1
|
Wu R, Prachyathipsakul T, Zhuang J, Liu H, Han Y, Liu B, Gong S, Qiu J, Wong S, Ribbe A, Medeiros J, Bhagabati J, Gao J, Wu P, Dutta R, Herrera R, Faraci S, Xiao H, Thayumanavan S. Conferring liver selectivity to a thyromimetic using a novel nanoparticle increases therapeutic efficacy in a diet-induced obesity animal model. PNAS NEXUS 2023; 2:pgad252. [PMID: 37649581 PMCID: PMC10465086 DOI: 10.1093/pnasnexus/pgad252] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 07/21/2023] [Indexed: 09/01/2023]
Abstract
Optimization of metabolic regulation is a promising solution for many pathologies, including obesity, dyslipidemia, type 2 diabetes, and inflammatory liver disease. Synthetic thyroid hormone mimics-based regulation of metabolic balance in the liver showed promise but was hampered by the low biocompatibility and harmful effects on the extrahepatic axis. In this work, we show that specifically directing the thyromimetic to the liver utilizing a nanogel-based carrier substantially increased therapeutic efficacy in a diet-induced obesity mouse model, evidenced by the near-complete reversal of body weight gain, liver weight and inflammation, and cholesterol levels with no alteration in the thyroxine (T4) / thyroid stimulating hormone (TSH) axis. Mechanistically, the drug acts by binding to thyroid hormone receptor β (TRβ), a ligand-inducible transcription factor that interacts with thyroid hormone response elements and modulates target gene expression. The reverse cholesterol transport (RCT) pathway is specifically implicated in the observed therapeutic effect. Overall, the study demonstrates a unique approach to restoring metabolic regulation impacting obesity and related metabolic dysfunctions.
Collapse
Affiliation(s)
- Ruiling Wu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Theeraphop Prachyathipsakul
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jiaming Zhuang
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Hongxu Liu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Yanhui Han
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Bin Liu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Shuai Gong
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jingyi Qiu
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA
| | - Siu Wong
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Alexander Ribbe
- Department of Polymer Science and Engineering, University of Massachusetts, Amherst, MA 01003, USA
| | - Jewel Medeiros
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jayashree Bhagabati
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jingjing Gao
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Peidong Wu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Ranit Dutta
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | | | | | - Hang Xiao
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| | - S Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
2
|
New-Aaron M, Dagur RS, Koganti SS, Ganesan M, Wang W, Makarov E, Ogunnaike M, Kharbanda KK, Poluektova LY, Osna NA. Alcohol and HIV-Derived Hepatocyte Apoptotic Bodies Induce Hepatic Stellate Cell Activation. BIOLOGY 2022; 11:1059. [PMID: 36101437 PMCID: PMC9312505 DOI: 10.3390/biology11071059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/07/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022]
Abstract
Recently, we found that both HIV and acetaldehyde, an alcohol metabolite, induce hepatocyte apoptosis, resulting in the release of large extracellular vesicles called apoptotic bodies (ABs). The engulfment of these hepatocyte ABs by hepatic stellate cells (HSC) leads to their profibrotic activation. This study aims to establish the mechanisms of HSC activation after engulfment of ABs from acetaldehyde and HIV-exposed hepatocytes (ABAGS+HIV). In vitro experiments were performed on Huh7.5-CYP (RLW) cells to generate hepatocyte ABs and LX2 cells were used as HSC. To generate ABs, RLW cells were pretreated for 24 h with acetaldehyde, then exposed overnight to HIV1ADA and to acetaldehyde for 96 h. Thereafter, ABs were isolated from cell suspension by a differential centrifugation method and incubated with LX2 cells (3:1 ratio) for profibrotic genes and protein analyses. We found that HSC internalized ABs via the tyrosine kinase receptor, Axl. While the HIV gag RNA/HIV proteins accumulated in ABs elicited no productive infection in LX2 and immune cells, they triggered ROS and IL6 generation, which, in turn, activated profibrotic genes via the JNK-ERK1/2 and JAK-STAT3 pathways. Similarly, ongoing profibrotic activation was observed in immunodeficient NSG mice fed ethanol and injected with HIV-derived RLW ABs. We conclude that HSC activation by hepatocyte ABAGS+HIV engulfment is mediated by ROS-dependent JNK-ERK1/2 and IL6 triggering of JAK-STAT3 pathways. This can partially explain the mechanisms of liver fibrosis development frequently observed among alcohol abusing PLWH.
Collapse
Affiliation(s)
- Moses New-Aaron
- Department of Environmental Health, Occupational Health and Toxicology, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (R.S.D.); (S.S.K.); (M.G.); (M.O.); (K.K.K.)
| | - Raghubendra Singh Dagur
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (R.S.D.); (S.S.K.); (M.G.); (M.O.); (K.K.K.)
| | - Siva Sankar Koganti
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (R.S.D.); (S.S.K.); (M.G.); (M.O.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Murali Ganesan
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (R.S.D.); (S.S.K.); (M.G.); (M.O.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Weimin Wang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68105, USA; (W.W.); (E.M.); (L.Y.P.)
| | - Edward Makarov
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68105, USA; (W.W.); (E.M.); (L.Y.P.)
| | - Mojisola Ogunnaike
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (R.S.D.); (S.S.K.); (M.G.); (M.O.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Kusum K. Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (R.S.D.); (S.S.K.); (M.G.); (M.O.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Larisa Y. Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68105, USA; (W.W.); (E.M.); (L.Y.P.)
| | - Natalia A. Osna
- Department of Environmental Health, Occupational Health and Toxicology, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (R.S.D.); (S.S.K.); (M.G.); (M.O.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68105, USA; (W.W.); (E.M.); (L.Y.P.)
| |
Collapse
|
3
|
Vargas-Pozada EE, Ramos-Tovar E, Acero-Hernández C, Cardoso-Lezama I, Galindo-Gómez S, Tsutsumi V, Muriel P. Caffeine mitigates experimental nonalcoholic steatohepatitis and the progression of thioacetamide-induced liver fibrosis by blocking the MAPK and TGF-β/Smad3 signaling pathways. Ann Hepatol 2022; 27:100671. [PMID: 35065262 DOI: 10.1016/j.aohep.2022.100671] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/13/2022] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND OBJECTIVES Caffeine consumption is associated with beneficial effects on hepatic disorders. The objectives of this study were to evaluate the antifibrotic effects of caffeine on experimental nonalcoholic steatohepatitis (NASH) induced with a high-fat, high-sucrose, high-cholesterol diet (HFSCD), as well as to evaluate the ability of caffeine to prevent the progression of experimental liver fibrosis induced by the administration of thioacetamide (TAA) in rats and explore the mechanisms of action. METHODS NASH and fibrosis were induced in rats by the administration of an HFSCD for 15 weeks, and liver fibrosis was induced by intraperitoneal administration of 200 mg/kg TAA 3 times per week, for 6 weeks. Caffeine was administered at a dose of 50 mg/kg body weight. The effects of diet, TAA, and caffeine on fibrosis were evaluated by biochemical and histological examinations. The profibrotic pathways were analyzed by western blotting and immunohistochemistry. RESULTS Rats exhibited liver fibrosis after HFSCD feeding and the administration of TAA. Caffeine could reduce the hepatic level of collagen and the fibrotic area in the liver. Caffeine prevented the progression of liver fibrosis by decreasing transforming growth factor-beta (TGF-β), connective tissue growth factor (CTGF), and alpha-smooth muscle actin (α-SMA) expression and by inhibiting the activation of mitogen-activated protein kinases (MAPKs) and Smad3 phosphorylation. CONCLUSIONS Caffeine attenuates NASH and the progression of liver fibrosis due to its antifibrotic effects and modulating the MAPK and TGF-β pathways. Therefore, caffeine could be a suitable candidate for treating liver diseases associated with fibrosis.
Collapse
Affiliation(s)
- Eduardo E Vargas-Pozada
- Laboratory of Experimental Hepatology, Department of Pharmacology, Cinvestav-IPN, Apartado 14-740 Mexico City, Mexico
| | - Erika Ramos-Tovar
- Postgraduate Studies and Research Section, School of Higher Education in Medicine-IPN, Apartado 11340 Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, Mexico City, Mexico
| | - Consuelo Acero-Hernández
- Laboratory of Experimental Hepatology, Department of Pharmacology, Cinvestav-IPN, Apartado 14-740 Mexico City, Mexico
| | - Irina Cardoso-Lezama
- Laboratory of Experimental Hepatology, Department of Pharmacology, Cinvestav-IPN, Apartado 14-740 Mexico City, Mexico
| | - Silvia Galindo-Gómez
- Department of Infectomics and Molecular Pathogenesis, Cinvestav-IPN, Apartado 14-740 Mexico City, Mexico
| | - Víctor Tsutsumi
- Department of Infectomics and Molecular Pathogenesis, Cinvestav-IPN, Apartado 14-740 Mexico City, Mexico
| | - Pablo Muriel
- Laboratory of Experimental Hepatology, Department of Pharmacology, Cinvestav-IPN, Apartado 14-740 Mexico City, Mexico.
| |
Collapse
|
4
|
Gong X, Shan L, Cao S, Li K, Wu Y, Zhang Q. Notoginsenoside R1, An Active Compound from Panax notoginseng, Inhibits Hepatic Stellate Cell Activation and Liver Fibrosis via MAPK Signaling Pathway. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:511-523. [PMID: 35114912 DOI: 10.1142/s0192415x22500197] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Activation of the hepatic stellate cell is implicated in pathological vascularization during development of liver fibrosis. MAPK signaling is involved in the activation of hepatic stellate cell. Oxidative stress and inflammation are also involved in the pathogenesis of liver fibrosis. Notoginsenoside R1 is an effective saponin isolated from the roots of Panax notoginseng (Burk) F. H. Chen and exerts anti-oxidant, anti-inflammatory and anti-fibrotic roles in various diseases. However, the role of Notoginsenoside R1 in liver fibrosis has not been investigated yet. First, a rat model with liver fibrosis was established through oral gavage administration with carbon tetrachloride. Data from hematoxylin and eosin (H&E) and Masson's trichrome stainings showed that carbon tetrachloride induced severe hepatic damages, including inflammatory cell infiltration, lipid droplets deposition in hepatocytes and liver centrilobular necrosis. Meanwhile, the rats were also intraperitoneal injected with different concentrations of Notoginsenoside R1. Results demonstrated that Notoginsenoside R1 treatment suppressed the pathological changes in the livers with enhanced levels of ALB and TP, and reduced levels of ALP, AST and ALT. Second, Notoginsenoside R1 also significantly attenuated carbon tetrachloride-induced decrease in PPAR-[Formula: see text] and increase in Coll-a1, [Formula: see text]-SMA and TIMP1 in liver tissues ([Formula: see text][Formula: see text] 0.001). Third, the decrease in GSH, SOD and GST and increase in MDA, IL-1[Formula: see text], IL-6 and TNF-[Formula: see text] induced by carbon tetrachloride were markedly restored by Notoginsenoside R1 ([Formula: see text][Formula: see text] 0.001). Lastly, Notoginsenoside R1 counteracted with the promotive effects of carbon tetrachloride on levels of proteins involved in MAPK signaling, including phosphorylated p65 (p-p65), p-ERK, p-JNK and p-p38. In conclusion, Notoginsenoside R1 suppressed the activation of hepatic stellate cells and exerted anti- oxidant and anti-inflammatory to attenuate carbon tetrachloride-induced liver fibrosis through inactivation of NF-[Formula: see text]B and MAPK signaling.
Collapse
Affiliation(s)
- Xu Gong
- Department of Infectious Diseases, Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang, Jiangsu Province 222004, P. R. China
| | - Linlin Shan
- Department of Infectious Diseases, Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang, Jiangsu Province 222004, P. R. China
| | - Sisi Cao
- Department of Infectious Diseases, Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang, Jiangsu Province 222004, P. R. China
| | - Kaitao Li
- Department of Infectious Diseases, Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang, Jiangsu Province 222004, P. R. China
| | - Yanli Wu
- Department of Infectious Diseases, Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang, Jiangsu Province 222004, P. R. China
| | - Qing Zhang
- Department of Gastroenterology, Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang, Jiangsu Province 222004, P. R. China
| |
Collapse
|
5
|
Dahuang Danshen Decoction Inhibits Pancreatic Fibrosis by Regulating Oxidative Stress and Endoplasmic Reticulum Stress. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6629729. [PMID: 34422078 PMCID: PMC8371665 DOI: 10.1155/2021/6629729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 12/13/2022]
Abstract
Background In Traditional Chinese Medicine (TCM), Dahuang Danshen decoction (DD) is used to treat pancreatic fibrosis. Pancreatic fibrosis is a typical manifestation of chronic pancreatitis (CP), which affects the digestive system. The therapeutic mechanisms of DD in pancreatic fibrosis are unclear. Aim This study aimed to investigate the regulatory mechanisms of DD on oxidative stress and endoplasmic reticulum stress in CP. Materials and Methods Experimental rats were intraperitoneally injected with 500 mg/kg BW of diethyldithiocarbamate (DDC) twice a week for six weeks to induce CP. At the same time, DD was administered orally at daily doses of 1.37 g/kg BW, 2.74 g/kg BW, and 5.48 g/kg BW to evaluate its treatment effects on CP. After all treatments, pancreatic tissues were harvested and subjected to H&E staining. Transmission electron microscopy (TEM) was also performed to show the endoplasmic reticulum structure in the pancreatic tissues. Immunohistochemistry was used to detect the α-SMA expression level in the pancreatic tissues. Metabolomics analysis of the serum and proteomics analysis of the pancreatic tissues were performed to reveal the changes of endogenous metabolites and proteins, respectively. Concentrations of GSH, MDA, SOD, ROS, col-1, and col-3 were determined using corresponding kits. The western blotting method was used to determine the protein levels of Keap-1, HO-1, NQO1, Nrf2, GRP, JNK, and caspase 12. The pancreatic mRNA levels of NQO1, GPX1, HO-1, GST-π, GRP, JNK, and caspase 12 were also determined by quantitative PCR. The interactions between TCM components and Keap-1 were investigated by molecular docking modeling. Results The pathohistological results demonstrated that DD could ameliorate DDC-induced CP in vivo, indicated by reduction of α-SMA, col-1, col-3, TNF-α, and IL-6. DD increased serum levels of GSH and SOD but reduced pancreatic ROS. DD decreased cytoplasmic Keap-1 and increased Nrf2 nuclear localization. Correspondingly, DD increased the expression levels of Nrf2 downstream antioxidant genes NQO1, GPX1, HO-1, and GST-π. DD also decreased ERS hallmarks caspase 12 cleavage and GRP expression. Eventually, DD inhibited PSC activation by reducing JNK phosphorylation and MMK-3/p38 expression. Molecular docking analysis showed that salvianolic acid B and emodin had a good binding affinity toward Keap-1. Conclusions These results demonstrated that DD could ameliorate the oxidative and endoplasmic reticulum stress through releasing Nrf2 from Keap-1 binding and inducing the downstream antioxidant enzymes. As a result, DD could thwart pancreatic fibrosis by inhibiting PSCs activation, which was induced by OS and ERS through JNK and MMK3/p38 pathways.
Collapse
|
6
|
Antifibrotic Effects of Kangxian Ruangan Capsule on Rats with Nonalcoholic Fatty Liver Fibrosis and Hepatic Stellate Cells through Regulation of TGF- β and TLR4 Signaling Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5649575. [PMID: 34422075 PMCID: PMC8371615 DOI: 10.1155/2021/5649575] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 05/07/2021] [Accepted: 07/22/2021] [Indexed: 01/18/2023]
Abstract
Kangxian ruangan (KXRG) is a traditional Chinese medicine (TCM) formula consisting of 12 herbs. TCM syndrome differentiation proposes that KXRG exerts pharmacological effects against nonalcoholic fatty liver disease (NAFLD) fibrosis. This work investigates the effect of KXRG on NAFLD fibrosis in vivo and in vitro. In vivo, the NAFLD fibrosis model was constructed in Wistar rats using methionine- and choline-deficient (MCD) diet, followed by KXRG (0.92 g/kg/d) treatment for 8 weeks. In vitro, primary hepatic stellate cells (HSCs) were activated using platelet-derived growth factor (PDGF) and treated with KXRG. Molecular mechanisms underlying fibrosis were investigated. After 8 weeks, compared with the control groups, the histological lesions, degree of fibrosis, and inflammatory reaction increased with the MCD diet as demonstrated by histological changes and increased fibrosis-related (α-SMA, TGF-β, COL1A1, and desmin, P < 0.01) and inflammation-related factors (TNF-α, MCP-1, and F4/80, P < 0.01), whereas they decreased with KXRG treatment (P < 0.01). KXRG not only inhibited the proliferation of activated HSCs and promoted their apoptosis but also resulted in G0-G1 arrest. Furthermore, KXRG suppressed HSC activation (P < 0.01), collagen synthesis (P < 0.01), and α-SMA expression (P < 0.01) with PDGF stimulation. In both the MCD diet-induced animal model and PDGF-induced cell model, KXRG inhibited TGF-β and TLR4 signaling (P < 0.01), similar to corresponding small-molecule inhibitors. These results demonstrated that KXRG might exert suppressive effects against NAFLD fibrosis via regulating TGF-β and TLR4 signaling. KXRG may act as a natural and potent therapeutic agent against NAFLD.
Collapse
|
7
|
Taurine ameliorates thioacetamide induced liver fibrosis in rats via modulation of toll like receptor 4/nuclear factor kappa B signaling pathway. Sci Rep 2021; 11:12296. [PMID: 34112866 PMCID: PMC8192756 DOI: 10.1038/s41598-021-91666-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis is a significant health problem that can cause serious illness and death. Unfortunately, a standard treatment for liver fibrosis has not been approved yet due to its complicated pathogenesis. The current study aimed at assessing the anti-fibrotic effect of taurine against thioacetamide induced liver fibrosis in rats through the modulation of toll like receptor 4/nuclear factor kappa B signaling pathway. Both concomitant and late taurine treatment (100 mg/kg, IP, daily) significantly reduced the rise in serum ALT and AST activities and significantly reversed the decrease in serum albumin and total protein. These results were confirmed by histopathological examinations and immunehistochemical inspection of α-SMA, caspase-3 and NF-κB. The antioxidant potential of taurine was verified by a marked increase of GSH content and a reduction of MDA level in liver tissue. The anti-fibrotic effects of taurine were evaluated by investigating the expression of TLR4, NF-κB. The protein levels of IL-6, LPS, MyD88, MD2, CD14, TGF-β1 and TNF-α were determined. Docking studies were carried out to understand how taurine interacts inside TLR4-MD2 complex and it showed good binding with the hydrophobic binding site of MD2. We concluded that the anti-fibrotic effect of taurine was attributable to the modulation of the TLR4/NF-κB signaling.
Collapse
|
8
|
Fan Y, Yin X. Potential therapeutic targets and biological mechanisms of Centella asiatica on hepatic fibrosis: a study of network pharmacology. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:932. [PMID: 34350247 PMCID: PMC8263891 DOI: 10.21037/atm-21-2253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 05/26/2021] [Indexed: 11/26/2022]
Abstract
Background Liver fibrosis is a common result of the repair process of various chronic liver diseases. This study is a network pharmacology study on the potential therapeutic targets and biological mechanisms of Centella asiatica for liver fibrosis. Methods The chemical components and potential targets of Centella asiatica were screened through TCMSP, PubChem database, and Swiss Target Prediction database. The DisGeNET and GeneCards databases were used to obtain targets of HF. Venn diagrams were used to find key targets, and draw protein interaction maps. Cytoscape software was used to construct an interaction network map of drug-component-target-disease-pathway. The mechanisms of action were predicted through enrichment analysis and KEGG analysis. Results In total, 6 main components, 297 drug targets, 337 HF targets, and 48 drug-disease targets were obtained in Centella asiatica. The key targets involved IL6, TNF, VEGFA, TP53, IL1β, MMP9, CXCL8, EGFR, JUN, SRC, MMP2, and TGF-β, among others. A total of 1293 entries were obtained by Gene Ontology (GO) enrichment analysis, which mainly involved the regulation of reactive oxygen species metabolic process, the regulation of smooth muscle cells, and the regulation of DNA-binding transcription factor activity. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment mainly screened 191 pathways, including the MAPK signaling pathway, the relaxin signaling pathway, and the Toll-like receptor signaling pathway, among others. Conclusions Centella asiatica may have a therapeutic effect on HF through multiple targets and pathways. Its mechanism is mainly related to the MAPK signaling pathway and the relaxin signaling pathway.
Collapse
Affiliation(s)
- Yuli Fan
- Department of Clinical Infectious Disease, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyan Yin
- Department of Emergency, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Wang W, Huang X, Fan X, Yan J, Luan J. Progress in evaluating the status of hepatitis C infection based on the functional changes of hepatic stellate cells (Review). Mol Med Rep 2020; 22:4116-4124. [PMID: 33000255 DOI: 10.3892/mmr.2020.11516] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 08/18/2020] [Indexed: 11/06/2022] Open
Abstract
Hepatitis C virus (HCV) infection is a global public health problem. Cirrhosis and hepatocellular carcinoma are the main causes of death in patients with chronic hepatitis C (CHC) infection. Liver fibrosis is an important cause of cirrhosis and end‑stage liver disease after CHC infection. Along with the course of infection, liver fibrosis exhibits a progressive exacerbation. Hepatic stellate cells (HSCs) are involved in both physiological and pathological processes of the liver. During the chronic liver injury process, the activated HSCs transform into myofibroblasts, which are important cells in the development of liver fibrosis. At present, HCV infection still lacks specific markers for the accurate detection of the disease condition and progression. Therefore, the present review focused on HSCs, which are closely related to HCV‑infected liver fibrosis, and analyzed the changes in the HSCs, including their surface‑specific markers, cytokine production, activation, cell function and morphological structure. The present review aimed to propose novel diagnostic markers, at both the cellular and molecular level, which would be of great significance for the timely diagnosis of the disease. According to this aim, the characteristic changes of HSCs during HCV infection were reviewed in the present article.
Collapse
Affiliation(s)
- Wei Wang
- Department of Blood Transfusion Medicine, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Xuelian Huang
- Department of Blood Transfusion Medicine, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Xuzhou Fan
- Department of Blood Transfusion Medicine, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Jingmei Yan
- Department of Blood Transfusion Medicine, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Jianfeng Luan
- Department of Blood Transfusion Medicine, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| |
Collapse
|
10
|
He X, Chen J, Mu Y, Zhang H, Chen G, Liu P, Liu W. The effects of inhibiting the activation of hepatic stellate cells by lignan components from the fruits of Schisandra chinensis and the mechanism of schisanhenol. J Nat Med 2020; 74:513-524. [PMID: 32193805 DOI: 10.1007/s11418-020-01394-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 03/06/2020] [Indexed: 12/15/2022]
Abstract
Liver fibrosis is a pathological manifestation induced by chronic liver injury and may cause cirrhosis and liver cancer with the chronic progression of fibrosis. During the onset and progression of liver fibrosis, the activation of hepatic stellate cells (HSCs) is the core mechanism for the secretion of many extracellular matrices to induce fibrosis. Lignans are reportedly the main effective components of Schisandra chinensis with good anti-fibrosis effects. In this study, we compared the inhibiting effects of the seven lignan components from S. chinensis on HSC activation. We found that the seven lignans inhibited the activation of human HSCs (LX-2) in various degrees. Among all lignans, schisanhenol showed the best effect in inhibiting the activation of LX-2 with a dose-effect relationship. Sal also inhibited the phosphorylations of Smad1, Smad2, Smad3, extracellular regulated protein kinase (ERK), c-Jun N-terminal kinase (JNK), p38, and nuclear transcription factor-κB (NF-κB), as well as downregulated Smad4. All these findings suggested that schisanhenol may ameliorate liver fibrosis by inhibiting the transforming growth factor β (TGF-β)/Smad and mitogen-activated protein kinase (MAPK) signaling pathways. Remarkably, schisanhenol may be a potential anti-liver fibrosis drug and warrants further research.
Collapse
Affiliation(s)
- Xiaoli He
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Shanghai, 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, 528 Zhangheng Road, Shanghai, 201203, China
- TCM Department, Ningbo Huamei Hospital Affiliated to Chinese Academy of Sciences, 41 Xibei Road, Ningbo, 315010, China
| | - Jiamei Chen
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Shanghai, 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, 528 Zhangheng Road, Shanghai, 201203, China
| | - Yongping Mu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Shanghai, 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, 528 Zhangheng Road, Shanghai, 201203, China
| | - Hua Zhang
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Shanghai, 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, 528 Zhangheng Road, Shanghai, 201203, China
| | - Gaofeng Chen
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Shanghai, 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, 528 Zhangheng Road, Shanghai, 201203, China
| | - Ping Liu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Shanghai, 201203, China.
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, 528 Zhangheng Road, Shanghai, 201203, China.
| | - Wei Liu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Shanghai, 201203, China.
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, 528 Zhangheng Road, Shanghai, 201203, China.
| |
Collapse
|
11
|
Jo YH, Lee H, Oh MH, Lee GH, Lee YJ, Lee JS, Kim MJ, Kim WY, Kim JS, Yoo DS, Cho SW, Cha SW, Pyo MK. Antioxidant and hepatoprotective effects of Korean ginseng extract GS-KG9 in a D-galactosamine-induced liver damage animal model. Nutr Res Pract 2020; 14:334-351. [PMID: 32765814 PMCID: PMC7390743 DOI: 10.4162/nrp.2020.14.4.334] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/04/2020] [Accepted: 03/24/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND/OBJECTIVES This study was designed to investigate the improvement effect of white ginseng extract (GS-KG9) on D-galactosamine (Ga1N)-induced oxidative stress and liver injury. SUBJECTS/METHODS Sixty Sprague-Dawley rats were divided into 6 groups. Rats were orally administrated with GS-KG9 (300, 500, or 700 mg/kg) or silymarin (25 mg/kg) for 2 weeks. The rats of the GS-KG9- and silymarin-treated groups and a control group were then intraperitoneally injected Ga1N at a concentration of 650 mg/kg for 4 days. To investigate the protective effect of GS-KG9 against GalN-induced liver injury, blood liver function indicators, anti-oxidative stress indicators, and histopathological features were analyzed. RESULTS Serum biochemical analysis indicated that GS-KG9 ameliorated the elevation of aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (ALP), and lactate dehydrogenase (LDH) in GalN-treated rats. The hepatoprotective effects of GS-KG9 involved enhancing components of the hepatic antioxidant defense system, including glutathione, glutathione peroxidase (GPx), superoxide dismutase (SOD), and catalase (CAT). In addition, GS-KG9 treatment inhibited reactive oxygen species (ROS) production induced by GalN treatment in hepatocytes and significantly increased the expression levels of nuclear factor erythroid-2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) proteins, which are antioxidant proteins. In particular, by histological analyses bases on hematoxylin and eosin, Masson's trichrome, α-smooth muscle actin, and transforming growth factor-β1 staining, we determined that the administration of 500 mg/kg GS-KG9 inhibited hepatic inflammation and fibrosis due to the excessive accumulation of collagen. CONCLUSIONS These findings demonstrate that GS-KG9 improves GalN-induced liver inflammation, necrosis, and fibrosis by attenuating oxidative stress. Therefore, GS-KG9 may be considered a useful candidate in the development of a natural preventive agent against liver injury.
Collapse
Affiliation(s)
- Yun Ho Jo
- International Ginseng & Herb Research Institute, Geumsan 32724, Korea
| | - Hwan Lee
- International Ginseng & Herb Research Institute, Geumsan 32724, Korea
| | - Myeong Hwan Oh
- International Ginseng & Herb Research Institute, Geumsan 32724, Korea
| | - Gyeong Hee Lee
- International Ginseng & Herb Research Institute, Geumsan 32724, Korea
| | - You Jin Lee
- International Ginseng & Herb Research Institute, Geumsan 32724, Korea
| | - Ji Sun Lee
- International Ginseng & Herb Research Institute, Geumsan 32724, Korea
| | - Min Jung Kim
- International Ginseng & Herb Research Institute, Geumsan 32724, Korea
| | - Won Yong Kim
- International Ginseng & Herb Research Institute, Geumsan 32724, Korea
| | - Jin Seong Kim
- International Ginseng & Herb Research Institute, Geumsan 32724, Korea
| | - Dae Seok Yoo
- International Ginseng & Herb Research Institute, Geumsan 32724, Korea
| | - Sang Won Cho
- International Ginseng & Herb Research Institute, Geumsan 32724, Korea
| | - Seon Woo Cha
- International Ginseng & Herb Research Institute, Geumsan 32724, Korea
| | - Mi Kyung Pyo
- International Ginseng & Herb Research Institute, Geumsan 32724, Korea
| |
Collapse
|
12
|
Kim JY, Kim KM, Yang JH, Cho SS, Kim SJ, Park SJ, Ahn SG, Lee GH, Yang JW, Lim SC, Kang KW, Ki SH. Induction of E6AP by microRNA-302c dysregulation inhibits TGF-β-dependent fibrogenesis in hepatic stellate cells. Sci Rep 2020. [PMID: 31949242 DOI: 10.1038/s41598-019-57322-w.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Hepatic stellate cells (HSCs) are essential for liver fibrosis. E6 associated protein (E6AP) is one of the E3-ubiquitin-protein ligase and has been studied in proliferation and cellular stress. Currently, no information is available on the role of E6AP on transforming growth factor-β (TGF-β) signaling and hepatic fibrogenesis. This study examined whether E6AP is overexpressed in activated HSCs, and if so, its effect on hepatic fibrogenesis and the molecular mechanism. E6AP was expressed higher in HSCs than hepatocytes, and was up-regulated in activated HSCs, HSCs from the livers of carbon tetrachloride-injected mice, or TGF-β-treated LX-2 cells. The TGF-β-mediated E6AP up-regulation was not due to altered mRNA level nor protein stability. Thus, we performed microRNA (miRNA, miR) analysis and found that miR-302c was dysregulated in TGF-β-treated LX-2 cells or activated primary HSCs. We revealed that miR-302c was a modulator of E6AP. E6AP overexpression inhibited TGF-β-induced expression of plasminogen activator inhibitor-1 in LX-2 cells, albeit it was independent of Smad pathway. Additionally, E6AP inhibited TGF-β-mediated phosphorylation of mitogen-activated protein kinases. To conclude, E6AP overexpression due to decreased miR-302c in HSCs attenuated hepatic fibrogenesis through inhibition of the TGF-β-induced mitogen-activated protein kinase signaling pathway, implying that E6AP and other molecules may contribute to protection against liver fibrosis.
Collapse
Affiliation(s)
- Ji Young Kim
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Kyu Min Kim
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Ji Hye Yang
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea.,College of Korean Medicine, Dongshin University, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Sam Seok Cho
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Seung Jung Kim
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Su Jung Park
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Sang-Gun Ahn
- Department of Pathology, College of Dentistry, Chosun University, Gwangju, 61452, Republic of Korea
| | - Gum Hwa Lee
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Jin Won Yang
- College of Pharmacy, Woosuk University, Wanju, Jeonbuk, 55338, Republic of Korea
| | - Sung Chul Lim
- College of Medicine, Chosun University, Gwangju, 61452, Republic of Korea
| | - Keon Wook Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sung Hwan Ki
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea.
| |
Collapse
|
13
|
Induction of E6AP by microRNA-302c dysregulation inhibits TGF-β-dependent fibrogenesis in hepatic stellate cells. Sci Rep 2020; 10:444. [PMID: 31949242 PMCID: PMC6965100 DOI: 10.1038/s41598-019-57322-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 12/27/2019] [Indexed: 12/13/2022] Open
Abstract
Hepatic stellate cells (HSCs) are essential for liver fibrosis. E6 associated protein (E6AP) is one of the E3-ubiquitin-protein ligase and has been studied in proliferation and cellular stress. Currently, no information is available on the role of E6AP on transforming growth factor-β (TGF-β) signaling and hepatic fibrogenesis. This study examined whether E6AP is overexpressed in activated HSCs, and if so, its effect on hepatic fibrogenesis and the molecular mechanism. E6AP was expressed higher in HSCs than hepatocytes, and was up-regulated in activated HSCs, HSCs from the livers of carbon tetrachloride-injected mice, or TGF-β-treated LX-2 cells. The TGF-β-mediated E6AP up-regulation was not due to altered mRNA level nor protein stability. Thus, we performed microRNA (miRNA, miR) analysis and found that miR-302c was dysregulated in TGF-β-treated LX-2 cells or activated primary HSCs. We revealed that miR-302c was a modulator of E6AP. E6AP overexpression inhibited TGF-β-induced expression of plasminogen activator inhibitor-1 in LX-2 cells, albeit it was independent of Smad pathway. Additionally, E6AP inhibited TGF-β-mediated phosphorylation of mitogen-activated protein kinases. To conclude, E6AP overexpression due to decreased miR-302c in HSCs attenuated hepatic fibrogenesis through inhibition of the TGF-β-induced mitogen-activated protein kinase signaling pathway, implying that E6AP and other molecules may contribute to protection against liver fibrosis.
Collapse
|
14
|
Huang X, Wang L, Meng M, Zhang S, Pham TTH, Jiang L, Chen L, Li Y, Zhou X, Qin L, Wu X, Zou C, Huang R. Extract of Averrhoacarambola L. (Oxalidaceae) roots ameliorates carbon tetrachloride-induced hepatic fibrosis in rats. Biomed Pharmacother 2020; 121:109516. [DOI: 10.1016/j.biopha.2019.109516] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/20/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023] Open
|
15
|
Ramos-Tovar E, Buendia-Montaño LD, Galindo-Gómez S, Hernández-Aquino E, Tsutsumi V, Muriel P. Stevia prevents experimental cirrhosis by reducing hepatic myofibroblasts and modulating molecular profibrotic pathways. Hepatol Res 2019; 49:212-223. [PMID: 30338893 DOI: 10.1111/hepr.13275] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/17/2018] [Accepted: 10/10/2018] [Indexed: 12/21/2022]
Abstract
AIM The aims of the present study were to investigate the capacity of stevia leaves to prevent experimental cirrhosis induced by chronic administration of carbon tetrachloride (CCl4 ) in rats and to explore the action mechanism involved. METHODS Liver cirrhosis was established by CCl4 treatment (400 mg/kg i.p. three times a week for 12 weeks); stevia powder was administered (100 mg/kg by gavage daily) during the CCl4 treatment. Serum markers of liver damage and hydroxyproline were evaluated and histopathological analyses were carried out. The profibrotic pathways were analyzed by western blot and immunohistochemistry. RESULTS We found for the first time that stevia cotreatment prevented the elevation of serum markers of necrosis and cholestasis and the occurrence of liver fibrosis. It is worth noting that stevia downregulated several profibrogenic pathways, including the reduction of hepatic myofibroblasts and decreased matrix metalloproteinase (MMP)2 and MMP13 expression, thereby blocking the liberation of transforming growth factor-β from the extracellular matrix. Notably, stevia reduced the phosphorylation of pSmad3L, the most profibrogenic and mitogenic Smad, by inhibiting the activation of c-Jun N-terminal kinase and extracellular signal-regulated kinase. Interestingly, Smad7, an important antifibrotic molecule, was upregulated by stevia treatment in cirrhotic rats. These multitarget mechanisms led to the prevention of experimental cirrhosis. CONCLUSIONS Because stevia possesses a reasonable safety profile, our results indicate that it could be useful in the clinical setting to treat chronic liver diseases.
Collapse
Affiliation(s)
- Erika Ramos-Tovar
- Laboratory of Experimental Hepatology, Department of Pharmacology, Cinvestav-IPN, Mexico City, Mexico
| | - Laura D Buendia-Montaño
- Laboratory of Experimental Hepatology, Department of Pharmacology, Cinvestav-IPN, Mexico City, Mexico
| | - Silvia Galindo-Gómez
- Department of Infectomics and Molecular Pathogenesis, Cinvestav-IPN, Mexico City, Mexico
| | - Erika Hernández-Aquino
- Laboratory of Experimental Hepatology, Department of Pharmacology, Cinvestav-IPN, Mexico City, Mexico
| | - Víctor Tsutsumi
- Department of Infectomics and Molecular Pathogenesis, Cinvestav-IPN, Mexico City, Mexico
| | - Pablo Muriel
- Laboratory of Experimental Hepatology, Department of Pharmacology, Cinvestav-IPN, Mexico City, Mexico
| |
Collapse
|
16
|
Zhu H, Chai Y, Dong D, Zhang N, Liu W, Ma T, Wu R, Lv Y, Hu L. AICAR-Induced AMPK Activation Inhibits the Noncanonical NF-κB Pathway to Attenuate Liver Injury and Fibrosis in BDL Rats. Can J Gastroenterol Hepatol 2018; 2018:6181432. [PMID: 30662889 PMCID: PMC6314002 DOI: 10.1155/2018/6181432] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/17/2018] [Accepted: 11/29/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND To evaluate the AMP-activated protein kinase- (AMPK-) mediated signaling and NF-κB-related inflammatory pathways that contribute to cholestatic diseases in the bile duct ligation (BDL) rat model of chronic cholestasis and verify the protective role of 5-Aminoimidazole-4-carboxamide1-β-D-ribofuranoside (AICAR) against hepatic injury and fibrosis triggered by cholestasis-related inflammation. METHODS Animals were randomly divided into three groups: sham-operated group, BDL group, and BDL+ AICAR group. Cholestatic liver injury was induced by common BDL. Two weeks later, rats in BDL+AICAR group started receiving AICAR treatment. Hepatic pathology was examined by haematoxylin and eosin (H&E) and sirius red staining and hydroxyproline assay was performed in evaluating the severity of hepatic cirrhosis. Real-time PCR and Western blot were performed for RNA gene expression of RNA and protein levels, respectively. RESULTS The BDL group showed liver injury as evidenced by histological changes and elevation in serum biochemicals, ductular reaction, fibrosis, and inflammation. The mRNA expression of canonical NF-κB inflammatory cytokines such as TNF-α, IL-1β, TGF-β, and the protein of noncanonical NF-κB, P100, and P52 was upregulated in the livers of BDL rats. The BDL rats with the administration of AICAR could induce AMPK activation inhibiting the noncanonical NF-κB pathway to attenuate liver injury and fibrosis in BDL rats. CONCLUSION The BDL model of hepatic cholestatic injury resulting in activation of Kupffer cells and recruitment of immune cells might initiate an inflammatory response through activation of the NF-κB pathway. The AMPK activator AICAR significantly alleviated BDL-induced inflammation in rats by mainly inhibiting the noncanonical NF-κB pathway and thus protecting against hepatic injury and fibrosis triggered by BDL.
Collapse
Affiliation(s)
- Haoyang Zhu
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Xi'an 710061, Shaanxi Province, China
| | - Yichao Chai
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Xi'an 710061, Shaanxi Province, China
| | - Dinghui Dong
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Xi'an 710061, Shaanxi Province, China
| | - Nana Zhang
- Institute for Cancer Research School of Basic Medical Science of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Wenyan Liu
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Xi'an 710061, Shaanxi Province, China
| | - Tao Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Xi'an 710061, Shaanxi Province, China
| | - Rongqian Wu
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Xi'an 710061, Shaanxi Province, China
| | - Yi Lv
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Xi'an 710061, Shaanxi Province, China
| | - Liangshuo Hu
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Xi'an 710061, Shaanxi Province, China
| |
Collapse
|
17
|
Liu Q, Lu J, Lin J, Tang Y, Pu W, Shi X, Jiang S, Liu J, Ma Y, Li Y, Xu J, Jin L, Wang J, Wu W. Salvianolic acid B attenuates experimental skin fibrosis of systemic sclerosis. Biomed Pharmacother 2018; 110:546-553. [PMID: 30530290 DOI: 10.1016/j.biopha.2018.12.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 11/28/2018] [Accepted: 12/02/2018] [Indexed: 02/02/2023] Open
Abstract
Systemic sclerosis (SSc) is a connective tissue disease characterized mainly by fibrosis of skin and internal organs. Our previous study has shown that salvianolic acid B (SAB), a bioactive component extracted from Salvia miltiorrhiza (SM), was one of the essential ingredients in the traditional Chinese medicine Yiqihuoxue formula, which has been used to treat SSc-related dermal and pulmonary fibrosis. The aim of the present study was to evaluate the effect of SAB on skin fibrosis and explore its underlying anti-fibrotic mechanism. We found that SAB was capable of alleviating skin fibrosis in a bleomycin-induced SSc mouse model, alleviating skin thickness and reducing collagen deposition. in vitro studies indicated that SAB reduced SSc skin fibroblast proliferation and downregulated extracellular matrix gene transcription and collagen protein expression. TGF-β/SMAD and MAPK/ERK pathway activation were also shown to be suppressed in SAB treated fibroblasts. Moreover, RNA-seq revealed that the anti-fibrotic effect of SAB might be related to antioxidant activity, the cell cycle, and the p53 signaling pathway. Taken together, our results suggest that SAB has the ability to alleviate SSc-related skin fibrosis both in vivo and in vitro.
Collapse
Affiliation(s)
- Qingmei Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Jiaying Lu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinran Lin
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yulong Tang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Weilin Pu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiangguang Shi
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Shuai Jiang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Jing Liu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Yanyun Ma
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Yuan Li
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Jinhua Xu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China; Human Phenome Institute, Fudan University, Shanghai, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China; Human Phenome Institute, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China.
| | - Wenyu Wu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China; Department of dermatology, Jing'an District Central Hospital, Shanghai, China.
| |
Collapse
|
18
|
Nahar S, Nakashima Y, Miyagi-Shiohira C, Kinjo T, Toyoda Z, Kobayashi N, Saitoh I, Watanabe M, Noguchi H, Fujita J. Cytokines in adipose-derived mesenchymal stem cells promote the healing of liver disease. World J Stem Cells 2018; 10:146-159. [PMID: 30631390 PMCID: PMC6325075 DOI: 10.4252/wjsc.v10.i11.146] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/07/2018] [Accepted: 10/11/2018] [Indexed: 02/06/2023] Open
Abstract
Adipose-derived mesenchymal stem cells (ADSCs) are a treatment cell source for patients with chronic liver injury. ADSCs are characterized by being harvested from the patient's own subcutaneous adipose tissue, a high cell yield (i.e., reduced immune rejection response), accumulation at a disease nidus, suppression of excessive immune response, production of various growth factors and cytokines, angiogenic effects, anti-apoptotic effects, and control of immune cells via cell-cell interaction. We previously showed that conditioned medium of ADSCs promoted hepatocyte proliferation and improved the liver function in a mouse model of acute liver failure. Furthermore, as found by many other groups, the administration of ADSCs improved liver tissue fibrosis in a mouse model of liver cirrhosis. A comprehensive protein expression analysis by liquid chromatography with tandem mass spectrometry showed that the various cytokines and chemokines produced by ADSCs promote the healing of liver disease. In this review, we examine the ability of expressed protein components of ADSCs to promote healing in cell therapy for liver disease. Previous studies demonstrated that ADSCs are a treatment cell source for patients with chronic liver injury. This review describes the various cytokines and chemokines produced by ADSCs that promote the healing of liver disease.
Collapse
Affiliation(s)
- Saifun Nahar
- Department of Infectious, Respiratory, and Digestive Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Yoshiki Nakashima
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Chika Miyagi-Shiohira
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Takao Kinjo
- Department of Basic Laboratory Sciences, School of Health Sciences in the Faculty of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Zensei Toyoda
- Department of Basic Laboratory Sciences, School of Health Sciences in the Faculty of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | | | - Issei Saitoh
- Division of Pediatric Dentistry, Graduate School of Medical and Dental Science, Niigata University, Niigata 951-8514, Japan
| | - Masami Watanabe
- Department of Urology, Okayama Univer sity Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Hirofumi Noguchi
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| | - Jiro Fujita
- Department of Infectious, Respiratory, and Digestive Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| |
Collapse
|
19
|
Wang ZF, Wang MY, Yu DH, Zhao Y, Xu HM, Zhong S, Sun WY, He YF, Niu JQ, Gao PJ, Li HJ. Therapeutic effect of chitosan on CCl4‑induced hepatic fibrosis in rats. Mol Med Rep 2018; 18:3211-3218. [PMID: 30085342 PMCID: PMC6102732 DOI: 10.3892/mmr.2018.9343] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 06/19/2018] [Indexed: 12/15/2022] Open
Abstract
Chitosan is a linear polysaccharide that is made by treating the chitin shells of shrimp and crustaceans with an alkaline substance, for example sodium hydroxide. Due to its unique physical and chemical properties, chitosan has a wide range of applications in the medical field. Currently, there are no effective treatments for liver fibrosis; therefore, the aim of the present study was to investigate the therapeutic effect of chitosan in a CCl4‑induced hepatic fibrosis (HF) rat model. The serum levels of aspartate transaminase (AST), alanine transaminase (ALT) and alkaline phosphatase (ALP) were measured by ELISA. Collagen (COL) 3 and α‑smooth muscle actin (SMA) expression levels in the rat liver were detected by reverse transcription‑semiquantitative polymerase chain reaction and western blotting, respectively. The results demonstrated that treatment with chitosan significantly improved HF, by decreasing the serum levels of AST, ALT, and ALP; improving liver histology; and decreasing the expression levels of COL3 and α‑SMA. Chitosan may offer an alternative approach for the clinical treatment of HF.
Collapse
Affiliation(s)
- Zhong-Feng Wang
- Department of Geriatrics, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Mao-Yu Wang
- Department of CCU, The First People's Hospital of Aksu Prefecture in Xinjiang, Aksu, Xinjiang 843000, P.R. China
| | - De-Hai Yu
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130061, P.R. China
| | - Yan Zhao
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, Jilin 130061, P.R. China
| | - Hong-Mei Xu
- Department of Obstetrics, The First Hospital, Jilin University, Changchun, Jilin 130061, P.R. China
| | - Sheng Zhong
- Department of Geriatrics, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wen-Yi Sun
- Department of Clinical Pharmacy and Pharmaceutical Management, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yu-Fang He
- Institute of Phytochemistry, Jilin Academy of Chinese Medicine Sciences, Changchun, Jilin 130012, P.R. China
| | - Jun-Qi Niu
- Department of Geriatrics, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Pu-Jun Gao
- Department of Geriatrics, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hai-Jun Li
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
20
|
Wang L, Pan MH, Lo CY, Zhao H, Li S, Ho CT, Yang G. Anti-fibrotic activity of polyphenol-enriched sugarcane extract in rats via inhibition of p38 and JNK phosphorylation. Food Funct 2018; 9:951-958. [DOI: 10.1039/c7fo01617d] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Sugarcane (Saccharum officinarum L.), which is one of the most important sources of sugar, is also rich in polyphenolic compounds.
Collapse
Affiliation(s)
- Liwen Wang
- Hubei Key Laboratory for Processing & Application of Catalytic Materials
- College of Chemistry & Chemical Engineering
- Huanggang Normal University
- China
- Tianjin Key Laboratory of Food and Biotechnology
| | - Min-Hsiung Pan
- Institute of Food Science and Technology
- National Taiwan University
- Taipei 10617
- Taiwan
- Department of Medical Research
| | - Chih-Yu Lo
- Department of Food Science
- National Chiayi University
- Chiayi 60004
- Taiwan
| | - Hui Zhao
- Tianjin Key Laboratory of Food and Biotechnology
- School of Biotechnology and Food Science
- Tianjin University of Commerce
- Tianjin
- China
| | - Shiming Li
- Hubei Key Laboratory for Processing & Application of Catalytic Materials
- College of Chemistry & Chemical Engineering
- Huanggang Normal University
- China
| | - Chi-Tang Ho
- Department of Food Science
- Rutgers University
- New Brunswick
- USA
| | - Guliang Yang
- Hubei Key Laboratory for Processing & Application of Catalytic Materials
- College of Chemistry & Chemical Engineering
- Huanggang Normal University
- China
| |
Collapse
|
21
|
Que R, Shen Y, Ren J, Tao Z, Zhu X, Li Y. Estrogen receptor‑β‑dependent effects of saikosaponin‑d on the suppression of oxidative stress‑induced rat hepatic stellate cell activation. Int J Mol Med 2017; 41:1357-1364. [PMID: 29286085 PMCID: PMC5819932 DOI: 10.3892/ijmm.2017.3349] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/06/2017] [Indexed: 11/22/2022] Open
Abstract
Saikosaponin-d (SSd) is one of the major triterpenoid saponins derived from Bupleurum falcatum L., which has been reported to possess antifibrotic activity. At present, there is little information regarding the potential target of SSd in hepatic stellate cells (HSCs), which serve an important role in excessive extracellular matrix (ECM) deposition during the pathogenesis of hepatic fibrosis. Our recent study indicated that SSd may be considered a novel type of phytoestrogen with estrogen-like actions. Therefore, the present study aimed to investigate the effects of SSd on the proliferation and activation of HSCs, and the underlying mechanisms associated with estrogen receptors. In the present study, a rat HSC line (HSC-T6) was used and cultured with dimethyl sulfoxide, SSd, or estradiol (E2; positive control), in the presence or absence of three estrogen receptor (ER) antagonists [ICI-182780, methylpiperidinopyrazole (MPP) or (R,R)-tetrahydrochrysene (THC)], for 24 h as pretreatment. Oxidative stress was induced by exposure to hydrogen peroxide for 4 h. Cell proliferation was assessed by MTT growth assay. Malondialdehyde (MDA), CuZn-superoxide dismutase (CuZn-SOD), tissue inhibitor of metalloproteinases-1 (TIMP- 1), matrix metalloproteinase-1 (MMP-1), transforming growth factor-β1 (TGF-β1), hydroxyproline (Hyp) and collagen-1 (COL1) levels in cell culture supernatants were determined by ELISA. Reactive oxygen species (ROS) was detected by flow cytometry. Total and phosphorylated mitogen-activated protein kinases (MAPKs) and α-smooth muscle actin (α-SMA) were examined by western blot analysis. TGF-β1 mRNA expression was determined by RT-quantitative (q)PCR. SSd and E2 were able to significantly suppress oxidative stress-induced proliferation and activation of HSC-T6 cells. Furthermore, SSd and E2 were able to reduce ECM deposition, as demonstrated by the decrease in transforming growth factor-β1, hydroxyproline, collagen-1 and tissue inhibitor of metalloproteinases-1, and by the increase in matrix metalloproteinase-1. These results suggested that the possible molecular mechanism could involve downregulation of the reactive oxygen species/mitogen-activated protein kinases signaling pathway. Finally, the effects of SSd and E2 could be blocked by co-incubation with ICI-182780 or THC, but not MPP, thus indicating that ERβ may be the potential target of SSd in HSC-T6 cells. In conclusion, these findings suggested that SSd may suppress oxidative stress-induced activation of HSCs, which relied on modulation of ERβ.
Collapse
Affiliation(s)
- Renye Que
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Yanting Shen
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Jianlin Ren
- Department of Scientific Research, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Zhihui Tao
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Xiaoyan Zhu
- Department of Physiology, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Yong Li
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| |
Collapse
|
22
|
Verjans R, Peters T, Beaumont FJ, van Leeuwen R, van Herwaarden T, Verhesen W, Munts C, Bijnen M, Henkens M, Diez J, de Windt LJ, van Nieuwenhoven FA, van Bilsen M, Goumans MJ, Heymans S, González A, Schroen B. MicroRNA-221/222 Family Counteracts Myocardial Fibrosis in Pressure Overload-Induced Heart Failure. Hypertension 2017; 71:280-288. [PMID: 29255073 DOI: 10.1161/hypertensionaha.117.10094] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 08/21/2017] [Accepted: 11/29/2017] [Indexed: 02/06/2023]
Abstract
Pressure overload causes cardiac fibroblast activation and transdifferentiation, leading to increased interstitial fibrosis formation and subsequently myocardial stiffness, diastolic and systolic dysfunction, and eventually heart failure. A better understanding of the molecular mechanisms underlying pressure overload-induced cardiac remodeling and fibrosis will have implications for heart failure treatment strategies. The microRNA (miRNA)-221/222 family, consisting of miR-221-3p and miR-222-3p, is differentially regulated in mouse and human cardiac pathology and inversely associated with kidney and liver fibrosis. We investigated the role of this miRNA family during pressure overload-induced cardiac remodeling. In myocardial biopsies of patients with severe fibrosis and dilated cardiomyopathy or aortic stenosis, we found significantly lower miRNA-221/222 levels as compared to matched patients with nonsevere fibrosis. In addition, miRNA-221/222 levels in aortic stenosis patients correlated negatively with the extent of myocardial fibrosis and with left ventricular stiffness. Inhibition of both miRNAs during AngII (angiotensin II)-mediated pressure overload in mice led to increased fibrosis and aggravated left ventricular dilation and dysfunction. In rat cardiac fibroblasts, inhibition of miRNA-221/222 derepressed TGF-β (transforming growth factor-β)-mediated profibrotic SMAD2 (mothers against decapentaplegic homolog 2) signaling and downstream gene expression, whereas overexpression of both miRNAs blunted TGF-β-induced profibrotic signaling. We found that the miRNA-221/222 family may target several genes involved in TGF-β signaling, including JNK1 (c-Jun N-terminal kinase 1), TGF-β receptor 1 and TGF-β receptor 2, and ETS-1 (ETS proto-oncogene 1). Our findings show that heart failure-associated downregulation of the miRNA-221/222 family enables profibrotic signaling in the pressure-overloaded heart.
Collapse
Affiliation(s)
- Robin Verjans
- From the Department of Cardiology (R.V., T.P., R.v.L., W.V., M.H., L.J.d.W., S.H., B.S.), Department of Physiology (C.M., F.A.v.N., M.v.B.), and Department of Internal Medicine (M.B.), CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands; Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain (F.J.B., J.D., A.G.); Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (F.J.B., J.D., A.G.); CIBERCV, Carlos III Institute of Health, Madrid, Spain (F.J.B., J.D., A.G.); Department of Molecular Cell Biology, Leiden University Medical Biology Center, The Netherlands (T.v.H., M.J.G.); Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain (J.D.); Netherlands Heart Institute (ICIN), Utrecht; and Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (CMVB), KU Leuven, Belgium (S.H.)
| | - Tim Peters
- From the Department of Cardiology (R.V., T.P., R.v.L., W.V., M.H., L.J.d.W., S.H., B.S.), Department of Physiology (C.M., F.A.v.N., M.v.B.), and Department of Internal Medicine (M.B.), CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands; Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain (F.J.B., J.D., A.G.); Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (F.J.B., J.D., A.G.); CIBERCV, Carlos III Institute of Health, Madrid, Spain (F.J.B., J.D., A.G.); Department of Molecular Cell Biology, Leiden University Medical Biology Center, The Netherlands (T.v.H., M.J.G.); Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain (J.D.); Netherlands Heart Institute (ICIN), Utrecht; and Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (CMVB), KU Leuven, Belgium (S.H.)
| | - Francisco Javier Beaumont
- From the Department of Cardiology (R.V., T.P., R.v.L., W.V., M.H., L.J.d.W., S.H., B.S.), Department of Physiology (C.M., F.A.v.N., M.v.B.), and Department of Internal Medicine (M.B.), CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands; Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain (F.J.B., J.D., A.G.); Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (F.J.B., J.D., A.G.); CIBERCV, Carlos III Institute of Health, Madrid, Spain (F.J.B., J.D., A.G.); Department of Molecular Cell Biology, Leiden University Medical Biology Center, The Netherlands (T.v.H., M.J.G.); Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain (J.D.); Netherlands Heart Institute (ICIN), Utrecht; and Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (CMVB), KU Leuven, Belgium (S.H.)
| | - Rick van Leeuwen
- From the Department of Cardiology (R.V., T.P., R.v.L., W.V., M.H., L.J.d.W., S.H., B.S.), Department of Physiology (C.M., F.A.v.N., M.v.B.), and Department of Internal Medicine (M.B.), CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands; Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain (F.J.B., J.D., A.G.); Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (F.J.B., J.D., A.G.); CIBERCV, Carlos III Institute of Health, Madrid, Spain (F.J.B., J.D., A.G.); Department of Molecular Cell Biology, Leiden University Medical Biology Center, The Netherlands (T.v.H., M.J.G.); Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain (J.D.); Netherlands Heart Institute (ICIN), Utrecht; and Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (CMVB), KU Leuven, Belgium (S.H.)
| | - Tessa van Herwaarden
- From the Department of Cardiology (R.V., T.P., R.v.L., W.V., M.H., L.J.d.W., S.H., B.S.), Department of Physiology (C.M., F.A.v.N., M.v.B.), and Department of Internal Medicine (M.B.), CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands; Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain (F.J.B., J.D., A.G.); Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (F.J.B., J.D., A.G.); CIBERCV, Carlos III Institute of Health, Madrid, Spain (F.J.B., J.D., A.G.); Department of Molecular Cell Biology, Leiden University Medical Biology Center, The Netherlands (T.v.H., M.J.G.); Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain (J.D.); Netherlands Heart Institute (ICIN), Utrecht; and Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (CMVB), KU Leuven, Belgium (S.H.)
| | - Wouter Verhesen
- From the Department of Cardiology (R.V., T.P., R.v.L., W.V., M.H., L.J.d.W., S.H., B.S.), Department of Physiology (C.M., F.A.v.N., M.v.B.), and Department of Internal Medicine (M.B.), CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands; Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain (F.J.B., J.D., A.G.); Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (F.J.B., J.D., A.G.); CIBERCV, Carlos III Institute of Health, Madrid, Spain (F.J.B., J.D., A.G.); Department of Molecular Cell Biology, Leiden University Medical Biology Center, The Netherlands (T.v.H., M.J.G.); Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain (J.D.); Netherlands Heart Institute (ICIN), Utrecht; and Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (CMVB), KU Leuven, Belgium (S.H.)
| | - Chantal Munts
- From the Department of Cardiology (R.V., T.P., R.v.L., W.V., M.H., L.J.d.W., S.H., B.S.), Department of Physiology (C.M., F.A.v.N., M.v.B.), and Department of Internal Medicine (M.B.), CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands; Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain (F.J.B., J.D., A.G.); Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (F.J.B., J.D., A.G.); CIBERCV, Carlos III Institute of Health, Madrid, Spain (F.J.B., J.D., A.G.); Department of Molecular Cell Biology, Leiden University Medical Biology Center, The Netherlands (T.v.H., M.J.G.); Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain (J.D.); Netherlands Heart Institute (ICIN), Utrecht; and Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (CMVB), KU Leuven, Belgium (S.H.)
| | - Mitchell Bijnen
- From the Department of Cardiology (R.V., T.P., R.v.L., W.V., M.H., L.J.d.W., S.H., B.S.), Department of Physiology (C.M., F.A.v.N., M.v.B.), and Department of Internal Medicine (M.B.), CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands; Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain (F.J.B., J.D., A.G.); Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (F.J.B., J.D., A.G.); CIBERCV, Carlos III Institute of Health, Madrid, Spain (F.J.B., J.D., A.G.); Department of Molecular Cell Biology, Leiden University Medical Biology Center, The Netherlands (T.v.H., M.J.G.); Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain (J.D.); Netherlands Heart Institute (ICIN), Utrecht; and Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (CMVB), KU Leuven, Belgium (S.H.)
| | - Michiel Henkens
- From the Department of Cardiology (R.V., T.P., R.v.L., W.V., M.H., L.J.d.W., S.H., B.S.), Department of Physiology (C.M., F.A.v.N., M.v.B.), and Department of Internal Medicine (M.B.), CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands; Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain (F.J.B., J.D., A.G.); Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (F.J.B., J.D., A.G.); CIBERCV, Carlos III Institute of Health, Madrid, Spain (F.J.B., J.D., A.G.); Department of Molecular Cell Biology, Leiden University Medical Biology Center, The Netherlands (T.v.H., M.J.G.); Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain (J.D.); Netherlands Heart Institute (ICIN), Utrecht; and Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (CMVB), KU Leuven, Belgium (S.H.)
| | - Javier Diez
- From the Department of Cardiology (R.V., T.P., R.v.L., W.V., M.H., L.J.d.W., S.H., B.S.), Department of Physiology (C.M., F.A.v.N., M.v.B.), and Department of Internal Medicine (M.B.), CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands; Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain (F.J.B., J.D., A.G.); Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (F.J.B., J.D., A.G.); CIBERCV, Carlos III Institute of Health, Madrid, Spain (F.J.B., J.D., A.G.); Department of Molecular Cell Biology, Leiden University Medical Biology Center, The Netherlands (T.v.H., M.J.G.); Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain (J.D.); Netherlands Heart Institute (ICIN), Utrecht; and Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (CMVB), KU Leuven, Belgium (S.H.)
| | - Leon J de Windt
- From the Department of Cardiology (R.V., T.P., R.v.L., W.V., M.H., L.J.d.W., S.H., B.S.), Department of Physiology (C.M., F.A.v.N., M.v.B.), and Department of Internal Medicine (M.B.), CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands; Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain (F.J.B., J.D., A.G.); Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (F.J.B., J.D., A.G.); CIBERCV, Carlos III Institute of Health, Madrid, Spain (F.J.B., J.D., A.G.); Department of Molecular Cell Biology, Leiden University Medical Biology Center, The Netherlands (T.v.H., M.J.G.); Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain (J.D.); Netherlands Heart Institute (ICIN), Utrecht; and Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (CMVB), KU Leuven, Belgium (S.H.)
| | - Frans A van Nieuwenhoven
- From the Department of Cardiology (R.V., T.P., R.v.L., W.V., M.H., L.J.d.W., S.H., B.S.), Department of Physiology (C.M., F.A.v.N., M.v.B.), and Department of Internal Medicine (M.B.), CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands; Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain (F.J.B., J.D., A.G.); Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (F.J.B., J.D., A.G.); CIBERCV, Carlos III Institute of Health, Madrid, Spain (F.J.B., J.D., A.G.); Department of Molecular Cell Biology, Leiden University Medical Biology Center, The Netherlands (T.v.H., M.J.G.); Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain (J.D.); Netherlands Heart Institute (ICIN), Utrecht; and Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (CMVB), KU Leuven, Belgium (S.H.)
| | - Marc van Bilsen
- From the Department of Cardiology (R.V., T.P., R.v.L., W.V., M.H., L.J.d.W., S.H., B.S.), Department of Physiology (C.M., F.A.v.N., M.v.B.), and Department of Internal Medicine (M.B.), CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands; Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain (F.J.B., J.D., A.G.); Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (F.J.B., J.D., A.G.); CIBERCV, Carlos III Institute of Health, Madrid, Spain (F.J.B., J.D., A.G.); Department of Molecular Cell Biology, Leiden University Medical Biology Center, The Netherlands (T.v.H., M.J.G.); Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain (J.D.); Netherlands Heart Institute (ICIN), Utrecht; and Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (CMVB), KU Leuven, Belgium (S.H.)
| | - Marie José Goumans
- From the Department of Cardiology (R.V., T.P., R.v.L., W.V., M.H., L.J.d.W., S.H., B.S.), Department of Physiology (C.M., F.A.v.N., M.v.B.), and Department of Internal Medicine (M.B.), CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands; Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain (F.J.B., J.D., A.G.); Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (F.J.B., J.D., A.G.); CIBERCV, Carlos III Institute of Health, Madrid, Spain (F.J.B., J.D., A.G.); Department of Molecular Cell Biology, Leiden University Medical Biology Center, The Netherlands (T.v.H., M.J.G.); Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain (J.D.); Netherlands Heart Institute (ICIN), Utrecht; and Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (CMVB), KU Leuven, Belgium (S.H.)
| | - Stephane Heymans
- From the Department of Cardiology (R.V., T.P., R.v.L., W.V., M.H., L.J.d.W., S.H., B.S.), Department of Physiology (C.M., F.A.v.N., M.v.B.), and Department of Internal Medicine (M.B.), CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands; Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain (F.J.B., J.D., A.G.); Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (F.J.B., J.D., A.G.); CIBERCV, Carlos III Institute of Health, Madrid, Spain (F.J.B., J.D., A.G.); Department of Molecular Cell Biology, Leiden University Medical Biology Center, The Netherlands (T.v.H., M.J.G.); Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain (J.D.); Netherlands Heart Institute (ICIN), Utrecht; and Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (CMVB), KU Leuven, Belgium (S.H.)
| | - Arantxa González
- From the Department of Cardiology (R.V., T.P., R.v.L., W.V., M.H., L.J.d.W., S.H., B.S.), Department of Physiology (C.M., F.A.v.N., M.v.B.), and Department of Internal Medicine (M.B.), CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands; Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain (F.J.B., J.D., A.G.); Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (F.J.B., J.D., A.G.); CIBERCV, Carlos III Institute of Health, Madrid, Spain (F.J.B., J.D., A.G.); Department of Molecular Cell Biology, Leiden University Medical Biology Center, The Netherlands (T.v.H., M.J.G.); Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain (J.D.); Netherlands Heart Institute (ICIN), Utrecht; and Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (CMVB), KU Leuven, Belgium (S.H.)
| | - Blanche Schroen
- From the Department of Cardiology (R.V., T.P., R.v.L., W.V., M.H., L.J.d.W., S.H., B.S.), Department of Physiology (C.M., F.A.v.N., M.v.B.), and Department of Internal Medicine (M.B.), CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands; Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain (F.J.B., J.D., A.G.); Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (F.J.B., J.D., A.G.); CIBERCV, Carlos III Institute of Health, Madrid, Spain (F.J.B., J.D., A.G.); Department of Molecular Cell Biology, Leiden University Medical Biology Center, The Netherlands (T.v.H., M.J.G.); Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain (J.D.); Netherlands Heart Institute (ICIN), Utrecht; and Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (CMVB), KU Leuven, Belgium (S.H.).
| |
Collapse
|
23
|
Tsai ML, Tsai SP, Ho CT. Tetrahydrocurcumin attenuates carbon tetrachloride-induced hepatic fibrogenesis by inhibiting the activation and autophagy of hepatic stellate cells. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.07.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
24
|
Diethylcarbamazine attenuates the expression of pro-fibrogenic markers and hepatic stellate cells activation in carbon tetrachloride-induced liver fibrosis. Inflammopharmacology 2017; 26:599-609. [DOI: 10.1007/s10787-017-0329-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 02/17/2017] [Indexed: 01/26/2023]
|
25
|
Xu M, Wang X, Zou Y, Zhong Y. Key role of liver sinusoidal endothelial cells in liver fibrosis. Biosci Trends 2017; 11:163-168. [PMID: 28250338 DOI: 10.5582/bst.2017.01007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Because of the prevalence of viral hepatitis and nonalcoholic fatty liver disease (NAFLD), liver fibrosis has become a very common disease in Asia and elsewhere in the world, constantly increasing the burden of care borne by society. Hepatic sinusoidal capillarization, characterized by gradually shrinking fenestrae on the surface of liver sinusoidal endothelial cells (LSECs) and the formation of an organized basement membrane, is an initial pathologic change associated with liver fibrosis. Basic and clinical studies have indicated that LSECs play a key role in hepatic sinusoidal capillarization by affecting various aspects of the development and progression of liver fibrosis. Reviewing studies on the effect of LSECs on liver fibrosis is essential to better understanding the pathogenesis of liver fibrosis and its mechanism of progression. Moreover, such a review will provide a theoretical basis for identifying new methods to promote the regression or even inhibition of fibrosis. This review will focus on structural and functional changes in LSECs during hepatic sinusoidal capillarization and the interaction between the micro-environment of the liver and the body's immune system.
Collapse
Affiliation(s)
- Mingxing Xu
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Sun Yat-Sen University
| | - Xuehua Wang
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Sun Yat-Sen University
| | - Yong Zou
- Department of Blood Transfusion, Third Affiliated Hospital of Sun Yat-Sen University
| | - Yuesi Zhong
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Sun Yat-Sen University
| |
Collapse
|
26
|
Hui J, Gao J, Wang Y, Zhang J, Han Y, Wei L, Wu J. Panax notoginseng saponins ameliorates experimental hepatic fibrosis and hepatic stellate cell proliferation by inhibiting the Jak2/ Stat3 pathways. J TRADIT CHIN MED 2016; 36:217-24. [PMID: 27400477 DOI: 10.1016/s0254-6272(16)30030-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To investigate the inhibitory effect of Panax notoginseng saponins (PNS) on liver fibrosis and explore the underlying mechanisms. METHODS Carbon tetrachloride (CCl4)-treated rats and hepatic stellate cells (HSCs) were used. The effect of PNS on CCl4-induced liver fibrosis was studied with histochemical and biochemical analysis. Transforming growth factor (TGF)-β1, α-smooth muscle actin (α-SMA), and collagen I mRNA expression were determined by reverse transcripwhile, the protein expression levels of α-SMA, collagen I, phosphorylation-Janus activated kinase signal transducer (p-Jak2)/Jak2, and phosphorylation-activator of transcription (p-Stat)3/Stat3 were determined by immunohistochemistry and/or immunoblotting. RESULTS PNS treatment significantly improved the liver function of rats as indicated by decreased serum enzymatic activities of alanine aminotransferase and aspartate aminotransferase. Histopathological results indicated that PNS alleviated liver damage and reduced the formation of fibrous septa. Moreover, PNS significantly decreased liver hydroxyproline and significantly attenuated expressions of collagen I, α-SMA, TGF-β1, p-Jak2 / Jak2, and p-Stat3/Stat3 in the rat liver fibrosis model and HSCs. CONCLUSION PNS can relieve liver fibrosis by modulating Jak2/Stat3 signaling transduction pathway, which may be one of its mechanisms to suppress hepatic fibrosis.
Collapse
|
27
|
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
|
28
|
Ebrahimi H, Naderian M, Sohrabpour AA. New Concepts on Pathogenesis and Diagnosis of Liver Fibrosis; A Review Article. Middle East J Dig Dis 2016; 8:166-178. [PMID: 27698966 PMCID: PMC5045669 DOI: 10.15171/mejdd.2016.29] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Liver fibrosis is a potentially reversible response to hepatic insults, triggered by different chronic diseases most importantly viral hepatitis, alcoholic, and nonalcoholic fatty liver disease. In the course of the chronic liver disease, hepatic fibrogenesis may develop, which is attributed to various types of cells, molecules, and pathways. Activated hepatic stellate cell (HSC), the primary source of extracellular matrix (ECM), is fundamental in pathophysiology of fibrogenesis, and thus is the most attractable target for reversing liver fibrosis. Although, liver biopsy has long been considered as the gold standard for diagnosis and staging of hepatic fibrosis, assessing progression and regression by biopsy is hampered by its limitations. We provide recent views on noninvasive approaches including serum biomarkers and radiologic techniques.
Collapse
Affiliation(s)
- Hedyeh Ebrahimi
- Liver and Pancreaticobiliary Diseases Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran. Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Naderian
- Liver and Pancreaticobiliary Diseases Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran. Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ali Sohrabpour
- Assistant Professor, Liver and Pancreaticobiliary Diseases Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
TGF-β in Hepatic Stellate Cell Activation and Liver Fibrogenesis: Updated. CURRENT PATHOBIOLOGY REPORTS 2015. [DOI: 10.1007/s40139-015-0089-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
30
|
Abstract
Hepatocyte death, inflammation, and liver fibrosis are the hallmarks of chronic liver disease. Tumor necrosis factor-α (TNFα) is an inflammatory cytokine involved in liver inflammation and sustained liver inflammation leads to liver fibrosis. TNFα exerts inflammation, proliferation, and apoptosis. However, the role of TNFα signaling in liver fibrosis is not fully understood. This review highlights the recent findings demonstrating the molecular mechanisms of TNFα and its downstream signaling in liver fibrosis. During the progression of liver fibrosis, hepatic stellate cells play a pivotal role in a dynamic process of production of extracellular matrix proteins and modulation of immune response. Hepatic stellate cells transdifferentiate into activated myofibroblasts in response to damaged hepatocyte-derived mediators and immune cell-derived cytokines/chemokines. Here, we will discuss the role of TNFα in hepatic stellate cell survival and activation and the crosstalk between hepatic stellate cells and hepatocytes or other immune cells, such as macrophages, dendritic cells, and B cells in the development of liver fibrosis.
Collapse
Affiliation(s)
- Yoon Mee Yang
- Department of Medicine, Division of Gastroenterology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Ekihiro Seki
- Department of Medicine, Division of Gastroenterology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| |
Collapse
|
31
|
Harris TR, Bettaieb A, Kodani S, Dong H, Myers R, Chiamvimonvat N, Haj FG, Hammock BD. Inhibition of soluble epoxide hydrolase attenuates hepatic fibrosis and endoplasmic reticulum stress induced by carbon tetrachloride in mice. Toxicol Appl Pharmacol 2015; 286:102-11. [PMID: 25827057 DOI: 10.1016/j.taap.2015.03.022] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Revised: 03/18/2015] [Accepted: 03/20/2015] [Indexed: 12/26/2022]
Abstract
Liver fibrosis is a pathological condition in which chronic inflammation and changes to the extracellular matrix lead to alterations in hepatic tissue architecture and functional degradation of the liver. Inhibitors of the enzyme soluble epoxide hydrolase (sEH) reduce fibrosis in the heart, pancreas and kidney in several disease models. In this study, we assess the effect of sEH inhibition on the development of fibrosis in a carbon tetrachloride (CCl4)-induced mouse model by monitoring changes in the inflammatory response, matrix remolding and endoplasmic reticulum stress. The sEH inhibitor 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU) was administered in drinking water. Collagen deposition in the liver was increased five-fold in the CCl4-treated group, and this was returned to control levels by TPPU treatment. Hepatic expression of Col1a2 and 3a1 mRNA was increased over fifteen-fold in the CCl4-treated group relative to the Control group, and this increase was reduced by 50% by TPPU treatment. Endoplasmic reticulum (ER) stress observed in the livers of CCl4-treated animals was attenuated by TPPU treatment. In order to support the hypothesis that TPPU is acting to reduce the hepatic fibrosis and ER stress through its action as a sEH inhibitor we used a second sEH inhibitor, trans-4-{4-[3-(4-trifluoromethoxy-phenyl)-ureido]-cyclohexyloxy}-benzoic acid (t-TUCB), and sEH null mice. Taken together, these data indicate that the sEH may play an important role in the development of hepatic fibrosis induced by CCl4, presumably by reducing endogenous fatty acid epoxide chemical mediators acting to reduce ER stress.
Collapse
Affiliation(s)
- Todd R Harris
- Department of Entomology and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - Ahmed Bettaieb
- Department of Nutrition, University of California, Davis, CA 95616, USA
| | - Sean Kodani
- Department of Entomology and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - Hua Dong
- Department of Entomology and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - Richard Myers
- Department of Internal Medicine: Cardiovascular, University of California, Davis, CA 95616, USA
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine: Cardiovascular, University of California, Davis, CA 95616, USA
| | - Fawaz G Haj
- Department of Nutrition, University of California, Davis, CA 95616, USA; Department of Internal Medicine: Endocrinology, Diabetes and Metabolism, University of California, Davis, CA 95616, USA
| | - Bruce D Hammock
- Department of Entomology and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA.
| |
Collapse
|