1
|
Ibn ElAchaouia Y, Mahmoudi A, Dallali D, Marrakchi R, Marti G, Chamkha M, Sayadi S, Noureddine A. UHPLC-HRMS-MS analysis, antioxidant and hepatoprotective effects of methanolic extracts from Ficus carica L., leaves and stem barks against acute CCl 4-induced liver damage. Nat Prod Res 2025:1-10. [PMID: 40314183 DOI: 10.1080/14786419.2025.2498075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 04/10/2025] [Accepted: 04/18/2025] [Indexed: 05/03/2025]
Abstract
Methanolic leaf (LME) and stem bark (SBME) extracts from Ficus carica L., were analysed using UHPLC-HRMS-MS analysis. Fifty-two compounds were identified belonging to flavonoids, phenolic acids and alkaloids. Their hepatoprotective activity was tested in a CCl4 rat model. In the CCl4-treated groups, the investigated extracts improved body weight (LME: 245.7 and SBME: 236.5 g) and relative liver weight (LME: 3.9 and SBME: 3.6 mg/g). They also reduced levels of aspartate aminotransferase (LME: 102.6 and SBME: 205.3.0 (UI/L), alanine aminotransferase (LME: 170.3 and SBME: 260.6 UI/L), lactate dehydrogenase (LME: 281.6 and SBME: 445.3 UI/L) and MDA (LME: 1.4 and SBME: 2.1 U/mg protein), while enhancing superoxide dismutase (LME: 65.1 and 46.2 U/mg protein) and catalase (LME: 11.0 and SBME: 8.3 U/mg protein) activities. These findings, together with histological examination, imply that the studied methanolic extracts protect the liver against steatosis, fibrosis and inflammation by increasing the antioxidative defense.
Collapse
Affiliation(s)
- Yosra Ibn ElAchaouia
- Laboratory of Organic Chemistry LR17-ES08 (Natural Substances Team), Faculty of Sciences of Sfax, University of Sfax, Sfax, Tunisia
| | - Asma Mahmoudi
- Laboratory of Environmental Bioprocesses, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Dhouha Dallali
- Laboratory of Organic Chemistry LR17-ES08 (Natural Substances Team), Faculty of Sciences of Sfax, University of Sfax, Sfax, Tunisia
| | - Rim Marrakchi
- Biochemistry Laboratory, CHU Hédi Chaker Sfax, Sfax, Tunisia
| | - Guillaume Marti
- Laboratoire de Recherche en Sciences Végétales, Metatoul-AgromiX Platform, Université de Toulouse, CNRS, INP, Auzeville, Auzeville-Tolosane, France
- MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
| | - Mohamed Chamkha
- Laboratory of Environmental Bioprocesses, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Sami Sayadi
- Center for Sustainable Development, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Allouche Noureddine
- Laboratory of Organic Chemistry LR17-ES08 (Natural Substances Team), Faculty of Sciences of Sfax, University of Sfax, Sfax, Tunisia
| |
Collapse
|
2
|
Bowen CM, Ditmars F, Liu N, Abril JM, Ajasin D, Russell WK, Stevenson H, Eugenin EA, Fair JH, Fagg WS. Amniotic Fluid Reduces Liver Fibrosis By Attenuating Hepatic Stellate Cell Activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.20.639215. [PMID: 40027749 PMCID: PMC11870538 DOI: 10.1101/2025.02.20.639215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Regardless of the source of injury or metabolic dysfunction, fibrosis is a frequent driver of liver pathology. Excessive liver fibrosis is caused by persistent activation of hepatic stellate cells (HSCs), which is defined by myofibroblast activation (MFA) and the epithelial-mesenchymal transition (EMT). Strategies to prevent or reverse this HSC phenotype will be critical for successful treatment of liver fibrosis. We have previously shown that full-term, cell-free human amniotic fluid (cfAF) inhibits MFA and EMT in fibroblasts in vitro. We hypothesize that cfAF treatment can attenuate HSC activation and limit liver fibrosis. We tested if cfAF could prevent liver fibrosis or HSC activation in murine models of liver damage, three-dimensional hepatic spheroids, and HSC cultures. Administering cfAF prevented weight loss and the extent of fibrosis in mice with chronic liver damage without stimulating deleterious immune responses. Gene expression profiling and immunostaining indicated that cfAF administration in carbon tetrachloride-treated mice reduced EMT- and MFA-related biomarker abundance and modulated transcript levels associated with liver metabolism, immune regulatory pathways, and cell signaling. cfAF treatment lowered MFA biomarker levels in a dose-dependent manner in hepatic spheroids exposed to ethanol. Treating HSCs with cfAF in vitro strongly repressed EMT. Multi-omics analyses revealed that it also attenuates TGFβ-induced MFA and inflammation-associated processes. Thus, cfAF treatment prevents liver fibrosis by safeguarding against persistent HSC activation. These findings suggest that cfAF may be a safe and effective therapy for reducing liver fibrosis and preventing the development of cirrhosis and/or hepatocellular carcinoma.
Collapse
Affiliation(s)
- Charles M. Bowen
- Division of Transplant, Department of Surgery, University of Texas Medical Branch, Galveston, Texas, 77555, USA
- John Sealy School of Medicine, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Frederick Ditmars
- Division of Transplant, Department of Surgery, University of Texas Medical Branch, Galveston, Texas, 77555, USA
- John Sealy School of Medicine, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Naiyou Liu
- Division of Transplant, Department of Surgery, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Jose Marri Abril
- Division of Transplant, Department of Surgery, University of Texas Medical Branch, Galveston, Texas, 77555, USA
- John Sealy School of Medicine, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - David Ajasin
- Department of Neurobiology, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - William K. Russell
- Deparment of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Heather Stevenson
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Eliseo A. Eugenin
- Department of Neurobiology, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Jeffrey H. Fair
- Division of Transplant, Department of Surgery, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - W. Samuel Fagg
- Division of Transplant, Department of Surgery, University of Texas Medical Branch, Galveston, Texas, 77555, USA
- Deparment of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, 77555, USA
- Merakris Therapeutics, RTP Frontier, Research Triangle Park, North Carolina, 27709, USA
| |
Collapse
|
3
|
Sohail A, Shams F, Nawaz A, Ain QU, Ijaz B. Antifibrotic potential of reserpine (alkaloid) targeting Keap1/Nrf2; oxidative stress pathway in CCl 4-induced liver fibrosis. Chem Biol Interact 2025; 407:111384. [PMID: 39800144 DOI: 10.1016/j.cbi.2025.111384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/24/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
The death rate due to liver cancer approaches 2 million annually, the majority is attributed to fibrosis. Currently, there is no efficient, safe, non-toxic, and anti-fibrotic drug available, suggesting room for better drug discovery. The current study aims to evaluate the anti-fibrotic role of reserpine, an alkaloid plant compound against CCl4-induced liver fibrosis. In-silico docking analysis showed the interaction of reserpine with keap1 protein with the binding energy -9.0 kcal/mol. In-vitro, biochemical analysis, anti-oxidative indexes, and inflammatory cytokines analysis were performed in HepG2 cells. The non-toxic nature of the compound (<100 μg/ml) was evaluated through MTT assay in HepG2 and Vero cell lines. The antifibrotic potential of the reserpine compound (dose of 0.5 mg/kg) was assessed in CCl4-administered C57BL/6J mice models. Hematoxylin & Eosin and Masson staining were performed to study the morphological changes of liver tissues. Immune histochemistry (IHC) analysis was performed to evaluate the effect of reserpine on the liver fibrosis marker. The biochemical assay indicated a significant decrease in ALT, AST, and MDA levels and increased catalase enzyme post-6-week reserpine treatment in mice models. Gene expression analysis revealed that the reserpine targets oxidative stress Keap1/Nrf2 pathway and down-regulated Keap1 expression by 5-fold and up-regulated Nrf2 and Nqo1 expression by 6 and 4.5-fold respectively showing its antioxidant response. It suppressed the expression of Cyp2e1 by 2.2-fold, illustrating the compound's ability to block lipid peroxidation. Histological and immunostaining exhibited improved hepatocyte morphology and reduced collagen deposition in liver tissues due to reserpine. Reserpine treatment lowered the fibrotic markers α-SMA and Col-1 by 1.3 and 1.5 folds respectively as compared to the control group and increased the expression of miR-200a and miR-29b by 15.5 and 8.2 folds (p < 0.05) while decreased miR-128-1-5p expression by 5-fold. A comprehensive In-silico, In-vitro, and In-vivo analysis revealed that reserpine has a strong anti-fibrotic effect against the CCl4-induced liver fibrosis in C57BL/6J mice model by targeting the Keap1/Nrf2 pathway.
Collapse
Affiliation(s)
- Aamir Sohail
- Centre of Excellence in Molecular Biology, University of the Punjab, Thokar Niaz Baig, Lahore, 53700, Pakistan
| | - Faiza Shams
- Centre of Excellence in Molecular Biology, University of the Punjab, Thokar Niaz Baig, Lahore, 53700, Pakistan
| | - Aleeza Nawaz
- Centre of Excellence in Molecular Biology, University of the Punjab, Thokar Niaz Baig, Lahore, 53700, Pakistan
| | - Qurrat Ul Ain
- Department of Medical Laboratory Technology, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Bushra Ijaz
- Centre of Excellence in Molecular Biology, University of the Punjab, Thokar Niaz Baig, Lahore, 53700, Pakistan; Applied and Functional Genomics Lab, Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan.
| |
Collapse
|
4
|
Bae AN, Lee H, Yang H, Mukherjee S, Im SS, Lee JH, Park JH. Enhancing Regulatory T cell function by mevalonate pathway inhibition prevents liver fibrosis. Biochem Biophys Res Commun 2025; 742:151094. [PMID: 39632293 DOI: 10.1016/j.bbrc.2024.151094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Liver fibrosis is a well-established risk factor for liver cancer development. Despite extensive mechanistic studies on liver fibrosis, the role of the immune cell network in fibrotic disease remains poorly understood. In this study, we demonstrate that regulatory T cells (Tregs) are involved in preventing liver fibrosis by regulating the mevalonate pathway. Blocking the mevalonate pathway increased the granzyme B secretion from Tregs, while restoring the pathway reduced it. Statin treatment, which inhibits the mevalonate pathway, alleviated liver fibrosis progression and enhanced the immunosuppressive function of Tregs in vivo. Mechanistically, mevalonate products, including geranylgeranyl pyrophosphate, inhibited the phosphorylation and activation of LKB1, that is a key regulator of Treg homeostasis. Furthermore, these products disrupted the interaction between LKB1 and cAMP-dependent protein kinase (PKA), leading to further reduction of LKB1 phosphorylation. These findings suggest that targeting LKB1 in Tregs through statin treatment prevents the progression of liver fibrosis, offering a promising and safe therapeutic strategy for liver disease and liver cancer.
Collapse
Affiliation(s)
- An-Na Bae
- Department of Anatomy, School of Medicine, Keimyung University, Daegu, South Korea
| | - Hajin Lee
- Department of Anatomy, School of Medicine, Keimyung University, Daegu, South Korea
| | - Huiseong Yang
- Department of Anatomy, School of Medicine, Keimyung University, Daegu, South Korea
| | - Sulagna Mukherjee
- Department of Physiology, School of Medicine, Keimyung University, Daegu, South Korea
| | - Seung-Soon Im
- Department of Physiology, School of Medicine, Keimyung University, Daegu, South Korea
| | - Jae-Ho Lee
- Department of Anatomy, School of Medicine, Keimyung University, Daegu, South Korea
| | - Jong Ho Park
- Department of Anatomy, School of Medicine, Keimyung University, Daegu, South Korea.
| |
Collapse
|
5
|
Benderski K, Schneider P, Kordeves P, Fichter M, Schunke J, De Lorenzi F, Durak F, Schrörs B, Akilli Ö, Kiessling F, Bros M, Diken M, Grabbe S, Schattenberg JM, Lammers T, Sofias AM, Kaps L. A hepatocellular carcinoma model with and without parenchymal liver damage that integrates technical and pathophysiological advantages for therapy testing. Pharmacol Res 2025; 211:107560. [PMID: 39730106 DOI: 10.1016/j.phrs.2024.107560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Hepatocellular Carcinoma (HCC) is the most common form of primary liver cancer, with cirrhosis being its strongest risk factor. Interestingly, an increasing number of HCC cases is also observed without cirrhosis. We developed an HCC model via intrasplenic injection of highly tumorigenic HCC cells, which, due to cellular tropism, invade the liver and allow for a controllable disease progression. Specifically, C57BL/6JRj mice were intrasplenically inoculated with Dt81Hepa1-6 HCC cells, with a subgroup pre-treated with CCl4 to induce cirrhosis (C-HCC). At four weeks post-inoculation, mice were sacrificed, and diseased livers were analyzed via histology, flow cytometry, and RT-qPCR to profile the extracellular matrix (ECM), angiogenesis, and immune cells. In addition, tumor-bearing mice were treated with the first-line therapy, AtezoBev, to assess therapeutic responsiveness of the model. Dt81Hepa1-6 cells displayed similar gene expression as human HCC. After intrasplenic injection, all mice developed multifocal disease. C-HCC mice had a significantly higher tumor load than non-cirrhotic HCC mice. Both HCC and C-HCC models displayed extensive ECM formation, increased levels of vascularization, and immune cell infiltration compared to healthy and non-cancerous cirrhotic livers. AtezoBev treatment produced robust antitumor efficacy, validating the model's suitability for therapy testing. In conclusion, we established a rapidly developing and high-yield HCC model through a simple intrasplenic injection, with or without cirrhotic damage. The model overexpressed key human HCC genes and showed high responsiveness to first-line treatment. Our model uniquely combines all the above-mentioned features, promoting its use towards HCC therapy testing.
Collapse
Affiliation(s)
- Karina Benderski
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Forckenbeckstrasse 55, Aachen 52074, Germany
| | - Paul Schneider
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, Mainz 55131, Germany
| | - Panayiotis Kordeves
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Forckenbeckstrasse 55, Aachen 52074, Germany
| | - Michael Fichter
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, Mainz 55131, Germany; Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz 55128, Germany
| | - Jenny Schunke
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, Mainz 55131, Germany; TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University Mainz GmbH, Freiligrathstrasse 12, Mainz 55131, Germany
| | - Federica De Lorenzi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Forckenbeckstrasse 55, Aachen 52074, Germany; Mildred Scheel School of Oncology (MSSO), Center for Integrated Oncology Aachen (CIOA), RWTH Aachen University Hospital, Pauwelsstrasse 30, Aachen 52074, Germany
| | - Feyza Durak
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University Mainz GmbH, Freiligrathstrasse 12, Mainz 55131, Germany
| | - Barbara Schrörs
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University Mainz GmbH, Freiligrathstrasse 12, Mainz 55131, Germany
| | - Özlem Akilli
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University Mainz GmbH, Freiligrathstrasse 12, Mainz 55131, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Forckenbeckstrasse 55, Aachen 52074, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, Mainz 55131, Germany
| | - Mustafa Diken
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University Mainz GmbH, Freiligrathstrasse 12, Mainz 55131, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, Mainz 55131, Germany
| | - Jörn M Schattenberg
- Department of Medicine II, Saarland University Medical Center, Saarland University, Kirrberger Strasse 100, Saarbrücken 66123, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Forckenbeckstrasse 55, Aachen 52074, Germany; Mildred Scheel School of Oncology (MSSO), Center for Integrated Oncology Aachen (CIOA), RWTH Aachen University Hospital, Pauwelsstrasse 30, Aachen 52074, Germany.
| | - Alexandros Marios Sofias
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Forckenbeckstrasse 55, Aachen 52074, Germany; Mildred Scheel School of Oncology (MSSO), Center for Integrated Oncology Aachen (CIOA), RWTH Aachen University Hospital, Pauwelsstrasse 30, Aachen 52074, Germany.
| | - Leonard Kaps
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, Mainz 55131, Germany; Department of Medicine II, Saarland University Medical Center, Saarland University, Kirrberger Strasse 100, Saarbrücken 66123, Germany.
| |
Collapse
|
6
|
Kim D, Sung M, Park M, Sun E, Yoon S, Yoo KH, Radhakrishnan K, Jung SY, Bae W, Cho S, Chung I. Galectin 3-binding protein (LGALS3BP) depletion attenuates hepatic fibrosis by reducing transforming growth factor-β1 (TGF-β1) availability and inhibits hepatocarcinogenesis. Cancer Commun (Lond) 2024; 44:1106-1129. [PMID: 39073023 PMCID: PMC11483554 DOI: 10.1002/cac2.12600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 07/04/2024] [Accepted: 07/19/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Increased Galectin 3-binding protein (LGALS3BP) serum levels have been used to assess hepatic fibrosis stages and the severity of hepatocellular carcinoma (HCC). Considering the crucial role of transforming growth factor-β1 (TGF-β1) in the emergence of these diseases, the present study tested the hypothesis that LGALS3BP regulates the TGF-β1 signaling pathway. METHODS The expression levels of LGALS3BP and TGFB1 were analyzed in patients with metabolic dysfunction-associated steatohepatitis (MASH) and HCC. Multiple omics techniques, such as RNA-sequencing, transposase-accessible chromatin-sequencing assay, and liquid chromatography-tandem mass spectrometry proteomics, were used to identify the regulatory mechanisms for the LGALS3BP-TGF-β1 axis. The effects of altered TGF-β1 signaling by LGALS3BP were investigated in conditional LGALS3BP-knockin and LGALS3BP-knockout mice. RESULTS In patients with MASH and HCC, the levels of LGALS3BP and TGFB1 exhibited positive correlations. Stimulation of LGALS3BP by the inflammatory cytokine interferon α in HCC cells or ectopic overexpression of LGALS3BP in hepatocytes promoted the expression levels of TGFB1. Aggravated fibrosis was observed in the livers of hepatocyte-specific LGALS3BP-knockin mice, with increased TGFB1 levels. LGALS3BP directly bound to and assembled integrin αV, an integral mediator required for releasing active TGF-β1 from extracellular latent complex with the rearranged F-actin cytoskeleton. The released TGF-β1 activated JunB transcription factor, which in turn promoted the TGF-β1 positive feedback loop. LGALS3BP deletion in the hepatocytes downregulated TGF-β1 signaling and CCl4 induced fibrosis. Moreover, LGALS3BP depletion hindered hepatocarcinogenesis by limiting the availability of fibrogenic TGF-β1. CONCLUSION LGALS3BP plays a crucial role in hepatic fibrosis and carcinogenesis by controlling the TGF-β1 signaling pathway, making it a promising therapeutic target in TGF-β1-related diseases.
Collapse
Affiliation(s)
- Dae‐Hwan Kim
- Department of Internal MedicineDivision of Hematology and OncologyChonnam National University Medical SchoolHwasunSouth Korea
- Department of Internal MedicineDivision of Hematology and OncologyChonnam National University Hwasun HospitalHwasunSouth Korea
- Combinatorial Tumor Immunotherapy MRC CenterChonnam National University Medical SchoolHwasunSouth Korea
- National Immunotherapy Innovation CenterHwasunSouth Korea
| | - Minjeong Sung
- Department of Internal MedicineDivision of Hematology and OncologyChonnam National University Medical SchoolHwasunSouth Korea
- Department of Internal MedicineDivision of Hematology and OncologyChonnam National University Hwasun HospitalHwasunSouth Korea
- Combinatorial Tumor Immunotherapy MRC CenterChonnam National University Medical SchoolHwasunSouth Korea
- National Immunotherapy Innovation CenterHwasunSouth Korea
- BioMedical Sciences Graduate ProgramChonnam National UniversityHwasunSouth Korea
| | - Myong‐Suk Park
- Department of Internal MedicineDivision of Hematology and OncologyChonnam National University Medical SchoolHwasunSouth Korea
- Department of Internal MedicineDivision of Hematology and OncologyChonnam National University Hwasun HospitalHwasunSouth Korea
- Combinatorial Tumor Immunotherapy MRC CenterChonnam National University Medical SchoolHwasunSouth Korea
- National Immunotherapy Innovation CenterHwasunSouth Korea
| | - Eun‐Gene Sun
- Department of Internal MedicineDivision of Hematology and OncologyChonnam National University Medical SchoolHwasunSouth Korea
- Department of Internal MedicineDivision of Hematology and OncologyChonnam National University Hwasun HospitalHwasunSouth Korea
- National Immunotherapy Innovation CenterHwasunSouth Korea
| | - Sumin Yoon
- Department of Biological ScienceSookmyung Women's UniversitySeoulSouth Korea
| | - Kyung Hyun Yoo
- Department of Biological ScienceSookmyung Women's UniversitySeoulSouth Korea
| | | | - Sung Yun Jung
- Department of Biochemistry and Molecular PharmacologyBaylor College of MedicineHoustonTexasUSA
| | - Woo‐Kyun Bae
- Department of Internal MedicineDivision of Hematology and OncologyChonnam National University Medical SchoolHwasunSouth Korea
- Department of Internal MedicineDivision of Hematology and OncologyChonnam National University Hwasun HospitalHwasunSouth Korea
- Combinatorial Tumor Immunotherapy MRC CenterChonnam National University Medical SchoolHwasunSouth Korea
- National Immunotherapy Innovation CenterHwasunSouth Korea
- BioMedical Sciences Graduate ProgramChonnam National UniversityHwasunSouth Korea
| | - Sang‐Hee Cho
- Department of Internal MedicineDivision of Hematology and OncologyChonnam National University Medical SchoolHwasunSouth Korea
- Department of Internal MedicineDivision of Hematology and OncologyChonnam National University Hwasun HospitalHwasunSouth Korea
- Combinatorial Tumor Immunotherapy MRC CenterChonnam National University Medical SchoolHwasunSouth Korea
- National Immunotherapy Innovation CenterHwasunSouth Korea
| | - Ik‐Joo Chung
- Department of Internal MedicineDivision of Hematology and OncologyChonnam National University Medical SchoolHwasunSouth Korea
- Department of Internal MedicineDivision of Hematology and OncologyChonnam National University Hwasun HospitalHwasunSouth Korea
- Combinatorial Tumor Immunotherapy MRC CenterChonnam National University Medical SchoolHwasunSouth Korea
- National Immunotherapy Innovation CenterHwasunSouth Korea
| |
Collapse
|
7
|
Smith ML, Wade JB, Wolstenholme J, Bajaj JS. Gut microbiome-brain-cirrhosis axis. Hepatology 2024; 80:465-485. [PMID: 36866864 PMCID: PMC10480351 DOI: 10.1097/hep.0000000000000344] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/10/2023] [Indexed: 03/04/2023]
Abstract
Cirrhosis is characterized by inflammation, degeneration, and fibrosis of liver tissue. Along with being the most common cause of liver failure and liver transplant, cirrhosis is a significant risk factor for several neuropsychiatric conditions. The most common of these is HE, which is characterized by cognitive and ataxic symptoms, resulting from the buildup of metabolic toxins with liver failure. However, cirrhosis patients also show a significantly increased risk for neurodegenerative diseases such as Alzheimer and Parkinson diseases, and for mood disorders such as anxiety and depression. In recent years, more attention has been played to communication between the ways the gut and liver communicate with each other and with the central nervous system, and the way these organs influence each other's function. This bidirectional communication has come to be known as the gut-liver-brain axis. The gut microbiome has emerged as a key mechanism affecting gut-liver, gut-brain, and brain-liver communication. Clinical studies and animal models have demonstrated the significant patterns of gut dysbiosis when cirrhosis is present, both with or without concomitant alcohol use disorder, and have provided compelling evidence that this dysbiosis also influences the cognitive and mood-related behaviors. In this review, we have summarized the pathophysiological and cognitive effects associated with cirrhosis, links to cirrhosis-associated disruption of the gut microbiome, and the current evidence from clinical and preclinical studies for the modulation of the gut microbiome as a treatment for cirrhosis and associated neuropsychiatric conditions.
Collapse
Affiliation(s)
- Maren L Smith
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
- Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - James B Wade
- Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jennifer Wolstenholme
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
- Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jasmohan S Bajaj
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University and Central Virginia Veterans Healthcare System, Richmond, Virginia, USA
| |
Collapse
|
8
|
Xin S, Liu X, He C, Gao H, Wang B, Hua R, Gao L, Shang H, Sun F, Xu J. Inflammation accelerating intestinal fibrosis: from mechanism to clinic. Eur J Med Res 2024; 29:335. [PMID: 38890719 PMCID: PMC11184829 DOI: 10.1186/s40001-024-01932-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/08/2024] [Indexed: 06/20/2024] Open
Abstract
Intestinal fibrosis is a prevalent complication of IBD that that can frequently be triggered by prolonged inflammation. Fibrosis in the gut can cause a number of issues, which continue as an ongoing challenge to healthcare systems worldwide. The primary causes of intestinal fibrosis are soluble molecules, G protein-coupled receptors, epithelial-to-mesenchymal or endothelial-to-mesenchymal transition, and the gut microbiota. Fresh perspectives coming from in vivo and in vitro experimental models demonstrate that fibrogenic pathways might be different, at least to some extent, independent of the ones that influence inflammation. Understanding the distinctive procedures of intestinal fibrogenesis should provide a realistic foundation for targeting and blocking specific fibrogenic pathways, estimating the risk of fibrotic consequences, detecting early fibrotic alterations, and eventually allowing therapy development. Here, we first summarize the inflammatory and non-inflammatory components of fibrosis, and then we elaborate on the underlying mechanism associated with multiple cytokines in fibrosis, providing the framework for future clinical practice. Following that, we discuss the relationship between modernization and disease, as well as the shortcomings of current studies. We outline fibrosis diagnosis and therapy, as well as our recommendations for the future treatment of intestinal fibrosis. We anticipate that the global review will provides a wealth of fresh knowledge and suggestions for future fibrosis clinical practice.
Collapse
Affiliation(s)
- Shuzi Xin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xiaohui Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Chengwei He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Han Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Department of Clinical Laboratory, Aerospace Clinical Medical College, Aerospace Central Hospital, Beijing, 100039, China
| | - Boya Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Rongxuan Hua
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Lei Gao
- Department of Intelligent Medical Engineering, School of Biomedical Engineering, Capital Medical University, Beijing, 100069, China
| | - Hongwei Shang
- Experimental Center for Morphological Research Platform, Capital Medical University, Beijing, 100069, China
| | - Fangling Sun
- Department of Laboratory Animal Research, Xuan Wu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Jingdong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
9
|
Wahid RM, Hassan NH, Samy W, Abdelhadi AA, Saadawy SF, Elsayed SF, Seada SG, Mohamed SRA. Unraveling the hepatic stellate cells mediated mechanisms in aging's influence on liver fibrosis. Sci Rep 2024; 14:13473. [PMID: 38866800 PMCID: PMC11169484 DOI: 10.1038/s41598-024-63644-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/30/2024] [Indexed: 06/14/2024] Open
Abstract
Aging enhances numerous processes that compromise homeostasis and pathophysiological processes. Among these, activated HSCs play a pivotal role in advancing liver fibrosis. This research delved into how aging impacts liver fibrosis mechanisms. The study involved 32 albino rats categorized into four groups: Group I (young controls), Group II (young with liver fibrosis), Group III (old controls), and Group IV (old with liver fibrosis). Various parameters including serum ALT, adiponectin, leptin, and cholesterol levels were evaluated. Histopathological analysis was performed, alongside assessments of TGF-β, FOXP3, and CD133 gene expressions. Markers of fibrosis and apoptosis were the highest in group IV. Adiponectin levels significantly decreased in Group IV compared to all other groups except Group II, while cholesterol levels were significantly higher in liver fibrosis groups than their respective control groups. Group III displayed high hepatic expression of desmin, α-SMA, GFAP and TGF- β and in contrast to Group I. Increased TGF-β and FOXP3 gene expressions were observed in Group IV relative to Group II, while CD133 gene expression decreased in Group IV compared to Group II. In conclusion, aging modulates immune responses, impairs regenerative capacities via HSC activation, and influences adipokine and cholesterol levels, elevating the susceptibility to liver fibrosis.
Collapse
Affiliation(s)
- Reham M Wahid
- Medical Physiology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Nancy Husseiny Hassan
- Human Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Walaa Samy
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Amina A Abdelhadi
- Medical Microbiology and Immunology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Sara F Saadawy
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Sherein F Elsayed
- Medical Physiology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Sara G Seada
- Medical Physiology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | | |
Collapse
|
10
|
Reches G, Khoon L, Ghanayiem N, Malka A, Piran R. Controlling autoimmune diabetes onset by targeting Protease-Activated Receptor 2. Biomed Pharmacother 2024; 175:116622. [PMID: 38653114 DOI: 10.1016/j.biopha.2024.116622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is a challenging autoimmune disease, characterized by an immune system assault on insulin-producing β-cells. As insulin facilitates glucose absorption into cells and tissues, β-cell deficiency leads to elevated blood glucose levels on one hand and target-tissues starvation on the other. Despite efforts to halt β-cell destruction and stimulate recovery, success has been limited. Our recent investigations identified Protease-Activated Receptor 2 (Par2) as a promising target in the battle against autoimmunity. We discovered that Par2 activation's effects depend on its initial activation site: exacerbating the disease within the immune system but fostering regeneration in affected tissue. METHODS We utilized tissue-specific Par2 knockout mice strains with targeted Par2 mutations in β-cells, lymphocytes, and the eye retina (as a control) in the NOD autoimmune diabetes model, examining T1D onset and β-cell survival. RESULTS We discovered that Par2 expression within the immune system accelerates autoimmune processes, while its presence in β-cells offers protection against β-cell destruction and T1D onset. This suggests a dual-strategy treatment for T1D: inhibiting Par2 in the immune system while activating it in β-cells, offering a promising strategy for T1D. CONCLUSIONS This study highlights Par2's potential as a drug target for autoimmune diseases, particularly T1D. Our results pave the way for precision medicine approaches in treating autoimmune conditions through targeted Par2 modulation.
Collapse
Affiliation(s)
- Gal Reches
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Lynn Khoon
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | | | - Assaf Malka
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Ron Piran
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.
| |
Collapse
|
11
|
Reches G, Piran R. Par2-mediated responses in inflammation and regeneration: choosing between repair and damage. Inflamm Regen 2024; 44:26. [PMID: 38816842 PMCID: PMC11138036 DOI: 10.1186/s41232-024-00338-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 05/19/2024] [Indexed: 06/01/2024] Open
Abstract
The protease activated receptor 2 (Par2) plays a pivotal role in various damage models, influencing injury, proliferation, inflammation, and regeneration. Despite extensive studies, its binary roles- EITHER aggravating injury or promoting recovery-make a conclusive translational decision on its modulation strategy elusive. Analyzing two liver regeneration models, autoimmune hepatitis and direct hepatic damage, we discovered Par2's outcome depends on the injury's nature. In immune-mediated injury, Par2 exacerbates damage, while in direct tissue injury, it promotes regeneration. Subsequently, we evaluated the clinical significance of this finding by investigating Par2's expression in the context of autoimmune diabetes. We found that the absence of Par2 in all lymphocytes provided full protection against the autoimmune destruction of insulin-producing β-cells in mice, whereas the introduction of a β-cell-specific Par2 null mutation accelerated the onset of autoimmune diabetes. This pattern led us to hypothesize whether these observations are universal. A comprehensive review of recent Par2 publications across tissues and systems confirms the claim drafted above: Par2's initial activation in the immune system aggravates inflammation, hindering recovery, whereas its primary activation in the damaged tissue fosters regeneration. As a membrane-anchored receptor, Par2 emerges as an attractive drug target. Our findings highlight a crucial translational modulation strategy in regenerative medicine based on injury type.
Collapse
Affiliation(s)
- Gal Reches
- The Azrieli Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed, Israel
| | - Ron Piran
- The Azrieli Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed, Israel.
| |
Collapse
|
12
|
Repáraz D, Casares N, Fuentes A, Navarro F. Establishment of a murine hepatocellular carcinoma model by hydrodynamic injection and characterization of the immune tumor microenvironment. Methods Cell Biol 2024; 185:79-97. [PMID: 38556453 DOI: 10.1016/bs.mcb.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent malignant neoplasms. Current treatments for HCC, such as tyrosine kinase inhibitors, have limited efficacy, highlighting the urgent need for better therapies. Immunotherapies, including anti-programmed death receptor 1 (PD-1) and anti-Cytotoxic T-lymphocyte associated protein 4 (CTLA-4), and more recently, the combination of anti-PD-L1 and anti-vascular endothelial growth factor (VEGF) monoclonal antibodies, have shown efficacy against HCC, resulting in Food and Drug Administration (FDA) approval. However, these immunotherapies only show efficiency in a small proportion of patients, meaning there is a great need to improve and optimize treatments against HCC. Accurate animal models that mimic human HCC are necessary to help better understand the nature of these tumors, which in turn will allow the development and testing of new treatments. Existing pre-clinical HCC models can be divided into non-genetic and genetic models. Non-genetic models involve implanting human or murine HCC cell lines or inducing tumors using chemical compounds or dietary modifications. These models have limitations, including slow tumor development and a lack of resemblance to human HCC. Genetic models, on the other hand, manipulate gene expression to induce HCC in mice and provide a better understanding of the effects of specific genes on tumor development. One method commonly used to generate HCC is hydrodynamic tail vein injection (HTVI), which consists of the delivery of oncogenes directly to the liver, resulting in expression and subsequent hepatocyte transformation. Usually, Sleeping Beauty transposase-containing plasmids are used to achieve stable and long-term gene expression. Once the HCC tumor is generated, and a proper tumor microenvironment (TME) is established, it is important to study the immune compartment of the TME, which plays a crucial role in HCC development and response to treatment. Techniques like flow cytometry can be used to analyze the immune cell populations in HCC tumors and assess their impact on tumor development and survival in mice. In this article, we thoroughly describe an example of the methodology to successfully generate HCC murine models via HTVI, and we propose a way to characterize the immune TME by flow cytometry.
Collapse
Affiliation(s)
- David Repáraz
- Radio-Oncology Department, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland.
| | - Noelia Casares
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Navarra, Spain
| | - Andrea Fuentes
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Navarra, Spain
| | - Flor Navarro
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Navarra, Spain.
| |
Collapse
|
13
|
Hong JG, Carbajal Y, Trotman J, Glass M, Sclar V, Alter IL, Zhang P, Wang L, Chen L, Petitjean M, Friedman SL, DeRossi C, Chu J. Mannose Supplementation Curbs Liver Steatosis and Fibrosis in Murine MASH by Inhibiting Fructose Metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.17.576067. [PMID: 38293175 PMCID: PMC10827199 DOI: 10.1101/2024.01.17.576067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) can progress to cirrhosis and liver cancer. There are no approved medical therapies to prevent or reverse disease progression. Fructose and its metabolism in the liver play integral roles in MASH pathogenesis and progression. Here we focus on mannose, a simple sugar, which dampens hepatic stellate cell activation and mitigates alcoholic liver disease in vitro and in vivo . In the well-validated FAT-MASH murine model, oral mannose supplementation improved both liver steatosis and fibrosis at low and high doses, whether administered either at the onset of the model ("Prevention") or at week 6 of the 12-week MASH regimen ("Reversal"). The in vivo anti-fibrotic effects of mannose supplementation were validated in a second model of carbon tetrachloride-induced liver fibrosis. In vitro human and mouse primary hepatocytes revealed that the anti-steatotic effects of mannose are dependent on the presence of fructose, which attenuates expression of ketohexokinase (KHK), the main enzyme in fructolysis. KHK is decreased with mannose supplementation in vivo and in vitro, and overexpression of KHK abrogated the anti-steatotic effects of mannose. Our study identifies mannose as a simple, novel therapeutic candidate for MASH that mitigates metabolic dysregulation and exerts anti-fibrotic effects.
Collapse
|
14
|
Kim YE, Bak SB, Kim MJ, Bae SJ, Lee WY, Kim YW. Forsythiaside A Activates AMP-Activated Protein Kinase and Regulates Oxidative Stress via Nrf2 Signaling. Int J Mol Sci 2023; 24:17033. [PMID: 38069356 PMCID: PMC10707188 DOI: 10.3390/ijms242317033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Forsythiaside A (FA) is an active constituent isolated from Forsythia suspensa, a beneficial herb used in traditional medicine known for its antioxidant and anti-inflammatory properties. Although various studies have suggested that FA has the protective effects, its impacts on arachidonic acid (AA) plus iron in vitro models and carbon tetrachloride (CCl₄)-induced mouse liver damage in vivo have not been explored. In this study, HepG2 cells were subjected to AA + iron treatment to induce apoptosis and mitochondrial impairment and determine the molecular mechanisms. FA exhibited protective effects by inhibiting cell damage and reactive oxygen species (ROS) production induced by AA + iron, as assessed via immunoblot and flow cytometry analyses. Further molecular investigations revealed that FA resulted in the activation of extracellular-signal-related protein kinase (ERK), which subsequently triggered the activation of AMP-activated protein kinase (AMPK), a critical regulator of cellular oxidative stress. Additionally, FA modulated the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway, which is a significant antioxidant transcription factor regulated by the AMPK pathway. For in vivo studies, mice were orally administered FA and then subjected to induction of CCl₄-based hepatotoxicity. The protective effect of FA was confirmed via blood biochemistry and immunohistochemical analyses. In conclusion, our findings demonstrated the protective effects of FA against oxidative stress both in vitro and in vivo, thus indicating that FA is a potential candidate for liver protection. Our study sheds light on the mechanistic pathways involved in the antioxidant effects of FA, highlighting the hepatoprotective potential of naturally occurring compounds in traditional herbs, such as FA.
Collapse
Affiliation(s)
- Young Eun Kim
- AI-Bio Convergence DDI Basic Research Lab., School of Korean Medicine, Dongguk University, Gyeongju 38066, Republic of Korea; (Y.E.K.); (S.-B.B.); (M.-J.K.); (S.-J.B.); (W.-Y.L.)
| | - Seon-Been Bak
- AI-Bio Convergence DDI Basic Research Lab., School of Korean Medicine, Dongguk University, Gyeongju 38066, Republic of Korea; (Y.E.K.); (S.-B.B.); (M.-J.K.); (S.-J.B.); (W.-Y.L.)
| | - Min-Jin Kim
- AI-Bio Convergence DDI Basic Research Lab., School of Korean Medicine, Dongguk University, Gyeongju 38066, Republic of Korea; (Y.E.K.); (S.-B.B.); (M.-J.K.); (S.-J.B.); (W.-Y.L.)
| | - Su-Jin Bae
- AI-Bio Convergence DDI Basic Research Lab., School of Korean Medicine, Dongguk University, Gyeongju 38066, Republic of Korea; (Y.E.K.); (S.-B.B.); (M.-J.K.); (S.-J.B.); (W.-Y.L.)
- College of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea
| | - Won-Yung Lee
- AI-Bio Convergence DDI Basic Research Lab., School of Korean Medicine, Dongguk University, Gyeongju 38066, Republic of Korea; (Y.E.K.); (S.-B.B.); (M.-J.K.); (S.-J.B.); (W.-Y.L.)
- College of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea
| | - Young Woo Kim
- AI-Bio Convergence DDI Basic Research Lab., School of Korean Medicine, Dongguk University, Gyeongju 38066, Republic of Korea; (Y.E.K.); (S.-B.B.); (M.-J.K.); (S.-J.B.); (W.-Y.L.)
| |
Collapse
|
15
|
Jing H, Ren Y, Zhou Y, Xu M, Krizkova S, Heger Z, Lu Q, Wang S, Liang X, Adam V, Li N. Remodeling of the liver fibrosis microenvironment based on nilotinib-loaded multicatalytic nanozymes with boosted antifibrogenic activity. Acta Pharm Sin B 2023; 13:5030-5047. [PMID: 38045041 PMCID: PMC10692490 DOI: 10.1016/j.apsb.2023.08.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/07/2023] [Accepted: 07/15/2023] [Indexed: 12/05/2023] Open
Abstract
Liver fibrosis is a reversible pathological process caused by chronic liver damage and a major risk factor for hepatocellular carcinoma (HCC). Hepatic stellate cell (HSC) activation is considered the main target for liver fibrosis therapy. However, the efficiency of this strategy is limited due to the complex microenvironment of liver fibrosis, including excessive extracellular matrix (ECM) deposition and hypoxia-induced imbalanced ECM metabolism. Herein, nilotinib (NIL)-loaded hyaluronic acid (HA)-coated Ag@Pt nanotriangular nanozymes (APNH NTs) were developed to inhibit HSCs activation and remodel the microenvironment of liver fibrosis. APNH NTs efficiently eliminated intrahepatic reactive oxygen species (ROS) due to their inherent superoxide dismutase (SOD) and catalase (CAT) activities, thereby downregulating the expression of NADPH oxidase-4 (NOX-4) and inhibiting HSCs activation. Simultaneously, the oxygen produced by the APNH NTs further alleviated the hypoxic microenvironment. Importantly, the released NIL promoted collagen depletion by suppressing the expression of tissue inhibitor of metalloproteinase-1 (TIMP-1), thus synergistically remodeling the microenvironment of liver fibrosis. Notably, an in vivo study in CCl4-induced mice revealed that APNH NTs exhibited significant antifibrogenic effects without obvious long-term toxicity. Taken together, the data from this work suggest that treatment with the synthesized APNH NTs provides an enlightening strategy for remodeling the microenvironment of liver fibrosis with boosted antifibrogenic activity.
Collapse
Affiliation(s)
- Huaqing Jing
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Yingzi Ren
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Yue Zhou
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Min Xu
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Sona Krizkova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno 61300, Czech Republic
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno 61300, Czech Republic
| | - Qiang Lu
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Siyu Wang
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Xiaoyang Liang
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno 61300, Czech Republic
| | - Nan Li
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| |
Collapse
|
16
|
Kim MJ, Song YR, Kim YE, Bae SJ, Lee WY, Bak SB, Kim YW. Kaempferol stimulation of autophagy regulates the ferroptosis under the oxidative stress as mediated with AMP-activated protein kinase. Free Radic Biol Med 2023; 208:630-642. [PMID: 37703935 DOI: 10.1016/j.freeradbiomed.2023.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/06/2023] [Accepted: 09/10/2023] [Indexed: 09/15/2023]
Abstract
Recent studies have highlighted the positive effects of Kaempferol (KP), including its anti-inflammatory and antioxidant properties. However, its impact on oxidative damage induced by heavy metals and pro-inflammatory mediators, such as arachidonic acid (AA), has not yet been identified. Our objective was to specifically evaluate liver damage due to AA + iron-induced oxidative stress, both in vitro and in vivo. In HepG2 cells, KP activated the AMP-activated protein kinase (AMPK), suggesting a hepatoprotective effect through AMPK inhibition, as assessed by immunoblot and FACS analysis (EC50 = 10 μM). KP also stimulated autophagy, a degradation process that eliminates aged, damaged, and unnecessary components, via mTOR inhibition and ULK1 phosphorylation. This activation was further validated by the upregulation of autophagy-related genes (ATG5) and Beclin-1, along with the conversion of LC3BI to LC3BII. Ferroptosis, a non-apoptotic type of cell death characterized by oxidative stress from the production of reactive oxygen species (ROS) and excessive iron accumulation, was linked to the activation of autophagy, as confirmed through the protein expression of deferoxamine (DFO) and ferrostatin-1 (Fer-1), the representative ferroptosis inhibitors (positive controls). In mice, oral administration of KP demonstrated protective effects against CCl4-induced hepatotoxicity. In conclusion, KP provides hepatoprotective effects against oxidative stress induced by AA + iron treatment in vitro and CCl4 treatment in vivo.
Collapse
Affiliation(s)
- Min-Jin Kim
- AI-Bio Convergence DDI Basic Research Lab., School of Korean Medicine, Dongguk University, Goyang-si, Gyeonggi-do, South Korea
| | - Yu-Rim Song
- AI-Bio Convergence DDI Basic Research Lab., School of Korean Medicine, Dongguk University, Goyang-si, Gyeonggi-do, South Korea
| | - Young Eun Kim
- AI-Bio Convergence DDI Basic Research Lab., School of Korean Medicine, Dongguk University, Goyang-si, Gyeonggi-do, South Korea
| | - Su-Jin Bae
- AI-Bio Convergence DDI Basic Research Lab., School of Korean Medicine, Dongguk University, Goyang-si, Gyeonggi-do, South Korea
| | - Won-Yung Lee
- AI-Bio Convergence DDI Basic Research Lab., School of Korean Medicine, Dongguk University, Goyang-si, Gyeonggi-do, South Korea; College of Korean Medicine, Wonkwang University, Iksan-si, South Korea
| | - Seon-Been Bak
- AI-Bio Convergence DDI Basic Research Lab., School of Korean Medicine, Dongguk University, Goyang-si, Gyeonggi-do, South Korea
| | - Young Woo Kim
- AI-Bio Convergence DDI Basic Research Lab., School of Korean Medicine, Dongguk University, Goyang-si, Gyeonggi-do, South Korea.
| |
Collapse
|
17
|
Sun C, Zhou C, Daneshvar K, Kratkiewicz AJ, Saad AB, Hess A, Chen JY, Pondick JV, York SR, Li W, Moran S, Gentile S, Rahman RU, Li Z, Sparks R, Habboub T, Kim BM, Choi MY, Affo S, Schwabe RF, Popov YV, Mullen AC. Conserved long noncoding RNA TILAM promotes liver fibrosis through interaction with PML in hepatic stellate cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.29.551032. [PMID: 37546982 PMCID: PMC10402143 DOI: 10.1101/2023.07.29.551032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Background & Aims Fibrosis is the common endpoint for all forms of chronic liver injury, and progression of fibrosis leads to the development of end-stage liver disease. Activation of hepatic stellate cells (HSCs) and their transdifferentiation to myofibroblasts results in the accumulation of extracellular matrix (ECM) proteins that form the fibrotic scar. Long noncoding (lnc) RNAs regulate the activity of HSCs and may provide targets for fibrotic therapies. Methods We identified lncRNA TILAM as expressed near COL1A1 in human HSCs and performed loss-of-function studies in human HSCs and liver organoids. Transcriptomic analyses of HSCs isolated from mice defined the murine ortholog of TILAM . We then generated Tilam -deficient GFP reporter mice and quantified fibrotic responses to carbon tetrachloride (CCl 4 ) and choline-deficient L-amino acid defined high fat diet (CDA-HFD). Co-precipitation studies, mass spectrometry, and gene expression analyses identified protein partners of TILAM . Results TILAM is conserved between human and mouse HSCs and regulates expression of ECM proteins, including collagen. Tilam is selectively induced in HSCs during the development of fibrosis in vivo . In both male and female mice, loss of Tilam results in reduced fibrosis in the setting of CCl 4 and CDA-HFD injury models. TILAM interacts with promyelocytic leukemia protein (PML) to stabilize PML protein levels and promote the fibrotic activity of HSCs. Conclusion TILAM is activated in HSCs and interacts with PML to drive the development of liver fibrosis. Depletion of TILAM may serve as a therapeutic approach to combat the development of end stage liver disease.
Collapse
|
18
|
Krishnan RP, Pandiar D, Ramani P. Sclerosing Variant of Adenoid Cystic Carcinoma - A Case Report on the Role of Sclerosis in the Prognostic Outcome. Ann Maxillofac Surg 2023; 13:248-251. [PMID: 38405569 PMCID: PMC10883206 DOI: 10.4103/ams.ams_116_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/22/2023] [Accepted: 10/19/2023] [Indexed: 02/27/2024] Open
Abstract
Rationale Sclerosing variant of adenoid cystic carcinoma (SACC) is a rare variant of adenoid cystic carcinoma (ACC) with only two cases in the literature. In this article, we report two cases of SACCs and attempt to explain the reason for sclerosis and its association with the disease outcome. Patient Concerns The first patient is a 43-year-old Dravidian Indian female with a chief complaint of pain and swelling in the right posterior cheek region of four months' duration. The second patient is an 8-year-old boy with a well-defined swelling in the left cheek region. Diagnosis Both these cases were diagnosed as sclerosing variants of ACC. Treatment The tumour was surgically excised for both patients. Outcomes Both the patients had no signs of residual disease/recurrence. Take-away Lessons We opine that the dense sclerotic stroma may have a preventing role in tumour cell growth and progression.
Collapse
Affiliation(s)
- Reshma Poothakulath Krishnan
- Department of Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Deepak Pandiar
- Department of Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Pratibha Ramani
- Department of Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| |
Collapse
|
19
|
Tong H, Wang L, Zhang K, Shi J, Wu Y, Bao Y, Wang C. S100A6 Activates Kupffer Cells via the p-P38 and p-JNK Pathways to Induce Inflammation, Mononuclear/macrophage Infiltration Sterile Liver Injury in Mice. Inflammation 2023; 46:534-554. [PMID: 36484925 DOI: 10.1007/s10753-022-01750-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/17/2022] [Accepted: 09/30/2022] [Indexed: 12/13/2022]
Abstract
Noninfectious liver injury, including the effects of chemical material, drugs and diet, is a major cause of liver diseases worldwide. In chemical and drugs-induced liver injury, innate inflammatory responses are mediated by extracellular danger signals. The S100 protein can act as danger signals, which can promote the migration and chemotaxis of immune cells, promote the release of various inflammatory cytokines, and regulate the body's inflammatory and immune responses. However, the role of S100A6 in inflammatory response in chemical and drugs-induced sterile liver injury remains unclear. We constructed the model of sterile liver injury induced by carbon tetrachloride (CCl4)/Paracetamol (APAP) and performed RNA sequencing (RNA-seq) on the liver tissues after injury (days 2 and 5). We analyzed inflammatory protein secretion in the liver tissue supernatant by enzyme-linked immunosorbent assay (ELISA), determined the inflammation response by bioinformatic analysis during sterile liver injury, and assessed mononuclear/macrophage infiltration by immunohistochemistry and flow cytometry. Immunohistochemistry was used to analyze the location of S100A6. We conducted inflammatory factor expression analysis and molecular mechanistic studies in Kupffer cells (KCs) induced by S100A6 using quantitative reverse transcription-polymerase chain reaction (qRT-PCR), ELISA, and western blot in vitro experiments. We performed chemokine CCL2 expression analysis and molecular mechanism studies using the same method. We used a Transwell assay to show the infiltration of mononuclear/macrophage. We here observed that aggravated inflammatory response was shown in CCl4 and APAP-administrated mice, as evidenced by enhanced production of inflammatory cytokines (TNF-α, IL-1β), and elevated mononuclear/macrophage infiltration and activation of immunity. The expression of S100A6 was significantly increased on day 2 after sterile liver injury, which is primarily produced by injured liver cells. Mechanistic studies established that S100A6 activates Kupffer cells (KCs) via the p-P38, p-JNK and P65 pathways to induce inflammation in vitro. Furthermore, TNF-α can stimulate liver cells via the p-P38 and p-JNK pathways to produce CCL2 and promote the infiltration of mononuclear/macrophage. In summary, we showed that S100A6 plays an important role in regulating inflammation, thus influencing sterile liver injury. Our findings provide novel evidence that S100A6 can as a danger signal that contributes to pro-inflammatory activation through p-P38 and p-JNK pathways in CCl4 and APAP-induced sterile liver injury in mice. In addition, the inflammatory factor TNF-α induces a large amount of CCL2 production in normal liver cells surrounding the injured area through a paracrine action, which is chemotactic for blood mononuclear/macrophage infiltration.
Collapse
Affiliation(s)
- He Tong
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China
| | - Li Wang
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, Inner Mongolia, China
| | - Kefan Zhang
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China
| | - Jing Shi
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China
| | - Yongshuai Wu
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China
| | - Yulong Bao
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, Inner Mongolia, China.
| | - Changshan Wang
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China.
| |
Collapse
|
20
|
Abdelsalam MM, El-Mahdy N, Abou-Saif S. Direct-acting antivirals sofosbuvir and daclatasvir attenuate carbon tetrachloride-induced liver fibrosis in mice. LIVER RESEARCH 2023; 7:71-81. [PMID: 39959700 PMCID: PMC11791913 DOI: 10.1016/j.livres.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/14/2022] [Accepted: 02/02/2023] [Indexed: 03/16/2023]
Abstract
Background and aim Advanced liver fibrosis is a major risk for developing hepatocellular carcinoma (HCC) in chronic hepatitis C virus (HCV) patients. Direct-acting antivirals (DAAs) which are used for treating HCV infection, produce more than 90% cure rate but do not seem to diminish the rate of occurrence or recurrence of HCC. This study aimed to investigate the effect of DAAs sofosbuvir (SOF) and daclatasvir (DAC) on carbon tetrachloride (CCl4)-induced fibrotic changes in mice. Methods Eighty adult male Swiss albino mice were randomly allocated into 8 groups (10 mice/group): normal control group, SOF group (receiving SOF 80 mg/kg body weight (BW), oral gavage, daily), DAC group (receiving DAC 30 mg/kg BW, oral gavage, daily), SOF + DAC group (receiving a combination of both, daily), CCl4 model group (receiving CCl4 2 mL/kg BW, intraperitoneal twice weekly) and three CCl4-intoxicated groups receiving either SOF or DAC or their combination. All CCl4 groups received CCl4 for 12 weeks followed by DAAs for another 12 weeks. Results CCl4-induced a significant elevation of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), and produced histopathological evidence of fibrosis and liver degeneration along with a significant increase (P ≤ 0.001) of the proliferation markers (proliferating cell nuclear antigen (PCNA) and Ki-67), hepatic stellate cells (HSCs) activation markers (alpha-smooth muscle actin (α-SMA) and glial fibrillary acidic protein (GFAP)), fibrosis marker (matrix metalloproteinase-9 (MMP-9)) and pro-inflammatory cytokine (tumor necrosis factor-alpha (TNF-α)). CCl4-intoxicated mice treated with SOF, DAC, or their combination revealed a significant amelioration (P ≤ 0.001) of CCl4-induced elevation of liver enzymes, fibrotic changes, and liver degeneration along with a significant attenuation (P ≤ 0.001) of CCl4-induced upregulation of all tested markers. The effects of SOF, DAC, and their combination on liver enzymes were comparable while the effect of SOF + DAC combination on mitigating CCl4-induced upregulation of the proliferation and HSCs activation markers was significantly stronger than either SOF or DAC alone. As for MMP-9 and TNF-α, the effects of DAC and SOF + DAC combination were comparable and both were more significant than that of SOF alone. Conclusions SOF and DAC may possess an antifibrotic effect that is independent of their role as antiviral agents against CCl4-induced liver injury. This might exclude the role of DAAs in early occurrence or accelerated recurrence of HCC through the progression of the HCV patients' pre-existing fibrosis. However, HCC patients treated with DAAs should be closely monitored with continuous HCC surveillance during and post-therapy.
Collapse
Affiliation(s)
- Mayadah M. Abdelsalam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Nageh El-Mahdy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Sabry Abou-Saif
- Department of Tropical Medicine and Infectious Diseases, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
21
|
The anti-toxic effect of the date palm fruit extract loaded on chitosan nanoparticles against CCl 4-induced liver fibrosis in a mouse model. Int J Biol Macromol 2023; 235:123804. [PMID: 36842736 DOI: 10.1016/j.ijbiomac.2023.123804] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 02/12/2023] [Accepted: 02/18/2023] [Indexed: 02/28/2023]
Abstract
The liver is the most important organ in the body. Hepatocyte oxidative damage occurs to excess ROS. Liver fibrosis is a mechanism that the immune system uses to treat extreme inflammation by repairing damaged tissue with the creation of a scar. The outcome of fibrosis may be reversed by consuming natural plant extracts with high ROS-scavenging ability. The date palm fruits contain caffeic acid, gallic acid, syringic acid, and ferulic acid, which have anti-inflammatory, antioxidant, and hepatoprotective properties. This study aimed to prepare a date fruit extract, load it onto chitosan nanoparticles, and compare its anti-fibrotic activity with the unloaded crude extract in the CCl4-mouse model. Our findings show that nanocomposite (Cs@FA/DEx) has anti-fibrotic properties and can improve liver function enzymes and endogenous antioxidant enzymes by inhibiting cell apoptosis caused by CCl4-induction in mice. Furthermore, significantly reduced CD95 and ICAM1 levels and down-regulation of TGFβ-1 and collagen-α-1 expression demonstrated the anti-fibrotic effects of the Cs@FA/DEx. Therefore, the Cs@FA/DEx might be an innovative supplement for inhibiting liver fibrosis and hepatocyte inflammation induced by chemical toxins. Besides, this nano-supplement could be a promising anti-hepatocellular carcinoma agent as it has potent in vitro anticancer activity against the HePG2 cell line.
Collapse
|
22
|
Oates JR, Sawada K, Giles DA, Alarcon PC, Damen MS, Szabo S, Stankiewicz TE, Moreno-Fernandez ME, Divanovic S. Thermoneutral housing shapes hepatic inflammation and damage in mouse models of non-alcoholic fatty liver disease. Front Immunol 2023; 14:1095132. [PMID: 36875069 PMCID: PMC9982161 DOI: 10.3389/fimmu.2023.1095132] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/27/2023] [Indexed: 02/19/2023] Open
Abstract
Introduction Inflammation is a common unifying factor in experimental models of non-alcoholic fatty liver disease (NAFLD) progression. Recent evidence suggests that housing temperature-driven alterations in hepatic inflammation correlate with exacerbated hepatic steatosis, development of hepatic fibrosis, and hepatocellular damage in a model of high fat diet-driven NAFLD. However, the congruency of these findings across other, frequently employed, experimental mouse models of NAFLD has not been studied. Methods Here, we examine the impact of housing temperature on steatosis, hepatocellular damage, hepatic inflammation, and fibrosis in NASH diet, methionine and choline deficient diet, and western diet + carbon tetrachloride experimental models of NAFLD in C57BL/6 mice. Results We show that differences relevant to NAFLD pathology uncovered by thermoneutral housing include: (i) augmented NASH diet-driven hepatic immune cell accrual, exacerbated serum alanine transaminase levels and increased liver tissue damage as determined by NAFLD activity score; (ii) augmented methionine choline deficient diet-driven hepatic immune cell accrual and increased liver tissue damage as indicated by amplified hepatocellular ballooning, lobular inflammation, fibrosis and overall NAFLD activity score; and (iii) dampened western diet + carbon tetrachloride driven hepatic immune cell accrual and serum alanine aminotransferase levels but similar NAFLD activity score. Discussion Collectively, our findings demonstrate that thermoneutral housing has broad but divergent effects on hepatic immune cell inflammation and hepatocellular damage across existing experimental NAFLD models in mice. These insights may serve as a foundation for future mechanistic interrogations focused on immune cell function in shaping NAFLD progression.
Collapse
Affiliation(s)
- Jarren R. Oates
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Keisuke Sawada
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Daniel A. Giles
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Pablo C. Alarcon
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Michelle S.M.A. Damen
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Sara Szabo
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Pathology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Traci E. Stankiewicz
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Maria E. Moreno-Fernandez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
23
|
Reches G, Blondheim Shraga NR, Carrette F, Malka A, Saleev N, Gubbay Y, Ertracht O, Haviv I, Bradley LM, Levine F, Piran R. Resolving the conflicts around Par2 opposing roles in regeneration by comparing immune-mediated and toxic-induced injuries. Inflamm Regen 2022; 42:52. [PMID: 36447218 PMCID: PMC9706915 DOI: 10.1186/s41232-022-00238-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 11/09/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Different factors may lead to hepatitis. Among which are liver inflammation and poisoning. We chose two hepatitis models, typical for these two underlying causes. Thus, we aimed to characterize the role of protease-activated receptor 2 (Par2) in liver regeneration and inflammation to reconcile Par2 conflicting role in many damage models, which sometimes aggravates the induced damage and sometimes alleviates it. METHODS WT and knockout (Par2KO) mice were injected with concanavalin A (ConA) to induce immune-mediated hepatitis or with carbon tetrachloride (CCl4) to elicit direct hepatic damage. To distinguish the immune component from the liver regenerative response, we conducted bone marrow (BM) replacements of WT and Par2KO mice and repeated the damage models. RESULTS ConA injection caused limited damage in Par2KO mice livers, while in the WT mice severe damage followed by leukocyte infiltration was evident. Reciprocal BM replacement of WT and Par2KO showed that WT BM-reconstituted Par2KO mice displayed marked liver damage, while in Par2KO BM-reconstituted WT mice, the tissue was generally protected. In the CCl4 direct damage model, hepatocytes regenerated in WT mice, whereas Par2KO mice failed to recover. Reciprocal BM replacement did not show significant differences in hepatic regeneration. In Par2KO mice, hepatitis was more apparent, while WT recovered regardless of the BM origin. CONCLUSIONS We conclude that Par2 activation in the immune system aggravates hepatitis and that Par2 activation in the damaged tissue promotes liver regeneration. When we incorporate this finding and revisit the literature reports, we reconciled the conflicts surrounding Par2's role in injury, recovery, and inflammation.
Collapse
Affiliation(s)
- Gal Reches
- grid.22098.310000 0004 1937 0503The Azrieli Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed, Israel
| | - Netta R. Blondheim Shraga
- grid.22098.310000 0004 1937 0503The Azrieli Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed, Israel
| | - Florent Carrette
- grid.479509.60000 0001 0163 8573Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Assaf Malka
- grid.22098.310000 0004 1937 0503The Azrieli Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed, Israel
| | - Natalia Saleev
- grid.22098.310000 0004 1937 0503The Azrieli Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed, Israel
| | - Yehuda Gubbay
- grid.22098.310000 0004 1937 0503The Azrieli Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed, Israel
| | - Offir Ertracht
- grid.415839.2Eliachar Research Laboratory, Galilee Medical Center, Nahariya, Israel
| | - Izhak Haviv
- grid.22098.310000 0004 1937 0503The Azrieli Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed, Israel
| | - Linda M. Bradley
- grid.479509.60000 0001 0163 8573Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Fred Levine
- grid.479509.60000 0001 0163 8573Sanford Children’s Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037 USA
| | - Ron Piran
- grid.22098.310000 0004 1937 0503The Azrieli Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed, Israel
| |
Collapse
|
24
|
Elzoheiry A, Ayad E, Omar N, Elbakry K, Hyder A. Anti-liver fibrosis activity of curcumin/chitosan-coated green silver nanoparticles. Sci Rep 2022; 12:18403. [PMID: 36319750 PMCID: PMC9626641 DOI: 10.1038/s41598-022-23276-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 10/27/2022] [Indexed: 11/24/2022] Open
Abstract
Liver fibrosis results from the hepatic accumulation of the extracellular matrix accompanied by a failure of the mechanisms responsible for matrix dissolution. Pathogenesis of liver fibrosis is associated with many proteins from different cell types. In the present study, in silico molecular docking analysis revealed that curcumin may inhibit the fibrosis-mediating proteins PDGF, PDGFRB, TIMP-1, and TLR-9 by direct binding. Nano-formulation can overcome curcumin problems, increasing the efficacy of curcumin as a drug by maximizing its solubility and bioavailability, enhancing its membrane permeability, and improving its pharmacokinetics, pharmacodynamics and biodistribution. Therefore, green silver nanoparticles (AgNPs) were synthesized in the presence of sunlight by means of the metabolite of Streptomyces malachiticus, and coated with curcumin-chitosan mixture to serve as a drug delivery tool for curcumin to target CCl4-induced liver fibrosis mouse model. Fibrosis induction significantly increased hepatic gene expression of COL1A1, α-SMA, PDGFRB, and TIMP1, elevated hepatic enzymes, increased histopathological findings, and increased collagen deposition as determined by Mason's trichrome staining. Treatment with naked AgNPs tended to increase these inflammatory effects, while their coating with chitosan, similar to treatment with curcumin only, did not prevent the fibrogenic effect of CCl4. The induction of liver fibrosis was reversed by concurrent treatment with curcumin/chitosan-coated AgNPs. In this nano form, curcumin was found to be efficient as anti-liver fibrosis drug, maintaining the hepatic architecture and function during fibrosis development. This efficacy can be attributed to its inhibitory role through a direct binding to fibrosis-mediating proteins such as PDGFRB, TIMP-1, TLR-9 and TGF-β.
Collapse
Affiliation(s)
- Alya Elzoheiry
- Zoology Department, Faculty of Science, Damietta University, New Damietta, Egypt
| | - Esraa Ayad
- Zoology Department, Faculty of Science, Damietta University, New Damietta, Egypt
| | - Nahed Omar
- Zoology Department, Faculty of Science, Damietta University, New Damietta, Egypt
| | - Kadry Elbakry
- Zoology Department, Faculty of Science, Damietta University, New Damietta, Egypt
| | - Ayman Hyder
- Faculty of Science, Damietta University, New Damietta, 34517, Egypt.
| |
Collapse
|
25
|
Ramani K, Mavila N, Abeynayake A, Tomasi ML, Wang J, Matsuda M, Seki E. Targeting A-kinase anchoring protein 12 phosphorylation in hepatic stellate cells regulates liver injury and fibrosis in mouse models. eLife 2022; 11:e78430. [PMID: 36193675 PMCID: PMC9531947 DOI: 10.7554/elife.78430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 08/03/2022] [Indexed: 12/24/2022] Open
Abstract
Trans-differentiation of hepatic stellate cells (HSCs) to activated state potentiates liver fibrosis through release of extracellular matrix (ECM) components, distorting the liver architecture. Since limited antifibrotics are available, pharmacological intervention targeting activated HSCs may be considered for therapy. A-kinase anchoring protein 12 (AKAP12) is a scaffolding protein that directs protein kinases A/C (PKA/PKC) and cyclins to specific locations spatiotemporally controlling their biological effects. It has been shown that AKAP12's scaffolding functions are altered by phosphorylation. In previously published work, observed an association between AKAP12 phosphorylation and HSC activation. In this work, we demonstrate that AKAP12's scaffolding activity toward the endoplasmic reticulum (ER)-resident collagen chaperone, heat-shock protein 47 (HSP47) is strongly inhibited by AKAP12's site-specific phosphorylation in activated HSCs. CRISPR-directed gene editing of AKAP12's phospho-sites restores its scaffolding toward HSP47, inhibiting HSP47's collagen maturation functions, and HSC activation. AKAP12 phospho-editing dramatically inhibits fibrosis, ER stress response, HSC inflammatory signaling, and liver injury in mice. Our overall findings suggest a pro-fibrogenic role of AKAP12 phosphorylation that may be targeted for therapeutic intervention in liver fibrosis.
Collapse
Affiliation(s)
- Komal Ramani
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical CenterLos AngelesUnited States
- Applied Cell Biology Division, Department of Biomedical Sciences, Cedars-Sinai Medical CenterLos AngelesUnited States
| | - Nirmala Mavila
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical CenterLos AngelesUnited States
- Applied Cell Biology Division, Department of Biomedical Sciences, Cedars-Sinai Medical CenterLos AngelesUnited States
| | - Aushinie Abeynayake
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical CenterLos AngelesUnited States
| | - Maria Lauda Tomasi
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical CenterLos AngelesUnited States
- Applied Cell Biology Division, Department of Biomedical Sciences, Cedars-Sinai Medical CenterLos AngelesUnited States
| | - Jiaohong Wang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical CenterLos AngelesUnited States
| | - Michitaka Matsuda
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical CenterLos AngelesUnited States
| | - Eki Seki
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical CenterLos AngelesUnited States
- Applied Cell Biology Division, Department of Biomedical Sciences, Cedars-Sinai Medical CenterLos AngelesUnited States
| |
Collapse
|
26
|
Wang Z, Zhao Z, Xia Y, Cai Z, Wang C, Shen Y, Liu R, Qin H, Jia J, Yuan G. Potential biomarkers in the fibrosis progression of nonalcoholic steatohepatitis (NASH). J Endocrinol Invest 2022; 45:1379-1392. [PMID: 35226336 DOI: 10.1007/s40618-022-01773-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/17/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE Fibrosis is the only histological feature reflecting the severity and prognosis of nonalcoholic steatohepatitis (NASH). We aim to explore novel genes associated with fibrosis progression in NASH. METHODS Two human RNA-seq datasets were downloaded from the public database. Weighted gene co-expression network analysis (WGCNA) was used to identify their co-expressed modules and further bioinformatics analysis was performed to identify hub genes within the modules. Finally, based on two single-cell RNA-seq datasets from mice and one microarray dataset from human, we further observed the expression of hub genes in different cell clusters and liver tissues. RESULTS 7 hub genes (SPP1, PROM1, SOX9, EPCAM, THY1, CD34 and MCAM) associated with fibrosis progression were identified. Single-cell RNA-seq analysis revealed that those hub genes were expressed by different cell clusters such as cholangiocytes, natural killer (NK) cells, and hepatic stellate cells (HSCs). We also found that SPP1 and CD34 serve as markers of different HSCs clusters, which are associated with inflammatory response and fibrogenesis, respectively. Further study suggested that SPP1, SOX9, MCAM and THY1 might be related to NASH-associated hepatocellular carcinoma (HCC). Receiver operating characteristic (ROC) analysis showed that the high expression of these genes could well predict the occurrence of HCC. At the same time, there were significant differences in metabolism-related pathway changes between different HCC subtypes, and SOX9 may be involved in these changes. CONCLUSIONS The present study identified novel genes associated with NASH fibrosis and explored their effects on fibrosis from a single-cell perspective that might provide new ideas for the early diagnosis, monitoring, evaluation, and prediction of fibrosis progression in NASH.
Collapse
Affiliation(s)
- Z Wang
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Z Zhao
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Y Xia
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Z Cai
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - C Wang
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Y Shen
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - R Liu
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - H Qin
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - J Jia
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China.
| | - G Yuan
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China.
| |
Collapse
|
27
|
Zhang D, Zheng J, Qiu G, Niu T, Gong Y, Cui S. CCl 4 inhibits the expressions of hepatic taurine biosynthetic enzymes and taurine synthesis in the progression of mouse liver fibrosis. Hum Exp Toxicol 2022; 41:9603271221135033. [DOI: 10.1177/09603271221135033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Carbon tetrachloride (CCl4) is a widely used hepatotoxin for the studies of liver fibrosis and cirrhosis, and taurine has function to abate liver fibrosis induced by CCl4. But the interacting mechanisms between taurine and CCl4 in liver are still largely unknown. These made us to hypothesize that CCl4 may induce liver fibrosis by affecting the expressions of taurine biosynthetic enzymes and taurine synthesis. We thus assayed the expressions of hepatic cysteine dioxygenase (CDO), cysteine sulfonate acid decarboxylase (CSAD) and taurine transporter (TauT) in the progression of mouse liver fibrosis induced by CCl4. The results demonstrated that CCl4 treatment markedly decreased hepatic CSAD, CDO expressions, and taurine levels in hepatic tissue, although TauT expression did not exhibit significant decline. It was contrasting that hepatic α-SMA, serum AST, ALT, ALP kept increasing, which were accompanied by the pathological characters of liver, whereas taurine supplement attenuated the progression of liver fibrosis induced by CCl4. These results demonstrate that CCl4 may induce liver fibrosis by inhibiting hepatic CSAD and CDO expressions and taurine synthesis, which are crucial for our understanding the mechanisms of liver fibrosis induced by CCl4, and also potential for establishing therapeutic strategies of liver fibrosis and related diseases.
Collapse
Affiliation(s)
- Di Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, PR China
| | - Jiaming Zheng
- College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China
| | - Guobin Qiu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China
| | - Tongjuan Niu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China
| | - Yuneng Gong
- College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China
| | - Sheng Cui
- College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, PR China
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, PR China
| |
Collapse
|
28
|
Li J, Deng X, Wang S, Jiang Q, Xu K. Resolvin D1 attenuates CCl4 Induced Liver Fibrosis by Inhibiting Autophagy-Mediated HSC activation via AKT/mTOR Pathway. Front Pharmacol 2021; 12:792414. [PMID: 34987404 PMCID: PMC8721195 DOI: 10.3389/fphar.2021.792414] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/22/2021] [Indexed: 01/30/2023] Open
Abstract
Resolvin D1 (RvD1) was previously reported to relieve inflammation and liver damage in several liver diseases, but its potential role in liver fibrosis remains elusive. The aim of our study was to investigate the effects and underlying mechanisms of RvD1 in hepatic autophagy in liver fibrosis. In vivo, male C57BL/6 mice were intraperitoneally injected with 20% carbon tetrachloride (CCl4, 5 ml/kg) twice weekly for 6 weeks to establish liver fibrosis model. RvD1 (100 ng or 300 ng/mouse) was added daily in the last 2 weeks of the modeling period. In vitro, lipopolysaccharide (LPS)-activated LX-2 cells were co-treated with increasing concentrations (2.5-10 nM) of RvD1. The degree of liver injury was measured by detecting serum AST and ALT contents and H&E staining. Hepatic fibrosis was assessed by masson's trichrome staining and metavir scoring. The qRT-PCR, western blot, immunohistochemistry, and immunofluorescence were applied to liver tissues or LPS-activated LX-2 cells to explore the protective effects of RvD1 in liver fibrosis. Our findings reported that RvD1 significantly attenuated CCl4 induced liver injury and fibrosis by decreasing plasma AST and ALT levels, reducing collagen I and α-SMA accumulation and other pro-fibrotic genes (CTGF, TIMP-1 and Vimentin) expressions in mouse liver, restoring damaged histological architecture and improving hepatic fibrosis scores. In vitro, RvD1 also repressed the LPS induced LX-2 cells activation and proliferation. These significant improvements mainly attributed to the inhibiting effect of RvD1 on autophagy in the process of hepatic stellate cell (HSC) activation, as demonstrated by decreased ratio of LC3-II/I and elevated p62 after RvD1 treatment. In addition, using AZD5363 (an AKT inhibitor that activates autophagy) and AZD8055 (an mTOR inhibitor, another autophagy activator), we further verified that RvD1 suppressed autophagy-mediated HSC activation and alleviated CCl4 induced liver fibrosis partly through AKT/mTOR pathway. Overall, these results demonstrate that RvD1 treatment is expected to become a novel therapeutic strategy against liver fibrosis.
Collapse
Affiliation(s)
- Jiahuan Li
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoling Deng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuhan Wang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qianqian Jiang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Keshu Xu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Bellassoued K, Hamed H, Ghrab F, Kallel R, Van Pelt J, Makni Ayadi F, Elfeki A. Antioxidant and hepatopreventive effects of Cassia angustifolia extract against carbon tetrachloride-induced hepatotoxicity in rats. Arch Physiol Biochem 2021; 127:486-496. [PMID: 31397186 DOI: 10.1080/13813455.2019.1650778] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The present study aimed to investigate the preventive effects of methanol fraction from Cassia angustifolia leaf extract (MECA), associated with its phytochemical content, on CCl4-induced hepatic toxicity in adult rats. In the controls, CCl4 induced an increase of serum biochemical parameters and triggered oxidative stress in the liver. MECA caused significant reductions in CCl4-elevated levels of hepatic markers, total cholesterol, triglycerides, and low-density lipoprotein and increased the level of high-density lipoprotein compared to the CCl4 group. Moreover, pretreatment with the MECA produced significant reductions in lipid peroxidation (thiobarbituric acid reactive substances) and protein carbonyl level in liver tissues as compared with the untreated group. The formation of pathological hepatic lesions was strongly prevented by MECA. Overall, this study suggests that administration of MECA has a high potential to quench free radicals and alleviate CCl4-induced hepatotoxicity in rats.
Collapse
Affiliation(s)
- Khaled Bellassoued
- Department of Life Sciences, Animal Ecophysiology Laboratory, University of Sfax, Sfax, Tunisia
| | - Houda Hamed
- Department of Life Sciences, Animal Ecophysiology Laboratory, University of Sfax, Sfax, Tunisia
| | - Ferdaws Ghrab
- Department of Coastal and Urban Environments, National Engineering School of Sfax, University of Sfax, Sfax, Tunisia
| | - Rim Kallel
- Faculty of Medicine of Sfax, Anatomopathology Laboratory, Habib Bourguiba University Hospital, University of Sfax, Sfax, Tunisia
| | - Jos Van Pelt
- Department of Oncology, Laboratory of Clinical Digestive Oncology, KU Leuven, Belgium
| | - Fatma Makni Ayadi
- Biochemistry Laboratory, Habib Bourguiba University Hospital, Sfax, Tunisia
| | - Abdelfattah Elfeki
- Department of Life Sciences, Animal Ecophysiology Laboratory, University of Sfax, Sfax, Tunisia
| |
Collapse
|
30
|
Hu Q, Liu M, You Y, Zhou G, Chen Y, Yuan H, Xie L, Han S, Zhu K. Dual inhibition of reactive oxygen species and spleen tyrosine kinase as a therapeutic strategy in liver fibrosis. Free Radic Biol Med 2021; 175:193-205. [PMID: 34492311 DOI: 10.1016/j.freeradbiomed.2021.08.241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/10/2021] [Accepted: 08/30/2021] [Indexed: 02/06/2023]
Abstract
Hepatic stellate cells (HSCs) play key roles in liver fibrosis (LF) and hepatocellular carcinoma (HCC). We previously reported that spleen tyrosine kinase (SYK) is critical for HSCs activation, however, the mechanisms are insufficiently understood. In the present study, we found that SYK facilitated autophagy to promote HSCs activation by enhancing reactive oxygen species (ROS) generation. However, SYK inhibitor GS-9973 could efficiently reduce HSCs ROS generation in vitro but not in vivo. Mechanistically, hepatocytes (HCs) would release ROS outside and then diffuse into HSCs to promote autophagy and activation in vitro in the context of inflammation. We then further examined the ROS contents in liver sections and primary liver cells of carbon tetrachloride (CCl4) induced mice treated with or without different doses of Silybin, a natural compound characterized by a well-established antioxidant and hepatoprotective properties, and found that ROS intensities in both liver sections and their deprived primary cells were efficiently inhibited in a dose-dependent fashion. Lastly, we evaluated the rational combination of Silybin and GS-9973 in the treatment of CCl4 induced mice and found that this combination is well tolerated and acts synergistically against HSCs activity, LF and HCC. The combinational use of Silybin and GS-9973 could be a promising therapeutic strategy in patients suffering from LF and even HCC.
Collapse
Affiliation(s)
- Qiaoting Hu
- Department of Medical Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou, Fujian, 350014, China
| | - Mingyu Liu
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510260, China.
| | - Yundan You
- Department of Emergency Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China
| | - Guo Zhou
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510260, China
| | - Ye Chen
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510260, China
| | - Hui Yuan
- Department of Gastroenterology, Huizhou Municipal Central Hospital, Huizhou, Guangdong, 516001, China
| | - Lulu Xie
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510260, China
| | - Shisong Han
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510260, China
| | - Kangshun Zhu
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510260, China.
| |
Collapse
|
31
|
Pterostilbene is more efficacious than hydroxystilbenes in protecting liver fibrogenesis in a carbon tetracholride-induced rat model. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104604] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
32
|
Uehara T, Pogribny IP, Rusyn I. The DEN and CCl 4 -Induced Mouse Model of Fibrosis and Inflammation-Associated Hepatocellular Carcinoma. Curr Protoc 2021; 1:e211. [PMID: 34370903 PMCID: PMC8744072 DOI: 10.1002/cpz1.211] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human hepatocellular carcinoma (HCC) develops most often as a complication of fibrosis or cirrhosis. Although most human studies of HCC provide crucial insights into the molecular signatures of HCC, they seldom address its etiology. Mouse models provide essential tools for investigating the pathogenesis of HCC, but the majority of rodent cancer models do not feature liver fibrosis. Detailed here is a protocol for an experimental mouse model of HCC that arises in association with advanced liver fibrosis. The disease model is induced by a single injection of N-nitrosodiethylamine (DEN) at 2 weeks of age followed by repeated administration of carbon tetrachloride (CCl4 ) from 8 weeks of age for up to 14 consecutive weeks. A dramatic potentiation of liver tumor incidence is observed following administration of DEN and CCl4 , with 100% of mice developing liver tumors at 5 months of age. This model has been employed for studying the molecular mechanisms of fibrogenesis and HCC development, as well as for cancer hazard/chemotherapy testing of drug candidates. © 2021 Wiley Periodicals LLC. Basic Protocol: The DEN and CCl4 -induced mouse model of fibrosis and inflammation-associated hepatocellular carcinoma.
Collapse
Affiliation(s)
- Takeki Uehara
- Strategic Development Department, Shionogi & Co., Osaka, Japan
| | - Igor P. Pogribny
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| |
Collapse
|
33
|
Bao YL, Wang L, Pan HT, Zhang TR, Chen YH, Xu SJ, Mao XL, Li SW. Animal and Organoid Models of Liver Fibrosis. Front Physiol 2021; 12:666138. [PMID: 34122138 PMCID: PMC8187919 DOI: 10.3389/fphys.2021.666138] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/03/2021] [Indexed: 12/12/2022] Open
Abstract
Liver fibrosis refers to the process underlying the development of chronic liver diseases, wherein liver cells are repeatedly destroyed and regenerated, which leads to an excessive deposition and abnormal distribution of the extracellular matrix such as collagen, glycoprotein and proteoglycan in the liver. Liver fibrosis thus constitutes the pathological repair response of the liver to chronic injury. Hepatic fibrosis is a key step in the progression of chronic liver disease to cirrhosis and an important factor affecting the prognosis of chronic liver disease. Further development of liver fibrosis may lead to structural disorders of the liver, nodular regeneration of hepatocytes and the formation of cirrhosis. Hepatic fibrosis is histologically reversible if treated aggressively during this period, but when fibrosis progresses to the stage of cirrhosis, reversal is very difficult, resulting in a poor prognosis. There are many causes of liver fibrosis, including liver injury caused by drugs, viral hepatitis, alcoholic liver, fatty liver and autoimmune disease. The mechanism underlying hepatic fibrosis differs among etiologies. The establishment of an appropriate animal model of liver fibrosis is not only an important basis for the in-depth study of the pathogenesis of liver fibrosis but also an important means for clinical experts to select drugs for the prevention and treatment of liver fibrosis. The present study focused on the modeling methods and fibrosis characteristics of different animal models of liver fibrosis, such as a chemical-induced liver fibrosis model, autoimmune liver fibrosis model, cholestatic liver fibrosis model, alcoholic liver fibrosis model and non-alcoholic liver fibrosis model. In addition, we also summarize the research and application prospects concerning new organoids in liver fibrosis models proposed in recent years. A suitable animal model of liver fibrosis and organoid fibrosis model that closely resemble the physiological state of the human body will provide bases for the in-depth study of the pathogenesis of liver fibrosis and the development of therapeutic drugs.
Collapse
Affiliation(s)
- Yu-long Bao
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Li Wang
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Hai-ting Pan
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Tai-ran Zhang
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Ya-hong Chen
- Health Management Center, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Shan-jing Xu
- School of Medicine, Shaoxing University, Shaoxing, Chian
| | - Xin-li Mao
- School of Medicine, Shaoxing University, Shaoxing, Chian
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Shao-wei Li
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| |
Collapse
|
34
|
Zhao Z, Hou Y, Zhou W, Keerthiga R, Fu A. Mitochondrial transplantation therapy inhibit carbon tetrachloride-induced liver injury through scavenging free radicals and protecting hepatocytes. Bioeng Transl Med 2021; 6:e10209. [PMID: 34027095 PMCID: PMC8126821 DOI: 10.1002/btm2.10209] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/02/2020] [Accepted: 12/06/2020] [Indexed: 12/14/2022] Open
Abstract
Carbon tetrachloride (CCl4)-induced liver injury is predominantly caused by free radicals, in which mitochondrial function of hepatocytes is impaired, accompanying with the production of ROS and decreased ATP energy supply in animals intoxicated with CCl4. Here we explored a novel therapeutic approach, mitochondrial transplantation therapy, for treating the liver injury. The results showed that mitochondria entered hepatocytes through macropinocytosis pathway, and thereby cell viability was recovered in a concentration-dependent manner. Mitochondrial therapy could increase ATP supply and reduce free radical damage. In liver injury model of mice, mitochondrial therapy significantly improved liver function and prevented tissue fibrogenesis. Transcriptomic data revealed that mitochondrial unfold protein response (UPRmt), a protective transcriptional response of mitochondria-to-nuclear retrograde signaling, would be triggered after mitochondrial administration. Then the anti-oxidant genes were up-regulated to scavenge free radicals. The mitochondrial function was rehabilitated through the transcriptional activation of respiratory chain enzyme and mitophage-associated genes. The protective response re-balanced the cellular homeostasis, and eventually enhanced stress resistance that is linked to cell survival. The efficacy of mitochondrial transplantation therapy in the animals would suggest a novel approach for treating liver injury caused by toxins.
Collapse
Affiliation(s)
- Zizhen Zhao
- College of Pharmaceutical Sciences, Southwest UniversityChongqingChina
| | - Yixue Hou
- College of Pharmaceutical Sciences, Southwest UniversityChongqingChina
| | - Wei Zhou
- College of Pharmaceutical Sciences, Southwest UniversityChongqingChina
| | | | - Ailing Fu
- College of Pharmaceutical Sciences, Southwest UniversityChongqingChina
| |
Collapse
|
35
|
Balasubramanian K. Combinatorial enumeration of stereo, chiral and position isomers of polysubstituted halocarbons: applications to machine learning of proton and 35Cl NMR spectroscopy of halocarbons. Theor Chem Acc 2021. [DOI: 10.1007/s00214-021-02744-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
36
|
Zhu Y, Zhang C, Huang M, Lin J, Fan X, Ni T. TRIM26 Induces Ferroptosis to Inhibit Hepatic Stellate Cell Activation and Mitigate Liver Fibrosis Through Mediating SLC7A11 Ubiquitination. Front Cell Dev Biol 2021; 9:644901. [PMID: 33869196 PMCID: PMC8044755 DOI: 10.3389/fcell.2021.644901] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatic stellate cells (HSCs) are activated by inflammatory mediators to secrete extracellular matrix for collagen deposition, leading to liver fibrosis. Ferroptosis is iron- and lipid hydroperoxide-dependent programmed cell death, which has recently been targeted for inhibiting liver fibrogenic processes. Tripartite motif-containing protein 26 (TRIM26) is an E3 ubiquitin ligase that functions as a tumor suppressor in hepatocellular carcinoma, while little is known about its function in liver fibrosis. In the present study, the differential expression of TRIM26 in normal and fibrotic liver tissues was examined based on both online databases and specimens collected from patient cohort. The effects of TRIM26 on HSCs ferroptosis were examined in vitro through evaluating cell proliferation, lipid peroxidation, and expression of key ferroptosis-related factors. In vivo function of TRIM26 in liver fibrosis was examined based on CCl4-induced mice model. We found that TRIM26 was downregulated in fibrotic liver tissues. The overexpression of TRIM26 inhibited HSCs proliferation, promoted lipid peroxidation, manipulated ferroptosis-related factor expressions, and counteracted the effect of iron inhibitor deferoxamine. Moreover, TRIM26 physically interacted with solute carrier family-7 member-11 (SLC7A11), a critical protein for lipid reactive oxygen species (ROS) scavenging, and mediated its ubiquitination. In addition, TRIM26 overexpression induced HSCs ferroptosis and mitigated CCl4-induced liver fibrosis in mice. In conclusion, TRIM26 promotes HSCs ferroptosis to suppress liver fibrosis through mediating the ubiquitination of SLC7A11. The TRIM26-targeted SLC7A11 suppression can be a novel therapeutic strategy for liver fibrosis.
Collapse
Affiliation(s)
- Yiming Zhu
- Department of General Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chihao Zhang
- Department of General Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Mingzhe Huang
- Department of General Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiayun Lin
- Department of General Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao Fan
- Department of General Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Ni
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
37
|
Dashek RJ, Diaz C, Chandrasekar B, Rector RS. The Role of RECK in Hepatobiliary Neoplasia Reveals Its Therapeutic Potential in NASH. Front Endocrinol (Lausanne) 2021; 12:770740. [PMID: 34745017 PMCID: PMC8564138 DOI: 10.3389/fendo.2021.770740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multimorbidity disorder ranging from excess accumulation of fat in the liver (steatosis) to steatohepatitis (NASH) and end-stage cirrhosis, and the development of hepatocellular carcinoma (HCC) in a subset of patients. The defining features of NASH are inflammation and progressive fibrosis. Currently, no pharmaceutical therapies are available for NAFLD, NASH and HCC; therefore, developing novel treatment strategies is desperately needed. Reversion Inducing Cysteine Rich Protein with Kazal motifs (RECK) is a well-known modifier of the extracellular matrix in hepatic remodeling and transition to HCC. More recently, its role in regulating inflammatory and fibrogenic processes has emerged. Here, we summarize the most relevant findings that extend our current understanding of RECK as a regulator of inflammation and fibrosis, and its induction as a potential strategy to blunt the development and progression of NASH and HCC.
Collapse
Affiliation(s)
- Ryan J. Dashek
- Research Service, Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, United States
- Comparative Medicine Program, Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Connor Diaz
- School of Medicine, University of Missouri, Columbia, MO, United States
| | - Bysani Chandrasekar
- Research Service, Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, United States
- Division of Cardiology, Department of Medicine, University of Missouri, Columbia, MO, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States
| | - R. Scott Rector
- Research Service, Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, United States
- *Correspondence: R. Scott Rector,
| |
Collapse
|
38
|
Wang Z, Cheng ZX, Abrams ST, Lin ZQ, Yates E, Yu Q, Yu WP, Chen PS, Toh CH, Wang GZ. Extracellular histones stimulate collagen expression in vitro and promote liver fibrogenesis in a mouse model via the TLR4-MyD88 signaling pathway. World J Gastroenterol 2020; 26:7513-7527. [PMID: 33384551 PMCID: PMC7754552 DOI: 10.3748/wjg.v26.i47.7513] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/08/2020] [Accepted: 12/06/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Liver fibrosis progressing to liver cirrhosis and hepatic carcinoma is very common and causes more than one million deaths annually. Fibrosis develops from recurrent liver injury but the molecular mechanisms are not fully understood. Recently, the TLR4-MyD88 signaling pathway has been reported to contribute to fibrosis. Extracellular histones are ligands of TLR4 but their roles in liver fibrosis have not been investigated. AIM To investigate the roles and potential mechanisms of extracellular histones in liver fibrosis. METHODS In vitro, LX2 human hepatic stellate cells (HSCs) were treated with histones in the presence or absence of non-anticoagulant heparin (NAHP) for neutralizing histones or TLR4-blocking antibody. The resultant cellular expression of collagen I was detected using western blotting and immunofluorescent staining. In vivo, the CCl4-induced liver fibrosis model was generated in male 6-week-old ICR mice and in TLR4 or MyD88 knockout and parental mice. Circulating histones were detected and the effect of NAHP was evaluated. RESULTS Extracellular histones strongly stimulated LX2 cells to produce collagen I. Histone-enhanced collagen expression was significantly reduced by NAHP and TLR4-blocking antibody. In CCl4-treated wild type mice, circulating histones were dramatically increased and maintained high levels during the duration of fibrosis-induction. Injection of NAHP not only reduced alanine aminotransferase and liver injury scores, but also significantly reduced fibrogenesis. Since the TLR4-blocking antibody reduced histone-enhanced collagen I production in HSC, the CCl4 model with TLR4 and MyD88 knockout mice was used to demonstrate the roles of the TLR4-MyD88 signaling pathway in CCl4-induced liver fibrosis. The levels of liver fibrosis were indeed significantly reduced in knockout mice compared to wild type parental mice. CONCLUSION Extracellular histones potentially enhance fibrogenesis via the TLR4-MyD88 signaling pathway and NAHP has therapeutic potential by detoxifying extracellular histones.
Collapse
Affiliation(s)
- Zhi Wang
- Department of Pathology and Pathophysiology, Medical School, Southeast University, Nanjing 210009, Jiangsu Province, China
- Department of Gastroenterology, Zhongda Hospital, Nanjing 210009, Jiangsu Province, China
| | - Zhen-Xing Cheng
- Department of Pathology and Pathophysiology, Medical School, Southeast University, Nanjing 210009, Jiangsu Province, China
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, United Kingdom
- Department of Gastroenterology, The First Affiliated Hospital, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Simon T Abrams
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, United Kingdom
| | - Zi-Qi Lin
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Edwin Yates
- Department of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, United Kingdom
| | - Qian Yu
- Department of Gastroenterology, Zhongda Hospital, Nanjing 210009, Jiangsu Province, China
| | - Wei-Ping Yu
- Department of Pathology and Pathophysiology, Medical School, Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Ping-Sheng Chen
- Department of Pathology and Pathophysiology, Medical School, Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, United Kingdom
- Roald Dahl Haemostasis & Thrombosis Ctr, Royal Liverpool University Hospital, Liverpool L69 7BE, United Kingdom
| | - Guo-Zheng Wang
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, United Kingdom
| |
Collapse
|
39
|
Sedum sarmentosum Total Flavonoids Alleviate Schistosomiasis-Induced Liver Fibrosis by Altering TGF- β1 and Smad7 Expression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:2083697. [PMID: 33293986 PMCID: PMC7714578 DOI: 10.1155/2020/2083697] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/01/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023]
Abstract
Objectives Schistosomiasis is a parasitic disease that affects over 142 million people worldwide. The main causes of death of schistosomiasis include liver granuloma and secondary hepatic cirrhosis resulting from severe fibrosis. Despite intensive research, controlling liver fibrosis associated with schistosomiasis remains challenging. Sedum sarmentosum total flavonoid (SSTF) is a promising agent to reduce liver fibrosis with an unknown mechanism. Thus, the objectives of this study are to validate its effect on the liver fibrosis caused by schistosomiasis and to explore the underlying molecular mechanism. Methods Sixty male Sprague-Dawley rats were randomly divided into six groups: one group of normal control and five groups of liver fibrosis induced by schistosomiasis japonica with or without SSTF or colchicine treatment, the latter serving as the positive control. Liver tissues from each animal were harvested to observe the degree and grade of hepatic fibrosis. We also measured the expression of transforming growth factor-beta 1 (TGF-β1) and Smad7 using RT-qPCR, Western blot, and immunohistochemistry. Results Compared with the untreated model group, groups treated with SSTF at all three tested doses had significantly reduced hepatic fibrosis (P < 0.05). Each dose of SSTF also significantly reduced TGF-β1 protein expression and mRNA levels in the liver tissues (P < 0.05). In contrast, the middle and high doses of SSTF significantly increased Smad7 protein expression and mRNA levels (P < 0.05). Immunohistochemistry showed that each dose of SSTF reduced TGF-β1 protein expression (P < 0.05). Conclusion Our results demonstrated that SSTF alleviated schistosomiasis japonica-induced hepatic fibrosis by inhibiting the TGF-β1/Smad7 pathway.
Collapse
|
40
|
Almatroodi SA, Almatroudi A, Anwar S, Yousif Babiker A, Khan AA, Alsahli MA, Rahmani AH. Antioxidant, anti-inflammatory and hepatoprotective effects of olive fruit pulp extract: in vivo and in vitro study. JOURNAL OF TAIBAH UNIVERSITY FOR SCIENCE 2020. [DOI: 10.1080/16583655.2020.1848761] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Saleh A. Almatroodi
- Department of Medical Laboratories, College of Applied Medical Science, Qassim University, Buraidah, Saudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Science, Qassim University, Buraidah, Saudi Arabia
| | - Shehwaz Anwar
- Department of Medical Laboratories, College of Applied Medical Science, Qassim University, Buraidah, Saudi Arabia
| | - Ali Yousif Babiker
- Department of Medical Laboratories, College of Applied Medical Science, Qassim University, Buraidah, Saudi Arabia
| | - Amjad Ali Khan
- Department of Basic Health Science, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Mohammed A. Alsahli
- Department of Medical Laboratories, College of Applied Medical Science, Qassim University, Buraidah, Saudi Arabia
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Science, Qassim University, Buraidah, Saudi Arabia
| |
Collapse
|
41
|
Hayashi M, Yamada S, Kurimoto K, Tanabe H, Hirabayashi S, Sonohara F, Inokawa Y, Takami H, Kanda M, Tanaka C, Nakayama G, Koike M, Kodera Y. miR-23b-3p Plays an Oncogenic Role in Hepatocellular Carcinoma. Ann Surg Oncol 2020; 28:3416-3426. [PMID: 33140250 DOI: 10.1245/s10434-020-09283-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 10/06/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Reports show miR-23b to be a cancer-related biomarker in various cancer types. Interestingly, it has a dual role of oncogenic and tumor-suppressive functions, depending on the cancer type. This study focused on the unknown association of miR-23b-3p with hepatocellular carcinoma (HCC). METHODS Expression of miR-23b-3p was measured in nine HCC cell lines and 125 resected human HCC samples by TaqMan microRNA assays. To detect its downstream target, miR-23b-3p mimic and inhibitor constructs were transfected and analyzed. RESULTS HepG2, a high miR-23b-3p-expressing cell line, was transfected with a miR-23b-3p inhibitor construct, whereas SK-Hep1, a low miR-23b-3p-expressing cell line, was transfected with a mimic construct. Proliferation of HCC cells was activated by miR-23b-3p overexpression and diminished by its knockdown. Then, 125 clinical HCC samples were examined to measure miR-23b-3p expression. Tumor expression of miR-23b-3p was upregulated in 48 cases (38%) and downregulated in 77 cases (62%). The upregulated cases were correlated with elderly patients (P = 0.015). These patients also showed significantly poor overall survival [hazard ratio (HR), 3.10; 95% conflidence interval (CI), 1.57-6.29; P = 0.001] in a multivariate analysis. Furthermore, mitochondrial metabolism-related genes (MICU3 and AUH) were detected as specific binding targets. CONCLUSION The study showed that miR-23b-3p functions as an oncogenic microRNA in HCC cell lines. Its overexpression in resected HCC tissues was a significant prognostic factor of overall survival. Both MICU3 and AUH may be candidate gene targets of miR-23b-3p.
Collapse
Affiliation(s)
- Masamichi Hayashi
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Suguru Yamada
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Keisuke Kurimoto
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Tanabe
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sho Hirabayashi
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fuminori Sonohara
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshikuni Inokawa
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideki Takami
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Chie Tanaka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Goro Nakayama
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiko Koike
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
42
|
Intestinal Clostridioides difficile Can Cause Liver Injury through the Occurrence of Inflammation and Damage to Hepatocytes. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7929610. [PMID: 33005688 PMCID: PMC7503108 DOI: 10.1155/2020/7929610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/19/2020] [Accepted: 08/06/2020] [Indexed: 12/14/2022]
Abstract
This study investigated if intestinal Clostridioides difficile (CD) causes liver injury. Four-week-old male C3H/HeN mice were treated with phosphate-buffered solution (control), CD, diethylnitrosamine (DEN) to induce liver injury with PBS (DEN+PBS), and DEN with CD (DEN+CD) for nine weeks. After sacrifice, livers and mesenteric lymph nodes (MLNs) were removed and bacterial translocation, transcriptomes, and proteins were analysed. CD was found in 20% of MLNs from the control and DEN+PBS groups, in 30% of MLNs from the CD group, and in 75% of MLNs from the DEN+CD groups, which had injured livers. Also, CD was detected in 50% of the livers in the DEN+CD group with CD-positive MLNs. Elevated IL-1β, HB-EGF, EGFR, TGF-α, PCNA, DES, HMGB1, and CRP expressions were observed in the CD and DEN+CD groups as compared to the control and DEN+PBS groups. Protein levels of IL-6 and HMGB1 were higher in the CD and DEN+CD groups than in the control and DEN+PBS groups. These results indicate that intestinal CD can initiate and aggravate liver injury, and the mechanism of pathogenesis for liver injury should be investigated in further studies.
Collapse
|
43
|
Munakarmi S, Chand L, Shin HB, Jang KY, Jeong YJ. Indole-3-Carbinol Derivative DIM Mitigates Carbon Tetrachloride-Induced Acute Liver Injury in Mice by Inhibiting Inflammatory Response, Apoptosis and Regulating Oxidative Stress. Int J Mol Sci 2020; 21:E2048. [PMID: 32192079 PMCID: PMC7139345 DOI: 10.3390/ijms21062048] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/12/2020] [Accepted: 03/16/2020] [Indexed: 02/06/2023] Open
Abstract
3,3'-Diindolylmethane (DIM), a metabolic product of indole-3-carbinol extracted from cruciferous vegetables exhibits anti-inflammatory and anti-cancer properties. Earlier, the product has been demonstrated to possess anti-fibrotic properties; however, its protective effects on liver injury have not been clearly elucidated. In this study, we postulated the effects and molecular mechanisms of action of DIM on carbon tetrachloride (CCl4)-induced liver injury in mice. Acute liver injury was induced by a single intraperitoneal administration of CCl4 (1 ml/kg) into mice. DIM was injected via subcutaneous route for three days at various doses (2.5, 5 and 10 mg/kg) before CCl4 injection. Mice were sacrificed and serum was collected for quantification of serum transaminases. The liver was collected and weighed. Treatment with DIM significantly reduced serum transaminases levels (AST and ALT), tumor necrosis factor-α (TNF-α) and reactive oxygen species (ROS). CCl4- induced apoptosis was inhibited by DIM treatment by the reduction in the levels of cleaved caspase-3 and Bcl2 associated X protein (Bax). DIM treated mice significantly restored Cytochrome P450 2E1, nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) expression in CCl4 treated mice. In addition, DIM downregulated overexpression of hepatic nuclear factor kappa B (NF-κB) and inhibited CCl4 mediated apoptosis. Our results suggest that the protective effects of DIM against CCl4- induced liver injury are due to the inhibition of ROS, reduction of pro-inflammatory mediators and apoptosis.
Collapse
Affiliation(s)
- Suvesh Munakarmi
- Laboratory of Liver Regeneration, Biomedical Research Institute, Chonbuk National University Medical School, Jeonju 54907, Korea; (S.M.); (L.C.)
| | - Lokendra Chand
- Laboratory of Liver Regeneration, Biomedical Research Institute, Chonbuk National University Medical School, Jeonju 54907, Korea; (S.M.); (L.C.)
| | - Hyun Beak Shin
- Department of Surgery, Chonbuk National University Hospital, Jeonju 54907, Korea;
| | - Kyu Yun Jang
- Department of Pathology, Chonbuk National University Hospital, Jeonju 54907, Korea;
| | - Yeon Jun Jeong
- Laboratory of Liver Regeneration, Biomedical Research Institute, Chonbuk National University Medical School, Jeonju 54907, Korea; (S.M.); (L.C.)
- Department of Surgery, Chonbuk National University Hospital, Jeonju 54907, Korea;
| |
Collapse
|
44
|
Padmanabhan J, Maan ZN, Kwon SH, Kosaraju R, Bonham CA, Gurtner GC. In Vivo Models for the Study of Fibrosis. Adv Wound Care (New Rochelle) 2019; 8:645-654. [PMID: 31827979 PMCID: PMC6904938 DOI: 10.1089/wound.2018.0909] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/28/2019] [Indexed: 02/04/2023] Open
Abstract
Significance: Fibrosis and scar formation pose a substantial physiological and psychological burden on patients and a significant public health burden on the economy, estimated to be up to $12 billion a year. Fibrosis research is heavily reliant on in vivo models, but variations in animal models and differences between animal and human fibrosis necessitates careful selection of animal models to study fibrosis. There is also an increased need for improved animal models that recapitulate human pathophysiology. Recent Advances: Several murine and porcine models, including xenograft, drug-induced fibrosis, and mechanical load-induced fibrosis, for different types of fibrotic disease have been described in the literature. Recent findings have underscored the importance of mechanical forces in the pathophysiology of scarring. Critical Issues: Differences in skin, properties of subcutaneous tissue, and modes of fibrotic healing in animal models and humans provide challenges toward investigating fibrosis with in vivo models. While porcine models are typically better suited to study cutaneous fibrosis, murine models are preferred because of the ease of handling and availability of transgenic strains. Future Directions: There is a critical need to develop novel murine models that recapitulate the mechanical cues influencing fibrosis in humans, significantly increasing the translational value of fibrosis research. We advocate a translational pipeline that begins in mouse models with modified biomechanical environments for foundational molecular and cellular research before validation in porcine models that closely mimic the human condition.
Collapse
Affiliation(s)
- Jagannath Padmanabhan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Zeshaan N. Maan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Sun Hyung Kwon
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Revanth Kosaraju
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Clark A. Bonham
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Geoffrey C. Gurtner
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
45
|
Yan J, Huang H, Liu Z, Shen J, Ni J, Han J, Wang R, Lin D, Hu B, Jin L. Hedgehog signaling pathway regulates hexavalent chromium-induced liver fibrosis by activation of hepatic stellate cells. Toxicol Lett 2019; 320:1-8. [PMID: 31756458 DOI: 10.1016/j.toxlet.2019.11.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/18/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022]
Abstract
With the spread of hexavalent chromium [Cr(VI)] contamination, risk of exposure in non-occupational populations is increasing. The liver is the main target organ for Cr(VI) accumulation; however, the effect of long-term Cr(VI) exposure on liver toxicity is largely unknown. In this study, we investigated the effect of chronic Cr(VI) exposure on liver fibrosis and its possible mechanism. Mice were injected with Cr(VI) for two months, and our results showed Cr(VI) treatment caused liver toxicity characterized by liver structure disorganization, liver dysfunction, and antioxidant enzyme system inhibition. The development of liver fibrosis was also found via the emergence of collagen fibril deposition, increased expression of extracellular matrix-related genes, activation of hepatic stellate cells (HSCs) and increase the expression levels of Hedgehog (Hh) signaling pathway-related molecules. To demonstrate the role of Hh signaling in the regulation of Cr(VI)-induced liver fibrosis, genetically modified mice with heterozygous deficiency of Shh (Shh+/-) were used. In the Shh+/- mice, Hh signaling, HSCs activation and liver fibrosis development were all ameliorated. In conclusion, we demonstrated that Cr(VI)-induced liver fibrosis development resulted from Hh pathway-mediated HSCs activation. Our findings strongly suggest that inhibition of Hh pathway may help in the development of new strategies for Cr(VI)-associated liver fibrosis.
Collapse
Affiliation(s)
- Junyan Yan
- College of Life Science, Shaoxing University, Shaoxing, Zhejiang, China
| | - Huarong Huang
- College of Life and Environmental Science, Hangzhou Normal University, Hangzhou, China
| | - Zuping Liu
- Department of Pathology, Affliliated Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Jiayuan Shen
- Department of Pathology, Affliliated Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Jian Ni
- College of Life Science, Shaoxing University, Shaoxing, Zhejiang, China
| | - Jiwei Han
- Department of Pathology, Affliliated Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Renjun Wang
- College of Life Science, Qufu Normal University, Qufu City, Shandong, China
| | - Derong Lin
- The Second Hospital of Shaoxing, Shaoxing, Zhejiang, China
| | - Baowei Hu
- College of Life Science, Shaoxing University, Shaoxing, Zhejiang, China.
| | - Lifang Jin
- College of Life Science, Shaoxing University, Shaoxing, Zhejiang, China.
| |
Collapse
|
46
|
Animal Models of Hepatocellular Carcinoma Prevention. Cancers (Basel) 2019; 11:cancers11111792. [PMID: 31739536 PMCID: PMC6895981 DOI: 10.3390/cancers11111792] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/08/2019] [Accepted: 11/10/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a deadly disease and therapeutic efficacy in advanced HCC is limited. Since progression of chronic liver disease to HCC involves a long latency period of a few decades, a significant window of therapeutic opportunities exists for prevention of HCC and improve patient prognosis. Nonetheless, there has been no clinical advancement in instituting HCC chemopreventive strategies. Some of the major challenges are heterogenous genetic aberrations of HCC, significant modulation of tumor microenvironment and incomplete understanding of HCC tumorigenesis. To this end, animal models of HCC are valuable tools to evaluate biology of tumor initiation and progression with specific insight into molecular and genetic mechanisms involved. In this review, we describe various animal models of HCC that facilitate effective ways to study therapeutic prevention strategies that have translational potential to be evaluated in a clinical context.
Collapse
|
47
|
Bozhkov AI, Ohiienko SL, Bondar AY, Klimova EM, Ivanov EG. Induced Liver Fibrosis Is Accompanied in Young and Old Animals by Age-Dependent Changes in Bone Marrow Cells. ADVANCES IN GERONTOLOGY 2019. [DOI: 10.1134/s2079057019030032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
48
|
Oct4 and Hnf4α-induced hepatic stem cells ameliorate chronic liver injury in liver fibrosis model. PLoS One 2019; 14:e0221085. [PMID: 31404112 PMCID: PMC6690533 DOI: 10.1371/journal.pone.0221085] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 07/30/2019] [Indexed: 12/20/2022] Open
Abstract
Direct conversion from fibroblasts to generate hepatocyte like-cells (iHeps) bypassing the pluripotent state has been described in previous reports as an attractive method acquiring hepatocytes for cell-based therapy. The limited proliferation of iHeps, however, has hampered it uses in cell-based therapy. Since hepatic stem cells (HepSCs) possess self-renewal and bipotency with the capacity to differentiate into both hepatocytes and cholangiocytes, they have therapeutic potential for treating liver disease. Here, we investigated the therapeutic effects of induced HepSCs (iHepSCs) on a carbon tetrachloride (CCl4)-induced liver fibrosis model. We demonstrate that Oct4 and Hnf4a are sufficient to convert fibroblasts into expandable iHepSCs. Hepatocyte-like cells derived from iHepSCs (iHepSC-HEPs) exhibit the typical morphology of hepatocytes and hepatic functions, including glycogen storage, low-density lipoprotein (LDL) uptake, Indocyanine green (ICG) detoxification, drug metabolism, urea production, and albumin secretion. iHepSCs-derived cholangiocyte-like cells (iHepSC-CLCs) expressed cholangiocyte-specific markers and formed cysts and tubule-like structures with apical-basal polarity and secretory function in three-dimensional culture condition. Furthermore, iHepSCs showed anti-inflammatory and anti-fibrotic effects in CCl4-induced liver fibrosis. This study demonstrates that Oct4 and Hnf4α-induced HepSCs show typical hepatic and biliary functionality in vitro. It also presents the therapeutic effect of iHepSCs in liver fibrosis. Therefore, directly converting iHepSCs from somatic cells may facilitate the development of patient-specific cell-based therapy for chronic liver damage.
Collapse
|
49
|
Al-Jawad FH, Al-Attar Z, Abbood MS. The Protective Effect of Nitroglycerin, N-Acetyl Cysteine and Metoprolol in CCL4 Induced Animal Model of Acute Liver Injury. Open Access Maced J Med Sci 2019; 7:1739-1743. [PMID: 31316651 PMCID: PMC6614250 DOI: 10.3889/oamjms.2019.469] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE: The current study was designed to determine the hepatoprotective effect of well-known drugs. Nitroglycerin, N-acetyl cysteine and Metoprolol in acute liver injury induced by CCL4. The antioxidant effects of b-blockers, especially carvedilol, have been described by several investigators. However, for metoprolol, the effect is a bit query as there is only one in-vitro study showing a little hepatoprotective effect. Thus, it is worthy to re-study the hepatoprotective effect of metoprolol. AIM: To explore the possible hepatoprotective effect of Nitroglycerin, N-acetyl cysteine and Metoprolol Tartrate MATERIAL AND METHODS: The normal serum values of ALP, AST, ALT, TSB and TSP were determined in 35 healthy rabbits allocated to 5 groups before CCL4 induction and at three occasions 24, 72, 120 hrs after induction by CCL4 and treatment with the tested drugs: Nitroglycerin, N-acetyl cysteine and Metoprolol for five successive days. RESULTS: Showed significant decrease in serum levels of ALP, AST, ALT and TSB with a significant increase in TSP level of all the tested drugs measured at 120 hrs compared with the control and their levels measured at 24, 72 hrs. CONCLUSION: All the tested drugs proved in having a hepatoprotective effect when they are given orally to animals. The histopathological sections of the liver tissue supported the real effect of these drugs in the management of ALI.
Collapse
Affiliation(s)
- Faruk H Al-Jawad
- Department of Pharmacology & Therapeutics, Al-Nahrain College of Medicine, Al-Nahrain University, Baghdad, Iraq
| | - Zaid Al-Attar
- Department of Pharmacology, Al-Kindy College of Medicine, University of Baghdad, Baghdad, Iraq
| | - Muayyad S Abbood
- The High Institute for Infertility Diagnosis and Assisted Reproductive Technologies, Al-Nahrain University, Baghdad, Iraq
| |
Collapse
|
50
|
Ko J, Yeh WJ, Huang WC, Yang HY. Camellia Oleifera Seed Extract Mildly Ameliorates Carbon Tetrachloride-Induced Hepatotoxicity in Rats by Suppressing Inflammation. J Food Sci 2019; 84:1586-1591. [PMID: 31116885 DOI: 10.1111/1750-3841.14645] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 04/05/2019] [Accepted: 04/11/2019] [Indexed: 11/25/2022]
Abstract
The objective of this study was to evaluate the effects of a hot-water extract of defatted Camellia oleifera seeds (CSE) in carbon tetrachloride (CCl4 )-induced liver damage in rats. Wistar rats were separated into four groups including the normal (N) and CCl4 control (C) groups, which are fed a control diet, and the CCL (low-dose CSE) and CCH (high-dose CSE) groups, which are fed with a control diet plus different amount of CSE for an 8-week experimental period. Liver injury in the C, CCL, and CCH groups was induced by injecting CCl4 (i.p.) twice a week from the 5th week to the end of the study. In CCl4 -treated rats, the alanine transaminase (ALT) and aspartate transaminase (AST) activities and malondialdehyde (MDA) concentration significantly increased compared to the normal group. Lower antioxidative enzyme activities and higher proinflammatory cytokines, transforming growth factor-β (TGF-β) and hydroxyproline concentrations in the liver were also found in the CCl4 -treated group compared to the normal group. In contrast, the administration of CSE alleviated the biochemical and histopathological changes including inflammation, liver cell damage, and fibrosis caused by CCl4 in rats. Our results indicated that CSE exhibited hepatoprotective effects in CCl4 -induced liver hepatotoxicity through alleviating hepatic inflammation and fibrosis in rats. PRACTICAL APPLICATION: Camellia oleifera are widely used for edible oil production while the defatted seeds pomace is often discarded. We found that extract of C. oleifera pomace containing phenolic compounds, saponins, and polysaccharides showed protective effects chemical-driven liver damage and, therefore, may be used in further studies and developing functional foods.
Collapse
Affiliation(s)
- Jung Ko
- School of Nutrition and Health Sciences, Taipei Medical Univ., 250 Wuxing St, Taipei, 11031, Taiwan
| | - Wan-Ju Yeh
- Dept. of Food Science, College of Agriculture, Tunghai Univ., Taichung, Taiwan
| | - Wen-Chih Huang
- Dept. of Anatomic Pathology, Far Eastern Memorial Hospital, No.21, Sec. 2, Nanya S. Rd., Banciao District, New Taipei City, Taiwan.,College of Nursing, National Taipei Univ. of Nursing and Health Sciences, No.365, Mingte Road, Peitou District, Taipei City, Taiwan
| | - Hsin-Yi Yang
- Dept. of Nutritional Science, Fu Jen Catholic Univ., No.510, Zhongzheng Rd., Xinzhuang Dist., New Taipei City, 24205, Taiwan
| |
Collapse
|