1
|
Dohi N, Yamaguchi M, Iwami K, Kaneko YK, Saito SY, Ishikawa T. Mouse liver blood flow is regulated by hepatic stellate cells in response to the sympathetic neurotransmitter norepinephrine. Life Sci 2024; 359:123214. [PMID: 39491770 DOI: 10.1016/j.lfs.2024.123214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND There is no clear information on the regulation of liver blood flow by the autonomic nervous system. We conducted this study to investigate whether quiescent hepatic stellate cells (qHSCs) regulate liver blood flow in response to the sympathetic neurotransmitter norepinephrine (NE). METHODS qHSCs isolated from mice were cultured in Dulbecco's modified Eagle medium without fetal bovine serum for 1 day on collagen gel. NE-induced qHSC contraction was evaluated using the quantitative single-cell contraction measurement method that we had developed previously. For the measurement of liver perfusion pressure in situ, a buffer solution was perfused from the portal vein in mice. RESULTS NE-induced a reversible contraction of qHSCs. This contraction was suppressed by the nonmuscle myosin II inhibitor blebbistatin, the myosin light chain kinase inhibitor ML-9, the Rho kinase inhibitor H-1152, the calmodulin inhibitor W-7, the store-operated calcium channel inhibitor YM-58483, and the IP3 receptor inhibitor xestospongin C. In contrast, the transient receptor potential C channel inhibitor SKF96365 did not affect the NE-induced contraction. CONCLUSION These results suggest that qHSCs contract in response to NE. NEW & NOTEWORTHY The present study provides direct evidence for the first time that norepinephrine (NE) induces a reversible contraction of isolated single quiescent hepatic stellate cells (qHSCs) and further suggests that the NE-mediated qHSC contraction participates in the regulation of liver blood flow in vivo.
Collapse
Affiliation(s)
- Naoki Dohi
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Japan
| | - Momoka Yamaguchi
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Japan.
| | - Kyosuke Iwami
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Japan
| | - Yukiko K Kaneko
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Japan
| | - Shin-Ya Saito
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Japan; Department of Drug Discovery and Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science, Japan
| | - Tomohisa Ishikawa
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Japan
| |
Collapse
|
2
|
Mishra F, Yuan Y, Yang JJ, Li B, Chan P, Liu Z. Depletion of Activated Hepatic Stellate Cells and Capillarized Liver Sinusoidal Endothelial Cells Using a Rationally Designed Protein for Nonalcoholic Steatohepatitis and Alcoholic Hepatitis Treatment. Int J Mol Sci 2024; 25:7447. [PMID: 39000553 PMCID: PMC11242029 DOI: 10.3390/ijms25137447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
Nonalcoholic steatohepatitis (NASH) and alcoholic hepatitis (AH) affect a large part of the general population worldwide. Dysregulation of lipid metabolism and alcohol toxicity drive disease progression by the activation of hepatic stellate cells and the capillarization of liver sinusoidal endothelial cells. Collagen deposition, along with sinusoidal remodeling, alters sinusoid structure, resulting in hepatic inflammation, portal hypertension, liver failure, and other complications. Efforts were made to develop treatments for NASH and AH. However, the success of such treatments is limited and unpredictable. We report a strategy for NASH and AH treatment involving the induction of integrin αvβ3-mediated cell apoptosis using a rationally designed protein (ProAgio). Integrin αvβ3 is highly expressed in activated hepatic stellate cells (αHSCs), the angiogenic endothelium, and capillarized liver sinusoidal endothelial cells (caLSECs). ProAgio induces the apoptosis of these disease-driving cells, therefore decreasing collagen fibril, reversing sinusoid remodeling, and reducing immune cell infiltration. The reversal of sinusoid remodeling reduces the expression of leukocyte adhesion molecules on LSECs, thus decreasing leukocyte infiltration/activation in the diseased liver. Our studies present a novel and effective approach for NASH and AH treatment.
Collapse
Affiliation(s)
- Falguni Mishra
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Yi Yuan
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Jenny J Yang
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | - Bin Li
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Payton Chan
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Zhiren Liu
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
3
|
Thomaz MDL, Vieira CP, Caris JA, Marques MP, Rocha A, Paz TA, Rezende REF, Lanchote VL. Liver Fibrosis Stages Affect Organic Cation Transporter 1/2 Activities in Hepatitis C Virus-Infected Patients. Pharmaceuticals (Basel) 2024; 17:865. [PMID: 39065716 PMCID: PMC11280093 DOI: 10.3390/ph17070865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
This study aims to evaluate the impact of liver fibrosis stages of chronic infection with hepatitis C virus (HCV) on the in vivo activity of organic cation transporters (hepatic OCT1 and renal OCT2) using metformin (MET) as a probe drug. Participants allocated in Group 1 (n = 15, mild to moderate liver fibrosis) or 2 (n = 13, advanced liver fibrosis and cirrhosis) received a single MET 50 mg oral dose before direct-acting antiviral (DAA) drug treatment (Phase 1) and 30 days after achieving sustained virologic response (Phase 2). OCT1/2 activity (MET AUC0-24) was found to be reduced by 25% when comparing the two groups in Phase 2 (ratio 0.75 (0.61-0.93), p < 0.05) but not in Phase 1 (ratio 0.81 (0.66-0.98), p > 0.05). When Phases 1 and 2 were compared, no changes were detected in both Groups 1 (ratio 1.10 (0.97-1.24), p > 0.05) and 2 (ratio 1.03 (0.94-1.12), p > 0.05). So, this study shows a reduction of approximately 25% in the in vivo activity of OCT1/2 in participants with advanced liver fibrosis and cirrhosis after achieving sustained virologic response and highlights that OCT1/2 in vivo activity depends on the liver fibrosis stage of chronic HCV infection.
Collapse
Affiliation(s)
- Matheus De Lucca Thomaz
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; (M.D.L.T.); (C.P.V.); (J.A.C.); (M.P.M.); (A.R.); (T.A.P.)
| | - Carolina Pinto Vieira
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; (M.D.L.T.); (C.P.V.); (J.A.C.); (M.P.M.); (A.R.); (T.A.P.)
| | - Juciene Aparecida Caris
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; (M.D.L.T.); (C.P.V.); (J.A.C.); (M.P.M.); (A.R.); (T.A.P.)
| | - Maria Paula Marques
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; (M.D.L.T.); (C.P.V.); (J.A.C.); (M.P.M.); (A.R.); (T.A.P.)
| | - Adriana Rocha
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; (M.D.L.T.); (C.P.V.); (J.A.C.); (M.P.M.); (A.R.); (T.A.P.)
| | - Tiago Antunes Paz
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; (M.D.L.T.); (C.P.V.); (J.A.C.); (M.P.M.); (A.R.); (T.A.P.)
| | - Rosamar Eulira Fontes Rezende
- Division of Gastroenterology, Department of Internal Medicine, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14049-900, Brazil;
- Reference Center, Hepatitis Outpatient Clinic, Municipal Health Secretary, Ribeirão Preto 14049-900, Brazil
| | - Vera Lucia Lanchote
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; (M.D.L.T.); (C.P.V.); (J.A.C.); (M.P.M.); (A.R.); (T.A.P.)
| |
Collapse
|
4
|
Fattahi N, Gorgannezhad L, Masoule SF, Babanejad N, Ramazani A, Raoufi M, Sharifikolouei E, Foroumadi A, Khoobi M. PEI-based functional materials: Fabrication techniques, properties, and biomedical applications. Adv Colloid Interface Sci 2024; 325:103119. [PMID: 38447243 DOI: 10.1016/j.cis.2024.103119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/15/2024] [Accepted: 02/22/2024] [Indexed: 03/08/2024]
Abstract
Cationic polymers have recently attracted considerable interest as research breakthroughs for various industrial and biomedical applications. They are particularly interesting due to their highly positive charges, acceptable physicochemical properties, and ability to undergo further modifications, making them attractive candidates for biomedical applications. Polyethyleneimines (PEIs), as the most extensively utilized polymers, are one of the valuable and prominent classes of polycations. Owing to their flexible polymeric chains, broad molecular weight (MW) distribution, and repetitive structural units, their customization for functional composites is more feasible. The specific beneficial attributes of PEIs could be introduced by purposeful functionalization or modification, long service life, biocompatibility, and distinct geometry. Therefore, PEIs have significant potential in biotechnology, medicine, and bioscience. In this review, we present the advances in PEI-based nanomaterials, their transfection efficiency, and their toxicity over the past few years. Furthermore, the potential and suitability of PEIs for various applications are highlighted and discussed in detail. This review aims to inspire readers to investigate innovative approaches for the design and development of next-generation PEI-based nanomaterials possessing cutting-edge functionalities and appealing characteristics.
Collapse
Affiliation(s)
- Nadia Fattahi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran; Department of Chemistry, Faculty of Science, University of Zanjan, Zanjan 45371-38791, Iran
| | - Lena Gorgannezhad
- Queensland Micro- and Nanotechnology Centre, Nathan Campus, Griffith University, 170 Kessels Road, Brisbane, QLD 4111, Australia
| | - Shabnam Farkhonde Masoule
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Niloofar Babanejad
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Ali Ramazani
- Department of Chemistry, Faculty of Science, University of Zanjan, Zanjan 45371-38791, Iran.
| | - Mohammad Raoufi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 13169-43551, Iran
| | - Elham Sharifikolouei
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129, Turin (TO), Italy
| | - Alireza Foroumadi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran; Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
| | - Mehdi Khoobi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran; Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Hou C, Wang D, Zhao M, Ballar P, Zhang X, Mei Q, Wang W, Li X, Sheng Q, Liu J, Wei C, Shen Y, Yang Y, Wang P, Shao J, Xu S, Wang F, Sun Y, Shen Y. MANF brakes TLR4 signaling by competitively binding S100A8 with S100A9 to regulate macrophage phenotypes in hepatic fibrosis. Acta Pharm Sin B 2023; 13:4234-4252. [PMID: 37799387 PMCID: PMC10547964 DOI: 10.1016/j.apsb.2023.07.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 05/18/2023] [Accepted: 06/13/2023] [Indexed: 10/07/2023] Open
Abstract
The mesencephalic astrocyte-derived neurotrophic factor (MANF) has been recently identified as a neurotrophic factor, but its role in hepatic fibrosis is unknown. Here, we found that MANF was upregulated in the fibrotic liver tissues of the patients with chronic liver diseases and of mice treated with CCl4. MANF deficiency in either hepatocytes or hepatic mono-macrophages, particularly in hepatic mono-macrophages, clearly exacerbated hepatic fibrosis. Myeloid-specific MANF knockout increased the population of hepatic Ly6Chigh macrophages and promoted HSCs activation. Furthermore, MANF-sufficient macrophages (from WT mice) transfusion ameliorated CCl4-induced hepatic fibrosis in myeloid cells-specific MANF knockout (MKO) mice. Mechanistically, MANF interacted with S100A8 to competitively block S100A8/A9 heterodimer formation and inhibited S100A8/A9-mediated TLR4-NF-κB signal activation. Pharmacologically, systemic administration of recombinant human MANF significantly alleviated CCl4-induced hepatic fibrosis in both WT and hepatocytes-specific MANF knockout (HKO) mice. This study reveals a mechanism by which MANF targets S100A8/A9-TLR4 as a "brake" on the upstream of NF-κB pathway, which exerts an impact on macrophage differentiation and shed light on hepatic fibrosis treatment.
Collapse
Affiliation(s)
- Chao Hou
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Dong Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Mingxia Zhao
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Petek Ballar
- Department of Biochemistry, Faculty of Pharmacy, Ege University, Izmir 35100, Turkey
| | - Xinru Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Qiong Mei
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Wei Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Xiang Li
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Qiang Sheng
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Jun Liu
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Chuansheng Wei
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Yujun Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Yi Yang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Peng Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Juntang Shao
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Sa Xu
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Fuyan Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Yuxian Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
6
|
Yin KL, Li M, Song PP, Duan YX, Ye WT, Tang W, Kokudo N, Gao Q, Liao R. Unraveling the Emerging Niche Role of Hepatic Stellate Cell-derived Exosomes in Liver Diseases. J Clin Transl Hepatol 2023; 11:441-451. [PMID: 36643031 PMCID: PMC9817040 DOI: 10.14218/jcth.2022.00326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/16/2022] [Accepted: 09/23/2022] [Indexed: 01/18/2023] Open
Abstract
Hepatic stellate cells (HSCs) play an essential role in various liver diseases, and exosomes are critical mediators of intercellular communication in local and distant microenvironments. Cellular crosstalk between HSCs and surrounding multiple tissue-resident cells promotes or inhibits the activation of HSCs. Substantial evidence has revealed that HSC-derived exosomes are involved in the occurrence and development of liver diseases through the regulation of retinoid metabolism, lipid metabolism, glucose metabolism, protein metabolism, and mitochondrial metabolism. HSC-derived exosomes are underpinned by vehicle molecules, such as mRNAs and microRNAs, that function in, and significantly affect, the processes of various liver diseases, such as acute liver injury, alcoholic liver disease, nonalcoholic fatty liver disease, viral hepatitis, fibrosis, and cancer. As such, numerous exosomes derived from HSCs or HSC-associated exosomes have attracted attention because of their biological roles and translational applications as potential targets for therapeutic targets. Herein, we review the pathophysiological and metabolic processes associated with HSC-derived exosomes, their roles in various liver diseases and their potential clinical application.
Collapse
Affiliation(s)
- Kun-Li Yin
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ming Li
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Pei-Pei Song
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Yu-Xin Duan
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wen-Tao Ye
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Tang
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Norihiro Kokudo
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
- Correspondence to: Qiang Gao, Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, 180 Fenglin Road, Shanghai 200032, China. ORCID: https://orcid.org/0000-0002-6695-9906. ; Rui Liao, Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, 1 Youyi Road, Chongqing 400016, China. ORCID: https://orcid.org/0000-0002-0057-2792. E-mail:
| | - Rui Liao
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Correspondence to: Qiang Gao, Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, 180 Fenglin Road, Shanghai 200032, China. ORCID: https://orcid.org/0000-0002-6695-9906. ; Rui Liao, Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, 1 Youyi Road, Chongqing 400016, China. ORCID: https://orcid.org/0000-0002-0057-2792. E-mail:
| |
Collapse
|
7
|
Song Z, Liu N, He Y, Chen J, Li J, Wang F, Wu Z. Knockout of ICAT in Adipose Tissue Alleviates Fibro-inflammation in Obese Mice. Inflammation 2023; 46:404-417. [PMID: 36181623 DOI: 10.1007/s10753-022-01742-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/08/2022] [Accepted: 09/14/2022] [Indexed: 11/24/2022]
Abstract
The E2 promoter binding factor 1 (E2F1) and the Wnt/β-catenin signaling are crucial in regulating metabolic homeostasis including obesity. The β-catenin interacting protein 1 (CTNNBIP1), also known as the inhibitor of β-catenin and TCF4 (ICAT), is required for E2F1 to inhibit the activity of β-catenin. However, the role of ICAT in E2F1 regulating obesity-related metabolic disorders remains unknown. In the present study, male adipose tissue-specific ICAT knockout (ICATadi-/-) C57BL/6 J mice and control littermates aged 6-8 weeks were fed with high-fat diet (HFD) for 12 weeks to explore the effect of ICAT on lipid metabolism and obesity-related disorders. Results showed that the adipose tissue-specific ICAT knockout had negligible effect on lipid metabolism, reflected by no difference in body weight, fat mass, and the expression of proteins involved in lipid metabolism in white adipose tissue (WAT) and the liver between the ICATadi-/- mice and the control littermate (ICATfl/fl) mice. However, the knockout of ICAT reduced inflammatory response in WAT and the liver. Additionally, Sirius red staining results showed that deletion of ICAT attenuated fibrosis and reduced mRNA levels of transforming growth factor β1(TGF-β1), matrix metallopeptidase 2 (Mmp2), Mmp3, and collagen, type V, alpha 1 (Col5a1) in WAT and the liver. These results suggested that knockout of ICAT improved the metabolic abnormalities of obese mice through attenuating adipose tissue and the liver inflammation as well as fibrosis. Our findings may provide a new insight to understand the role of ICAT in inflammation and fibrosis.
Collapse
Affiliation(s)
- Zhuan Song
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Ning Liu
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, China
| | - Yu He
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Jingqing Chen
- Laboratory Animal Center of the Academy of Military Medical Sciences, Beijing, 100193, China
| | - Jun Li
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Fengchao Wang
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
| | - Zhenlong Wu
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China. .,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
8
|
Chu Y, Bao L, Teng Y, Yuan B, Ma L, Liu Y, Kang H. The Fibrotic Effects of LINC00663 in Human Hepatic Stellate LX-2 Cells and in Bile Duct-Ligated Cholestasis Mice Are Mediated through the Splicing Factor 2-Fibronectin. Cells 2023; 12:cells12020215. [PMID: 36672150 PMCID: PMC9857260 DOI: 10.3390/cells12020215] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/25/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
Hepatic fibrosis can develop into cirrhosis or even cancer without active therapy at an early stage. Long non-coding RNAs (lncRNAs) have been shown to be involved in the regulation of a wide variety of important biological processes. However, lncRNA mechanism(s) involved in cholestatic liver fibrosis remain unclear. RNA sequence data of hepatic stellate cells from bile duct ligation (BDL) mice or controls were analyzed by weighted gene co-expression network analysis (WGCNA). Based on WGCNA analysis, a competing endogenous RNA network was constructed. We identified LINC00663 and evaluated its function using a panel of assays, including a wound healing assay, a dual-luciferase reporter assay, RNA binding protein immunoprecipitation and chromatin immunoprecipitation. Functional research showed that LINC00663 promoted the activation, migration and epithelial-mesenchymal transition (EMT) of LX-2 cells and liver fibrosis in BDL mice. Mechanistically, LINC00663 regulated splicing factor 2 (SF2)-fibronectin (FN) alternative splicing through the sponging of hsa-miR-3916. Moreover, forkhead box A1 (FOXA1) specifically interacted with the promoter of LINC00663. In summary, we elaborated the fibrotic effects of LINC00663 in human hepatic stellate LX-2 cells and in bile duct-ligated cholestasis mice. We established a FOXA1/LINC00663/hsa-miR-3916/SF2-FN axis that provided a potential target for the diagnosis and targeted therapy of cholestatic liver fibrosis.
Collapse
Affiliation(s)
- Yang Chu
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Linan Bao
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Yun Teng
- Department of Laboratory Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Bo Yuan
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Lijie Ma
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Ying Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Hui Kang
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
- Correspondence:
| |
Collapse
|
9
|
20-Hydroxytetraenoic acid induces hepatic fibrosis via the TGF-β1/Smad3 signaling pathway. Toxicol Lett 2022; 373:1-12. [PMID: 36368619 DOI: 10.1016/j.toxlet.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/24/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022]
Abstract
Hepatic fibrosis is caused by excessive accumulation of extracellular matrix (ECM) due to repeated liver injury. Hepatic stellate cells (HSCs) play a key role in the pathogenesis and progression of hepatic fibrosis. A study showed that CYP4A14 gene defect can inhibit hepatic fibrosis, but the specific mechanism was not clear. In this experiment, patients with hepatic fibrosis, LX-2 cells (a human HSCs line), and mice with liver fibrosis induced by carbon tetrachloride (CCl4) were used to study the effect of 20-Hydroxytetraenoic acid (20-HETE), one of the main metabolites of arachidonic acid (AA) catalyzed by CYP4A enzyme, on hepatic fibrosis and its mechanism. Our experimental results showed that the 20-HETE of patients with hepatic fibrosis is significantly higher than that of normal people and is closely related to the degree of fibrosis. 20-HETE could induce activation of LX-2 cells and 20-HETE antagonist could inhibit the induction of 20-HETE. 20-HETE was significantly increased in CCl4-induced liver fibrosis mice and inhibition of 20-HETE production could attenuate hepatic fibrosis. 20-HETE induced hepatic fibrosis mainly via the TGF- β1/Smad3 signal pathway. In conclusion, the results suggest that 20-HETE plays an important role in hepatic fibrosis and may be a possible target for the clinical treatment of hepatic fibrosis.
Collapse
|
10
|
Wang S, Zhang J, Chen H, Zhan X, Nie H, Wang C, Zhang Y, Zheng B, Gong Q. MicroRNA-181b promotes schistosomiasis-induced hepatic fibrosis by targeting Smad7. Mol Biochem Parasitol 2022; 252:111523. [PMID: 36195241 DOI: 10.1016/j.molbiopara.2022.111523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 09/08/2022] [Accepted: 09/29/2022] [Indexed: 12/31/2022]
Abstract
Schistosomiasis is a common parasitic disease. Hepatosplenic schistosomiasis, caused by Schistosoma japonicum and Schistosoma mansoni, involves pathological changes, including worm egg-induced hepatic granuloma and fibrosis, which can markedly affect the liver's physiological functions. Although the drug praziquantel (PZQ) is used to treat schistosomiasis, drugs against schistosomiasis-induced liver fibrosis are rare in the clinical setting. Therefore, developing effective strategies to prevent and treat schistosomiasis-induced liver fibrosis is crucial. Previous studies have shown that miRNAs are involved in various liver diseases. In this study, we found a gradual increase in miR-181b expression in the murine liver as S. japonicum infection progressed, while the expression of Smad7 decreased. Down-regulating miR-181b significantly alleviated S. japonicum-induced hepatic granuloma and liver fibrosis. In vitro experiments showed that treatment with TGF-β1 upregulated miR-181b levels in the hepatic stellate cell (HSC) line LX2 in a concentration- and time-dependent manner. Downregulation of miR-181b significantly decreased collagen type I alpha 1 chain (COL1A1) expression in TGF-β1-stimulated LX2 cells. These findings indicate that miR-181b promotes HSC activation by down-regulating Smad7 expression, activating the TGF-β1/Smad signaling pathway, and leading to excess collagen expression and deposition. Our findings suggest that miR-181b might be a potentially novel therapeutic target for schistosomiasis-induced liver fibrosis.
Collapse
Affiliation(s)
- Shu Wang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Jianqiang Zhang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China; Department of Nephrology, Ezhou Central Hospital, Ezhou, China
| | - Hui Chen
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China; Department of Laboratory Medicine, First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Xiang Zhan
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Hao Nie
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Chao Wang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Yanxiang Zhang
- Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Bing Zheng
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China.
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China.
| |
Collapse
|
11
|
Mesenchymal Stem Cells Influence Activation of Hepatic Stellate Cells, and Constitute a Promising Therapy for Liver Fibrosis. Biomedicines 2021; 9:biomedicines9111598. [PMID: 34829827 PMCID: PMC8615475 DOI: 10.3390/biomedicines9111598] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 12/12/2022] Open
Abstract
Liver fibrosis is a common feature of chronic liver disease. Activated hepatic stellate cells (HSCs) are the main drivers of extracellular matrix accumulation in liver fibrosis. Hence, a strategy for regulating HSC activation is crucial in treating liver fibrosis. Mesenchymal stem cells (MSCs) are multipotent stem cells derived from various post-natal organs. Therapeutic approaches involving MSCs have been studied extensively in various diseases, including liver disease. MSCs modulate hepatic inflammation and fibrosis and/or differentiate into hepatocytes by interacting directly with immune cells, HSCs, and hepatocytes and secreting modulators, thereby contributing to reduced liver fibrosis. Cell-free therapy including MSC-released secretomes and extracellular vesicles has elicited extensive attention because they could overcome MSC transplantation limitations. Herein, we provide basic information on hepatic fibrogenesis and the therapeutic potential of MSCs. We also review findings presenting the effects of MSC itself and MSC-based cell-free treatments in liver fibrosis, focusing on HSC activation. Growing evidence supports the anti-fibrotic function of either MSC itself or MSC modulators, although the mechanism underpinning their effects on liver fibrosis has not been established. Further studies are required to investigate the detailed mechanism explaining their functions to expand MSC therapies using the cell itself and cell-free treatments for liver fibrosis.
Collapse
|
12
|
Han H, Zhang Y, Peng G, Li L, Yang J, Yuan Y, Xu Y, Liu ZR. Extracellular PKM2 facilitates organ-tissue fibrosis progression. iScience 2021; 24:103165. [PMID: 34693222 PMCID: PMC8517170 DOI: 10.1016/j.isci.2021.103165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/26/2021] [Accepted: 09/21/2021] [Indexed: 01/06/2023] Open
Abstract
Persistent activation of fibroblasts and resistance of myofibroblasts to turnover play important roles in organ-tissue fibrosis development and progression. The mechanism that mediates apoptosis resistance of myofibroblasts is not understood. Here, we report that myofibroblasts express and secrete PKM2. Extracellular PKM2 (EcPKM2) facilitates progression of fibrosis by protecting myofibroblasts from apoptosis. EcPKM2 upregulates arginase-1 expression in myofibroblasts and therefore facilitates proline biosynthesis and subsequent collagen production. EcPKM2 interacts with integrin αvβ3 on myofibroblasts to activate FAK-PI3K signaling axis. Activation of FAK-PI3K by EcPKM2 activates downstream NF-κB survival pathway to prevent myofibroblasts from apoptosis. On the other hand, activation of FAK- PI3K by EcPKM2 suppresses PTEN to subsequently upregulate arginase-1 in myofibroblasts. Our studies uncover an important mechanism for organ fibrosis progression. More importantly, an antibody disrupting the interaction between PKM2 and integrin αvβ3 is effective in reversing fibrosis, suggesting a possible therapeutic strategy and target for treatment of organ fibrosis.
Collapse
Affiliation(s)
- Hongwei Han
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Yinwei Zhang
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Guangda Peng
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Liangwei Li
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Jenny Yang
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | - Yi Yuan
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Yiting Xu
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | - Zhi-Ren Liu
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
13
|
Turaga RC, Satyanarayana G, Sharma M, Yang JJ, Wang S, Liu C, Li S, Yang H, Grossniklaus H, Farris AB, Gracia-Sancho J, Liu ZR. Targeting integrin αvβ3 by a rationally designed protein for chronic liver disease treatment. Commun Biol 2021; 4:1087. [PMID: 34531529 PMCID: PMC8445973 DOI: 10.1038/s42003-021-02611-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/26/2021] [Indexed: 12/22/2022] Open
Abstract
Chronic Liver Diseases (CLD) are characterized by abnormal accumulation of collagen fibrils, neo-angiogenesis, and sinusoidal remodeling. Collagen deposition along with intrahepatic angiogenesis and sinusoidal remodeling alters sinusoid structure resulting in portal hypertension, liver failure, and other complications. Efforts were made to develop treatments for CLDs. However, the success of such treatments is limited and unpredictable. We report a strategy for CLD treatment by induction of integrin αvβ3 mediated cell apoptosis using a rationally designed protein (ProAgio). ProAgio is designed to target integrin αvβ3 at a novel site. Integrin αvβ3 is highly expressed in activated Hepatic Stellate Cells (HSC), angiogenic endothelium, and capillarized Liver Sinusoidal Endothelial Cells (LSEC). ProAgio induces apoptosis of these disease causative cells. Tests with liver fibrosis mouse models demonstrate that ProAgio reverses liver fibrosis and relieves blood flow resistance by depleting activated HSC and capillarized LSEC. Our studies demonstrate an effective approach for CLD treatment.
Collapse
Affiliation(s)
- Ravi Chakra Turaga
- Department of Biology, Georgia State University, Atlanta, GA, 30324, USA
| | | | - Malvika Sharma
- Department of Biology, Georgia State University, Atlanta, GA, 30324, USA
| | - Jenny J Yang
- Department of Chemistry, Georgia State University, Atlanta, USA
| | | | | | - Sun Li
- Department of Chemistry, Georgia State University, Atlanta, USA
| | - Hua Yang
- Department Ophthalmology, Emory University, Atlanta, GA, 30322, USA
| | | | | | | | - Zhi-Ren Liu
- Department of Biology, Georgia State University, Atlanta, GA, 30324, USA.
| |
Collapse
|
14
|
Role of the sympathetic nervous system in cardiometabolic control: implications for targeted multiorgan neuromodulation approaches. J Hypertens 2021; 39:1478-1489. [PMID: 33657580 DOI: 10.1097/hjh.0000000000002839] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sympathetic overdrive plays a key role in the perturbation of cardiometabolic homeostasis. Diet-induced and exercise-induced weight loss remains a key strategy to combat metabolic disorders, but is often difficult to achieve. Current pharmacological approaches result in variable responses in different patient cohorts and long-term efficacy may be limited by medication intolerance and nonadherence. A clinical need exists for complementary therapies to curb the burden of cardiometabolic diseases. One such approach may include interventional sympathetic neuromodulation of organs relevant to cardiometabolic control. The experience from catheter-based renal denervation studies clearly demonstrates the feasibility, safety and efficacy of such an approach. In analogy, denervation of the common hepatic artery is now feasible in humans and may prove to be similarly useful in modulating sympathetic overdrive directed towards the liver, pancreas and duodenum. Such a targeted multiorgan neuromodulation strategy may beneficially influence multiple aspects of the cardiometabolic disease continuum offering a holistic approach.
Collapse
|
15
|
Xie Z, Wu Y, Liu S, Lai Y, Tang S. LncRNA-SNHG7/miR-29b/DNMT3A axis affects activation, autophagy and proliferation of hepatic stellate cells in liver fibrosis. Clin Res Hepatol Gastroenterol 2021; 45:101469. [PMID: 32893175 DOI: 10.1016/j.clinre.2020.05.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/13/2020] [Accepted: 05/20/2020] [Indexed: 02/04/2023]
Abstract
OBJECTIVES To determine the relative expression of long non-coding small nucleolar RNA host gene 7 (lncRNA-SNHG7) in fibrotic liver and hepatic stellate cells, and investigate the biological effects and mechanisms of SNHG7 on liver fibrosis. METHODS Liver fibrosis model of mice was established, primary hepatic stellate cells (HSCs) were cultured from normal mice and induced to activate by TGF-β. Cell viability, proliferation, and autophagy were detected with MTT, BrdU, and MDC stain, respectively. Liver tissue stained with Masson and Sirius Red. The interaction between SNHG7 and miR-29b was investigated by immunoprecipitation, RNA pull-down and Dual-luciferase reporter gene assay. The effects of SNHG7 on the expression of miR-29b, DNMT3A and liver fibrosis related factors were detected in vitro or in vivo transfection experiments. RESULTS SNHG7 was signally increased in liver tissue and HSCs of liver fibrosis model of mice, and inhibition of SNHG7 expression in liver fibrosis mice can reduce liver fibrosis. We also found that SNHG7 could bind to miR-29b in HSCs and inhibit the expression of miR-29b. In TGF-β-stimulated normal HSCs, knockdown of SNHG7 expression after shSNHG7 transfection could restrain DNMT3A and HSCs activation factors α-SMA, Collα1 and autophagy-related factors LC3I/II, Beclin1. However, this shSNHG7 effect was reversed by the inhibition of miR-29b. CONCLUSION Inhibition of lncRNA-SNHG7 can inhibit liver fibrosis. This is partly due to SNHG7 acts as a competitive endogenous RNA (ceRNA) to affect the expression of DNMT3A, a downstream target gene of miR-29b, by binding to miR-29b, thereby affecting the activation, autophagy and proliferation of HSCs.
Collapse
Affiliation(s)
- Zhengyuan Xie
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No.1 Mingde Road, Nanchang 330006, People's Republic of China.
| | - Yuanhao Wu
- The Second Medical College Of Nanchang University, Nanchang, Jiangxi, China
| | - Sifu Liu
- Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Yueliang Lai
- Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Shenglan Tang
- Medical College of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
16
|
Optimization of the isolation procedure and culturing conditions for hepatic stellate cells obtained from mouse. Biosci Rep 2021; 41:227415. [PMID: 33350435 PMCID: PMC7823195 DOI: 10.1042/bsr20202514] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/28/2020] [Accepted: 12/18/2020] [Indexed: 12/28/2022] Open
Abstract
Liver fibrosis (LF) mortality rate is approximately 2 million per year. Irrespective of the etiology of LF, a key element in its development is the transition of hepatic stellate cells (HSCs) from a quiescent phenotype to a myofibroblast-like cell with the production of fibrotic proteins. It is necessary to define optimal isolation and culturing conditions for good HSCs yield and proper phenotype preservation for studying the activation of HSCs in vitro. In the present study, the optimal conditions of HSC isolation and culture were examined to maintain the HSC’s undifferentiated phenotype. HSCs were isolated from Balb/c mice liver using Nycodenz, 8, 9.6, and 11%. The efficiency of the isolation procedure was evaluated by cell counting and purity determination by flow cytometry. Quiescent HSCs were cultured in test media supplemented with different combinations of fetal bovine serum (FBS), glutamine (GLN), vitamin A (vitA), insulin, and glucose. The cells were assessed at days 3 and 7 of culture by evaluating the morphology, proliferation using cell counting kit-8, lipid storage using Oil Red O (ORO) staining, expression of a-smooth muscle actin, collagen I, and lecithin-retinol acyltransferase by qRT-PCR and immunocytochemistry (ICC). The results showed that Nycodenz, at 9.6%, yielded the best purity and quantity of HSCs. Maintenance of HSC undifferentiated phenotype was achieved optimizing culturing conditions (serum-free Dulbecco’s Modified Eagle’s Medium (DMEM) supplemented with glucose (100 mg/dl), GLN (0.5 mM), vitA (100 μM), and insulin (50 ng/ml)) with a certain degree of proliferation allowing their perpetuation in culture. In conclusion, we have defined optimal conditions for HSCs isolation and culture.
Collapse
|
17
|
Lee SW, Jung DJ, Jeong GS. Gaining New Biological and Therapeutic Applications into the Liver with 3D In Vitro Liver Models. Tissue Eng Regen Med 2020; 17:731-745. [PMID: 32207030 PMCID: PMC7710770 DOI: 10.1007/s13770-020-00245-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Three-dimensional (3D) cell cultures with architectural and biomechanical properties similar to those of natural tissue have been the focus for generating liver tissue. Microarchitectural organization is believed to be crucial to hepatic function, and 3D cell culture technologies have enabled the construction of tissue-like microenvironments, thereby leading to remarkable progress in vitro models of human tissue and organs. Recently, to recapitulate the 3D architecture of tissues, spheroids and organoids have become widely accepted as new practical tools for 3D organ modeling. Moreover, the combination of bioengineering approach offers the promise to more accurately model the tissue microenvironment of human organs. Indeed, the employment of sophisticated bioengineered liver models show long-term viability and functional enhancements in biochemical parameters and disease-orient outcome. RESULTS Various 3D in vitro liver models have been proposed as a new generation of liver medicine. Likewise, new biomedical engineering approaches and platforms are available to more accurately replicate the in vivo 3D microarchitectures and functions of living organs. This review aims to highlight the recent 3D in vitro liver model systems, including micropatterning, spheroids, and organoids that are either scaffold-based or scaffold-free systems. Finally, we discuss a number of challenges that will need to be addressed moving forward in the field of liver tissue engineering for biomedical applications. CONCLUSION The ongoing development of biomedical engineering holds great promise for generating a 3D biomimetic liver model that recapitulates the physiological and pathological properties of the liver and has biomedical applications.
Collapse
Affiliation(s)
- Sang Woo Lee
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-Ro 43 Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Da Jung Jung
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-Ro 43 Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Gi Seok Jeong
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-Ro 43 Gil, Songpa-Gu, Seoul, 05505, Republic of Korea.
- Department of Convergence Medicine, University of Ulsan College of Medicine, 88 Olympic-Ro 43 Gil, Songpa-Gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
18
|
Hamza AA, Lashin FM, Gamel M, Hassanin SO, Abdalla Y, Amin A. Hawthorn Herbal Preparation from Crataegus oxyacantha Attenuates In Vivo Carbon Tetrachloride -Induced Hepatic Fibrosis via Modulating Oxidative Stress and Inflammation. Antioxidants (Basel) 2020; 9:antiox9121173. [PMID: 33255507 PMCID: PMC7760839 DOI: 10.3390/antiox9121173] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/14/2020] [Accepted: 11/21/2020] [Indexed: 02/06/2023] Open
Abstract
Hawthorn (HAW) is a herbal preparation extracted from Crataegus oxyacantha. HAW has cardioprotective, antioxidants, anti-inflammatory, and anti-hypotensive effects. HAW’s effect on hepatic fibrosis remains, however, unknown. This study evaluated the impact of HAW on carbon tetrachloride (CCl4)-induced hepatic fibrosis in rats and elucidated its mechanisms. HAW reduced liver index and the serum liver enzyme markers and reduced liver damage, and fibrosis as confirmed by histopathological scoring of hematoxylin-eosin staining. Collagen deposition was reduced in HAW group compared to CCl4 group as confirmed by Masson staining, hydroxyproline content, and both mRNA and protein levels of alpha-smooth muscle actin, collagen 1 and 3. HAW also down regulated the gene expressions of inflammatory markers including interleukin-IL-1β, tumor necrosis factor-α, transforming growth factor-β 1, nuclear factor kappa-B, and cyclooxygenase-2 and decreased the myeloperoxidase activity. The effects of HAW was also associated with decreased levels of hepatic oxidative stress markers (malondialdehyde and P.Carbonyl) and with increased activity of superoxide dismutase. Those effects are possibly mediated by blocking the pro-oxidant machinery and down regulating the inflammatory and profibrotic responses. Finally, chlorogenic acid, epicatechin, rutin, vitexin quercetin, and iso quercetin were identified as the major species of polyphenols of the HAW herbal preparation used here. Therefore, HAW’s potent protecting effects against liver fibrosis predicts a significant beneficial application.
Collapse
Affiliation(s)
- Alaaeldin Ahmed Hamza
- Hormone Evaluation Department, National Organization for Drug Control and Research, Giza 12611, Egypt; (F.M.L.); (M.G.)
- Correspondence: (A.A.H.); (A.A.)
| | - Fawzy Mohamed Lashin
- Hormone Evaluation Department, National Organization for Drug Control and Research, Giza 12611, Egypt; (F.M.L.); (M.G.)
| | - Mona Gamel
- Hormone Evaluation Department, National Organization for Drug Control and Research, Giza 12611, Egypt; (F.M.L.); (M.G.)
| | - Soha Osama Hassanin
- Biochemistry Department, Modern University for Technology and information, Cairo 11585, Egypt;
| | - Youssef Abdalla
- Department of Kinesiology, Michigan State University, East Lansing, MI 48823, USA;
| | - Amr Amin
- Biological Sciences Collegiate Division, The University of Chicago, Chicago, IL 60637, USA
- Correspondence: (A.A.H.); (A.A.)
| |
Collapse
|
19
|
Ustuner D, Kolac UK, Ustuner MC, Tanrikut C, Ozdemir Koroglu Z, Burukoglu Donmez D, Ozen H, Ozden H. Naringenin Ameliorate Carbon Tetrachloride-Induced Hepatic Damage Through Inhibition of Endoplasmic Reticulum Stress and Autophagy in Rats. J Med Food 2020; 23:1192-1200. [PMID: 32125927 DOI: 10.1089/jmf.2019.0265] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hepatic fibrosis emerges upon exposure of liver to various chemicals and if not treated, it develops various diseases such as cirrhosis and cancer. Carbon tetrachloride (CCl4) is a widely used toxin in animal models to develop hepatic fibrosis. Accumulation of unfolded proteins in cells causes stress in the endoplasmic reticulum (ER) and various mechanisms are involved in the cell to reduce the damage caused by these unfolding proteins. The most well known of these is the unfolded protein response. Further, autophagy works to remove these proteins if the damage cannot be repaired and is permanent. In our study, we investigated the effects of naringenin (NRG), a flavanon abundant in citrus fruits, on ER stress and autophagy in CCl4-injured rat liver. The animals were given 0.2 mL/kg of CCl4 for 10 days and treatment group was administered 100 mg/kg of NRG for 14 days. Histopathological examination was performed to show liver damage and to determine the therapeutic properties of the active substance. Transmission electron microscopy (TEM) analysis was carried out to establish cell level damage and effect of treatment. In addition, levels of ER stress and autophagy markers of liver were measured. According to our findings, TEM demonstrated positive effect of NRG and histological examinations reported ameliorative effects. In addition, NRG reduced levels of ER stress markers and inhibited autophagy significantly compared to CCl4-treated group. As a result, NRG significantly reduced damage in hepatocytes and provided a significant amelioration.
Collapse
Affiliation(s)
- Derya Ustuner
- Department of Medical Laboratory, Vocational School of Health Services Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Umut Kerem Kolac
- Department of Medical Biology, Faculty of Medicine, Aydın Adnan Menderes University, Efeler, Turkey
| | - Mehmet Cengiz Ustuner
- Department of Medical Biology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Cihan Tanrikut
- Department of Medical Biology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Zeynep Ozdemir Koroglu
- Department of Medical Laboratory, Vocational School of Health Services Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Dilek Burukoglu Donmez
- Department of Histology and Embryology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Hulya Ozen
- Department of Biostatistics and Medical Informatics, and Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Hilmi Ozden
- Department of Anatomy, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| |
Collapse
|
20
|
Miao Y, Wu Y, Jin Y, Lei M, Nan J, Wu X. Benzoquinone derivatives with antioxidant activity inhibit activated hepatic stellate cells and attenuate liver fibrosis in TAA-induced mice. Chem Biol Interact 2020; 317:108945. [DOI: 10.1016/j.cbi.2020.108945] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 12/31/2019] [Accepted: 01/10/2020] [Indexed: 02/07/2023]
|
21
|
Sha M, Gao Y, Deng C, Wan Y, Zhuang Y, Hu X, Wang Y. Therapeutic effects of AdipoRon on liver inflammation and fibrosis induced by CCl 4 in mice. Int Immunopharmacol 2020; 79:106157. [PMID: 31911372 DOI: 10.1016/j.intimp.2019.106157] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/23/2019] [Accepted: 12/26/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The present work aimed to investigate the effects of AdipoRon against acute hepatitis and liver fibrosis induced by carbon tetrachloride (CCl4) in mice. METHODS C57BL/6 mice were randomly divided into five groups: control, model, AdipoRon groups (three different dosages), CCl4 was administered to induce acute hepatitis or liver fibrosis except for control group. The liver function, inflammatory and fibrotic profiles were evaluated by histology, immunohistochemistry and expression analysis, respectively. RESULTS AdipoRon pretreatment effectively attenuated oxidative stress and hepatocellular damage in acute CCl4 intoxication, demonstrated by marked reduction in peroxidation indexes [hepatic malonaldehyde (MDA), total nitric oxide synthase (tNOS), inducible nitric oxide synthase (iNOS)] and serum transaminases [alanine aminotransferase (ALT), aspartate transaminase (AST)]. Moreover, AdipoRon attenuated the severity of fibrosis induced by sustaining CCl4 challenge, with the alleviation of fibrous deposit and architecture distortion. The levels of canonical fibrosis markers (aminotransferases, hydroxyproline, hyaluronic acid, laminin) were also dose-dependently modulated by AdipoRon. Immunochemistry and expression analysis showed AdipoRon restrained the proinflammatory and profibrotic cytokines (TNF-α, TGF-β1, α-SMA, COL1A1), which somehow, ascribed the anti-fibrotic action to inhibiting hepatic stellate cells (HSCs) activation and quenching specific inflammation-fibrogenesis pathways. CONCLUSIONS AdipoRon demonstrates a remedial capacity against hepatitis and fibrosis induced by CCl4, potentially by inflammation restraint and HSC deactivation, which might pave the way for its therapeutical application in hepatic fibrosis.
Collapse
Affiliation(s)
- Min Sha
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Yaru Gao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Can Deng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Yuemeng Wan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Yuan Zhuang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaochuan Hu
- Department of Occupational Disease, Qingdao Central Hospital, Shandong, China
| | - Ying Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
22
|
Differentiation-inducing factor-1 prevents hepatic stellate cell activation through inhibiting GSK3β inactivation. Biochem Biophys Res Commun 2019; 520:140-144. [PMID: 31582219 DOI: 10.1016/j.bbrc.2019.09.117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 09/26/2019] [Indexed: 11/22/2022]
Abstract
Differentiation-inducing factor-1 (DIF-1), a morphogen produced by the cellular slime mold Dictyostelium discoideum, is a natural product that has attracted considerable attention for its antitumor properties. Here, we report a novel inhibitory effect of DIF-1 on the activation of hepatic stellate cells (HSCs) responsible for liver fibrosis. DIF-1 drastically inhibited transdifferentiation of quiescent HSCs into myofibroblastic activated HSCs in a concentration-dependent manner, thus conferring an antifibrotic effect against in the liver. Neither SQ22536, an adenylate cyclase inhibitor, nor ODQ, a guanylate cyclase inhibitor, showed any effect on the inhibition of HSC activation by DIF-1. In contrast, TWS119, a glycogen synthase kinase 3β (GSK3β) inhibitor, attenuated the inhibitory effect of DIF-1. Moreover, the level of inactive GSK3β (phosphorylated at Ser9) was significantly reduced by DIF-1. DIF-1 also inhibited nuclear translocation of β-catenin and reduced the level of non-phospho (active) β-catenin. These results suggest that DIF-1 inhibits HSC activation by disrupting the Wnt/β-catenin signaling pathway through dephosphorylation of GSK3β. We propose that DIF-1 is a possible candidate as a therapeutic agent for preventing liver fibrosis.
Collapse
|
23
|
Maia-Ceciliano TC, Dutra RR, Aguila MB, Mandarim-De-Lacerda CA. The deficiency and the supplementation of vitamin D and liver: Lessons of chronic fructose-rich diet in mice. J Steroid Biochem Mol Biol 2019; 192:105399. [PMID: 31175967 DOI: 10.1016/j.jsbmb.2019.105399] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 03/25/2019] [Accepted: 06/05/2019] [Indexed: 01/01/2023]
Abstract
The fructose added to soft drinks and processed food, as well as frequent detection of vitamin D deficiency in the body, are two insults increasingly considered to cause lesions in target organs. We studied the liver after a chronic high-fructose diet deficient and supplemented with vitamin D. Sixty C57BL/6 mature male mice were allocated into six groups (n = 10) for ten weeks: control (C), control deficient in vitamin D (CDD), control supplemented with vitamin D (CDS), fructose (F), fructose deficient in vitamin D (FDD), and fructose supplemented with vitamin D (FDS). The gene expressions of vitamin D receptor and CYP27B1 and 25 hydroxyvitamin D plasma level ensured that the diets caused vitamin D deficiency or supplementation. Body mass did not change, but blood pressure (BP) increased in CDD, F, and FDD, whereas BP was controlled in FDS. Insulinemia, insulin tolerance and resistance were seen in both vitamin D deficiency and fructose groups but improved with vitamin D supplementation. The steatosis and fibrosis were observed in the CDD, F and FDD groups. Also, F and FDD showed activation of stellate cells (HSC). Lipogenesis and inflammation gene expressions were enhanced in the CDD, F and FDD groups, but diminished with vitamin D supplementation. In conclusion, we demonstrated the adverse effects of vitamin D deficiency on metabolism, liver steatosis and, combined with fructose intake, liver interstitial fibrosis with hepatic stellate cell activation, and alteration of the lipogenesis, beta-oxidation, and liver inflammation. All these data improved when vitamin D was supplemented in the animals.
Collapse
Affiliation(s)
- Thais C Maia-Ceciliano
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Rafaela R Dutra
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Marcia B Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Carlos A Mandarim-De-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
24
|
Liang Y, Pan Q, Wang R, Ye Z, Li Z, Zeng L, Chen Y, Ma X, Li M, Miao H. Microvesicles Derived from TGF-β1 Stimulated Hepatic Stellate Cells Aggravate Hepatocellular Injury. Stem Cells Dev 2019; 28:1128-1139. [PMID: 31140359 DOI: 10.1089/scd.2019.0032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hepatic stellate cells (HSCs) are liver-specific cells playing critical roles in liver physiological and pathophysiological processes. Transforming growth factor-β1 (TGF-β1) is an inflammatory cytokine secreted by both hepatocytes and HSCs. We have previously shown that microvesicles (MVs) derived from quiescent HSCs protect hepatocyte functions. In this study, we investigated the effects of MVs released from TGF-β1-stimulated HSCs (HSC-MVs) on xenobiotic-injured hepatocytes. Two hepatocyte cell lines (BRL-3A and HL-7702) were treated with N-acetyl-p-aminophenol or H2O2 to build the injury models. Different concentrations of HSC-MVs were used to coculture with injured hepatocytes. MTT, Hochest33258 staining, and flow cytometry were used to determine their effects on the viability and apoptosis of hepatocytes. Liver injury indicators, alanine aminotransferase (ALT) and aspartate amino transferase (AST), were assessed by enzyme-linked immune sorbent assay kits. The phosphoinositide 3-kinase (PI3K) activator (740Y-P) and extracelluar signal regulated kinase (Erk)1/2 activator (platelet-derived growth factor-BB) were used for pathway analysis. The expression levels of p-PI3K/PI3K, p-Akt/Akt, and activated caspase-3 were measured by western blot. Results showed that (i) HSC-MVs dose dependently impaired the viability of hepatocytes in both injury models, (ii) moreover, HSC-MVs dose dependently increased the apoptosis in those cell models, (iii) HSC-MVs also elevated the levels of ALT and AST in the coculture media, and (iv) these effects were accompanied by a decrease in p-PI3K/PI3K and p-Akt/Akt, which could be partially abolished by 740Y-P. Meanwhile, the proapoptotic effect of HSC-MVs was associated with p-Erk1/2/Erk1/2 downregulation and activated caspase-3 upregulation, and could be inhibited by Erk1/2 activation. Our findings demonstrate that HSC-MVs are involved in inflammatory hepatocytes injury probably through the PI3K/Akt, Erk1/2, and caspase-3 pathways.
Collapse
Affiliation(s)
- Yaolong Liang
- 1Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Qunwen Pan
- 2Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Rongfeng Wang
- 1Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhirong Ye
- 1Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zitao Li
- 1Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lingdiao Zeng
- 1Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yanfang Chen
- 2Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaotang Ma
- 2Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Mingyi Li
- 1Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Huilai Miao
- 1Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
25
|
Integration of VEGF and α-SMA Expression Improves the Prediction Accuracy of Fibrosis in Chronic Hepatitis C Liver Biopsy. Appl Immunohistochem Mol Morphol 2019; 25:261-270. [PMID: 26990742 DOI: 10.1097/pai.0000000000000299] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The progression of fibrosis in chronic hepatitis C (CHC) is a multifactorial process. The high adverse effects and the cost of standard health care increase the demand to discover new predictors for the progression of fibrosis in CHC patients. Our study aims to establish the relation between the angiogenic marker [vascular endothelial growth factor (VEGF)] and activated hepatic stellate cells (HSCs) represented by the expression of α-smooth muscle actin (α-SMA) and whether these 2 markers can be used as predictors for the progression of fibrosis in patients with CHC. MATERIALS AND METHODS Histopathologic and immunohistochemical analyses were used for examining the morphology and the expression of VEGF and α-SMA in 60 CHC biopsies procured from CHC patients. Multivariate analysis was used to correlate the protein expression with staging and grading of liver fibrosis. Cutoff values of α-SMA and VEGF were determined by the receiver operating characteristics curve. RESULTS There was a positive correlation between VEGF and HSCs expressing α-SMA (ρ=0.287, P=0.026) and both factors were correlated with the stage of fibrosis (P<0.001). Using the receiver operating characteristics curve, both VEGF (area under the curve=0.71, P<0.006) and α-SMA (area under the curve=0.82, P<0.001) were positive predictors for moderate and severe fibrosis. CONCLUSIONS This study demonstrates the relation between VEGF expression and the activated HSCs denoted by the expression of α-SMA in CHC biopsies and together can be used as a predictor for the progression of fibrosis.
Collapse
|
26
|
Amiot L, Vu N, Drenou B, Scrofani M, Chalin A, Devisme C, Samson M. The anti-fibrotic role of mast cells in the liver is mediated by HLA-G and interaction with hepatic stellate cells. Cytokine 2019; 117:50-58. [PMID: 30825834 DOI: 10.1016/j.cyto.2019.02.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 12/15/2018] [Accepted: 02/02/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS We have reported a significant association between HLA-G expression or the number of hepatic mast cells and liver fibrosis. Here, we investigated the role of HLA-G and mast cells in liver fibrosis, focusing, in particular, on interactions between human mast and stellate cells. METHODS Human mast cells (HMC cell line, CD34-derived mast cells, or tissue-derived mast cells) were co-cultured with purified human hepatic stellate cells (HSCs), and collagen I production by HSCs was evaluated. Mast cells and HSCs were characterized by immunocytochemistry. Various conditions were tested: different times in direct or indirect contact, presence or absence of cytokines, addition or not of HLA-G, and presence or absence of specific protease inhibitors. RESULTS The reciprocal interaction between HSCs and mast cells led to the attraction of mast cells to HSCs in vivo and in vitro, and to a significant decrease in collagen production, at all times of co-culture, following the direct or indirect contact of mast cells with HSCs alone or in the presence of TGF-β, IFN-α or IL-10. We identified the diffusible factors involved in collagen I degradation as mast cell proteases. Moreover, HLA-G expression increased during the co-culture of HSCs and mast cells, with HLA-G acting on both mast cells and HSCs, to enhance collagen I degradation. CONCLUSIONS Mast cells play a beneficial, anti-fibrotic role in liver fibrosis, via the HLA-G-mediated decrease of collagen I. These findings are consistent with high levels of cross-communication between mast cells and hepatic stellate cells and the role of HLA-G.
Collapse
Affiliation(s)
- Laurence Amiot
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France; Univ Rennes, CHU Rennes, F-35000 Rennes, France.
| | - Nicolas Vu
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Bernard Drenou
- CH Emile Muller, F-68100 Mulhouse, France; Institut de Recherche en Hématologie et Transplantation (IRHT), F-68100 Mulhouse, France
| | - Maurice Scrofani
- Institut de Recherche en Hématologie et Transplantation (IRHT), F-68100 Mulhouse, France
| | - Arnaud Chalin
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Christelle Devisme
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Michel Samson
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| |
Collapse
|
27
|
Li H, Huang MH, Jiang JD, Peng ZG. Hepatitis C: From inflammatory pathogenesis to anti-inflammatory/hepatoprotective therapy. World J Gastroenterol 2018; 24:5297-5311. [PMID: 30598575 PMCID: PMC6305530 DOI: 10.3748/wjg.v24.i47.5297] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/27/2018] [Accepted: 12/01/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV) infection commonly causes progressive liver diseases that deteriorate from chronic inflammation to fibrosis, cirrhosis and even to hepatocellular carcinoma. A long-term, persistent and uncontrolled inflammatory response is a hallmark of these diseases and further leads to hepatic injury and more severe disease progression. The levels of inflammatory cytokines and chemokines change with the states of infection and treatment, and therefore, they may serve as candidate biomarkers for disease progression and therapeutic effects. The mechanisms of HCV-induced inflammation involve classic pathogen pattern recognition, inflammasome activation, intrahepatic inflammatory cascade response, and oxidative and endoplasmic reticulum stress. Direct-acting antivirals (DAAs) are the first-choice therapy for effectively eliminating HCV, but DAAs alone are not sufficient to block the uncontrolled inflammation and severe liver injury in HCV-infected individuals. Some patients who achieve a sustained virologic response after DAA therapy are still at a long-term risk for progression to liver cirrhosis and hepatocellular carcinoma. Therefore, coupling with anti-inflammatory/hepatoprotective agents with anti-HCV effects is a promising therapeutic regimen for these patients during or after treatment with DAAs. In this review, we discuss the relationship between inflammatory mediators and HCV infection, summarize the mechanisms of HCV-induced inflammation, and describe the potential roles of anti-inflammatory/hepatoprotective drugs with anti-HCV activity in the treatment of advanced HCV infection.
Collapse
Affiliation(s)
- Hu Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Meng-Hao Huang
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Jian-Dong Jiang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zong-Gen Peng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
28
|
Huang GR, Wei SJ, Huang YQ, Xing W, Wang LY, Liang LL. Mechanism of combined use of vitamin D and puerarin in anti-hepatic fibrosis by regulating the Wnt/β-catenin signalling pathway. World J Gastroenterol 2018; 24:4178-4185. [PMID: 30271082 PMCID: PMC6158481 DOI: 10.3748/wjg.v24.i36.4178] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/30/2018] [Accepted: 08/01/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To reveal the protective mechanism of the combined use of vitamin D and puerarin in the progression of hepatic fibrosis induced by carbon tetrachloride (CCl4).
METHODS Eight-week-old male Wistar rats were randomly divided into a normal control group (C group), a CCl4 group (CCl4 group), a vitamin D group (V group), a puerarin group (P group), and a combined group of vitamin D and puerarin (V + P group), each of which contained ten rats. In this way, we built a rat model of CCl4-induced hepatic fibrosis with intervention by vitamin D, puerarin, or a combination of the two. After eight weeks, the mice were sacrificed to collect serum and liver specimens. Blood was collected to detect the hyaluronic acid (HA). We also measured hydroxyproline (Hyp) and prepared paraffin sections of liver. After Sirius red staining, the liver specimens were observed under a microscope. RT-PCR and western blot analysis were adopted to detect the mRNA and the protein levels of Collagen I, Collagen III, Wnt1, and β-catenin in the liver tissues, respectively.
RESULTS Hepatic fibrosis was observed in the CCl4 group. In comparison, hepatic fibrosis was attenuated in the V, P, and V + P groups: the HA level in blood and the Hyp level in liver were reduced, and the mRNA levels of Collagen I, Collagen III, Wnt, and β-catenin in liver were also decreased, as well as the protein levels of Wnt1 and β-catenin. Among these groups, the V + P group demonstrated the greatest amelioration of hepatic fibrosis.
CONCLUSION The combined application of vitamin D and puerarin is capable of alleviating CCl4-induced hepatic fibrosis of rats. As to the mechanism, it is probably because the combined use is able to silence the Wnt1/β-catenin pathway, suppress the activation of hepatic stellate cells, and reduce the secretion of collagen fibers, therefore improving the anti-hepatic fibrosis effect.
Collapse
Affiliation(s)
- Gan-Rong Huang
- Youjiang Medical University for Nationalities, School of Basic Medical Sciences, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Si-Jun Wei
- Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Yan-Qiang Huang
- Youjiang Medical University for Nationalities, School of Basic Medical Sciences, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Wei Xing
- Youjiang Medical University for Nationalities, School of Basic Medical Sciences, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Lu-Yao Wang
- Youjiang Medical University for Nationalities, School of Basic Medical Sciences, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Ling-Ling Liang
- Youjiang Medical University for Nationalities, School of Basic Medical Sciences, Baise 533000, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
29
|
Yang L, Li LC, Wang X, Wang WH, Wang YC, Xu CR. The contributions of mesoderm-derived cells in liver development. Semin Cell Dev Biol 2018; 92:63-76. [PMID: 30193996 DOI: 10.1016/j.semcdb.2018.09.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 08/31/2018] [Accepted: 09/02/2018] [Indexed: 02/07/2023]
Abstract
The liver is an indispensable organ for metabolism and drug detoxification. The liver consists of endoderm-derived hepatobiliary lineages and various mesoderm-derived cells, and interacts with the surrounding tissues and organs through the ventral mesentery. Liver development, from hepatic specification to liver maturation, requires close interactions with mesoderm-derived cells, such as mesothelial cells, hepatic stellate cells, mesenchymal cells, liver sinusoidal endothelial cells and hematopoietic cells. These cells affect liver development through precise signaling events and even direct physical contact. Through the use of new techniques, emerging studies have recently led to a deeper understanding of liver development and its related mechanisms, especially the roles of mesodermal cells in liver development. Based on these developments, the current protocols for in vitro hepatocyte-like cell induction and liver-like tissue construction have been optimized and are of great importance for the treatment of liver diseases. Here, we review the roles of mesoderm-derived cells in the processes of liver development, hepatocyte-like cell induction and liver-like tissue construction.
Collapse
Affiliation(s)
- Li Yang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Lin-Chen Li
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xin Wang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China
| | - Wei-Hua Wang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Yan-Chun Wang
- Haidian Maternal & Child Health Hospital, Beijing, 100080, China
| | - Cheng-Ran Xu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China.
| |
Collapse
|
30
|
Palyu E, Harsfalvi J, Tornai T, Papp M, Udvardy M, Szekeres-Csiki K, Pataki L, Vanhoorelbeke K, Feys HB, Deckmyn H, Tornai I. Major Changes of von Willebrand Factor Multimer Distribution in Cirrhotic Patients with Stable Disease or Acute Decompensation. Thromb Haemost 2018; 118:1397-1408. [PMID: 29972862 PMCID: PMC6202934 DOI: 10.1055/s-0038-1661393] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Background
There is an unstable balance between pro- and anti-haemostatic processes in patients with cirrhosis. We hypothesized, that in patients with acute decompensation (AD) the major alterations of von Willebrand factor (VWF) could contribute to the pro-thrombotic situation as compared to patients with stable (ST) cirrhosis.
Patients and Methods
We analysed different parameters of VWF, including detailed multimer distribution by densitometry and platelet adhesion, together with
ad
isintegrin-like
a
nd
m
etalloproteinase with
t
hrombo
s
pondin type-1 motifs
13
(ADAMTS13) activity and antigen and C-reactive protein (CRP) levels in patients with ST cirrhosis (
n
= 99), with AD (
n
= 54) and controls (
n
= 92).
Results
VWF antigen, ristocetin co-factor as well as collagen-binding activities were elevated in both cirrhotic groups in a stepwise manner. There was a decrease in high and an increase in low molecular weight multimer ratios in the majority of ST cirrhosis. However, in 24 out of 54 AD patients, ultra-large VWF multimers (ultra-large molecular weight multimers [ULMWM]) were found. ADAMTS13 activity in ST and AD patients without ULMWM was similar to controls (median [interquartile range; IQR]%: 98 [67–132] and 91 [60–110] vs. 106 [88–117], respectively). The presence of ULMWM in AD patients was associated with low ADAMTS13 activity [33 (24–49)%] and high CRP level [23 (7.1–83.6) mg/L]. Adhesion of normal platelets showed a stepwise increase in the presence of cirrhotic plasmas, reaching the highest level in AD patients with ULMWM.
Conclusion
Characteristic changes of VWF parameters are seen in ST cirrhosis. In AD patients, highly increased VWF and reduced ADAMTS13 activity could be found, along with the presence of ULMWM, which are possible markers and contributors of the disease progression.
Collapse
Affiliation(s)
- Eszter Palyu
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Jolan Harsfalvi
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Tamas Tornai
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Maria Papp
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Miklos Udvardy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Katalin Szekeres-Csiki
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Lajos Pataki
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Karen Vanhoorelbeke
- Laboratory for Thrombosis Research, IRF Life Science, KU Leuven Kulak, Kortrijk, Belgium
| | - Hendrik B Feys
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium.,Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Hans Deckmyn
- Laboratory for Thrombosis Research, IRF Life Science, KU Leuven Kulak, Kortrijk, Belgium
| | - Istvan Tornai
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
31
|
Anuja GI, Shine VJ, Latha PG, Suja SR. Protective effect of ethyl acetate fraction of Drynaria quercifolia against CCl 4 induced rat liver fibrosis via Nrf2/ARE and NFκB signalling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2018; 216:79-88. [PMID: 29174446 DOI: 10.1016/j.jep.2017.11.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 10/30/2017] [Accepted: 11/11/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Drynaria quercifolia rhizome is traditionally used as hepatoprotective drug especially in chronic jaundice. AIM OF THE STUDY The present study was undertaken to scientifically evaluate the efficacy of D. quercifolia rhizome against liver fibrosis. MATERIALS AND METHODS D. quercifolia rhizome crude extract (DQ) and its fractions of hexane (HDQ), ethyl acetate (EDQ), butanol (BDQ) were evaluated in vitro using primary hepatocytes and RAW 264.7 cells. In vivo anti-liver fibrotic activity of EDQ was assessed using CCl4 induced liver fibrosis in Wistar rats and serum biochemical parameters (AST, ALT, ALP, SB, cholesterol), MDA, PT, INR, GSH, SOD, CAT, liver glycogen, serum albumin levels were monitored. qRT-PCR analysis of TNF-α, COX-2, iNOS were performed. ELISA method was used to estimate TNF-α, COX-1 & 2. Histopathological studies like H & E, Masson's trichrome, immunohistochemistry staining for α-SMA, TIMP-1, Nrf2 were conducted. LC-Q-TOF-MS analysis of EDQ was conducted. RESULTS In vitro activity guided fractionation of D. quercifolia revealed EDQ as active fraction when compared to other extracts. EDQ treatment significantly inhibited the expression of α-SMA, TIMP-1, COX-2, TNF-α, iNOS and increased the levels of Nrf2 in rat liver fibrosis. LC-Q-TOF-MS analysis of EDQ confirmed the presence of naringin and naringenin. CONCLUSION The anti-liver fibrotic activity of EDQ is via inhibition of NFκB signalling pathway, antioxidant response through Nrf2 activation and further inhibition of HSC activation.
Collapse
Affiliation(s)
- G I Anuja
- Ethnomedicine and Ethnopharmacology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute, Palode, Thiruvananthapuram 695562, Kerela, India.
| | - V J Shine
- Ethnomedicine and Ethnopharmacology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute, Palode, Thiruvananthapuram 695562, Kerela, India
| | - P G Latha
- Ethnomedicine and Ethnopharmacology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute, Palode, Thiruvananthapuram 695562, Kerela, India
| | - S R Suja
- Ethnomedicine and Ethnopharmacology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute, Palode, Thiruvananthapuram 695562, Kerela, India
| |
Collapse
|
32
|
Yamaguchi M, Saito SY, Nishiyama R, Nakamura M, Todoroki K, Toyo'oka T, Ishikawa T. Caffeine Suppresses the Activation of Hepatic Stellate Cells cAMP-Independently by Antagonizing Adenosine Receptors. Biol Pharm Bull 2018; 40:658-664. [PMID: 28458351 DOI: 10.1248/bpb.b16-00947] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
During liver injury, hepatic stellate cells (HSCs) are activated by various cytokines and transdifferentiated into myofibroblast-like activated HSCs, which produce collagen, a major source of liver fibrosis. Therefore, the suppression of HSC activation is regarded as a therapeutic target for liver fibrosis. Several epidemiological reports have revealed that caffeine intake decreases the risk of liver disease. In this study, therefore, we investigated the effect of caffeine on the activation of primary HSCs isolated from mice. Caffeine suppressed the activation of HSC in a concentration-dependent manner. BAPTA-AM, an intracellular Ca2+ chelator, had no effect on the caffeine-induced suppression of HSC activation. None of the isoform-selective inhibitors of phosphodiesterase1 to 5 affected changes in the morphology of HSC during activation, whereas CGS-15943, an adenosine receptor antagonist, inhibited them. Caffeine had no effect on intracellular cAMP level or on the phosphorylation of extracellular signal-regulated kinase (ERK)1/2. In contrast, caffeine significantly decreased the phosphorylation of Akt1. These results suggest that caffeine inhibits HSC activation by antagonizing adenosine receptors, leading to Akt1 signaling activation.
Collapse
Affiliation(s)
- Momoka Yamaguchi
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka
| | - Shin-Ya Saito
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka
| | - Ryota Nishiyama
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka
| | - Misuzu Nakamura
- Laboratory of Analytical and Bio-analytical Chemistry, School of Pharmaceutical Sciences, University of Shizuoka
| | - Kenichiro Todoroki
- Laboratory of Analytical and Bio-analytical Chemistry, School of Pharmaceutical Sciences, University of Shizuoka
| | - Toshimasa Toyo'oka
- Laboratory of Analytical and Bio-analytical Chemistry, School of Pharmaceutical Sciences, University of Shizuoka
| | - Tomohisa Ishikawa
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
33
|
Lebda MA, Sadek KM, Abouzed TK, Tohamy HG, El-Sayed YS. Melatonin mitigates thioacetamide-induced hepatic fibrosis via antioxidant activity and modulation of proinflammatory cytokines and fibrogenic genes. Life Sci 2017; 192:136-143. [PMID: 29180002 DOI: 10.1016/j.lfs.2017.11.036] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 11/18/2017] [Accepted: 11/23/2017] [Indexed: 12/11/2022]
Abstract
AIMS The potential antifibrotic effects of melatonin against induced hepatic fibrosis were explored. MAIN METHODS Rats were allocated into four groups: placebo; thioacetamide (TAA) (200mg/kg bwt, i.p twice weekly for two months); melatonin (5mg/kgbwt, i.p daily for a week before TAA and continued for an additional two months); and melatonin plus TAA. Hepatic fibrotic changes were evaluated biochemically and histopathologically. Hepatic oxidative/antioxidative indices were assessed. The expression of hepatic proinflammatory cytokines (tumor necrosis factor-α, and interleukin-1β), fibrogenic-related genes (transforming growth factor-1β, collagen I, collagen, III, laminin, and autotaxin) and an antioxidant-related gene (thioredoxin-1) were detected by qRT-PCR. KEY FINDINGS In fibrotic rats, melatonin lowered serum aspartate aminotransferase, alanine aminotransferase, and autotaxin activities, bilirubin, hepatic hydroxyproline and plasma ammonia levels. Melatonin displayed hepatoprotective and antifibrotic potential as indicated by mild hydropic degeneration of some hepatocytes and mild fibroplasia. In addition, TAA induced the depletion of glutathione, glutathione s-transferase, glutathione peroxidase, superoxide dismutase, catalase, and paraoxonase-1 (PON-1), while inducing the accumulation of malondialdehyde, protein carbonyl (C=O) and nitric oxide (NO), and DNA fragmentation. These effects were restored by melatonin pretreatment. Furthermore, melatonin markedly attenuated the expression of proinflammatory cytokines and fibrogenic genes via the upregulation of thioredoxin-1 mRNA transcripts. SIGNIFICANCE Melatonin exhibits potent anti-inflammatory, antioxidant and fibrosuppressive activities against TAA-induced hepatic fibrogenesis via the suppression of oxidative stress, DNA damage, proinflammatory cytokines and fibrogenic gene transcripts. In addition, we demonstrate that the antifibrotic activity of melatonin is mediated by the induction of thioredoxin-1 with attenuation of autotaxin expressions.
Collapse
Affiliation(s)
- Mohamed A Lebda
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Egypt.
| | - Kadry M Sadek
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhur University, Egypt
| | - Tarek K Abouzed
- Department of Biochemistry, Faculty of Veterinary Medicine, Kafr Elsheikh University, Egypt
| | - Hossam G Tohamy
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Egypt
| | - Yasser S El-Sayed
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Damanhur University, Egypt.
| |
Collapse
|
34
|
A three-dimensional cell culture device for simulation of hepatic hypertension. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2017; 2017:2076-2079. [PMID: 29060305 DOI: 10.1109/embc.2017.8037262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Hepatic stellate cells (HSCs) play a crucial role in the development of liver fibrosis which is characterized by massive tissue scarring, elevated hepatic sinusoidal pressure, and portal hypertension. However, while a multitude of chemical factors have been investigated in the pathogenesis of liver fibrosis, physical factors such as elevated hydrostatic pressure and shear stress caused by blood flow in sinusoids remain unclear. In this study, we developed a three-dimensional (3D) cell culture microfluidic platform that mimics the physical environments of hepatic sinusoids to investigate the effects of elevated hydrostatic pressure on HSCs phenotypes.
Collapse
|
35
|
Xie SR, An JY, Zheng LB, Huo XX, Guo J, Shih D, Zhang XL. Effects and mechanism of adenovirus-mediated phosphatase and tension homologue deleted on chromosome ten gene on collagen deposition in rat liver fibrosis. World J Gastroenterol 2017; 23:5904-5912. [PMID: 28932082 PMCID: PMC5583575 DOI: 10.3748/wjg.v23.i32.5904] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 06/19/2017] [Accepted: 07/24/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate the effects of phosphatase and tension homologue deleted on chromosome ten (PTEN) gene on collagen metabolism in hepatic fibrosis and the underlying mechanisms.
METHODS Rat primary hepatic stellate cells (HSCs) and human LX-2 cells were transfected with adenovirus containing cDNA constructs encoding wild-type PTEN (Ad-PTEN), PTEN mutant G129E gene (Ad-G129E), and RNA interference constructs targeting the PTEN sequence PTEN short hairpin RNA to up-regulate and down-regulate the expression of PTEN. HSCs were assayed using fluorescent microscopy, real-time polymerase chain reaction, and western blotting. Moreover, a CCl4-induced rat hepatic fibrosis model was established to investigate the in vivo effects. Hematoxylin and eosin, and Masson’s trichrome were used to assess the histological changes. The expression of collagen I and III was assessed using immunohistochemistry and western blot analysis.
RESULTS Elevated expression of PTEN gene reduced serum levels of alanine transaminase and aspartate transaminase, decreased collagen deposition in the liver, and reduced hepatocyte necrosis. In contrast, knockdown of PTEN expression had an opposite effect, such as increased collagen deposition in the liver, and was molecularly characterized by the increased expression of matrix metalloproteinase (MMP)-13 (P < 0.01) and MMP-2 (P < 0.01), as well as decreased expression of the tissue inhibitor of metalloproteinase (TIMP)-1 (P < 0.01) and TIMP-2 (P < 0.01).
CONCLUSION These data indicated that gene therapy using recombinant adenovirus encoding PTEN might be a novel way of treating hepatic fibrosis.
Collapse
Affiliation(s)
- Shu-Rui Xie
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang 050000, Hebei Province, China
- Department of Gastroenterology, Xingtai People's Hospital, Xingtai 054031, Hebei Province, China
| | - Jun-Yan An
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang 050000, Hebei Province, China
| | - Li-Bo Zheng
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang 050000, Hebei Province, China
| | - Xiao-Xia Huo
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang 050000, Hebei Province, China
| | - Jian Guo
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang 050000, Hebei Province, China
| | - David Shih
- Inflammatory Bowel and Immunobiology Research Institute, F. Widjaja Foundation, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Xiao-Lan Zhang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Shijiazhuang 050000, Hebei Province, China
| |
Collapse
|
36
|
Li J, Ghazwani M, Liu K, Huang Y, Chang N, Fan J, He F, Li L, Bu S, Xie W, Ma X, Li S. Regulation of hepatic stellate cell proliferation and activation by glutamine metabolism. PLoS One 2017; 12:e0182679. [PMID: 28797105 PMCID: PMC5552314 DOI: 10.1371/journal.pone.0182679] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/21/2017] [Indexed: 12/20/2022] Open
Abstract
Liver fibrosis is the excessive accumulation of extracellular matrix proteins, which is mainly caused by accumulation of activated hepatic stellate cells (HSCs). The mechanisms of activation and proliferation of HSCs, two key events after liver damage, have been studied for many years. Here we report a novel pathway to control HSCs by regulating glutamine metabolism. We demonstrated that the proliferation of HSCs is critically dependent on glutamine that is used to generate α-ketoglutarate (α-KG) and non-essential amino acid (NEAA). In addition, both culture- and in vivo-activated HSCs have increased glutamine utilization and increased expression of genes related to glutamine metabolism, including GLS (glutaminase), aspartate transaminase (GOT1) and glutamate dehydrogenase (GLUD1). Inhibition of these enzymes, as well as glutamine depletion, had a significant inhibitory effect on HSCs activation. In addition to providing energy expenditure, conversion of glutamine to proline is enhanced. The pool of free proline may also be increased via downregulation of POX expression. Hedgehog signaling plays an important role in the regulation of glutamine metabolism, as well as TGF-β1, c-Myc, and Ras signalings, via transcriptional upregulation and repression of key metabolic enzymes in this pathway. Finally, changes in glutamine metabolism were also found in mouse liver tissue following CCl4-induced acute injury. CONCLUSION Glutamine metabolism plays an important role in regulating the proliferation and activation of HSCs. Strategies that are targeted at glutamine metabolism may represent a novel therapeutic approach to the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Jiang Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States of America
- * E-mail: (JL); (SL)
| | - Mohammed Ghazwani
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Ke Liu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Yixian Huang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Na Chang
- Department of Cell Biology,Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Jie Fan
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Fengtian He
- Department of Biochemistry and Molecular Biology (F.H.), College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Liying Li
- Department of Cell Biology,Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Shizhong Bu
- Diabetes Research Center, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Wen Xie
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Xiaochao Ma
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Song Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States of America
- * E-mail: (JL); (SL)
| |
Collapse
|
37
|
Bai GP, Yan GH, Wang GJ, Wan P, Liu XF, Wang H, Zhang RH. Effects of lentiviral vector‑mediated shRNA silencing of TGFβ1 on the expression of Col1a1 in rat hepatic stellate cells. Mol Med Rep 2017; 16:2785-2790. [PMID: 28713909 DOI: 10.3892/mmr.2017.6945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 05/11/2017] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to construct a lentiviral RNA interference (RNAi) vector targeting the transforming growth factor β1 (TGFβ1) gene of rats, in order to examine its effect on silencing of the TGFβ1 gene and on the expression of collagen type 1 α1 (Col1a1) in HSC‑T6 rat hepatic stellate cells. Three RNAi sites of the TGFβ1 gene were selected according to its CDs sequence. Three pairs of small interfering RNA (siRNA) of these RNAi sites were synthesized and then transfected into HSC‑T6 cells, respectively, to confirm the optimal siRNA sequence via reverse transcription‑polymerase chain reaction analysis. Subsequently, shRNA targeting the sequence of the optimal siRNA was designed, synthesized and annealed to form a double‑stranded structure. The annealed oligonucleotide fragment was cloned into pGreenPuro plasmids to establish the pGreenPuro/TGFβ1 shRNA lentiviral vector, which was then transfected into 293T cells, following identification by restriction enzyme digestion and sequencing for the production of lentiviral particles exhibiting high reactivity. These particles were used to infect HSC‑T6 cells, following which the expression of GFP in the transfected cells was observed under an inverted microscope. The effects on TGFβ1 gene silencing and the expression levels of Colla1 were detected at the mRNA and protein levels. The results provided confirmation of the optimal siRNA sequence. Enzyme digestion and sequencing verified successful construction of the pGreenPuro/TGFβ1 shRNA lentiviral vector. This lentiviral vector effectively silenced the TGFβ1 gene in the HSC‑T6 cells, and inhibited the expression of Col1a1 at the mRNA and protein levels. Taken together, the lentiviral RNAi vector targeting the TGFβ1 gene of rats was successfully constructed, which effectively silenced the TGFβ1 gene of the HSC‑T6 cells and inhibited the expression of Col1a1.
Collapse
Affiliation(s)
- Gan-Ping Bai
- Department of Integrated Chinese and Western Medicine, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Guo-He Yan
- Institute of Combined Injuries, The State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Guo-Jian Wang
- Department of Integrated Chinese and Western Medicine, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Ping Wan
- Department of Integrated Chinese and Western Medicine, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Xiao-Fei Liu
- Department of Integrated Chinese and Western Medicine, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Heng Wang
- Department of Integrated Chinese and Western Medicine, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Rong-Hua Zhang
- Department of Integrated Chinese and Western Medicine, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
38
|
El-Lakkany NM, El-Maadawy WH, Seif El-Din SH, Hammam OA, Mohamed SH, Ezzat SM, Safar MM, Saleh S. Rosmarinic acid attenuates hepatic fibrogenesis via suppression of hepatic stellate cell activation/proliferation and induction of apoptosis. ASIAN PAC J TROP MED 2017. [PMID: 28647181 DOI: 10.1016/j.apjtm.2017.05.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To investigate the antifibrotic role of rosmarinic acid (RA), a natural polyphenolic compound, on HSCs activation/proliferation and apoptosis in vitro and in vivo. METHODS The impact of RA on stellate cell line (HSC-T6) proliferation, activation and apoptosis was assessed along with its safety on primary hepatocytes. In vivo, rats were divided into: (i) normal; (ii) thioacetamide (TAA)-intoxicated rats for 12 weeks; (iii) TAA + silymarin or (iv) TAA + RA. At the end of experiment, liver functions, oxidative stress, inflammatory and profibrogenic markers, tissue inhibitor metalloproteinases type-1 (TIMP-1) and hydroxyproline (HP) levels were evaluated. Additionally, liver histopathology and immunohistochemical examinations of alpha-smooth muscle actin (α-SMA), caspase-3 and proliferation cellular nuclear antigen (PCNA) were determined. RESULTS RA exhibited anti-proliferative effects on cultured HSCs in a time and concentration dependent manner showing an IC50 of 276 μg/mL and 171 μg/mL for 24 h and 48 h, respectively, with morphological reversion of activated stellate cell morphology to quiescent form. It significantly improved ALT, AST, oxidative stress markers and reduced TIMP-1, HP levels, inflammatory markers and fibrosis score (S1 vs S4). Furthermore, reduction in α-SMA plus elevation in caspase-3 expressions of HSCs in vitro and in vivo associated with an inhibition in proliferation of damaged hepatocytes were recorded. CONCLUSIONS RA impeded the progression of liver fibrosis through inhibition of HSCs activation/proliferation and induction of apoptosis with preservation of hepatic architecture.
Collapse
Affiliation(s)
- Naglaa M El-Lakkany
- Department of Pharmacology, Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba P.O. Box 30, Giza 12411, Egypt.
| | - Walaa H El-Maadawy
- Department of Pharmacology, Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba P.O. Box 30, Giza 12411, Egypt
| | - Sayed H Seif El-Din
- Department of Pharmacology, Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba P.O. Box 30, Giza 12411, Egypt
| | - Olfat A Hammam
- Department of Pathology, Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba P.O. Box 30, Giza 12411, Egypt
| | - Salwa H Mohamed
- Department of Immunology, Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba P.O. Box 30, Giza 12411, Egypt
| | - Shahira M Ezzat
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Marwa M Safar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Samira Saleh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
39
|
Lee GM, Kim YR, Ryu JH, Kim TH, Cho EY, Lee YH, Yoon KH. Quantitative Measurement of Hepatic Fibrosis with Gadoxetic Acid-Enhanced Magnetic Resonance Imaging in Patients with Chronic Hepatitis B Infection: A Comparative Study on Aspartate Aminotransferase to Platelet Ratio Index and Fibrosis-4 Index. Korean J Radiol 2017; 18:444-451. [PMID: 28458596 PMCID: PMC5390613 DOI: 10.3348/kjr.2017.18.3.444] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/19/2016] [Indexed: 12/21/2022] Open
Abstract
Objective To quantitatively measure hepatic fibrosis on gadoxetic acid-enhanced magnetic resonance (MR) in chronic hepatitis B (CHB) patients and identify the correlations with aspartate aminotransferase-to-platelet ratio index (APRI) and fibrosis-4 index (FIB-4) values. Materials and Methods This study on gadoxetic acid-enhanced 3T MR imaging included 81 patients with CHB infection. To quantitatively measure hepatic fibrosis, MR images were analyzed with an aim to identify inhomogeneous signal intensities calculated from a coefficient of variation (CV) map in the liver parenchyma. We also carried out a comparative analysis between APRI and FIB-4 based on metaregression results. The diagnostic performance of the CV map was evaluated using a receiver-operating characteristic (ROC) curve. Results In the MR images, the mean CV values in control, groups I, II, and III based on APRI were 4.08 ± 0.92, 4.24 ± 0.80, 5.64 ± 1.11, and 5.73 ± 1.28, respectively (p < 0.001). In CHB patients grouped by FIB-4, the mean CV values of groups A, B, and C were 4.22 ± 0.95, 5.40 ± 1.19, and 5.71 ± 1.17, respectively (p < 0.001). The mean CV values correlated well with APRI (r = 0.392, p < 0.001) and FIB-4 (r = 0.294, p < 0.001). In significant fibrosis group, ROC curve analysis yielded an area under the curve of 0.875 using APRI and 0.831 using FIB-4 in HB, respectively. Conclusion Gadoxetic acid-enhanced MR imaging for calculating a CV map showed moderate correlation with APRI and FIB-4 values and could be employed to quantitatively measure hepatic fibrosis in patients with CHB.
Collapse
Affiliation(s)
- Guy Mok Lee
- Department of Radiology, Wonkwang University School of Medicine, Iksan 54538, Korea
| | - Youe Ree Kim
- Department of Radiology, Wonkwang University School of Medicine, Iksan 54538, Korea
| | - Jong Hyun Ryu
- Imaging Science Research Center, Wonkwang University, Iksan 54538, Korea
| | - Tae-Hoon Kim
- Imaging Science Research Center, Wonkwang University, Iksan 54538, Korea
| | - Eun Young Cho
- Department of Internal Medicine, Wonkwang University School of Medicine, Iksan 54538, Korea
| | - Young Hwan Lee
- Department of Radiology, Wonkwang University School of Medicine, Iksan 54538, Korea
| | - Kwon-Ha Yoon
- Department of Radiology, Wonkwang University School of Medicine, Iksan 54538, Korea.,Imaging Science Research Center, Wonkwang University, Iksan 54538, Korea
| |
Collapse
|
40
|
Salas-Villalobos T, Lozano-Sepúlveda S, Rincón-Sánchez A, Govea-Salas M, Rivas-Estilla A. Mechanisms involved in liver damage resolution after hepatitis C virus clearance. MEDICINA UNIVERSITARIA 2017. [DOI: 10.1016/j.rmu.2017.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
41
|
Tang Y, Zhang Q, Zhu Y, Chen G, Yu F. Low concentrations of bilirubin inhibit activation of hepatic stellate cells in vitro. Mol Med Rep 2017; 15:1647-1653. [PMID: 28260031 PMCID: PMC5364989 DOI: 10.3892/mmr.2017.6201] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 12/02/2016] [Indexed: 01/11/2023] Open
Abstract
Hepatic stellate cell (HSC) activation serves a key role in liver fibrosis, and is associated with chronic liver diseases. Bilirubin, a product of heme degradation, has been demonstrated to have antioxidant properties. The present study investigated the effects of physiological concentrations of bilirubin on rat HSC activation. Rat HSCs were isolated and cultured for several generations to induce activation. The activated HSCs were subsequently treated with 0, 1, 10 or 20 mg/l bilirubin and assayed for parameters of cell activation. As the bilirubin concentration increased, HSCs demonstrated reduced production of reactive oxygen species, reduced protein expression levels of α-smooth muscle actin, a decreased mRNA expression ratio of tissue inhibitor of matrix metalloproteinase-1/matrix metalloproteinase-2, decreased proliferation and increased apoptosis. In conclusion, elevated bilirubin levels, within its physiological concentration range, appeared to inhibit HSC activation. These findings suggested a potential role for bilirubin in the treatment of fibrosis that requires further investigation.
Collapse
Affiliation(s)
- Yinhe Tang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Qiyu Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yefan Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Gang Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Fuxiang Yu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
42
|
Lambrecht J, Jan Poortmans P, Verhulst S, Reynaert H, Mannaerts I, van Grunsven LA. Circulating ECV-Associated miRNAs as Potential Clinical Biomarkers in Early Stage HBV and HCV Induced Liver Fibrosis. Front Pharmacol 2017; 8:56. [PMID: 28232800 PMCID: PMC5298975 DOI: 10.3389/fphar.2017.00056] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/26/2017] [Indexed: 12/12/2022] Open
Abstract
Introduction: Chronic hepatitis B (HBV) and C (HCV) virus infection is associated with the activation of hepatic stellate cells (HSCs) toward a myofibroblastic phenotype, resulting in excessive deposition of extracellular matrix, the development of liver fibrosis, and its progression toward cirrhosis. The gold standard for the detection and staging of liver fibrosis remains the liver biopsy, which is, however, associated with some mild and severe drawbacks. Other non-invasive techniques evade these drawbacks, but lack inter-stage specificity and are unable to detect early stages of fibrosis. We investigated whether circulating vesicle-associated miRNAs can be used in the diagnosis and staging of liver fibrosis in HBV and HCV patients. Methods: Plasma samples were obtained from 14 healthy individuals and 39 early stage fibrotic patients (F0–F2) with chronic HBV or HCV infection who underwent transient elastography (Fibroscan). Extracellular vesicles were extracted from the plasma and the level of miRNA-122, -150, -192, -21, -200b, and -92a was analyzed by qRT-PCR in total plasma and circulating vesicles. Finally, these same miRNAs were also quantified in vesicles extracted from in vitro activating primary HSCs. Results: In total plasma samples, only miRNA-200b (HBV: p = 0.0384; HCV: p = 0.0069) and miRNA-122 (HBV: p < 0.0001; HCV: p = 0.0007) were significantly up-regulated during early fibrosis. In circulating vesicles, miRNA-192 (HBV: p < 0.0001; HCV: p < 0.0001), -200b (HBV: p < 0.0001; HCV: p < 0.0001), -92a (HBV: p < 0.0001; HCV: p < 0.0001), and -150 (HBV: p = 0.0016; HCV: p = 0.004) displayed a significant down-regulation in both HBV and HCV patients. MiRNA expression profiles in vesicles isolated from in vitro activating primary mouse HSCs resembled the miRNA expression profile in circulating vesicles. Conclusion: Our analysis revealed a distinct miRNA expression pattern in total plasma and its circulating vesicles. The expression profile of miRNAs in circulating vesicles of fibrotic patients suggests the potential use of these vesicle-associated miRNAs as markers for early stages of liver fibrosis.
Collapse
Affiliation(s)
- Joeri Lambrecht
- Liver Cell Biology Lab, Department of Basic Biomedical Sciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Pieter Jan Poortmans
- Liver Cell Biology Lab, Department of Basic Biomedical Sciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Stefaan Verhulst
- Liver Cell Biology Lab, Department of Basic Biomedical Sciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Hendrik Reynaert
- Liver Cell Biology Lab, Department of Basic Biomedical Sciences, Vrije Universiteit BrusselBrussels, Belgium; Department of Gastroenterology and Hepatology, Universitair Ziekenhuis BrusselBrussels, Belgium
| | - Inge Mannaerts
- Liver Cell Biology Lab, Department of Basic Biomedical Sciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Leo A van Grunsven
- Liver Cell Biology Lab, Department of Basic Biomedical Sciences, Vrije Universiteit Brussel Brussels, Belgium
| |
Collapse
|
43
|
Zhou D, Wang J, He LN, Li BH, Ding YN, Chen YW, Fan JG. Prolyl oligopeptidase attenuates hepatic stellate cell activation through induction of Smad7 and PPAR-γ. Exp Ther Med 2017; 13:780-786. [PMID: 28352366 DOI: 10.3892/etm.2017.4033] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 10/21/2016] [Indexed: 12/30/2022] Open
Abstract
Prolyl oligopeptidase (POP) is a serine endopeptidase widely distributed in vivo with high activity in the liver. However, its biological functions in the liver have remained largely elusive. A previous study by our group has shown that POP produced N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP) and thereby exerted an anti-fibrogenic effect on hepatic stellate cells (HSCs) in vitro. It was therefore hypothesized that POP may affect the activation state of HSCs and has an important role in liver fibrosis. The HSC-T6 immortalized rat liver stellate cell line was treated with the POP inhibitor S17092 or transfected with recombinant lentivirus to overexpress POP. Cell proliferation and apoptosis were determined using a Cell Counting Kit-8 and flow cytometry, respectively. The activation status of HSCs was determined by examination of the expression of α-smooth muscle actin (α-SMA), collagen I, monocyte chemoattractant protein-1 (MCP-1), transforming growth factor (TGF)-β-Smad signaling and peroxisome proliferator activated receptor-γ (PPAR-γ). Inhibition by S17092 decreased, whereas lentiviral expression increased the activity of POP and cell proliferation, while neither of the treatments affected cell apoptosis. Of note, S17092 significantly increased, whereas POP overexpression decreased the expression of α-SMA and MCP-1 without affecting the expression of collagen I and TGF-β1. Furthermore, S17092 caused a reduction, whereas POP overexpression caused an upregulation of Smad7 protein and PPAR-γ, but not phosphorylated-Smad2/3 expression. In conclusion, POP attenuated the activation of HSCs through inhibition of TGF-β signaling and induction of PPAR-γ, which may have therapeutic potential in liver fibrosis.
Collapse
Affiliation(s)
- Da Zhou
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Jing Wang
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Ling-Nan He
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Bing-Hang Li
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Yong-Nian Ding
- Department of Gastroenterology, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830028, P.R. China
| | - Yuan-Wen Chen
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Jian-Gao Fan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| |
Collapse
|
44
|
Lee J, Choi B, No DY, Lee G, Lee SR, Oh H, Lee SH. A 3D alcoholic liver disease model on a chip. Integr Biol (Camb) 2016; 8:302-8. [PMID: 26857817 DOI: 10.1039/c5ib00298b] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Alcohol is one of the main causes of liver diseases, and the development of alcoholic liver disease (ALD) treatment methods has been one of the hottest issues. For this purpose, development of in vitro models mimicking the in vivo physiology is one of the critical requirements, and they help to determine the disease mechanisms and to discover the treatment method. Herein, a three-dimensional (3D) ALD model was developed and its superior features in mimicking the in vivo condition were demonstrated. A spheroid-based microfluidic chip was employed for the development of the 3D in vitro model of ALD progression. We co-cultured rat primary hepatocytes and hepatic stellate cells (HSCs) in a fluidic chip to investigate the role of HSCs in the recovery of liver with ALD. An interstitial level of flow derived by an osmotic pump was applied to the chip to provide in vivo mimicking of fluid activity. Using this in vitro tool, we were able to observe structural changes and decreased hepatic functions with the increase in ethanol concentration. The recovery process of liver injured by alcohol was observed by providing fresh culture medium to the damaged 3D liver tissue for few days. A reversibly- and irreversibly-injured ALD model was established. The proposed model can not only be used for the research of alcoholic disease mechanism, but also has the potential for use in studies of hepatotoxicity and drug screening applications.
Collapse
Affiliation(s)
- JaeSeo Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 136-713, Republic of Korea
| | - BongHwan Choi
- School of Biomedical Engineering, College of Health Science, Korea University, Seoul 136-713, Republic of Korea
| | - Da Yoon No
- Department of Bioengineering, School of Engineering and Medicine, Stanford University, 318 Campus Drive, Stanford, CA 94305, USA
| | - GeonHui Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 136-713, Republic of Korea
| | - Seung-Ri Lee
- Department of Biopharmaceutical Sciences College of Pharmacy University of Illinois at Chicago, USA
| | - HyunJik Oh
- Department of Biomedical Engineering, College of Health Science, Korea University, Seoul 136-713, Republic of Korea. and MicroFIT R&BD Institute, Dunchon-daero 457beon-gil, Jungwon-gu, Gyeonggi-do 462-806, Republic of Korea
| | - Sang-Hoon Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 136-713, Republic of Korea and Department of Biomedical Engineering, College of Health Science, Korea University, Seoul 136-713, Republic of Korea.
| |
Collapse
|
45
|
Denardin CC, Martins LAM, Parisi MM, Vieira MQ, Terra SR, Barbé-Tuana FM, Borojevic R, Vizzotto M, Emanuelli T, Guma FCR. Autophagy induced by purple pitanga (Eugenia uniflora L.) extract triggered a cooperative effect on inducing the hepatic stellate cell death. Cell Biol Toxicol 2016; 33:197-206. [PMID: 27744523 DOI: 10.1007/s10565-016-9366-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 10/07/2016] [Indexed: 12/18/2022]
Abstract
Activated hepatic stellate cells (HSC) are the major source of collagen I in liver fibrosis. Eugenia uniflora L. is a tree species that is widely distributed in South America. E. uniflora L. fruit-popularly known as pitanga-has been shown to exert beneficial properties. Autophagy contributes to the maintenance of cellular homeostasis and survival under stress situation, but it has also been suggested to be an alternative cell death pathway. Mitochondria play a pivotal role on signaling cell death. Mitophagy of damaged mitochondria is an important cell defense mechanism against organelle-mediated cell death signaling. We previously found that purple pitanga extract induced mitochondrial dysfunction, cell cycle arrest, and death by apoptosis and necrosis in GRX cells, a well-established activated HSC line. We evaluated the effects of 72-h treatment with crescent concentrations of purple pitanga extract (5 to 100 μg/mL) on triggering autophagy in GRX cells, as this is an important mechanism to cells under cytotoxic conditions. We found that all treated cells presented an increase in the mRNA expression of autophagy-related protein 7 (ATG7). Concomitantly, flow cytometry and ultrastructural analysis of treated cells revealed an increase of autophagosomes/autolysosomes that consequentially led to an increased mitophagy. As purple pitanga extract was previously found to be broadly cytotoxic to GRX cells, we postulated that autophagy contributes to this scenario, where cell death seems to be an inevitable fate. Altogether, the effectiveness on inducing activated HSC death can make purple pitanga extract a good candidate on treating liver fibrosis.
Collapse
Affiliation(s)
- Cristiane C Denardin
- Programa de Pós-Graduação em Ciências Biológicas-Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
- Curso de Farmácia, Universidade Federal do Pampa (UNIPAMPA), Campus Uruguaiana, Uruguaiana, RS, Brasil
| | - Leo A M Martins
- Programa de Pós-Graduação em Ciências Biológicas-Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - Mariana M Parisi
- Programa de Pós-Graduação em Ciências Biológicas-Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - Moema Queiroz Vieira
- Programa de Pós-Graduação em Ciências Biológicas-Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - Silvia R Terra
- Programa de Pós-Graduação em Ciências Biológicas-Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - Florencia M Barbé-Tuana
- Programa de Pós-Graduação em Ciências Biológicas-Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - Radovan Borojevic
- Departamento de Histologia e Embriologia, ICB, UFRJ, Rio de Janeiro, RJ, Brasil
| | - Márcia Vizzotto
- Empresa Brasileira de Pesquisa Agropecuária de Clima Temperado, Pelotas, RS, Brasil
| | - Tatiana Emanuelli
- Núcleo Integrado de Desenvolvimento em Análises Laboratoriais (NIDAL), Departamento de Tecnologia e Ciência de Alimentos, Universidade Federal de Santa Maria, Santa Maria, RS, Brasil
| | - Fátima Costa Rodrigues Guma
- Programa de Pós-Graduação em Ciências Biológicas-Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil.
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600-Anexo, Lab 21, CEP: 90035-003, Porto Alegre, RS, Brasil.
- Centro de Microscopia e Microanálise, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil.
| |
Collapse
|
46
|
Liu Y, Yang P, Chen N, Lin S, Liu M. Effects of recombinant human adenovirus-p53 on the regression of hepatic fibrosis. Int J Mol Med 2016; 38:1093-100. [PMID: 27572658 PMCID: PMC5029955 DOI: 10.3892/ijmm.2016.2716] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 08/10/2016] [Indexed: 01/28/2023] Open
Abstract
Hepatic fibrosis is a scarring process that may progress to hepatic cirrhosis and even hepatic carcinoma if left untreated. Hepatic stellate cells (HSCs) play essential roles in the development of hepatic fibrosis. The tumor suppressor protein p53 is a transcription factor that is involved in cell proliferation, cell cycle regulation, apoptosis and DNA repair. Recombinant human adenovirus-p53 (Ad-p53) has been demonstrated to act as a promising antitumor gene therapy in various types of cancer. However, there is limited infomration regarding the therapeutic effect of Ad-p53 on the regression of hepatic fibrosis. In order to examine the underlying molecular mechanism responsible for the effects of Ad-p53 on HSCs, a rat model of hepatic fibrosis was established and HSC-T6 cells were cultured under different conditions. The expression of p53, transforming growth factor (TGF-β1) and α-smooth muscle actin (α-SMA), which is a marker of activated HSCs, was detected by immunohistochemical assays and RT-qPCR. In vitro, five different concentrations (1×106, 5×106, 1×107, 2×107 and 5×107 PFU/ml) of Ad-p53 were selected for use in the MTT assay to analyze the proliferation of HSCs at 0, 24, 48 and 72 h. Flow cytometric analysis was applied to determine the effect of three different concentrations of Ad-p53 (5×106, 1×107 and 2×107 PFU/ml) on the cell cycle and the apoptosis of HSC-T6 cells at 24 and 48 h. The results of immunohistochemical studies and RT-qPCR showed that Ad-p53 upregulated the expression of p53, and downregulated the expression of TGF-β1 and α-SMA. The MTT assay revealed that when treated with various doses of Ad-p53, the proliferation of HSCs was inhibited within a certain range of concentrations and time periods. Analysis of flow cytometric data showed that Ad-p53 arrested the cell cycle in G1 phase and significantly induced apoptosis. Taken together, these findings suggest that Ad-p53 promotes apoptosis and inhibits the proliferation of HSCs in a time- and dose-dependent manner by modulating the expression of p53, TGF-β1 and α-SMA.
Collapse
Affiliation(s)
- Yehong Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Puye Yang
- Department of Infectious Diseases, Xi'an North Hospital of Xi'an Medical University, Xi'an, Shaanxi 710043, P.R. China
| | - Na Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Shumei Lin
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Min Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
47
|
Eom YW, Kim G, Baik SK. Mesenchymal stem cell therapy for cirrhosis: Present and future perspectives. World J Gastroenterol 2015; 21:10253-10261. [PMID: 26420953 PMCID: PMC4579873 DOI: 10.3748/wjg.v21.i36.10253] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 06/01/2015] [Accepted: 08/31/2015] [Indexed: 02/06/2023] Open
Abstract
Cirrhosis occurs as a result of various chronic liver injuries, which may be caused by viral infections, alcohol abuse and the administration of drugs and chemicals. Recently, bone marrow cells (BMCs), hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs) have been used for developing treatments for cirrhosis. Clinical trials have investigated the therapeutic potential of BMCs, HSCs and MSCs for the treatment of cirrhosis based on their potential to differentiate into hepatocytes. Although the therapeutic mechanisms of BMC, HSC and MSC treatments are still not fully characterized, the evidence thus far has indicated that the potential therapeutic mechanisms of MSCs are clearer than those of BMCs or HSCs with respect to liver regenerative medicine. MSCs suppress inflammatory responses, reduce hepatocyte apoptosis, increase hepatocyte regeneration, reverse liver fibrosis and enhance liver functionality. This paper summarizes the clinical studies that have used BMCs, HSCs and MSCs in patients with liver failure or cirrhosis. We also present the potential therapeutic mechanisms of BMCs, HSCs and MSCs for the improvement of liver function.
Collapse
|
48
|
Wang Y, Zhang X, Yang Y, Yang X, Ye B. Study on the Antifibrotic Effects of Recombinant Shark Hepatical Stimulator Analogue (r-sHSA) in Vitro and in Vivo. Mar Drugs 2015; 13:5201-18. [PMID: 26295240 PMCID: PMC4557020 DOI: 10.3390/md13085201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 05/21/2015] [Accepted: 08/10/2015] [Indexed: 12/18/2022] Open
Abstract
Hepatic fibrosis is an effusive wound healing process, characterized by an excessive deposition of extracellular matrix (ECM), as the consequence of chronic liver injury of any etiology. Current therapeutic repertoire for hepatic fibrosis is limited to withdrawal of the noxious agent, which is not always feasible. Hence, in this article, the antifibrotic effects and possible mechanisms of r-sHSA, a recombinant protein with hepatoprotection potential, were investigated. Using NIH/3T3 (mouse embro-fibroblast cell line), skin fibroblasts (human skin fibroblasts, SFBs) and HSC-T6 (rat hepatic stellate cell line), the in vitro effect of r-sHSA was evaluated by measuring the expression levels of alpha-1 Type I collagen (Col1A1) and α-smooth muscle actin (α-SMA). It turned out those fibrosis indicators were typically inhibited by r-sHSA, suggesting its capacity in HSCs inactivation. The antifibrotic activity of r-sHSA was further investigated in vivo on CCl4-induced hepatic fibrosis, in view of significant improvement of the biochemical and histological indicators. More specifically, CCl4-intoxication induced a significant increase in serological biomarkers, e.g., transaminase (AST, ALT), and alkaline phosphatase (ALP), as well as disturbed hepatic antioxidative status; most of the parameters were spontaneously ameliorated to a large extent by withdrawal of CCl4, although the fibrotic lesion was observed histologically. In contrast, r-sHSA treatment markedly eliminated fibrous deposits and restored architecture of the liver in a dose dependent manner, concomitantly with the phenomena of inflammation relief and HSCs deactivation. To sum up, these findings suggest a therapeutic potential for r-sHSA in hepatic fibrosis, though further studies are required.
Collapse
Affiliation(s)
- Ying Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| | - Xiaoyuan Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| | - Yang Yang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| | - Xiaohong Yang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| | - Boping Ye
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
49
|
Nicotinic acid prevents experimental liver fibrosis by attenuating the prooxidant process. Int Immunopharmacol 2015; 28:244-51. [PMID: 26093271 DOI: 10.1016/j.intimp.2015.05.045] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 05/12/2015] [Accepted: 05/27/2015] [Indexed: 12/11/2022]
Abstract
UNLABELLED Liver fibrosis is the excessive accumulation of extracellular matrix proteins that occurs in most chronic liver diseases. Nicotinamide treatment has been shown to prevent collagen accumulation and fibrogenesis in a bleomycin model of lung fibrosis. In this study, we evaluated the effects of nicotinic acid (NA) on experimental liver fibrosis and investigated its underlying mechanism. METHODS Fibrosis was induced by chronic TAA administration and the effects of co-administration with NA for 8 weeks were evaluated, including control groups. RESULTS TAA administration induced liver fibrosis, which was prevented by nicotinic acid. NA prevented the elevation of liver enzymes and prevented hepatic glycogen depletion. Liver histopathology and hydroxyproline levels were significantly lower in the rats treated with TAA plus NA compared with TAA only. NA demonstrated antioxidant properties by restoring the redox equilibrium (lipid peroxidation and GPx levels). Western blot assays showed decreased expression levels of TGF-β and its downstream inductor CTGF. Additionally, NA prevented hepatic stellate cell activation due by blocking the expression of α-SMA. Zymography assays showed that NA decreased the activity of matrix metalloproteinases 2 and 9. CONCLUSIONS NA prevents experimental fibrosis; the mechanisms of action are associated with its antioxidant properties and the reduction in TGF-β expression. The decrease in TGF-β levels may be associated with the attenuation of the oxidative processes, thus resulting in a reduction in HSC activation and ECM deposition. The findings suggest a possible role for NA as an antifibrotic agent for liver injury.
Collapse
|
50
|
Neuzillet C, de Gramont A, Tijeras-Raballand A, de Mestier L, Cros J, Faivre S, Raymond E. Perspectives of TGF-β inhibition in pancreatic and hepatocellular carcinomas. Oncotarget 2014; 5:78-94. [PMID: 24393789 PMCID: PMC3960190 DOI: 10.18632/oncotarget.1569] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Advanced pancreatic ductal adenocarcinoma (PDAC) and hepatocellular carcinoma (HCC) are non-curable diseases with a particularly poor prognosis. Over the last decade, research has increasingly focused on the microenvironment surrounding cancer cells, and its role in tumour development and progression. PDAC and HCC differ markedly regarding their pathological features: PDAC are typically stromal-predominant, desmoplastic, poorly vascularized tumours, whereas HCC are cellular and highly vascularized. Despite these very different settings, PDAC and HCC share transforming growth factor-β (TGF-β) as a common key-signalling mediator, involved in epithelial-to-mesenchymal transition, invasion, and stroma-tumour dialogue. Recently, novel drugs blocking the TGF-β pathway have entered clinical evaluation demonstrating activity in patients with advanced PDAC and HCC. TGF-β signalling is complex and mediates both pro- and anti-tumoural activities in cancer cells depending on their context, in space and time, and their microenvironment. In this review we provide a comprehensive overview of the role of the TGF-β pathway and its deregulation in PDAC and HCC development and progression at the cellular and microenvironment levels. We also summarize key preclinical and clinical data on the role of TGF-β as a target for therapeutic intervention in PDAC and HCC, and explore perspectives to optimize TGF-β inhibition therapy.
Collapse
Affiliation(s)
- Cindy Neuzillet
- INSERM U728 and U773 and Department of Medical Oncology, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 100 boulevard du Général Leclerc, Clichy, France
| | | | | | | | | | | | | |
Collapse
|