gunewardena
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, U.S.A
| | - Steven A. Weinman
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, U.S.A
- Liver Center, University of Kansas Medical Center, Kansas City, KS 66160, U.S.A
- Kansas City VA Medical Center, Kansas City, MO, USA
| | - Irina Tikhanovich
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, U.S.A
- Liver Center, University of Kansas Medical Center, Kansas City, KS 66160, U.S.A
| |
Collapse
|
11
|
Kim M, Hur S, Kim KH, Cho Y, Kim K, Kim HR, Nam KT, Lim KM. A New Murine Liver Fibrosis Model Induced by Polyhexamethylene Guanidine-Phosphate. Biomol Ther (Seoul) 2021; 30:126-136. [PMID: 34580237 PMCID: PMC8902451 DOI: 10.4062/biomolther.2021.120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 11/05/2022] Open
Abstract
Liver fibrosis is part of the wound healing process to help the liver recover from the injuries caused by various liver-damaging insults. However, liver fibrosis often progresses to life-threatening cirrhosis and hepatocellular carcinoma. To overcome the limitations of current in vivo liver fibrosis models for studying the pathophysiology of liver fibrosis and establishing effective treatment strategies, we developed a new mouse model of liver fibrosis using polyhexamethylene guanidine phosphate (PHMG-p), a humidifier sterilizer known to induce lung fibrosis in humans. Male C57/BL6 mice were intraperitoneally injected with PHMG-p (0.03% and 0.1%) twice a week for 5 weeks. Subsequently, liver tissues were examined histologically and RNA-sequencing was performed to evaluate the expression of key genes and pathways affected by PHMG-p. PHMG-p injection resulted in body weight loss of ~15% and worsening of physical condition. Necropsy revealed diffuse fibrotic lesions in the liver with no effect on the lungs. Histology, collagen staining, immunohistochemistry for smooth muscle actin and collagen, and polymerase chain reaction analysis of fibrotic genes revealed that PHMG-p induced liver fibrosis in the peri-central, peri-portal, and capsule regions. RNA-sequencing revealed that PHMG-p affected several pathways associated with human liver fibrosis, especially with upregulation of lumican and IRAK3, and downregulation of GSTp1 and GSTp2, which are closely involved in liver fibrosis pathogenesis. Collectively we demonstrated that the PHMG-p-induced liver fibrosis model can be employed to study human liver fibrosis.
Collapse
Affiliation(s)
- Minjeong Kim
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Sumin Hur
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| | - Kwang H Kim
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| | - Yejin Cho
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| | - Keunyoung Kim
- College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Ha Ryong Kim
- College of Pharmacy, Daegu Catholic University, Daegu 38430, Republic of Korea
| | - Ki Taek Nam
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| | - Kyung-Min Lim
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
12
|
Choi JH, Park S, Kim GD, Kim JY, Jun JH, Bae SH, Baik SK, Hwang SG, Kim GJ. Increased Phosphatase of Regenerating Liver-1 by Placental Stem Cells Promotes Hepatic Regeneration in a Bile-Duct-Ligated Rat Model. Cells 2021; 10:cells10102530. [PMID: 34685509 PMCID: PMC8533985 DOI: 10.3390/cells10102530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 11/16/2022] Open
Abstract
Phosphatase of regenerating liver-1 (PRL-1) controls various cellular processes and liver regeneration. However, the roles of PRL-1 in liver regeneration induced by chorionic-plate-derived mesenchymal stem cells (CP-MSCs) transplantation remain unknown. Here, we found that increased PRL-1 expression by CP-MSC transplantation enhanced liver regeneration in a bile duct ligation (BDL) rat model by promoting the migration and proliferation of hepatocytes. Engrafted CP-MSCs promoted liver function via enhanced hepatocyte proliferation through increased PRL-1 expression in vivo and in vitro. Moreover, higher increased expression of PRL-1 regulated CP-MSC migration into BDL-injured rat liver through enhancement of migration-related signals by increasing Rho family proteins. The dual effects of PRL-1 on proliferation of hepatocytes and migration of CP-MSCs were substantially reduced when PRL-1 was silenced with siRNA-PRL-1 treatment. These findings suggest that PRL-1 may serve as a multifunctional enhancer for therapeutic applications of CP-MSC transplantation.
Collapse
Affiliation(s)
- Jong Ho Choi
- Department of Oral Pathology, College of Dentistry, Gangneung-Wonju National University, Gangneung-si 25457, Korea;
| | - Sohae Park
- Department of Biomedical Science, CHA University, Seongnam-si 13488, Korea; (S.P.); (J.Y.K.); (J.H.J.)
- Research Institute of Placental Science, CHA University, Seongnam-si 13488, Korea
| | - Gi Dae Kim
- Department of Food and Nutrition, Kyungnam University, Changwon-si 51767, Korea;
| | - Jae Yeon Kim
- Department of Biomedical Science, CHA University, Seongnam-si 13488, Korea; (S.P.); (J.Y.K.); (J.H.J.)
| | - Ji Hye Jun
- Department of Biomedical Science, CHA University, Seongnam-si 13488, Korea; (S.P.); (J.Y.K.); (J.H.J.)
- Research Institute of Placental Science, CHA University, Seongnam-si 13488, Korea
| | - Si Hyun Bae
- Department of Internal Medicine, Catholic University Medical College, Seoul 03312, Korea;
| | - Soon Koo Baik
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Korea;
| | - Seong-Gyu Hwang
- CHA Bundang Medical Center, Department of Internal Medicine, Division of Gastroenterology, CHA University School of Medicine, Seongnam-si 13496, Korea;
| | - Gi Jin Kim
- Department of Biomedical Science, CHA University, Seongnam-si 13488, Korea; (S.P.); (J.Y.K.); (J.H.J.)
- Research Institute of Placental Science, CHA University, Seongnam-si 13488, Korea
- Correspondence: ; Tel.: +82-31-881-7145
| |
Collapse
|
13
|
Panter KE, Stegelmeier BL, Gardner DR, Stonecipher CA, Lee ST, Kitchen D, Brackett A, Davis C. Clinical, pathologic, and toxicologic characterization of Salvia reflexa (lance-leaf sage) poisoning in cattle fed contaminated hay. J Vet Diagn Invest 2021; 33:538-547. [PMID: 33719809 DOI: 10.1177/1040638721995784] [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] [Indexed: 11/15/2022] Open
Abstract
Salvia reflexa (lance-leaf sage)-contaminated alfalfa hay was fed to ~500 mixed-breed beef cattle. Within hours of exposure, nearly half of the cattle developed lethargy, anorexia, depression, and recumbency, followed by bellowing, colic, and death. Even though the uneaten contaminated hay was removed the first day, nearly 100 animals died within the first 48 h. Three of these cattle were examined postmortem, and tissues and hay samples were collected for microscopic and chemical analysis. Several days later, a smaller number of the clinically poisoned cattle developed neurologic disease with aberrant behavior, aggression, icterus, blindness, exhaustion, and death. A total of 165 cattle were fatally poisoned. Poisoned cattle had swollen, dark, mottled livers that had a prominent nutmeg-like lobular pattern on cut section. Histologically, there was severe centrilobular-to-panlobular hepatic necrosis with marked hepatocellular swelling, degeneration, and necrosis. The surviving cattle developed liver disease characterized by altered serum biochemical analyses and microscopic hepatocellular degeneration and necrosis. In subsequent biopsies and analysis, these lesions resolved within 6-7 mo. After confirming toxicity of the hay in cattle, goats, and mice, followed by a mouse bioassay-guided chemical fractionation process, Salvia reflexa was identified as the contaminant in the hay responsible for the hepatotoxicity. S. reflexa has not been reported previously to cause fatal hepatotoxicity in livestock in North America, to our knowledge.
Collapse
Affiliation(s)
- Kip E Panter
- USDA-Agricultural Research Service, Poisonous Plant Research Laboratory, North, Logan, UT
| | - Bryan L Stegelmeier
- USDA-Agricultural Research Service, Poisonous Plant Research Laboratory, North, Logan, UT
| | - Dale R Gardner
- USDA-Agricultural Research Service, Poisonous Plant Research Laboratory, North, Logan, UT
| | - Clinton A Stonecipher
- USDA-Agricultural Research Service, Poisonous Plant Research Laboratory, North, Logan, UT
| | - Stephen T Lee
- USDA-Agricultural Research Service, Poisonous Plant Research Laboratory, North, Logan, UT
| | | | | | - Charlie Davis
- Colorado State University, Veterinary Diagnostic Laboratory, Fort Collins, CO
| |
Collapse
|
14
|
Tang W, Chen X, Liu D, Xie J. Bioactive Components of Mesona Blume and Their Potential Health Benefits. FOOD REVIEWS INTERNATIONAL 2020. [DOI: 10.1080/87559129.2020.1849271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Wei Tang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
| | - Xianxiang Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
| | - Dan Liu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
| | - Jianhua Xie
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
15
|
Silva J, Yu X, Moradian R, Folk C, Spatz MH, Kim P, Bhatti AA, Davies DL, Liang J. Dihydromyricetin Protects the Liver via Changes in Lipid Metabolism and Enhanced Ethanol Metabolism. Alcohol Clin Exp Res 2020; 44:1046-1060. [PMID: 32267550 PMCID: PMC7211127 DOI: 10.1111/acer.14326] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/03/2020] [Indexed: 12/13/2022]
Abstract
Background Excess alcohol (ethanol, EtOH) consumption is a significant cause of chronic liver disease, accounting for nearly half of the cirrhosis‐associated deaths in the United States. EtOH‐induced liver toxicity is linked to EtOH metabolism and its associated increase in proinflammatory cytokines, oxidative stress, and the subsequent activation of Kupffer cells. Dihydromyricetin (DHM), a bioflavonoid isolated from Hovenia dulcis, can reduce EtOH intoxication and potentially protect against chemical‐induced liver injuries. But there remains a paucity of information regarding the effects of DHM on EtOH metabolism and liver protection. As such, the current study tests the hypothesis that DHM supplementation enhances EtOH metabolism and reduces EtOH‐mediated lipid dysregulation, thus promoting hepatocellular health. Methods The hepatoprotective effect of DHM (5 and 10 mg/kg; intraperitoneal injection) was evaluated using male C57BL/6J mice and a forced drinking ad libitum EtOH feeding model and HepG2/VL‐17A hepatoblastoma cell models. EtOH‐mediated lipid accumulation and DHM effects against lipid deposits were determined via H&E stains, triglyceride measurements, and intracellular lipid dyes. Protein expression of phosphorylated/total proteins and serum and hepatic cytokines was determined via Western blot and protein array. Total NAD+/NADH Assay of liver homogenates was used to detect NAD + levels. Results DHM reduced liver steatosis, liver triglycerides, and liver injury markers in mice chronically fed EtOH. DHM treatment resulted in increased activation of AMPK and downstream targets, carnitine palmitoyltransferase (CPT)‐1a, and acetyl CoA carboxylase (ACC)‐1. DHM induced expression of EtOH‐metabolizing enzymes and reduced EtOH and acetaldehyde concentrations, effects that may be partly explained by changes in NAD+. Furthermore, DHM reduced the expression of proinflammatory cytokines and chemokines in sera and cell models. Conclusion In total, these findings support the utility of DHM as a dietary supplement to reduce EtOH‐induced liver injury via changes in lipid metabolism, enhancement of EtOH metabolism, and suppressing inflammation responses to promote liver health.
Collapse
Affiliation(s)
- Joshua Silva
- From the, Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Xin Yu
- From the, Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Renita Moradian
- From the, Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Carson Folk
- From the, Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Maximilian H Spatz
- From the, Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Phoebe Kim
- From the, Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Adil A Bhatti
- From the, Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Daryl L Davies
- From the, Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Jing Liang
- From the, Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, California
| |
Collapse
|
16
|
Lelis DDF, Freitas DFD, Machado AS, Crespo TS, Santos SHS. Angiotensin-(1-7), Adipokines and Inflammation. Metabolism 2019; 95:36-45. [PMID: 30905634 DOI: 10.1016/j.metabol.2019.03.006] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/11/2019] [Accepted: 03/20/2019] [Indexed: 12/12/2022]
Abstract
Nowadays the adipose tissue is recognized as one of the most critical endocrine organs releasing many adipokines that regulate metabolism, inflammation and body homeostasis. There are several described adipokines, including the renin-angiotensin system (RAS) components that are especially activated in some diseases with increased production of angiotensin II and several pro-inflammatory hormones. On the other hand, RAS also expresses angiotensin-(1-7), which is now recognized as the main peptide on counteracting Ang II effects. New studies have shown that increased activation of ACE2/Ang-(1-7)/MasR arm can revert and prevent local and systemic dysfunctions improving lipid profile and insulin resistance by modulating insulin actions, and reducing inflammation. In this context, the present review shows the interaction and relevance of Ang-(1-7) effects on regulating adipokines, and as one adipokine itself, modulating body homeostasis, with emphasis on its anti-inflammatory properties, especially in the context of metabolic disorders with focus on obesity and type 2 diabetes mellitus pandemic.
Collapse
Affiliation(s)
- Deborah de Farias Lelis
- Laboratory of Health Sciences, Post Graduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Daniela Fernanda de Freitas
- Laboratory of Health Sciences, Post Graduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Amanda Souto Machado
- Laboratory of Health Sciences, Post Graduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Thaísa Soares Crespo
- Laboratory of Health Sciences, Post Graduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Sérgio Henrique Sousa Santos
- Institute of Agricultural Sciences, Food Engineering College, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil; Laboratory of Health Sciences, Post Graduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil.
| |
Collapse
|
17
|
Iron-Induced Liver Injury: A Critical Reappraisal. Int J Mol Sci 2019; 20:ijms20092132. [PMID: 31052166 PMCID: PMC6539962 DOI: 10.3390/ijms20092132] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/25/2019] [Accepted: 04/27/2019] [Indexed: 12/12/2022] Open
Abstract
Iron is implicated in the pathogenesis of a number of human liver diseases. Hereditary hemochromatosis is the classical example of a liver disease caused by iron, but iron is commonly believed to contribute to the progression of other forms of chronic liver disease such as hepatitis C infection and nonalcoholic fatty liver disease. In this review, we present data from cell culture experiments, animal models, and clinical studies that address the hepatotoxicity of iron. These data demonstrate that iron overload is only weakly fibrogenic in animal models and rarely causes serious liver damage in humans, calling into question the concept that iron overload is an important cause of hepatotoxicity. In situations where iron is pathogenic, iron-induced liver damage may be potentiated by coexisting inflammation, with the resulting hepatocyte necrosis an important factor driving the fibrogenic response. Based on the foregoing evidence that iron is less hepatotoxic than is generally assumed, claims that assign a causal role to iron in liver injury in either animal models or human liver disease should be carefully evaluated.
Collapse
|
18
|
Protective effect of cultured bear bile powder against dimethylnitrosamine-induced hepatic fibrosis in rats. Biomed Pharmacother 2019; 112:108701. [DOI: 10.1016/j.biopha.2019.108701] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 02/14/2019] [Accepted: 02/19/2019] [Indexed: 02/07/2023] Open
|
19
|
Yoshioka H, Nonogaki T, Fukaya S, Ichimaru Y, Nagatsu A, Yoshikawa M, Fujii H, Nakao M. Sasa veitchii extract protects against carbon tetrachloride-induced hepatic fibrosis in mice. Environ Health Prev Med 2018; 23:49. [PMID: 30322375 PMCID: PMC6190662 DOI: 10.1186/s12199-018-0739-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/01/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The current study aimed to investigate the hepatoprotective effects of Sasa veitchii extract (SE) on carbon tetrachloride (CCl4)-induced liver fibrosis in mice. METHODS Male C57BL/6J mice were intraperitoneally injected with CCl4 dissolved in olive oil (1 g/kg) twice per week for 8 weeks. SE (0.1 mL) was administered orally once per day throughout the study, and body weight was measured weekly. Seventy-two hours after the final CCl4 injection, mice were euthanized and plasma samples were collected. The liver and kidneys were collected and weighed. RESULTS CCl4 administration increased liver weight, decreased body weight, elevated plasma alanine aminotransferase, and aspartate aminotransferase and increased liver oxidative stress (malondialdehyde and glutathione). These increases were attenuated by SE treatment. Overexpression of tumor necrosis factor-α was also reversed following SE treatment. Furthermore, CCl4-induced increases in α-smooth muscle actin, a marker for hepatic fibrosis, were attenuated in mice treated with SE. Moreover, SE inhibited CCl4-induced nuclear translocation of hepatic nuclear factor kappa B (NF-κB) p65 and phosphorylation of mitogen-activated protein kinase (MAPK). CONCLUSION These results suggested that SE prevented CCl4-induced hepatic fibrosis by inhibiting the MAPK and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Hiroki Yoshioka
- College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori, Moriyamaku, Nagoya, Aichi, 463-8521, Japan.
| | - Tsunemasa Nonogaki
- College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori, Moriyamaku, Nagoya, Aichi, 463-8521, Japan
| | - Shiori Fukaya
- College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori, Moriyamaku, Nagoya, Aichi, 463-8521, Japan
| | - Yoshimi Ichimaru
- College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori, Moriyamaku, Nagoya, Aichi, 463-8521, Japan
| | - Akito Nagatsu
- College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori, Moriyamaku, Nagoya, Aichi, 463-8521, Japan
| | - Masae Yoshikawa
- College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori, Moriyamaku, Nagoya, Aichi, 463-8521, Japan
| | - Hirohisa Fujii
- College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori, Moriyamaku, Nagoya, Aichi, 463-8521, Japan
| | - Makoto Nakao
- College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori, Moriyamaku, Nagoya, Aichi, 463-8521, Japan
| |
Collapse
|
20
|
Königshofer P, Brusilovskaya K, Schwabl P, Reiberger T. Animal models of portal hypertension. Biochim Biophys Acta Mol Basis Dis 2018; 1865:1019-1030. [PMID: 30055295 DOI: 10.1016/j.bbadis.2018.07.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 07/14/2018] [Accepted: 07/16/2018] [Indexed: 12/12/2022]
Abstract
Chronic liver diseases ultimately lead to cirrhosis and portal hypertension (PHT). Indeed, PHT is a major cause of severe complications, while medical treatment is limited to non-selective beta blockers. Sophisticated animal models are needed to investigate novel treatment options for different etiologies of liver disease, effective anti-fibrotic agents as well as vasoactive drugs against PHT. In this review, we present some of the most common animal models of liver disease and PHT - including pre-hepatic, intra-hepatic and post-hepatic PHT in rodents. Methodology for induction, considerations for disease etiology, advantages and limitations and practical issues of these animal models are discussed. The appropriate and sensible use of animal models in preclinical research supporting the 3R concept of replacement, reduction and refinement is highlighted.
Collapse
Affiliation(s)
- P Königshofer
- Div. of Gastroenterology and Hepatology, Dept. of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Vienna Hepatic Hemodynamic Lab, Medical University of Vienna, Vienna, Austria
| | - K Brusilovskaya
- Div. of Gastroenterology and Hepatology, Dept. of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Vienna Hepatic Hemodynamic Lab, Medical University of Vienna, Vienna, Austria
| | - P Schwabl
- Div. of Gastroenterology and Hepatology, Dept. of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Vienna Hepatic Hemodynamic Lab, Medical University of Vienna, Vienna, Austria
| | - T Reiberger
- Div. of Gastroenterology and Hepatology, Dept. of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Vienna Hepatic Hemodynamic Lab, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
21
|
Yuan D, Xiang T, Huo Y, Liu C, Wang T, Zhou Z, Dun Y, Zhao H, Zhang C. Preventive effects of total saponins of Panax japonicus on fatty liver fibrosis in mice. Arch Med Sci 2018; 14:396-406. [PMID: 29593815 PMCID: PMC5868672 DOI: 10.5114/aoms.2016.63260] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 12/14/2015] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Nonalcoholic fatty liver disease (NAFLD) is a condition in which excess fat accumulates in the liver of a patient without a history of alcohol abuse. Fatty liver fibrosis, a severe form of NAFLD, is a key step which can be reversed by effective medical intervention. This paper aims to describe the protective role and mechanisms of action of total saponins of Panax japonicus (SPJ) against fatty liver fibrosis in mice. In this study, fatty liver fibrosis was induced by a high-fat (HF) diet combined with intraperitoneal injection of porcine serum. MATERIAL AND METHODS The fatty liver fibrosis model was induced by HF diet combined with intraperitoneal injection of porcine serum. The endoplasmic reticulum stress (ERS) response and C/EBP homologous protein (CHOP) and p-Jun N-terminal kinase (JNK)-mediated apoptosis and inflammation were assessed by serum biochemistry, hematoxylin-eosin (H + E), Masson and electronic microscopy staining, Hyp content detection, Western blotting and real time polymerase chain reaction (RT-PCR). RESULTS Saponins of Panax japonicus could significantly improve liver function and decrease the lipid level in the serum. The liver steatosis, collagen fibers and inflammatory cell infiltration were significantly improved in the SPJ group according to microscope observation. The RT-PCR analysis revealed that the collagen I (Coll), α smooth muscle actin (α-SMA), tissue inhibitors of MMPs (TIMP), CHOP and GRP78 mRNA expression levels were distinctly weakened by SPJ treatment; and western blotting analysis indicated that the phosphorylated JNK (p-JNK), Coll and 78 kD glucose-regulated protein (GRP78) protein expression levels were significantly alleviated, which might be associated with the inhibition of the ERS response and the CHOP and JNK-mediated apoptosis and inflammation pathway. CONCLUSIONS Based on this research, SPJ as a preventive medicine has great potential in prevention of liver fibrosis.
Collapse
Affiliation(s)
- Ding Yuan
- Medical College of China Three Gorges University, Yichang, China
- Renhe Hospital of China Three Gorges University, Yichang, China
| | - Tingting Xiang
- Medical College of China Three Gorges University, Yichang, China
| | - Yuanxiu Huo
- Medical College of China Three Gorges University, Yichang, China
| | - Chaoqi Liu
- Medical College of China Three Gorges University, Yichang, China
| | - Ting Wang
- Medical College of China Three Gorges University, Yichang, China
| | - Zhiyong Zhou
- Medical College of China Three Gorges University, Yichang, China
| | - Yaoyan Dun
- Medical College of China Three Gorges University, Yichang, China
| | - Haixia Zhao
- Medical College of China Three Gorges University, Yichang, China
| | - Changcheng Zhang
- Medical College of China Three Gorges University, Yichang, China
| |
Collapse
|
22
|
Analysis of Pathological Activities of CCN Proteins in Fibrotic Diseases: Liver Fibrosis. Methods Mol Biol 2018; 1489:445-463. [PMID: 27734396 DOI: 10.1007/978-1-4939-6430-7_37] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
Abstract
Hepatic fibrosis is a complex pathology arising from chronic injury. Pathological features are dominated by the excessive production of extracellular matrix proteins, particularly collagens which are deposited as insoluble scar material that can compromise tissue function. Fibrosis in the liver can often be assessed by staining for collagen in tissue sections and this is an approach that is widely used for grading of fibrosis in human biopsies. However, the recognition of the molecular components that drive fibrosis, including CCN proteins, and the involvement of hepatic stellate cells (HSC) as the principal collagen-producing cells in fibrosing liver, has resulted in a wide variety of molecular and cellular approaches to study the pathogenesis of fibrosis both in vivo and in vitro.
Collapse
|
23
|
Kan F, Ye L, Yan T, Cao J, Zheng J, Li W. Proteomic and transcriptomic studies of HBV-associated liver fibrosis of an AAV-HBV-infected mouse model. BMC Genomics 2017; 18:641. [PMID: 28830339 PMCID: PMC5568174 DOI: 10.1186/s12864-017-3984-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 08/01/2017] [Indexed: 01/05/2023] Open
Abstract
Background Human hepatitis B virus (HBV) infection is an important public health issue in the Asia-Pacific region and is associated with chronic hepatitis, liver fibrosis, cirrhosis and even liver cancer. However, the underlying mechanisms of HBV-associated liver fibrosis remain incompletely understood. Results In the present study, proteomic and transcriptomic approaches as well as biological network analyses were performed to investigate the differentially expressed molecular signature and key regulatory networks that were associated with HBV-mediated liver fibrosis. RNA sequencing and 2DE-MALDI-TOF/TOF were performed on liver tissue samples obtained from HBV-infected C57BL/6 mouse generated via AAV8-HBV virus. The results showed that 322 genes and 173 proteins were differentially expressed, and 28 HBV-specific proteins were identified by comprehensive proteomic and transcriptomic analysis. GO analysis indicated that the differentially expressed proteins were predominantly involved in oxidative stress, which plays a key role in HBV-related liver fibrosis. Importantly, CAT, PRDX1, GSTP1, NXN and BLVRB were shown to be associated with oxidative stress among the differentially expressed proteins. The most striking results were validated by Western blot and RT-qPCR. The RIG-I like receptor signaling pathway was found to be the major signal pathway that changed during HBV-related fibrosis. Conclusions This study provides novel insights into HBV-associated liver fibrosis and reveals the significant role of oxidative stress in liver fibrosis. Furthermore, CAT, BLVRB, NXN, PRDX1, and IDH1 may be candidates for detection of liver fibrosis or therapeutic targets for the treatment of liver fibrosis. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3984-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fangming Kan
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lei Ye
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tao Yan
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiaqi Cao
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jianhua Zheng
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Wuping Li
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
24
|
Study of Protective Effects of Gold Nano Particles on the Liver Toxicity Induced by Carbon-Tetrachloride (CCl4) in Male Rats. ACTA ACUST UNITED AC 2017. [DOI: 10.5812/zjrms.9293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
25
|
Han XY, Hu JN, Wang Z, Wei SN, Zheng SW, Wang YP, Li W. 5-HMF Attenuates Liver Fibrosis in CCl 4-Plus-Alcohol-Induced Mice by Suppression of Oxidative Stress. J Nutr Sci Vitaminol (Tokyo) 2017; 63:35-43. [PMID: 28367924 DOI: 10.3177/jnsv.63.35] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The aim of this study was to investigate the effects of 5-hydroxymethyl-2-furfural (5-HMF) on liver fibrosis induced by carbon tetrachloride (CCl4) and alcohol. Male ICR mice were treated with CCl4 dissolved in olive oil (10% v/v, 2.5 μg/L) intraperitoneally (i.p.), and given at a dose of 2.5×10-5 mg/kg B.W. twice a week for 7 wk. Concurrently, mice received drinking water with or without alcohol. The mice in treatment groups and positive control group were gavaged with 5-HMF (7.5, 15, and 30 mg/kg B.W.) or Huganpian (350 mg/kg B.W.) daily starting in the fourth week and lasting for 4 wk. The blood samples were analyzed for biochemical markers of hepatic injury and tissue samples were subjected for estimation of liver antioxidants and histopathological studies. The concentrations of HA (hyaluronic acid), LN (laminin), CIV (collagen type IV), and MDA (malondialdehyde), as well as the serum levels of ALT (alanine aminotransferase) and AST (aspartate aminotransferase) were markedly reduced by 5-HMF. On the other hand, enzymatic antioxidants SOD (superoxide dismutase), CAT (catalase) and GSH-Px (glutathione peroxidase) were markedly elevated in liver tissue treated with 5-HMF. Histopathological examination revealed that 5-HMF treatment noticeably prevented hepatocyte apoptosis, fatty degeneration and inflammatory cell infiltration on liver fibrosis induced by CCl4 and alcohol. Hoechst 33258 staining also revealed hepatocyte apoptosis. 5-HMF could exert protective effects against liver injury and reduce liver fibrosis induced by CCl4 and alcohol in mice.
Collapse
Affiliation(s)
- Xin-Yue Han
- College of Chinese Medicinal Materials, Jilin Agricultural University
| | | | | | | | | | | | | |
Collapse
|
26
|
Sokar SS, El-Sayad MES, Ghoneim MES, Shebl AM. Combination of Sitagliptin and Silymarin ameliorates liver fibrosis induced by carbon tetrachloride in rats. Biomed Pharmacother 2017; 89:98-107. [DOI: 10.1016/j.biopha.2017.02.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 01/19/2017] [Accepted: 02/07/2017] [Indexed: 12/30/2022] Open
|
27
|
Hung WL, Yang G, Wang YC, Chiou YS, Tung YC, Yang MJ, Wang BN, Ho CT, Wang Y, Pan MH. Protective effects of theasinensin A against carbon tetrachloride-induced liver injury in mice. Food Funct 2017; 8:3276-3287. [DOI: 10.1039/c7fo00700k] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
TSA markedly reduced the CCl4-induced liver injury in mice.
Collapse
Affiliation(s)
- Wei-Lun Hung
- Citrus Research and Education Center
- Department of Food Science and Human Nutrition
- University of Florida
- Lake Alfred
- USA
| | - Guliang Yang
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization; Hubei Collaborative Innovation Center for the Characteristic Resources Exploitation of Dabie Mountains; Huanggang Normal University
- Huanggang
- China
| | - Yu-Chuan Wang
- Institute of Food Science and Technology
- National Taiwan University
- Taipei 10617
- Taiwan
| | - Yi-Shiou Chiou
- Institute of Food Science and Technology
- National Taiwan University
- Taipei 10617
- Taiwan
| | - Yen-Chen Tung
- Institute of Food Science and Technology
- National Taiwan University
- Taipei 10617
- Taiwan
| | | | - Bi-Ni Wang
- College of Food Engineering and Nutritional Science
- Shaanxi Normal University
- Xi'an 710119
- China
| | - Chi-Tang Ho
- Department of Food Science
- Rutgers University
- New Brunswick
- USA
| | - Yu Wang
- Citrus Research and Education Center
- Department of Food Science and Human Nutrition
- University of Florida
- Lake Alfred
- USA
| | - Min-Hsiung Pan
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization; Hubei Collaborative Innovation Center for the Characteristic Resources Exploitation of Dabie Mountains; Huanggang Normal University
- Huanggang
- China
- Institute of Food Science and Technology
- National Taiwan University
| |
Collapse
|
28
|
miR-706 inhibits the oxidative stress-induced activation of PKCα/TAOK1 in liver fibrogenesis. Sci Rep 2016; 6:37509. [PMID: 27876854 PMCID: PMC5120320 DOI: 10.1038/srep37509] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 10/28/2016] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress induces the activation of liver fibrogenic cells (myofibroblasts), thus promoting the expression of fibrosis-related genes, leading to hepatic fibrogenesis. MicroRNAs (miRNAs) are a new class of small RNAs ~18–25 nucleotides in length involved in post-transcriptional regulation of gene expression. Wound-healing and remodeling processes in liver fibrosis have been associated with changes in hepatic miRNA expression. However, the role of miR-706 in liver fibrogenesis is currently unknown. In the present study, we show that miR-706 is abundantly expressed in hepatocytes. Moreover, oxidative stress leads to a significant downregulation of miR-706, and the further reintroduction of miR-706 inhibits oxidative stress-induced expression of fibrosis-related markers such as α-SMA. Subsequent studies revealed that miR-706 directly inhibits PKCα and TAOK1 expression via binding to the 3′-untranslated region, preventing epithelial mesenchymal transition. In vivo studies showed that intravenous injection of miR-706 agomir successfully increases hepatic miR-706 and decreases α-SMA, PKCα, and TAOK1 protein levels in livers of carbon tetrachloride (CCl4)-treated mice. In summary, this study reveals a protective role for miR-706 by blocking the oxidative stress-induced activation of PKCα/TAOK1. Our results further identify a major implication for miR-706 in preventing hepatic fibrogenesis and suggest that miR-706 may be a suitable molecular target for anti-fibrosis therapy.
Collapse
|
29
|
Tan H, He Q, Li R, Lei F, Lei X. Trillin Reduces Liver Chronic Inflammation and Fibrosis in Carbon Tetrachloride (CCl4) Induced Liver Injury in Mice. Immunol Invest 2016; 45:371-82. [PMID: 27219527 DOI: 10.3109/08820139.2015.1137935] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
UNLABELLED Trillin is an active ingredient isolated from Dioscorea nipponica Makino. This study investigated the anti-inflammatory and anti-fibrosis effects of trillin on CCl4-induced hepatotoxicity in C57BL/6 mice. Chronic inflammation and fibrosis were induced by intraperitoneal administration of CCl4 0.5 μL/g of body weight twice a week for 6 weeks. Trillin (50 mg/kg, 100 mg/kg) was administered by gavage for 12 days before finishing the CCl4 induction. Aspartate amino-transferase (AST) and glutamic-pyruvic transaminase (ALT) in serum were determined by AST and ALT kits. Superoxidase dismutase (SOD) activity and malondialdehyde (MDA) levels in serum were assayed by SOD and MDA kits. Meanwhile, the levels of inflammatory mediators including tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β) in serum were detected by enzyme-linked immunosorbent assay (ELISA) method. Pathological changes were observed by hematoxylin-eosin (HE) staining. The proteins of the NF-κB pathway and the TGF-β/Smad pathway were measured by western blot. The trillin-treated group exhibited reduced AST, ALT, MDA, IL-6, TNF-α, and IL-1β, and increased SOD. Histological analyses of the trillin-treated group exhibited reduced inflammatory process and prevented liver fibrosis. Western blot analyses of the trillin-treated group showed reduced NF-κB pathway and TGF-β/Smad pathway. SIGNIFICANCE Based on the results of the present study, trillin can be used as a potential anti-inflammatory drug for chronic hepatic inflammation.
Collapse
Affiliation(s)
- Huabing Tan
- a Department of Infectious Diseases and Lab of Liver Disease, Renmin Hospital , Hubei University of Medicine , Shiyan , Hubei Province , China
| | - Qin He
- a Department of Infectious Diseases and Lab of Liver Disease, Renmin Hospital , Hubei University of Medicine , Shiyan , Hubei Province , China
| | - Rugui Li
- a Department of Infectious Diseases and Lab of Liver Disease, Renmin Hospital , Hubei University of Medicine , Shiyan , Hubei Province , China
| | - Feifei Lei
- a Department of Infectious Diseases and Lab of Liver Disease, Renmin Hospital , Hubei University of Medicine , Shiyan , Hubei Province , China
| | - Xu Lei
- a Department of Infectious Diseases and Lab of Liver Disease, Renmin Hospital , Hubei University of Medicine , Shiyan , Hubei Province , China
| |
Collapse
|
30
|
Shrestha N, Chand L, Han MK, Lee SO, Kim CY, Jeong YJ. Glutamine inhibits CCl4 induced liver fibrosis in mice and TGF-β1 mediated epithelial-mesenchymal transition in mouse hepatocytes. Food Chem Toxicol 2016; 93:129-37. [PMID: 27137983 DOI: 10.1016/j.fct.2016.04.024] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 04/27/2016] [Accepted: 04/27/2016] [Indexed: 12/11/2022]
Abstract
Glutamine, traditionally a non-essential amino acid, now has been considered as essential in serious illness and injury. It is a major precursor for glutathione synthesis. However, the anti-fibrotic effect of glutamine and its molecular mechanism in experimental liver fibrosis have not been explored. In the present study we aimed to examine the potential role of glutamine in carbon tetrachloride (CCl4) induced liver fibrosis and TGF-β1 mediated epithelial mesenchymal transition (EMT) and apoptosis in mouse hepatocytes. Liver fibrosis was induced by intraperitoneal injection of CCl4 three times a week for 10 weeks. Glutamine treatment effectively attenuated liver injury and oxidative stress. Collagen content was significantly decreased in liver sections of glutamine treated mice compared to CCl4 model mice. Furthermore, glutamine decreased expression level of α-SMA and TGF-β in liver tissue. Our in vitro study showed that TGF-β1 treatment in hepatocytes resulted in loss of E-cadherin and increased expression of mesenchymal markers and EMT related transcription factor. In addition, TGF-β1 increased the expression of apoptotic markers. However, glutamine interestingly suppressed TGF-β1 mediated EMT and apoptosis. In conclusion, our results suggest that glutamine ameliorates CCl4 induced liver fibrosis and suppresses TGF-β1 induced EMT progression and apoptosis.
Collapse
Affiliation(s)
- Nirajan Shrestha
- Laboratory of Liver Regeneration, Biomedical Research Institute, Chonbuk National University Hospital, 561-712, Jeonju, South Korea
| | - Lokendra Chand
- Laboratory of Liver Regeneration, Biomedical Research Institute, Chonbuk National University Hospital, 561-712, Jeonju, South Korea
| | - Myung Kwan Han
- Department of Microbiology, Chonbuk National University Medical School, 561-712, Jeonju, South Korea
| | - Seung Ok Lee
- Department of Internal Medicine, Chonbuk National University Medical School, 561-712, Jeonju, South Korea
| | - Chan Young Kim
- Department of Surgery, Chonbuk National University Medical School, 561-712, Jeonju, South Korea
| | - Yeon Jun Jeong
- Laboratory of Liver Regeneration, Biomedical Research Institute, Chonbuk National University Hospital, 561-712, Jeonju, South Korea; Department of Surgery, Chonbuk National University Medical School, 561-712, Jeonju, South Korea.
| |
Collapse
|
31
|
Dietrich CG, Götze O, Geier A. Molecular changes in hepatic metabolism and transport in cirrhosis and their functional importance. World J Gastroenterol 2016; 22:72-88. [PMID: 26755861 PMCID: PMC4698509 DOI: 10.3748/wjg.v22.i1.72] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 09/24/2015] [Accepted: 11/13/2015] [Indexed: 02/06/2023] Open
Abstract
Liver cirrhosis is the common endpoint of many hepatic diseases and represents a relevant risk for liver failure and hepatocellular carcinoma. The progress of liver fibrosis and cirrhosis is accompanied by deteriorating liver function. This review summarizes the regulatory and functional changes in phase I and phase II metabolic enzymes as well as transport proteins and provides an overview regarding lipid and glucose metabolism in cirrhotic patients. Interestingly, phase I enzymes are generally downregulated transcriptionally, while phase II enzymes are mostly preserved transcriptionally but are reduced in their function. Transport proteins are regulated in a specific way that resembles the molecular changes observed in obstructive cholestasis. Lipid and glucose metabolism are characterized by insulin resistance and catabolism, leading to the disturbance of energy expenditure and wasting. Possible non-invasive tests, especially breath tests, for components of liver metabolism are discussed. The heterogeneity and complexity of changes in hepatic metabolism complicate the assessment of liver function in individual patients. Additionally, studies in humans are rare, and species differences preclude the transferability of data from rodents to humans. In clinical practice, some established global scores or criteria form the basis for the functional evaluation of patients with liver cirrhosis, but difficult treatment decisions such as selection for transplantation or resection require further research regarding the application of existing non-invasive tests and the development of more specific tests.
Collapse
|
32
|
Zheng QF, Li JS, Bai L, Zheng SJ. Protective effect of liver fibrosis against acute liver injury. Shijie Huaren Xiaohua Zazhi 2016; 24:3537. [DOI: 10.11569/wcjd.v24.i24.3537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
33
|
Yasin BR, El-Fawal HAN, Mousa SA. Date (Phoenix dactylifera) Polyphenolics and Other Bioactive Compounds: A Traditional Islamic Remedy's Potential in Prevention of Cell Damage, Cancer Therapeutics and Beyond. Int J Mol Sci 2015; 16:30075-90. [PMID: 26694370 PMCID: PMC4691153 DOI: 10.3390/ijms161226210] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 12/08/2015] [Accepted: 12/09/2015] [Indexed: 12/20/2022] Open
Abstract
This review analyzes current studies of the therapeutic effects of Phoenix dactylifera, or date palm fruit, on the physiologic system. Specifically, we sought to summarize the effects of its application in preventing cell damage, improving cancer therapeutics and reducing damage caused by conventional chemotherapy. Phoenix dactylifera exhibits potent anti-oxidative properties both in vitro and in vivo. This allows the fruit to prevent depletion of intrinsic protection from oxidative cell damage and assist these defense systems in reducing cell damage. Macroscopically, this mechanism may be relevant to the prevention of various adverse drug events common to chemotherapy including hepatotoxicity, nephrotoxicity, gastrotoxicity, and peripheral neuropathy. While such effects have only been studied in small animal systems, research suggests a potential application to more complex mammalian systems and perhaps a solution to some problems of chemotherapy in hepato-compromised and nephro-compromised patients.
Collapse
Affiliation(s)
- Bibi R Yasin
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| | - Hassan A N El-Fawal
- Neurotoxicology Laboratory, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA.
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| |
Collapse
|
34
|
Kanda T, Yokosuka O. The androgen receptor as an emerging target in hepatocellular carcinoma. J Hepatocell Carcinoma 2015; 2:91-9. [PMID: 27508198 PMCID: PMC4918288 DOI: 10.2147/jhc.s48956] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the male-dominant liver diseases with poor prognosis, although treatments for HCC have been progressing in the past decades. Androgen receptor (AR) is a member of the nuclear receptor superfamily. Previous studies reported that AR was expressed in human HCC and non-HCC tissues. AR is activated both ligand-dependently and ligand-independently. The latter is associated with a mitogen-activated protein kinase–, v-akt murine thymoma viral oncogene homolog 1–, or signal-transducer and activator of transcription–signaling pathway, which has been implicated in the development of HCC. It has been reported that more than 200 RNA expression levels are altered by androgen treatment. In the liver, androgen-responsive genes are cytochrome P450s, transforming growth factor β, vascular endothelial growth factor, and glucose-regulated protein 78 kDa, which are also associated with human hepatocarcinogenesis. Recent studies also revealed that AR plays a role in cell migration and metastasis. It is possible that cross-talk among AR-signaling, endoplasmic reticulum stress, and innate immune response is important for human hepatocarcinogenesis and HCC development. This review shows that AR could play a potential role in human HCC and represent one of the important target molecules for the treatment of HCC.
Collapse
Affiliation(s)
- Tatsuo Kanda
- Department of Gastroenterology and Nephrology, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Osamu Yokosuka
- Department of Gastroenterology and Nephrology, Chiba University, Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
35
|
Ye L, Yu H, Li C, Hirsch ML, Zhang L, Samulski RJ, Li W, Liu Z. Adeno-Associated Virus Vector Mediated Delivery of the HBV Genome Induces Chronic Hepatitis B Virus Infection and Liver Fibrosis in Mice. PLoS One 2015; 10:e0130052. [PMID: 26075890 PMCID: PMC4468063 DOI: 10.1371/journal.pone.0130052] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/15/2015] [Indexed: 01/04/2023] Open
Abstract
Liver cirrhosis and hepatocellular carcinomas are major health problems of chronic hepatitis B virus (HBV) infection. To date, rare model has reproduced liver fibrosis associated with long-term HBV infection which in turn has hindered both the understanding of HBV biology and the development of new treatment options. Here, using adeno-associated virus serotype 8 (AAV8) mediated delivery of a 1.2-kb HBV genome, we successfully generated a chronic HBV infectious mouse model that presents the associated liver fibrosis observed following human infection. After AAV8/HBV1.2 vector administration, mice demonstrated effective HBV replication and transcription which resulted in HBV antigen expression and viremia over 6 months. Although no obvious acute inflammatory response was noted, these mice still developed chronic liver disease and hepatic fibrogenesis as demonstrated by increased ground glass-like hepatocytes, an increasing trend of collagen deposition and upregulated fibrosis markers, including type I collagen, type III collagen, tissue inhibitor of metalloproteinase (TIMP), and transforming growth factor-β1(TGF-β1). Taken together, AAV-mediated HBV gene delivery to the mouse liver, induced HBV persistent infection accompanied by liver fibrosis which can serve as a model for investigating the precise mechanisms underlying liver fibrosis following chronic HBV infection as well as for the potential development of novel therapeutics.
Collapse
MESH Headings
- Animals
- Blotting, Northern
- Blotting, Southern
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/virology
- Cells, Cultured
- Dependovirus/genetics
- Disease Models, Animal
- Drug Delivery Systems
- Enzyme-Linked Immunosorbent Assay
- Genetic Vectors/administration & dosage
- Genome, Viral
- HEK293 Cells
- Hepatitis B virus/genetics
- Hepatitis B, Chronic/genetics
- Hepatitis B, Chronic/virology
- Humans
- Liver Cirrhosis/genetics
- Liver Cirrhosis/virology
- Liver Neoplasms/genetics
- Liver Neoplasms/virology
- Mice
- Mice, Inbred C57BL
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Viremia/genetics
- Viremia/virology
- Virus Replication
Collapse
Affiliation(s)
- Lei Ye
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - Haisheng Yu
- Key Laboratory of Immunity and Infection, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Chengwen Li
- Gene Therapy Center, Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Matthew L. Hirsch
- Gene Therapy Center, Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Liguo Zhang
- Key Laboratory of Immunity and Infection, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - R. Jude Samulski
- Gene Therapy Center, Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Wuping Li
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- * E-mail:
| | - Zhong Liu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| |
Collapse
|
36
|
Dong D, Yin L, Qi Y, Xu L, Peng J. Protective Effect of the Total Saponins from Rosa laevigata Michx Fruit against Carbon Tetrachloride-Induced Liver Fibrosis in Rats. Nutrients 2015; 7:4829-4850. [PMID: 26083117 PMCID: PMC4488818 DOI: 10.3390/nu7064829] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 06/05/2015] [Accepted: 06/05/2015] [Indexed: 02/07/2023] Open
Abstract
In this study, the protective effect of the total saponins from Rosa laevigata Michx (RLTS) against liver fibrosis induced by carbon tetrachloride (CCl4) in rats was evaluated. The results showed that RLTS significantly rehabilitated the levels of alanine aminotransferase, aspartate aminotransferase, malondialdehyde, glutathione, glutathione peroxidase, catalase, superoxide dismutase, hydroxyproline, α-smooth muscle actin, collagen I, collagen III and fibronectin, which were confirmed using H&E, Sirius Red and Masson histopathological assays. Further research indicated that RLTS markedly reduced cytochrome P450 2E1 activity, attenuated oxidative stress, and suppressed inflammation. In addition, RLTS facilitated matrix degradation through down-regulation of matrix metalloproteinase2, matrix metalloproteinase 9 and metalloproteinases1, and exerted the anti-fibrotic effects through affecting transforming growth factor β/Smad, focal adhesion kinase/phosphatidylinositol-3-kinase/amino kinase terminal/70-kDa ribosomal S6 Kinase (FAK-PI3K-Akt-p70(S6K)) and mitogen-activated protein kinase (MAPK) signaling pathways. Taken together, our data indicate that RLTS can be applied as one effective candidate for the treatment of liver fibrosis in the future.
Collapse
Affiliation(s)
- Deshi Dong
- College of Pharmacy, Dalian Medical University, No. 9 Western Lvshun South Road, Dalian 116044, China.
- The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, No. 9 Western Lvshun South Road, Dalian 116044, China.
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, No. 9 Western Lvshun South Road, Dalian 116044, China.
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, No. 9 Western Lvshun South Road, Dalian 116044, China.
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, No. 9 Western Lvshun South Road, Dalian 116044, China.
| |
Collapse
|
37
|
Park HJ, Kim HG, Wang JH, Choi MK, Han JM, Lee JS, Son CG. Comparison of TGF-β, PDGF, and CTGF in hepatic fibrosis models using DMN, CCl4, and TAA. Drug Chem Toxicol 2015; 39:111-118. [PMID: 26045230 DOI: 10.3109/01480545.2015.1052143] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 03/31/2015] [Accepted: 05/13/2015] [Indexed: 11/13/2022]
Abstract
Three chemotoxins including dimethylnitrosamine (DMN), carbon tetrachloride (CCl4), and thioacetamide (TAA) are commonly used in hepatofibrotic models. We aimed to draw characteristics of histopathology and pro-fibrogenic cytokines including TGF-β, PDGF and CTGF among three models. Rats were divided into six groups and intra-peritoneally injected with DMN (10 mg/kg, for three weeks, three consecutive days weekly), CCl4 (1.6 g/kg, for 10 weeks, twice weekly), TAA (200 mg/kg, for 12 weeks, twice weekly) or their corresponded treatment for each control group. The liver weights were decreased in DMN model, but not other models. Ascites were occurred as 3-, 2-, and 7-rats in DMN, CCl4, and TAA model, respectively. The lipid peroxidation was highest in CCl4 model, serum levels of liver enzymes were increased as similar severity. The hepatofibrotic alterations were remarkable in DMN and TAA model, but not CCl4 as evidenced by the Masson trichrome staining and hydroxyproline. The immunohistochemistry for α-SAM showed that the DMN model was most severely enhanced than other models. On the other hand, hepatic tissue levels of pro-fibrogenic cytokines including TGF-β, PDGF, and CTGF were generally increased in three models, but totally different among models or measurement resources. Especially, serum levels of three cytokines were remarkably increased by CCl4 injection and CTGF levels in both hepatic tissue and serum were highest in CCl4 group. Our results firstly demonstrated comparative study for features of morphological finding and pro-fibrogenic cytokines in serum and hepatic protein levels among three models. Above results would be a helpful reference for hepatofibrotic studies.
Collapse
Affiliation(s)
- Hye-Jung Park
- a Department of Liver and Immunology Research Center , Daejeon Oriental Hospital of Daejeon University , Republic of Korea and
| | - Hyeong-Geug Kim
- a Department of Liver and Immunology Research Center , Daejeon Oriental Hospital of Daejeon University , Republic of Korea and
| | - Jing-Hua Wang
- b Laboratory of Medical Science , Dongguk University Medical Center, Dongguk University , Republic of Korea
| | - Min-Kyung Choi
- a Department of Liver and Immunology Research Center , Daejeon Oriental Hospital of Daejeon University , Republic of Korea and
| | - Jong-Min Han
- a Department of Liver and Immunology Research Center , Daejeon Oriental Hospital of Daejeon University , Republic of Korea and
| | - Jin-Seok Lee
- a Department of Liver and Immunology Research Center , Daejeon Oriental Hospital of Daejeon University , Republic of Korea and
| | - Chang-Gue Son
- a Department of Liver and Immunology Research Center , Daejeon Oriental Hospital of Daejeon University , Republic of Korea and
| |
Collapse
|
38
|
Buko V, Belonovskaya E, Naruta E, Lukivskaya O, Kanyuka O, Zhuk O, Kranc R, Stoika R, Sybirna N. Pituitary tumor transforming gene as a novel regulatory factor of liver fibrosis. Life Sci 2015; 132:34-40. [PMID: 25936962 DOI: 10.1016/j.lfs.2015.04.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 03/16/2015] [Accepted: 04/16/2015] [Indexed: 12/12/2022]
Abstract
AIMS Pituitary tumor-transforming gene (PTTG) is involved in multiple cellular pathways. We studied the development of liver fibrosis induced by thioacetamide (TAA) in knockout (PTTG-/-) and wildtype (PTTG+/+) mice. MAIN METHODS Liver fibrosis in PTTG+/+ and PTTG-/- mice was induced by escalating dose TAA treatment (50-400mg/kg, i.p.) for 12 weeks and assessed by histochemistry, immunohistochemistry, liver hydroxyproline, serum fibrosis markers and fibrosis-related mRNA expression by real-time PCR determination. KEY FINDINGS Both PTTG+/+ and PTTG-/- mice treated with TAA developed signs of fibrosis and inflammatory cell infiltration. However, histological signs of bridging fibrosis and connective tissue square morphometry were significantly attenuated in mice lacking PTTG. α-SMA immunohistochemistry revealed that hepatic stellate cell activation was markedly reduced in PTTG-/- mice compared to wildtype controls. Hepatic hydroxyproline levels were significantly lower in fibrotic PTTG-/- group. The serum TNFα and hepatic TNFα mRNA expression were significantly lower in fibrotic PTTG-/- animals, as well as hepatic TGFβ and VEGF mRNA levels compared to TAA-treated wildtype controls. Serum hyaluronate and TGFβ levels were markedly elevated in fibrotic mice of both genotypes, but were not altered by the absence of PTTG. SIGNIFICANCE TAA-induced fibrosis development is significantly ameliorated in PTTG-/- mice. These animals demonstrated diminished stellate cell activation, suppressed circulating serum markers of inflammation, fibrogenesis and angiogenesis. The presented findings suggest that PTTG is functionally required for hepatic fibrosis progression in an animal model of chronic liver injury. PTTG can be considered as a new important target for prevention and treatment of liver fibrosis/cirrhosis.
Collapse
Affiliation(s)
- Vyacheslav Buko
- Institute of Biochemistry of Biologically Active Compounds, National Academy of Sciences, Grodno, Belarus; School of Medical Sciences, Bialystok, Poland.
| | - Elena Belonovskaya
- Institute of Biochemistry of Biologically Active Compounds, National Academy of Sciences, Grodno, Belarus
| | - Elena Naruta
- Institute of Biochemistry of Biologically Active Compounds, National Academy of Sciences, Grodno, Belarus
| | - Oxana Lukivskaya
- Institute of Biochemistry of Biologically Active Compounds, National Academy of Sciences, Grodno, Belarus
| | | | - Olga Zhuk
- Institute of Biochemistry of Biologically Active Compounds, National Academy of Sciences, Grodno, Belarus
| | | | - Rostislav Stoika
- Lviv National Ivan Franko University, Lviv, Ukraine; Institute of Cell Biology, National Academy of Sciences, Lviv, Ukraine
| | | |
Collapse
|
39
|
Sunami Y, von Figura G, Kleger A, Strnad P, Hüser N, Hartmann D. The role of telomeres in liver disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 125:159-72. [PMID: 24993702 DOI: 10.1016/b978-0-12-397898-1.00007-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Telomeres stabilize open chromosome ends and protect them against chromosomal end-to-end fusions, breakage, instability, and nonreciprocal translocations. Telomere dysfunction is known to lead to an impaired regenerative capacity of hepatocytes and an increased cirrhosis formation in the context of acute and chronic liver injury. In addition, telomere dysfunction and telomerase mutations have been associated with the induction of chromosomal instability and consequently with cirrhosis development and hepatocarcinogenesis. The identification of molecular mechanisms related to telomere dysfunction and telomerase activation might lead to new therapeutic strategies. In this chapter, we are reviewing the current knowledge about the importance of telomere dysfunction in liver diseases.
Collapse
Affiliation(s)
- Yoshiaki Sunami
- Department of General Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Guido von Figura
- Department of Internal Medicine II, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Alexander Kleger
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Pavel Strnad
- Department of Internal Medicine III and IZKF, University Hospital Aachen, Aachen, Germany
| | - Norbert Hüser
- Department of General Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Daniel Hartmann
- Department of General Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| |
Collapse
|
40
|
Delire B, Stärkel P, Leclercq I. Animal Models for Fibrotic Liver Diseases: What We Have, What We Need, and What Is under Development. J Clin Transl Hepatol 2015; 3:53-66. [PMID: 26357635 PMCID: PMC4542084 DOI: 10.14218/jcth.2014.00035] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 12/10/2014] [Accepted: 12/12/2014] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis is part of the wound-healing response to liver damage of various origins and represents a major health problem. Although our understanding of the pathogenesis of liver fibrosis has grown considerably over the last 20 years, effective antifibrotic therapies are still lacking. The use of animal models is crucial for determining mechanisms underlying initiation, progression, and resolution of fibrosis and for developing novel therapies. To date, no animal model can recapitulate all the hepatic and extra-hepatic features of liver disease. In this review, we will discuss the current rodent models of liver injuries. We will then focus on the available ways to target specifically particular compounds of fibrogenesis and on the new models of liver diseases like the humanized liver mouse model.
Collapse
Affiliation(s)
- Bénédicte Delire
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique (IREC), Catholic University of Louvain (UCL), Brussels, Belgium
| | - Peter Stärkel
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique (IREC), Catholic University of Louvain (UCL), Brussels, Belgium
- Department of Gastroenterology, Saint-Luc Academic Hospital and Institute of Clinical Research, Catholic University of Louvain, Brussels, Belgium
| | - Isabelle Leclercq
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique (IREC), Catholic University of Louvain (UCL), Brussels, Belgium
- Correspondence to: Isabelle Leclercq, Laboratoire d'Hépato-Gastro-Entérologie, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue E Mounier 53, Box B1.52.01, Brussels 1200, Belgium. Tel: +32-27645379, Fax: +32-27645346. E-mail:
| |
Collapse
|
41
|
Hepatoprotective properties of betulonic acid amide and heptral in toxic liver injury induced by carbon tetrachloride in combination with ethanol. Bull Exp Biol Med 2015; 158:336-41. [PMID: 25573364 DOI: 10.1007/s10517-015-2756-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Indexed: 01/11/2023]
Abstract
Toxic liver injury with the development of fibrosis and cirrhosis was modeled in Wistar rats by intragastric administration of 0.1 ml/kg CCl4 in combination with 5% ethanol with glucose 3 times a week for 6 weeks. The animals were treated with betulonic acid amide (50 mg/kg in Tween aqueous solution) and heptral (6 mg/kg) as hepatoprotective compounds. It was found that betulonic acid amide stimulated the regenerative response in hepatocytes under conditions of combined toxic exposure and promoted recovery of their qualitative and quantitative characteristics, which was accompanied by a significant decrease in the severity of liver fibrosis and the absence of cirrhotic transformation of the liver.
Collapse
|
42
|
Protective Effects of Norursodeoxycholic Acid Versus Ursodeoxycholic Acid on Thioacetamide-induced Rat Liver Fibrosis. J Clin Exp Hepatol 2014; 4:293-301. [PMID: 25755576 PMCID: PMC4298626 DOI: 10.1016/j.jceh.2014.02.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 02/02/2014] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND/OBJECTIVES Effects of norursodeoxycholic acid (norUDCA) and ursodeoxycholic acid (UDCA) on liver fibrosis progression and liver fibrosis reversal in thioacetamide (TAA)-treated rats were studied. METHODS Advanced liver fibrosis was induced by TAA treatment (200 mg/kg, i.p.) for 12 weeks. In the second experiment resolution of liver fibrosis was assessed after 8 weeks of TAA withdrawal. During 8 last weeks of each trial, fibrotic rats were daily administered with UDCA (80 mg/kg) and norUDCA (equimolar to 80 mg/kg of UDCA) by oral gavage. Liver fibrosis was assessed by Sirius red staining, liver hydroxyproline and serum fibrosis markers determination. RESULTS The TAA treatment resulted in advanced fibrosis and increase in liver hydroxyproline content and serum fibrosis markers. These signs of fibrosis were less pronounced in rats after TAA withdrawal. Treatment with of norUDCA significantly decreased the total and relative liver hydroxyproline contents in rats with fibrosis reversal, whereas UDCA did not change these parameters. Both compounds decreased serum TGFβ and type IV collagen contents, whereas other serum markers did not differ from the placebo group. In the fibrosis progression model the square of connective tissue was decreased by norUDCA. Serum type IV collagen and procollagen III-NT contents in these experiments were lowered by both UDCA and norUDCA, whereas rest of serum fibrosis markers were diminished only by norUDCA. CONCLUSIONS Both norUDCA and UDCA showed therapeutic and prophylactic antifibrotic effect in rats with TAA-induced liver fibrosis. For most of tested parameters norUDCA was more effective than UDCA, especially in the experiment with liver fibrosis regression.
Collapse
|
43
|
Jang YO, Kim MY, Cho MY, Baik SK, Cho YZ, Kwon SO. Effect of bone marrow-derived mesenchymal stem cells on hepatic fibrosis in a thioacetamide-induced cirrhotic rat model. BMC Gastroenterol 2014; 14:198. [PMID: 25425284 PMCID: PMC4251876 DOI: 10.1186/s12876-014-0198-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 11/06/2014] [Indexed: 01/01/2023] Open
Abstract
Background Cirrhosis is a long-term consequence of chronic hepatic injury with fibrosis. No effective therapy is currently available for decompensated cirrhosis except liver transplantation. Hence, we investigated the effect of bone marrow-derived mesenchymal stem cells (BM-MSCs) on hepatic fibrosis in a thioacetamide (TAA)-induced cirrhotic rat model. Methods The BM-MSCs were injected directly into the right liver lobe twice, at 6 and 8 weeks during the 12-week TAA administration, in thioacetamide (TAA)-induced cirrhotic rats model, and hepatic fibrosis was evaluated. At 12 weeks, the effect of BM-MSCs on hepatic fibrosis was analyzed histomorphologically using the Laennec fibrosis scoring system, and the collagen proportionate area was quantified. Cirrhosis-related factors, such as transforming growth factor β1 (TGF-β1), type 1 collagen (collagen-1), α-smooth muscle actin (α-SMA), and P-Smad3/Smad3 expression levels, were evaluated using real-time polymerase chain reaction and western blot assays. Results According to the Laennec fibrosis scoring system, histological improvement was observed in hepatic fibrosis after BM-MSC treatment (P <0.01). The percentage of the collagen proportionate area decreased from 16.72 ± 5.51 to 5.06 ± 1.27 after BM-MSC treatment (P <0.01). The content of hepatic hydroxyproline was significantly lower in the BM-MSC treated group (46.25 ± 13.19) compared to the untreated cirrhotic group (85.81 ± 17.62; P <0.01). BM-MSC administration significantly decreased TGF-β1, collagen-1, and α-SMA expression in TAA-induced cirrhotic rats (P <0.01). We also confirmed P-Smad3/Smad3, downstream effectors of the TGF-β1 signaling pathway, and found that MSC transplantation inhibited Smad3 phosphorylation. Conclusions BM-MSC treatment attenuated hepatic fibrosis in rats with TAA-induced cirrhosis, raising the possibility of the clinical use of BM-MSCs in the treatment of cirrhosis. Electronic supplementary material The online version of this article (doi:10.1186/s12876-014-0198-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yoon Ok Jang
- Department of Internal Medicine, Yonsei University, Wonju College of Medicine, 162, Ilsan-dong, Wonju, Republic of Korea.
| | - Moon Young Kim
- Department of Internal Medicine, Yonsei University, Wonju College of Medicine, 162, Ilsan-dong, Wonju, Republic of Korea.
| | - Mee Yon Cho
- Department of Pathology, Yonsei University, Wonju College of Medicine, 162, Ilsan-dong, Wonju, Republic of Korea.
| | - Soon Koo Baik
- Department of Internal Medicine, Yonsei University, Wonju College of Medicine, 162, Ilsan-dong, Wonju, Republic of Korea.
| | - Youn Zoo Cho
- Department of Internal Medicine, Yonsei University, Wonju College of Medicine, 162, Ilsan-dong, Wonju, Republic of Korea.
| | - Sang Ok Kwon
- Department of Internal Medicine, Yonsei University, Wonju College of Medicine, 162, Ilsan-dong, Wonju, Republic of Korea.
| |
Collapse
|
44
|
Diethylcarbamazine reduces chronic inflammation and fibrosis in carbon tetrachloride- (CCl₄-) induced liver injury in mice. Mediators Inflamm 2014; 2014:696383. [PMID: 25374445 PMCID: PMC4211150 DOI: 10.1155/2014/696383] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 08/15/2014] [Accepted: 08/27/2014] [Indexed: 02/07/2023] Open
Abstract
This study investigated the anti-inflammatory effects of DEC on the CCl4-induced hepatotoxicity in C57BL/6 mice. Chronic inflammation was induced by i.p. administration of CCl4 0.5 μL/g of body weight through two injections a week for 6 weeks. DEC (50 mg/kg) was administered by gavage for 12 days before finishing the CCl4 induction. Histological analyses of the DEC-treated group exhibited reduced inflammatory process and prevented liver necrosis and fibrosis. Immunohistochemical and immunofluorescence analyses of the DEC-treated group showed reduced COX-2, IL1β, MDA, TGF-β, and αSMA immunopositivity, besides exhibiting decreased IL1β, COX-2, NFκB, IFNγ, and TGFβ expressions in the western blot analysis. The DEC group enhanced significantly the IL-10 expression. The reduction of hepatic injury in the DEC-treated group was confirmed by the COX-2 and iNOS mRNA expression levels. Based on the results of the present study, DEC can be used as a potential anti-inflammatory drug for chronic hepatic inflammation.
Collapse
|
45
|
Yi HS, Lee YS, Byun JS, Seo W, Jeong JM, Park O, Duester G, Haseba T, Kim SC, Park KG, Gao B, Jeong WI. Alcohol dehydrogenase III exacerbates liver fibrosis by enhancing stellate cell activation and suppressing natural killer cells in mice. Hepatology 2014; 60:1044-53. [PMID: 24668648 PMCID: PMC4867000 DOI: 10.1002/hep.27137] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Accepted: 03/15/2014] [Indexed: 01/03/2023]
Abstract
UNLABELLED The important roles of retinols and their metabolites have recently been emphasized in the interactions between hepatic stellate cells (HSCs) and natural killer (NK) cells. Nevertheless, the expression and role of retinol metabolizing enzyme in both cell types have yet to be clarified. Thus, we investigated the expression of retinol metabolizing enzyme and its role in liver fibrosis. Among several retinol metabolizing enzymes, only alcohol dehydrogenase (ADH) 3 expression was detected in isolated HSCs and NK cells, whereas hepatocytes express all of them. In vitro treatment with 4-methylpyrazole (4-MP), a broad ADH inhibitor, or depletion of the ADH3 gene down-regulated collagen and transforming growth factor-β1 (TGF-β1) gene expression, but did not affect α-smooth muscle actin gene expression in cultured HSCs. Additionally, in vitro, treatments with retinol suppressed NK cell activities, whereas inhibition of ADH3 enhanced interferon-γ (IFN-γ) production and cytotoxicity of NK cells against HSCs. In vivo, genetic depletion of the ADH3 gene ameliorated bile duct ligation- and carbon tetrachloride-induced liver fibrosis, in which a higher number of apoptotic HSCs and an enhanced activation of NK cells were detected. Freshly isolated HSCs from ADH3-deficient mice showed reduced expression of collagen and TGF-β1, but enhanced expression of IFN-γ was detected in NK cells from these mice compared with those of control mice. Using reciprocal bone marrow transplantation of wild-type and ADH3-deficient mice, we demonstrated that ADH3 deficiency in both HSCs and NK cells contributed to the suppressed liver fibrosis. CONCLUSION ADH3 plays important roles in promoting liver fibrosis by enhancing HSC activation and inhibiting NK cell activity, and could be used as a potential therapeutic target for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Hyon-Seung Yi
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 305-701, Republic of Korea
| | - Young-Sun Lee
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 305-701, Republic of Korea
| | - Jin-Seok Byun
- Department of Oral Medicine, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - Wonhyo Seo
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 305-701, Republic of Korea
| | - Jong-Min Jeong
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 305-701, Republic of Korea
| | - Ogyi Park
- Laboratory of Liver Study, National Institute on Alcohol Abuse and Alcoholism, National Institute of Health, Bethesda, Maryland, USA
| | - Gregg Duester
- Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA
| | - Takeshi Haseba
- Department of Legal Medicine, Nippon Medical School, Tokyo 113-8602, Japan
| | - Sun Chang Kim
- Intelligent Synthetic Biology Center, 373-1, Guseong-dong, Yuseong-gu, Daejeon 305-701, Republic of Korea
,Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Keun-Gyu Park
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Bin Gao
- Laboratory of Liver Study, National Institute on Alcohol Abuse and Alcoholism, National Institute of Health, Bethesda, Maryland, USA
| | - Won-Il Jeong
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 305-701, Republic of Korea
| |
Collapse
|
46
|
Therapeutic effect of hepatocyte growth factor-secreting mesenchymal stem cells in a rat model of liver fibrosis. Exp Mol Med 2014; 46:e110. [PMID: 25145391 PMCID: PMC4150933 DOI: 10.1038/emm.2014.49] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Accepted: 06/08/2014] [Indexed: 12/19/2022] Open
Abstract
Bone marrow-derived mesenchymal stromal cells (MSCs) have been reported to be beneficial for the treatment of liver fibrosis. Here, we investigated the use of genetically engineered MSCs that overexpress hepatocyte growth factor (HGF) as a means to improve their therapeutic effect in liver fibrosis. Liver fibrosis was induced by intraperitoneal injection of dimethylnitrosamine. HGF-secreting MSCs (MSCs/HGF) were prepared by transducing MSCs with an adenovirus carrying HGF-encoding cDNA. MSCs or MSCs/HGF were injected directly into the spleen of fibrotic rats. Tissue fibrosis was assessed by histological analysis 12 days after stem cell injection. Although treatment with MSCs reduced fibrosis, treatment with MSCs/HGF produced a more significant reduction and was associated with elevated HGF levels in the portal vein. Collagen levels in the liver extract were decreased after MSC/HGF therapy, suggesting recovery from fibrosis. Furthermore, liver function was improved in animals receiving MSCs/HGF, indicating that MSC/HGF therapy resulted not only in reduction of liver fibrosis but also in improvement of hepatocyte function. Assessment of cell and biochemical parameters revealed that mRNA levels of the fibrogenic cytokines PDGF-bb and TGF-β1 were significantly decreased after MSC/HGF therapy. Subsequent to the decrease in collagen, expression of matrix metalloprotease-9 (MMP-9), MMP-13, MMP-14 and urokinase-type plasminogen activator was augmented following MSC/HGF, whereas tissue inhibitor of metalloprotease-1 (TIMP-1) expression was reduced. In conclusion, therapy with MSCs/HGF resulted in an improved therapeutic effect compared with MSCs alone, probably because of the anti-fibrotic activity of HGF. Thus, MSC/HGF represents a promising approach toward a cell therapy for liver fibrosis.
Collapse
|
47
|
Abdelaziz DHA, Ali SA. The protective effect of Phoenix dactylifera L. seeds against CCl4-induced hepatotoxicity in rats. JOURNAL OF ETHNOPHARMACOLOGY 2014; 155:736-743. [PMID: 24945397 DOI: 10.1016/j.jep.2014.06.026] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 04/18/2014] [Accepted: 06/06/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In traditional Egyptian medicine, Phoenix dactylifera L. (date palm) seeds are listed in folk remedies for the management of diabetes, liver diseases and gastrointestinal disorders. The present study was conducted to investigate the protective effect of Phoenix dactylifera L. seeds aqueous suspension against the chemically-induced hepatic injury in rats. METHODS Liver injury was achieved by exposing Wistar rats to CCl4 (10% in olive oil; 0.5 mL/rat; IP) twice a week for 4 weeks. Along with CCl4, aqueous suspensions of raw or roasted Phoenix dactylifera seeds (1.0 g/kg) were administered orally in a daily manner. RESULTS Our results demonstrated that Phoenix dactylifera seeds significantly improved the CCl4-induced alterations in liver function parameters (AST, ALT, ALP and albumin). Moreover, the CCl4-induced oxidative stress, represented by elevated thiobarbituric acid reactive substance (TBARS), nitric oxide and oxidative DNA damage, was ameliorated by Phoenix dactylifera seeds treatment. In addition, Phoenix dactylifera seeds restored the activities of hepatic antioxidant enzymes (superoxide dismutase and glutathione S-transferase) that were declined after CCl4 treatment. Examination of liver histopathology revealed that Phoenix dactylifera seeds attenuate the incidence of liver lesions (including vacuolization and fibroblast proliferation) triggered by CCl4 intoxication. CONCLUSION The Phoenix dactylifera seeds could be a promising candidate for protection against the CCl4-induced liver intoxication, and this hepatoprotective effect might be attributed to the antioxidant and free radical scavenging activities.
Collapse
Affiliation(s)
- Dalia H A Abdelaziz
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Cairo, Egypt.
| | - Sahar A Ali
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Cairo, Egypt.
| |
Collapse
|
48
|
Zhao G, Hatting M, Nevzorova YA, Peng J, Hu W, Boekschoten MV, Roskams T, Muller M, Gassler N, Liedtke C, Davis RJ, Cubero FJ, Trautwein C. Jnk1 in murine hepatic stellate cells is a crucial mediator of liver fibrogenesis. Gut 2014; 63:1159-1172. [PMID: 24037431 DOI: 10.1136/gutjnl-2013-305507] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE The c-Jun N-terminal kinase-1 (Jnk1) gene has been shown to be involved in liver fibrosis. Here, we aimed to investigate the molecular mechanism and define the cell type involved in mediating the Jnk1-dependent effect on liver fibrogenesis. DESIGN Jnk1(f/f) wildtype (WT), Jnk1(-/-) and Jnk1(Δhepa) (hepatocyte-specific deletion of Jnk1) mice were subjected to (i) bile duct ligation (BDL) and (ii) CCl4-induced liver fibrosis. Additionally, we performed bone marrow transplantations (BMT), isolated primary hepatic stellate cells (HSCs), studied their activation in vitro and investigated human diseased liver samples. RESULTS Phosphorylated Jnk was expressed in myofibroblasts, epithelial and inflammatory cells during the progression of fibrogenesis in humans and mice. In mice, liver transaminases, alkaline phosphatase, bilirubin and liver histology revealed reduced injury in Jnk1(-/-) compared with WT and Jnk1(Δhepa) mice correlating with lower hepatocyte cell death and proliferation. Consequently, parameters of liver fibrosis such as Sirius red staining and collagen IA1 and α-smooth muscle actin expression were downregulated in Jnk1(-/-) compared with WT and Jnk1(Δhepa) livers, 4 weeks after CCl4 or BDL. BMT experiments excluded bone marrow-derived cells from having a major impact on the Jnk1-dependent effect on fibrogenesis, while primary HSCs from Jnk1(-/-) livers showed reduced transdifferentiation and extracellular matrix production. Moreover, Jnk1 ablation caused a reduced lifespan and poor differentiation of HSCs into matrix-producing myofibroblasts. CONCLUSIONS Jnk1 in HSCs, but not in hepatocytes, significantly contribute to liver fibrosis development, identifying Jnk1 in HSCs as a profibrotic kinase and a promising cell-directed target for liver fibrosis.
Collapse
Affiliation(s)
- Gang Zhao
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Maximilian Hatting
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Yulia A Nevzorova
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Jin Peng
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Wei Hu
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Mark V Boekschoten
- Division of Human Nutrition, Nutrition, Metabolism & Genomics Group, Wageningen University, Wageningen, The Netherlands
| | - Tania Roskams
- Department of Morphology and Molecular Pathology, Liver Research Unit, University of Leuven, Leuven, Belgium
| | - Michael Muller
- Division of Human Nutrition, Nutrition, Metabolism & Genomics Group, Wageningen University, Wageningen, The Netherlands
| | - Nikolaus Gassler
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Christian Liedtke
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Roger J Davis
- Howard Hughes Medical Institute and University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | | - Christian Trautwein
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
49
|
Dutta-Moscato J, Solovyev A, Mi Q, Nishikawa T, Soto-Gutierrez A, Fox IJ, Vodovotz Y. A Multiscale Agent-Based in silico Model of Liver Fibrosis Progression. Front Bioeng Biotechnol 2014; 2:18. [PMID: 25152891 PMCID: PMC4126446 DOI: 10.3389/fbioe.2014.00018] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 05/14/2014] [Indexed: 01/06/2023] Open
Abstract
Chronic hepatic inflammation involves a complex interplay of inflammatory and mechanical influences, ultimately manifesting in a characteristic histopathology of liver fibrosis. We created an agent-based model (ABM) of liver tissue in order to computationally examine the consequence of liver inflammation. Our liver fibrosis ABM (LFABM) is comprised of literature-derived rules describing molecular and histopathological aspects of inflammation and fibrosis in a section of chemically injured liver. Hepatocytes are modeled as agents within hexagonal lobules. Injury triggers an inflammatory reaction, which leads to activation of local Kupffer cells and recruitment of monocytes from circulation. Portal fibroblasts and hepatic stellate cells are activated locally by the products of inflammation. The various agents in the simulation are regulated by above-threshold concentrations of pro- and anti-inflammatory cytokines and damage-associated molecular pattern molecules. The simulation progresses from chronic inflammation to collagen deposition, exhibiting periportal fibrosis followed by bridging fibrosis, and culminating in disruption of the regular lobular structure. The ABM exhibited key histopathological features observed in liver sections from rats treated with carbon tetrachloride (CCl4). An in silico “tension test” for the hepatic lobules predicted an overall increase in tissue stiffness, in line with clinical elastography literature and published studies in CCl4-treated rats. Therapy simulations suggested differential anti-fibrotic effects of neutralizing tumor necrosis factor alpha vs. enhancing M2 Kupffer cells. We conclude that a computational model of liver inflammation on a structural skeleton of physical forces can recapitulate key histopathological and macroscopic properties of CCl4-injured liver. This multiscale approach linking molecular and chemomechanical stimuli enables a model that could be used to gain translationally relevant insights into liver fibrosis.
Collapse
Affiliation(s)
- Joyeeta Dutta-Moscato
- Department of Biomedical Informatics, University of Pittsburgh , Pittsburgh, PA , USA ; Department of Surgery, University of Pittsburgh , Pittsburgh, PA , USA ; Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, PA , USA
| | - Alexey Solovyev
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, PA , USA ; Department of Mathematics, University of Pittsburgh , Pittsburgh, PA , USA
| | - Qi Mi
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, PA , USA ; Department of Sports Medicine and Nutrition, University of Pittsburgh , Pittsburgh, PA , USA
| | - Taichiro Nishikawa
- McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, PA , USA ; Department of Surgery, Children's Hospital of Pittsburgh , Pittsburgh, PA , USA
| | - Alejandro Soto-Gutierrez
- McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, PA , USA ; Department of Pathology, University of Pittsburgh , Pittsburgh, PA , USA ; Thomas E. Starzl Transplantation Institute, University of Pittsburgh , Pittsburgh, PA , USA
| | - Ira J Fox
- McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, PA , USA ; Department of Surgery, Children's Hospital of Pittsburgh , Pittsburgh, PA , USA ; Thomas E. Starzl Transplantation Institute, University of Pittsburgh , Pittsburgh, PA , USA
| | - Yoram Vodovotz
- Department of Surgery, University of Pittsburgh , Pittsburgh, PA , USA ; Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, PA , USA
| |
Collapse
|
50
|
Chheda TK, Shivakumar P, Sadasivan SK, Chanderasekharan H, Moolemath Y, Oommen AM, Madanahalli JR, Marikunte VV. Fast food diet with CCl4 micro-dose induced hepatic-fibrosis--a novel animal model. BMC Gastroenterol 2014; 14:89. [PMID: 24884574 PMCID: PMC4036109 DOI: 10.1186/1471-230x-14-89] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 05/06/2014] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is defined as a spectrum of conditions ranging from hepatocellular steatosis to steatohepatitis and fibrosis, progressing to cirrhosis, which occur in the absence of excessive alcohol use. Several animal models capture aspects of NAFLD but are limited either in their representation of the disease stages or use for development of therapeutics due to the extended periods of time required to develop full histological features. METHODS Here, we report the development of a novel rat model for NAFLD that addresses some of these limitations. We used a fast food diet (FFD) and a CCl4 micro dose (0.5 ml/kg B.wt) for 8 weeks in Wistar rats. Serological analyses, gene expression profiling and liver histology studies were conducted to investigate the development of steatosis, steatohepatitis and fibrosis in the FFD-CCl4 model when compared to the individual effects of a FFD or a micro dose of CCl4 in rats. RESULTS The serum biochemical profile of the FFD-CCl4 model showed an increase in liver injury and fibrosis. This was also accompanied by a significant increase in liver triglycerides (TG), inflammation and oxidative stress. Importantly, we observed extensive fibrosis confirmed by: i) increased gene expression of fibrosis markers and, ii) moderate to severe collagen deposition seen as perisinusoidal and bridging fibrosis using H&E, Trichome and Sirius Red staining. CONCLUSIONS In summary, we find that the FFD-CCl4 rat model developed NAFLD histological features including, steatosis, inflammation and fibrosis in 8 weeks showing promise as a model that can be used to develop NAFLD therapeutics and liver anti-fibrotics.
Collapse
|