1
|
Mohammad Omar J, Hai Y, Jin S. Hypoxia-induced factor and its role in liver fibrosis. PeerJ 2022; 10:e14299. [PMID: 36523459 PMCID: PMC9745792 DOI: 10.7717/peerj.14299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 10/04/2022] [Indexed: 12/12/2022] Open
Abstract
Liver fibrosis develops as a result of severe liver damage and is considered a major clinical concern throughout the world. Many factors are crucial for liver fibrosis progression. While advancements have been made to understand this disease, no effective pharmacological drug and treatment strategies have been established that can effectively prevent liver fibrosis or even could halt the fibrotic process. Most of those advances in curing liver fibrosis have been aimed towards mitigating the causes of fibrosis, including the development of potent antivirals to inhibit the hepatitis virus. It is not practicable for many individuals; however, a liver transplant becomes the only suitable alternative. A liver transplant is an expensive procedure. Thus, there is a significant need to identify potential targets of liver fibrosis and the development of such agents that can effectively treat or reverse liver fibrosis by targeting them. Researchers have identified hypoxia-inducible factors (HIFs) in the last 16 years as important transcription factors driving several facets of liver fibrosis, making them possible therapeutic targets. The latest knowledge on HIFs and their possible role in liver fibrosis, along with the cell-specific activities of such transcription factors that how they play role in liver fibrosis progression, is discussed in this review.
Collapse
Affiliation(s)
- Jan Mohammad Omar
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical, Harbin, Heilongjiang, China
| | - Yang Hai
- College of International Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Shizhu Jin
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical, Harbin, Heilongjiang, China
| |
Collapse
|
2
|
Flores Molina M, Abdelnabi MN, Mazouz S, Villafranca-Baughman D, Trinh VQH, Muhammad S, Bédard N, Osorio Laverde D, Hassan GS, Di Polo A, Shoukry NH. Distinct spatial distribution and roles of Kupffer cells and monocyte-derived macrophages in mouse acute liver injury. Front Immunol 2022; 13:994480. [PMID: 36248843 PMCID: PMC9562324 DOI: 10.3389/fimmu.2022.994480] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/05/2022] [Indexed: 11/18/2022] Open
Abstract
Macrophages are key regulators of inflammation and repair, but their heterogeneity and multiple roles in the liver are not fully understood. We aimed herein to map the intrahepatic macrophage populations and their function(s) during acute liver injury. We used flow cytometry, gene expression analysis, multiplex-immunofluorescence, 3D-reconstruction, and spatial image analysis to characterize the intrahepatic immune landscape in mice post-CCl4-induced acute liver injury during three distinct phases: necroinflammation, and early and late repair. We observed hepatocellular necrosis and a reduction in liver resident lymphocytes during necroinflammation accompanied by the infiltration of circulating myeloid cells and upregulation of inflammatory cytokines. These parameters returned to baseline levels during the repair phase while pro-repair chemokines were upregulated. We identified resident CLEC4F+ Kupffer cells (KCs) and infiltrating IBA1+CLEC4F- monocyte-derived macrophages (MoMFs) as the main hepatic macrophage populations during this response to injury. While occupying most of the necrotic area, KCs and MoMFs exhibited distinctive kinetics, distribution and morphology at the site of injury. The necroinflammation phase was characterized by low levels of KCs and a remarkable invasion of MoMFs suggesting their potential role in phagoctosing necrotic hepatocytes, while opposite kinetics/distribution were observed during repair. During the early repair phase, yolksac - derived KCs were restored, whereas MoMFs diminished gradually then dissipated during late repair. MoMFs interacted with hepatic stellate cells during the necroinflammatory and early repair phases, potentially modulating their activation state and influencing their fibrogenic and pro-repair functions that are critical for wound healing. Altogether, our study reveals novel and distinct spatial and temporal distribution of KCs and MoMFs and provides insights into their complementary roles during acute liver injury.
Collapse
Affiliation(s)
- Manuel Flores Molina
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Mohamed N. Abdelnabi
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Sabrina Mazouz
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Deborah Villafranca-Baughman
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de neurosciences, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Vincent Quoc-Huy Trinh
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Shafi Muhammad
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Nathalie Bédard
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - David Osorio Laverde
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Ghada S. Hassan
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Adriana Di Polo
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de neurosciences, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Naglaa H. Shoukry
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de médecine, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
- *Correspondence: Naglaa H. Shoukry,
| |
Collapse
|
3
|
Intrinsic and Extrinsic Control of Hepatocellular Carcinoma by TAM Receptors. Cancers (Basel) 2021; 13:cancers13215448. [PMID: 34771611 PMCID: PMC8582520 DOI: 10.3390/cancers13215448] [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: 10/01/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Tyro3, Axl, and MerTK are receptor tyrosine kinases of the TAM family, which are activated by their ligands Gas6 and Protein S. TAM receptors have large physiological implications, including the removal of dead cells, activation of immune cells, and prevention of bleeding. In the last decade, TAM receptors have been suggested to play a relevant role in liver fibrogenesis and the development of hepatocellular carcinoma. The understanding of TAM receptor functions in tumor cells and their cellular microenvironment is of utmost importance to advances in novel therapeutic strategies that conquer chronic liver disease including hepatocellular carcinoma. Abstract Hepatocellular carcinoma (HCC) is the major subtype of liver cancer, showing high mortality of patients due to limited therapeutic options at advanced stages of disease. The receptor tyrosine kinases Tyro3, Axl and MerTK—belonging to the TAM family—exert a large impact on various aspects of cancer biology. Binding of the ligands Gas6 or Protein S activates TAM receptors causing homophilic dimerization and heterophilic interactions with other receptors to modulate effector functions. In this context, TAM receptors are major regulators of anti-inflammatory responses and vessel integrity, including platelet aggregation as well as resistance to chemotherapy. In this review, we discuss the relevance of TAM receptors in the intrinsic control of HCC progression by modulating epithelial cell plasticity and by promoting metastatic traits of neoplastic hepatocytes. Depending on different etiologies of HCC, we further describe the overt role of TAM receptors in the extrinsic control of HCC progression by focusing on immune cell infiltration and fibrogenesis. Additionally, we assess TAM receptor functions in the chemoresistance against clinically used tyrosine kinase inhibitors and immune checkpoint blockade in HCC progression. We finally address the question of whether inhibition of TAM receptors can be envisaged for novel therapeutic strategies in HCC.
Collapse
|
4
|
Shojaei Jeshvaghani Z, Arefian E, Asgharpour S, Soleimani M. Latency-Associated Transcript-Derived MicroRNAs in Herpes Simplex Virus Type 1 Target SMAD3 and SMAD4 in TGF-β/Smad Signaling Pathway. IRANIAN BIOMEDICAL JOURNAL 2021; 25:169-79. [PMID: 33546553 DOI: 10.29252/ibj.25.3.169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Background During its latent infection, hepatic stellate cell (HSV-1) produces only a micro RNA (miRNA) precursor called latency-associated transcript (LAT), which encodes six distinct miRNAs. Recent studies have suggested that some of these miRNAs could target cellular mRNAs. One of the key cell signaling pathways that can be affected by HSV-1 is the TGF-β/Smad pathway. Herein, we investigated the potential role of the LAT as well as three LAT-derived miRNAs in targeting SMAD3 and SMAD4, as two main mediators in TGF-β/Smad. Methods The selection of LAT-derived miRNAs was based on the search results obtained from an online miRNA prediction tool. HEK293T cells were transfected with each miRNA-expressing lentivector and with the construct-expressing LAT. To survey the effect of LAT on the expression of pro-fibrotic markers, we transfected LX-2 cells with LAT construct. The impact of viral miRNA overexpression on SMADs and fibrotic markers was measured by quantitative PCR and luciferase assays. Results Among the LAT-derived miRNAs, miR-H2, miR-H3, and miR-H4 were selected for the study. Our results demonstrated that while miR-H2 binds to both SMAD mRNAs, miR-H3 and miR-H4 inhibit only the expression of the SMAD4 and SMAD3, respectively. Transfection of the LX-2 with LAT also decreased pro-fibrotic genes expression. Conclusion Our findings display that LAT negatively regulates TGF-β/Smad through targeting SMAD3 and SMAD4 by its miRNAs. These viral miRNAs can also contribute to the development of therapeutic interventions in diseases for which prevention or treatment can be achieved through targeting TGF-β pathway.
Collapse
Affiliation(s)
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | | | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
5
|
Latency-Associated Transcript-Derived MicroRNAs in Herpes Simplex Virus Type 1 Target SMAD3 and SMAD4 in TGF-β/Smad Signaling Pathway. IRANIAN BIOMEDICAL JOURNAL 2021. [PMID: 33546553 PMCID: PMC8183387 DOI: 10.52547/ibj.25.3.169] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Background: During its latent infection, HSV-1 produces only a miRNA precursor called LAT, which encodes six distinct miRNAs. Recent studies have suggested that some of these miRNAs could target cellular mRNAs. One of the key cell signaling pathways that can be affected by HSV-1 is the TGF-β/Smad pathway. Herein, we investigated the potential role of the LAT as well as three LAT-derived miRNAs in targeting SMAD3 and SMAD4, as two main mediators in TGF-β/Smad. Methods: The selection of LAT-derived miRNAs was based on the search results obtained from an online miRNA prediction tool. HEK293T cells were transfected with each miRNA-expressing lentivector and with the construct-expressing LAT. To survey the effect of LAT on the expression of pro-fibrotic markers, we transfected LX-2 cells with LAT construct. The impact of viral miRNA overexpression on SMADs and fibrotic markers was measured by qPCR and luciferase assays. Results: Among the LAT-derived miRNAs, miR-H2, miR-H3, and miR-H4 were selected for the study. Our results demonstrated that while miR-H2 binds to both SMAD mRNAs, miR-H3 and miR-H4 inhibit only the expression of the SMAD4 and SMAD3, respectively. Transfection of the LX-2 with LAT also decreased pro-fibrotic genes expression. Conclusion: Our findings display that LAT negatively regulates TGF-β/Smad through targeting SMAD3 and SMAD4 by its miRNAs. These viral miRNAs can also contribute to the development of therapeutic interventions in diseases for which prevention or treatment can be achieved through targeting TGF-β pathway.
Collapse
|
6
|
Huang M, Fu M, Wang J, Xia C, Zhang H, Xiong Y, He J, Liu J, Liu B, Pan S, Liu F. TGF-β1-activated cancer-associated fibroblasts promote breast cancer invasion, metastasis and epithelial-mesenchymal transition by autophagy or overexpression of FAP-α. Biochem Pharmacol 2021; 188:114527. [PMID: 33741330 DOI: 10.1016/j.bcp.2021.114527] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/03/2021] [Accepted: 03/12/2021] [Indexed: 02/07/2023]
Abstract
Cancer-associated fibroblasts (CAFs) play an important role in the initiation, metastasis, and invasion of breast cancer. However, whether autophagy acts as a tumor promotion mechanism by inducing epithelial-mesenchymal transition (EMT) is still controversial and remains undefined at the mechanistic levels. In this study, we investigated whether autophagy or FAP-α is required for the invasion, pulmonary metastasis and EMT of breast cancer cells and underlying mechanism. We employed an in vitro model of NIH3T3 fibroblasts treated with H2O2 and confirmed that TGF-β1 could convert fibroblasts into CAFs through autophagy under oxidative stress in the tumor microenvironment. Modulation of autophagy by rapamycin, 3-methyladenine or ATG-5 knockdown regulated the expression of CAFs markers, suggesting a role of autophagy in the tumor promotion mechanism of TGF-β1-induced CAFs activation. Furthermore, we established an indirect co-culture model and a mixed xenograft as a corresponding in vivo model. We demonstrated that TGF-β1-activated CAFs promote tumor invasion, pulmonary metastasis and EMT, which act through autophagy and overexpression of FAP-α in both models, while autophagy inhibitor 3-methyladenine blocked these effects induced by TGF-β1-activated CAFs. Moreover, the co-localization of LC3β and EMT marker vimentin in mixed xenograft also revealed that TGF-β1-activated CAFs promote tumor growth, pulmonary metastasis, and EMT program partly through autophagy. In addition, knockdown of FAP-α resulted in reversed EMT and abolished tumor invasion and pulmonary metastasis induced by TGF-β1-activated CAFs. Taken together, we conclude that both autophagy and FAP-α are required for breast cancer cell invasion and metastasis. Targeting autophagy or FAP-α rather than both can serve as a potential approach to improve the prognosis for human breast cancer.
Collapse
Affiliation(s)
- Minli Huang
- Clinical Pharmacology Institute, Nanchang University, Nanchang 330031, PR China
| | - Mengru Fu
- Clinical Pharmacology Institute, Nanchang University, Nanchang 330031, PR China
| | - Jia Wang
- Clinical Pharmacology Institute, Nanchang University, Nanchang 330031, PR China
| | - Chunhua Xia
- Clinical Pharmacology Institute, Nanchang University, Nanchang 330031, PR China
| | - Hong Zhang
- Clinical Pharmacology Institute, Nanchang University, Nanchang 330031, PR China
| | - Yuqing Xiong
- Clinical Pharmacology Institute, Nanchang University, Nanchang 330031, PR China
| | - Jiake He
- The Second Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Jianming Liu
- Clinical Pharmacology Institute, Nanchang University, Nanchang 330031, PR China
| | - Bingchen Liu
- Clinical Pharmacology Institute, Nanchang University, Nanchang 330031, PR China
| | - Siyi Pan
- Clinical Pharmacology Institute, Nanchang University, Nanchang 330031, PR China
| | - Fanglan Liu
- Clinical Pharmacology Institute, Nanchang University, Nanchang 330031, PR China.
| |
Collapse
|
7
|
Zhang Q, Chen K, Wu T, Song H. Swertiamarin ameliorates carbon tetrachloride-induced hepatic apoptosis via blocking the PI3K/Akt pathway in rats. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2018; 23:21-28. [PMID: 30627006 PMCID: PMC6315090 DOI: 10.4196/kjpp.2019.23.1.21] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 04/04/2018] [Accepted: 06/01/2018] [Indexed: 01/07/2023]
Abstract
Swertiamarin (STM) is an iridoid compound that is present in the Gentianaceae swertia genus. Here we investigated antiapoptotic effects of STM on carbon tetrachloride (CCl4)-induced liver injury and its possible mechanisms. Adult male Sprague Dawley rats were randomly divided into a control group, an STM 200 mg/kg group, a CCl4 group, a CCl4+STM 100 mg/kg group, and a CCl4+STM 200 mg/kg group. Rats in experimental groups were subcutaneously injected with 40% CCl4 twice weekly for 8 weeks. STM (100 and 200 mg/kg per day) was orally given to experimental rats by gavage for 8 consecutive weeks. Hepatocyte apoptosis was determined by TUNEL assay and the expression levels of Bcl-2, Bax, and cleaved caspase-3 proteins were evaluated by western blot analysis. The expression of TGF-β1, collagen I, collagen III, CTGF and fibronectin mRNA were estimated by qRT-PCR. The results showed that STM significantly reduced the number of TUNEL-positive cells compared with the CCl4 group. The levels of Bax and cleaved caspase-3 proteins, and TGF-β1, collagen I, collagen III, CTGF, and fibronectin mRNA were significantly reduced by STM compared with the CCl4 group. In addition, STM markedly abrogated the repression of Bcl-2 by CCl4. STM also attenuated the activation of the PI3K/Akt pathway in the liver. These results suggested that STM ameliorated CCl4-induced hepatocyte apoptosis in rats.
Collapse
Affiliation(s)
- Qianrui Zhang
- Department of Pharmacy, General Hospital of the Yangtze River Shipping, Wuhan 430022, China
| | - Kang Chen
- Department of Pharmacy, Huanggang Central Hospital, Huanggang 438000, China
| | - Tao Wu
- Department of Pharmacy, Wuhan NO.4 Hospital, Wuhan Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongping Song
- Department of Pharmacy, Wuhan NO.4 Hospital, Wuhan Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
8
|
Caja L, Dituri F, Mancarella S, Caballero-Diaz D, Moustakas A, Giannelli G, Fabregat I. TGF-β and the Tissue Microenvironment: Relevance in Fibrosis and Cancer. Int J Mol Sci 2018. [PMID: 29701666 DOI: 10.3390/ijms19051294.pmid:29701666;pmcid:pmc5983604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
Transforming growth factor-β (TGF-β) is a cytokine essential for the induction of the fibrotic response and for the activation of the cancer stroma. Strong evidence suggests that a strong cross-talk exists among TGF-β and the tissue extracellular matrix components. TGF-β is stored in the matrix as part of a large latent complex bound to the latent TGF-β binding protein (LTBP) and matrix binding of latent TGF-β complexes, which is required for an adequate TGF-β function. Once TGF-β is activated, it regulates extracellular matrix remodelling and promotes a fibroblast to myofibroblast transition, which is essential in fibrotic processes. This cytokine also acts on other cell types present in the fibrotic and tumour microenvironment, such as epithelial, endothelial cells or macrophages and it contributes to the cancer-associated fibroblast (CAF) phenotype. Furthermore, TGF-β exerts anti-tumour activity by inhibiting the host tumour immunosurveillance. Aim of this review is to update how TGF-β and the tissue microenvironment cooperate to promote the pleiotropic actions that regulate cell responses of different cell types, essential for the development of fibrosis and tumour progression. We discuss recent evidences suggesting the use of TGF-β chemical inhibitors as a new line of defence against fibrotic disorders or cancer.
Collapse
Affiliation(s)
- Laia Caja
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Box 582, 75123 Uppsala, Sweden.
| | - Francesco Dituri
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Serena Mancarella
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Daniel Caballero-Diaz
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Gran Via de l'Hospitalet, 199, 08908 Barcelona, Spain.
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Aristidis Moustakas
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Box 582, 75123 Uppsala, Sweden.
| | - Gianluigi Giannelli
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Gran Via de l'Hospitalet, 199, 08908 Barcelona, Spain.
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, L'Hospitalet, 08907 Barcelona, Spain.
| |
Collapse
|
9
|
Caja L, Dituri F, Mancarella S, Caballero-Diaz D, Moustakas A, Giannelli G, Fabregat I. TGF-β and the Tissue Microenvironment: Relevance in Fibrosis and Cancer. Int J Mol Sci 2018; 19:ijms19051294. [PMID: 29701666 PMCID: PMC5983604 DOI: 10.3390/ijms19051294] [Citation(s) in RCA: 227] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/17/2018] [Accepted: 04/24/2018] [Indexed: 12/14/2022] Open
Abstract
Transforming growth factor-β (TGF-β) is a cytokine essential for the induction of the fibrotic response and for the activation of the cancer stroma. Strong evidence suggests that a strong cross-talk exists among TGF-β and the tissue extracellular matrix components. TGF-β is stored in the matrix as part of a large latent complex bound to the latent TGF-β binding protein (LTBP) and matrix binding of latent TGF-β complexes, which is required for an adequate TGF-β function. Once TGF-β is activated, it regulates extracellular matrix remodelling and promotes a fibroblast to myofibroblast transition, which is essential in fibrotic processes. This cytokine also acts on other cell types present in the fibrotic and tumour microenvironment, such as epithelial, endothelial cells or macrophages and it contributes to the cancer-associated fibroblast (CAF) phenotype. Furthermore, TGF-β exerts anti-tumour activity by inhibiting the host tumour immunosurveillance. Aim of this review is to update how TGF-β and the tissue microenvironment cooperate to promote the pleiotropic actions that regulate cell responses of different cell types, essential for the development of fibrosis and tumour progression. We discuss recent evidences suggesting the use of TGF-β chemical inhibitors as a new line of defence against fibrotic disorders or cancer.
Collapse
Affiliation(s)
- Laia Caja
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Box 582, 75123 Uppsala, Sweden.
| | - Francesco Dituri
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Serena Mancarella
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Daniel Caballero-Diaz
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Gran Via de l'Hospitalet, 199, 08908 Barcelona, Spain.
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Aristidis Moustakas
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Box 582, 75123 Uppsala, Sweden.
| | - Gianluigi Giannelli
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Gran Via de l'Hospitalet, 199, 08908 Barcelona, Spain.
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, L'Hospitalet, 08907 Barcelona, Spain.
| |
Collapse
|
10
|
Protective effect of an L-type calcium channel blocker, amlodipine, on paracetamol-induced hepatotoxicity in rats. Hum Exp Toxicol 2018; 37:1169-1179. [DOI: 10.1177/0960327118758382] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Paracetamol (P), one of the most popular and commonly used analgesic and antipyretic agents, causes hepatotoxicity in overdoses. Amlodipine (AML), an L-type calcium channel blocker, has been shown to have anti-inflammatory activity by reversing the effect of calcium in the inflammation pathogenesis. In this study, the hepatoprotective activity of AML on P-induced hepatotoxicity was evaluated. Thirty male albino Wistar rats were divided into five groups: (1) control, (2) 2 g/kg of P, (3) 2 g/kg of P + 5 mg/kg of AML, (4) 2 g/kg of P + 10 mg/kg of AML, and (5) 10 mg/kg of AML. Some liver enzymes, oxidative parameters, cytokine mRNA expressions, histopathology, and immunohistochemical studies were performed in liver and blood samples. The serum levels of alanine aminotransferase and aspartate aminotransferase and the mRNA expression of tumor necrosis factor-alpha (TNF-α) and transforming growth factor-beta in the liver tissues were significantly increased in the group treated with P. The superoxide dismutase and glutathione parameters decreased and malondialdehyde levels increased in the livers of the rats treated with P. All these parameters were increased with both doses of the AML similar to the control group. A histopathological examination of the liver showed that AML administration ameliorated the P-induced inflammatory liver damage. In immunohistochemical staining, the expression of TNF-α in the cytoplasm of the hepatocytes was increased in the P group but not in other treatment groups when compared to the control. In conclusion, AML treatment showed significant protective effects against P-induced hepatotoxicity by increasing the activity of antioxidants and reducing inflammatory cytokines.
Collapse
|
11
|
Pingitore P, Dongiovanni P, Motta BM, Meroni M, Lepore SM, Mancina RM, Pelusi S, Russo C, Caddeo A, Rossi G, Montalcini T, Pujia A, Wiklund O, Valenti L, Romeo S. PNPLA3 overexpression results in reduction of proteins predisposing to fibrosis. Hum Mol Genet 2017; 25:5212-5222. [PMID: 27742777 PMCID: PMC5886043 DOI: 10.1093/hmg/ddw341] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 09/28/2016] [Indexed: 01/22/2023] Open
Abstract
Liver fibrosis is a pathological scarring response to chronic hepatocellular injury and hepatic stellate cells (HSCs) are key players in this process. PNPLA3 I148M is a common variant robustly associated with liver fibrosis but the mechanisms underlying this association are unknown. We aimed to examine a) the effect of fibrogenic and proliferative stimuli on PNPLA3 levels in HSCs and b) the role of wild type and mutant PNPLA3 overexpression on markers of HSC activation and fibrosis. Here, we show that PNPLA3 is upregulated by the fibrogenic cytokine transforming growth factor-beta (TGF-β), but not by platelet-derived growth factor (PDGF), and is involved in the TGF-β-induced reduction in lipid droplets in primary human HSCs. Furthermore, we show that retinol release from human HSCs ex vivo is lower in cells with the loss-of-function PNPLA3 148M compared with 148I wild type protein. Stable overexpression of PNPLA3 148I wild type, but not 148M mutant, in human HSCs (LX-2 cells) induces a reduction in the secretion of matrix metallopeptidase 2 (MMP2), tissue inhibitor of metalloproteinase 1 and 2 (TIMP1 and TIMP2), which is mediated by retinoid metabolism. In conclusion, we show a role for PNPLA3 in HSC activation in response to fibrogenic stimuli. Moreover, we provide evidence to indicate that PNPLA3-mediated retinol release may protect against liver fibrosis by inducing a specific signature of proteins involved in extracellular matrix remodelling.
Collapse
Affiliation(s)
- Piero Pingitore
- Department of Molecular and Clinical Medicine, University of Gothenburg, Sweden
| | - Paola Dongiovanni
- Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Policlinico Milano, Milan, Italy
| | | | - Marica Meroni
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Saverio Massimo Lepore
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | | | - Serena Pelusi
- Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Policlinico Milano, Milan, Italy
| | - Cristina Russo
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Andrea Caddeo
- Department of Molecular and Clinical Medicine, University of Gothenburg, Sweden
| | - Giorgio Rossi
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy.,Liver Surgery and Transplant Unit, Fondazione IRCCS Ca' Granda Ospedale Policlinico Milano, Milan, Italy
| | - Tiziana Montalcini
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Arturo Pujia
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Olov Wiklund
- Department of Molecular and Clinical Medicine, University of Gothenburg, Sweden.,Cardiology Department, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Luca Valenti
- Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Policlinico Milano, Milan, Italy.,Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, University of Gothenburg, Sweden.,Clinical Nutrition Unit, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy.,Cardiology Department, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
12
|
Kim JY, An HJ, Kim WH, Gwon MG, Gu H, Park YY, Park KK. Anti-fibrotic Effects of Synthetic Oligodeoxynucleotide for TGF-β1 and Smad in an Animal Model of Liver Cirrhosis. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 8:250-263. [PMID: 28918026 PMCID: PMC5511593 DOI: 10.1016/j.omtn.2017.06.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/29/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022]
Abstract
Liver fibrosis is characterized by changes in tissue architecture and extracellular matrix composition. Liver fibrosis affects not only hepatocytes but also the non-parenchymal cells such as hepatic stellate cells (HSCs), which are essential for maintaining an intact liver structure and function. Transforming growth factor β1 (TGF-β1) is a multifunctional cytokine that induces liver fibrosis through activation of Smad signaling pathways. To improve a new therapeutic approach, synthetic TGF-β1/Smad oligodeoxynucleotide (ODN) was used to suppress both TGF-β1 expression and Smad transcription factor using a combination of antisense ODN and decoy ODN. The aims of this study are to investigate the anti-fibrotic effects of TGF-β1/Smad ODN on simultaneous suppressions of both Smad transcription factor and TGF-β1 mRNA expression in the hepatic fibrosis model in vitro and in vivo. Synthetic TGF-β1/Smad ODN effectively inhibits Smad binding activity and TGF-β1 expression. TGF-β1/Smad ODN attenuated the epithelial mesenchymal transition (EMT) and activation of HSCs in TGF-β1-induced AML12 and HSC-T6 cells. TGF-β1/Smad ODN prevented the fibrogenesis and deposition of collagen in CCl4-treated mouse model. Synthetic TGF-β1/Smad ODN demonstrates anti-fibrotic effects that are mediated by the suppression of fibrogenic protein and inflammatory cytokines. Therefore, synthetic TGF-β1/Smad ODN has substantial therapeutic feasibility for the treatment of liver fibrotic diseases.
Collapse
Affiliation(s)
- Jung-Yeon Kim
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Hyun-Jin An
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Woon-Hae Kim
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Mi-Gyeong Gwon
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Hyemin Gu
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Yoon-Yub Park
- Department of Physiology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Kwan-Kyu Park
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea.
| |
Collapse
|
13
|
Rókusz A, Veres D, Szücs A, Bugyik E, Mózes M, Paku S, Nagy P, Dezső K. Ductular reaction correlates with fibrogenesis but does not contribute to liver regeneration in experimental fibrosis models. PLoS One 2017; 12:e0176518. [PMID: 28445529 PMCID: PMC5405957 DOI: 10.1371/journal.pone.0176518] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/12/2017] [Indexed: 12/13/2022] Open
Abstract
Background and aims Ductular reaction is a standard component of fibrotic liver tissue but its function is largely unknown. It is supposed to interact with the matrix producing myofibroblasts and compensate the declining regenerative capacity of hepatocytes. The relationship between the extent of fibrosis—ductular reaction, proliferative activity of hepatocytes and ductular reaction were studied sequentially in experimental hepatic fibrosis models. Methods Liver fibrosis/cirrhosis was induced in wild type and TGFβ overproducing transgenic mice by carbon tetrachloride and thioacetamide administration. The effect of thioacetamide was modulated by treatment with imatinib and erlotinib. The extent of ductular reaction and fibrosis was measured by morphometry following cytokeratin 19 immunofluorescent labeling and Picro Sirius staining respectively. The proliferative activity of hepatocytes and ductular reaction was evaluated by BrdU incorporation. The temporal distribution of the parameters was followed and compared within and between different experimental groups. Results There was a strong significant correlation between the extent of fibrosis and ductular reaction in each experimental group. Although imatinib and erlotinib temporarily decreased fibrosis this effect later disappeared. We could not observe negative correlation between the proliferation of hepatocytes and ductular reaction in any of the investigated models. Conclusions The stringent connection between ductular reaction and fibrosis, which cannot be influenced by any of our treatment regimens, suggests that there is a close mutual interaction between them instead of a unidirectional causal relationship. Our results confirm a close connection between DR and fibrogenesis. However, since the two parameters changed together we could not establish a causal relationship and were unable to reveal which was the primary event. The lack of inverse correlation between the proliferation of hepatocytes and ductular reaction questions that ductular reaction can compensate for the failing regenerative activity of hepatocytes. No evidences support the persistent antifibrotic property of imatinib or erlotinib.
Collapse
Affiliation(s)
- András Rókusz
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Dániel Veres
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Armanda Szücs
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Edina Bugyik
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Miklós Mózes
- Institute of Pathophysiology, Semmelweis University, Budapest, Hungary
| | - Sándor Paku
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary.,Tumor Progression Research Group, Joint Research Organization of the Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Péter Nagy
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Katalin Dezső
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| |
Collapse
|
14
|
Iracheta-Vellve A, Petrasek J, Gyongyosi B, Satishchandran A, Lowe P, Kodys K, Catalano D, Calenda CD, Kurt-Jones EA, Fitzgerald KA, Szabo G. Endoplasmic Reticulum Stress-induced Hepatocellular Death Pathways Mediate Liver Injury and Fibrosis via Stimulator of Interferon Genes. J Biol Chem 2016; 291:26794-26805. [PMID: 27810900 DOI: 10.1074/jbc.m116.736991] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 10/07/2016] [Indexed: 01/02/2023] Open
Abstract
Fibrosis, driven by inflammation, marks the transition from benign to progressive stages of chronic liver diseases. Although inflammation promotes fibrogenesis, it is not known whether other events, such as hepatocyte death, are required for the development of fibrosis. Interferon regulatory factor 3 (IRF3) regulates hepatocyte apoptosis and production of type I IFNs. In the liver, IRF3 is activated via Toll-like receptor 4 (TLR4) signaling or the endoplasmic reticulum (ER) adapter, stimulator of interferon genes (STING). We hypothesized that IRF3-mediated hepatocyte death is an independent determinant of chemically induced liver fibrogenesis. To test this, we performed acute or chronic CCl4 administration to WT and IRF3-, Toll/Interleukin-1R (TIR) domain-containing adapter-inducing interferon-β (TRIF)-, TRIF-related adaptor molecule (TRAM)-, and STING-deficient mice. We report that acute CCl4 administration to WT mice resulted in early ER stress, activation of IRF3, and type I IFNs, followed by hepatocyte apoptosis and liver injury, accompanied by liver fibrosis upon repeated administration of CCl4 Deficiency of IRF3 or STING prevented hepatocyte death and fibrosis both in acute or chronic CCl4 In contrast, mice deficient in type I IFN receptors or in TLR4 signaling adaptors, TRAM or TRIF, upstream of IRF3, were not protected from hepatocyte death and/or fibrosis, suggesting that the pro-apoptotic role of IRF3 is independent of TLR signaling in fibrosis. Hepatocyte death is required for liver fibrosis with causal involvement of STING and IRF3. Thus, our results identify that IRF3, by its association with STING in the presence of ER stress, couples hepatocyte apoptosis with liver fibrosis and indicate that innate immune signaling regulates outcomes of liver fibrosis via modulation of hepatocyte death in the liver.
Collapse
Affiliation(s)
- Arvin Iracheta-Vellve
- From the Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Jan Petrasek
- From the Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Benedek Gyongyosi
- From the Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Abhishek Satishchandran
- From the Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Patrick Lowe
- From the Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Karen Kodys
- From the Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Donna Catalano
- From the Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Charles D Calenda
- From the Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Evelyn A Kurt-Jones
- From the Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Katherine A Fitzgerald
- From the Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Gyongyi Szabo
- From the Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| |
Collapse
|
15
|
Vasconcellos R, Alvarenga ÉC, Parreira RC, Lima SS, Resende RR. Exploring the cell signalling in hepatocyte differentiation. Cell Signal 2016; 28:1773-88. [DOI: 10.1016/j.cellsig.2016.08.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/18/2016] [Accepted: 08/18/2016] [Indexed: 02/08/2023]
|
16
|
Fullár A, Firneisz G, Regős E, Dudás J, Szarvas T, Baghy K, Ramadori G, Kovalszky I. Response of Hepatic Stellate Cells to TGFB1 Differs from the Response of Myofibroblasts. Decorin Protects against the Action of Growth Factor. Pathol Oncol Res 2016; 23:287-294. [PMID: 27495255 DOI: 10.1007/s12253-016-0095-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/27/2016] [Indexed: 12/15/2022]
Abstract
Regardless to the exact nature of damage, hepatic stellate cells (HSCs) and other non-parenchymal liver cells transform to activated myofibroblasts, synthesizing the accumulating extracellular matrix (ECM) proteins, and transforming growth factor-β1 (TGF-β1) plays a crucial role in this process. Later it was discovered that decorin, member of the small leucin rich proteoglycan family is able to inhibit this action of TGF-β1. The aim of our present study was to clarify whether HSCs and activated myofibroblasts of portal region exert identical or different response to TGF-β1 exposure, and the inhibitory action of decorin against the growth factor is a generalized phenomenon on myofibroblast of different origin? To this end we measured mRNA expression and production of major collagen components (collagen type I, III and IV) of the liver after stimulation and co-stimulation with TGF-β1 and decorin in primary cell cultures of HSCs and myofibroblasts (MFs). Production of matrix proteins, decorin and members of the TGF-β1 signaling pathways were assessed on Western blots. Messenger RNA expression of collagens and TIEG was quantified by real-time RT-PCR. HSCs and MFs responded differently to TGF-β1 exposure. In contrast to HSCs in which TGF-β1 stimulated the synthesis of collagen type I, type III, and type IV, only the increase of collagen type IV was detected in portal MFs. However, in a combined treatment, decorin seemed to interfere with TGF-β1 and its stimulatory effect was abolished. The different mode of TGF-β1 action is mirrored by the different activation of signaling pathways in activated HSCs and portal fibroblasts. In HSCs the activation of pSMAD2 whereas in myofibroblasts the activation of MAPK pathway was detected. The inhibitory effect of decorin was neither related to the Smad-dependent nor to the Smad-independent signaling pathways.
Collapse
Affiliation(s)
- Alexandra Fullár
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Gábor Firneisz
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Eszter Regős
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - József Dudás
- Department of Otorhinolaryngology, Medical University Innsbruck, Innsbruck, Austria
- Department of Gastroenterology and Endocrinology, George August University, Göttingen, Germany
| | - Tibor Szarvas
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Kornélia Baghy
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Giuliano Ramadori
- Department of Gastroenterology and Endocrinology, George August University, Göttingen, Germany
| | - Ilona Kovalszky
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary.
| |
Collapse
|
17
|
Mandal A, Raju S, Viswanathan C. Cryopreserved hepatic progenitor cells derived from human embryonic stem cells can arrest progression of liver fibrosis in rats. Cell Biol Int 2016; 40:1107-15. [PMID: 27453189 DOI: 10.1002/cbin.10649] [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: 11/25/2015] [Accepted: 07/22/2016] [Indexed: 11/06/2022]
Abstract
Hepatocytes generated from human embryonic stem cells (hESCs) are considered to be an excellent candidate for restoring the liver function deficiencies. We have earlier standardized a three-step differentiation protocol to generate functional hepatocyte-like cells (HLCs) from hESCs, which expressed the major hepatic markers. We have also found that the HLCs remain stable and functional even after extended period of in vitro culture and cryopreservation. In the present study, we have aimed to investigate the therapeutic potential of cryopreserved-thawed hESC-derived hepatic progenitor cells following transplantation in carbon tetrachloride-induced fibrotic rat livers. Significant therapeutic effects, including improved hepatic histology and normal serum biochemistry of hepatic enzymes along with increased survival rate, were observed in the cell transplanted rats. This result is an encouraging indication to develop methods for clinical application of hESC-derived hepatic lineage cells.
Collapse
Affiliation(s)
- Arundhati Mandal
- Regenerative Medicine, Reliance Life Sciences Pvt. Ltd., Dhirubhai Ambani Life Sciences Centre, R-282, TTC Industrial Area of MIDC, Thane Belapur Road, Rabale, Navi Mumbai, 400701, India
| | - Sheena Raju
- Regenerative Medicine, Reliance Life Sciences Pvt. Ltd., Dhirubhai Ambani Life Sciences Centre, R-282, TTC Industrial Area of MIDC, Thane Belapur Road, Rabale, Navi Mumbai, 400701, India
| | - Chandra Viswanathan
- Regenerative Medicine, Reliance Life Sciences Pvt. Ltd., Dhirubhai Ambani Life Sciences Centre, R-282, TTC Industrial Area of MIDC, Thane Belapur Road, Rabale, Navi Mumbai, 400701, India.
| |
Collapse
|
18
|
Li H, You H, Fan X, Jia J. Hepatic macrophages in liver fibrosis: pathogenesis and potential therapeutic targets. BMJ Open Gastroenterol 2016; 3:e000079. [PMID: 27252881 PMCID: PMC4885270 DOI: 10.1136/bmjgast-2016-000079] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 04/16/2016] [Accepted: 04/18/2016] [Indexed: 12/20/2022] Open
Abstract
Hepatic macrophages account for the largest non-parenchymal cell population in the liver. Recent studies have found that hepatic macrophages have different functions in different stages of experimental liver fibrosis. Some studies found that there are different types of hepatic macrophages in the liver, although others have suggested that hepatic macrophages could switch to different phenotypes in different environments. Many studies demonstrated that while hepatic macrophages promoted fibrosis through the recruitment of proinflammatory immune cells, and the secretion of proinflammatory cytokines and chemokines in the early stages, these also promoted the resolution of hepatic fibrosis through the secretion of matrix metalloproteinases in the late stages. This article will review the current role played by hepatic macrophages in liver fibrosis and the potential therapeutic targets that modulate hepatic macrophages.
Collapse
Affiliation(s)
- Hai Li
- Department of Hepatopancreatobiliary and Splenic Medicine , Affiliated Hospital, Logistics University of People's Armed Police Force , Tianjin , People's Republic of China
| | - Hong You
- Liver Research Center, Beijing Friendship Hospital, Capital Medial University , Beijing , People's Republic of China
| | - Xu Fan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine , Beijing , People's Republic of China
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medial University , Beijing , People's Republic of China
| |
Collapse
|
19
|
Gruionu G, Bazou D, Maimon N, Onita-Lenco M, Gruionu LG, Huang P, Munn LL. Implantable tissue isolation chambers for analyzing tumor dynamics in vivo. LAB ON A CHIP 2016; 16:1840-1851. [PMID: 27128791 PMCID: PMC5155583 DOI: 10.1039/c6lc00237d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Recruitment of new blood vessels from the surrounding tissue is central to tumor progression and involves a fundamental transition of the normal, organized vasculature into a dense disarray of vessels that infiltrates the tumor. At present, studying the co-development of the tumor and recruited normal tissue is experimentally challenging because many of the important events occur rapidly and over short length scales in a dense three-dimensional space. To overcome these experimental limitations, we partially confined tumors within biocompatible and optically clear tissue isolation chambers (TICs) and implanted them in mice to create a system that is more amenable to microscopic analysis. Our goal was to integrate the tumor into a recruited host tissue - complete with vasculature - and demonstrate that the system recapitulates relevant features of the tumor microenvironment. We show that the TICs allow clear visualization of the cellular events associated with tumor growth and progression at the host-tumor interface including cell infiltration, matrix remodeling and angiogenesis. The tissue within the chamber is viable for more than a month, and the process is robust in both the skin and brain. Treatment with losartan, an angiotensin II receptor antagonist, decreased the collagen density and fiber length in the TIC, consistent with the known activity of this drug. We further show that collagen fibers display characteristic tumor signatures and play a central role in angiogenesis, guiding the migration of tethered endothelial sprouts. The methodology combines accessible methods of microfabrication with animal models and will enable more informative studies of the cellular mechanisms of tumor progression.
Collapse
Affiliation(s)
- Gabriel Gruionu
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | | | | | | | | | | | | |
Collapse
|
20
|
DerHovanessian A, Weigt SS, Palchevskiy V, Shino MY, Sayah DM, Gregson AL, Noble PW, Palmer SM, Fishbein MC, Kubak BM, Ardehali A, Ross DJ, Saggar R, Lynch JP, Elashoff RM, Belperio JA. The Role of TGF-β in the Association Between Primary Graft Dysfunction and Bronchiolitis Obliterans Syndrome. Am J Transplant 2016; 16:640-9. [PMID: 26461171 PMCID: PMC4946573 DOI: 10.1111/ajt.13475] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 06/09/2015] [Accepted: 07/03/2015] [Indexed: 01/25/2023]
Abstract
Primary graft dysfunction (PGD) is a possible risk factor for bronchiolitis obliterans syndrome (BOS) following lung transplantation; however, the mechanism for any such association is poorly understood. Based on the association of TGF-β with acute and chronic inflammatory disorders, we hypothesized that it might play a role in the continuum between PGD and BOS. Thus, the association between PGD and BOS was assessed in a single-center cohort of lung transplant recipients. Bronchoalveolar lavage fluid concentrations of TGF-β and procollagen collected within 24 h of transplantation were compared across the spectrum of PGD, and incorporated into Cox models of BOS. Immunohistochemistry localized expression of TGF-β and its receptor in early lung biopsies posttransplant. We found an association between PGD and BOS in both bilateral and single lung recipients with a hazard ratio of 3.07 (95% CI 1.76-5.38) for the most severe form of PGD. TGF-β and procollagen concentrations were elevated during PGD (p < 0.01), and associated with increased rates of BOS. Expression of TGF-β and its receptor localized to allograft infiltrating mononuclear and stromal cells, and the airway epithelium. These findings validate the association between PGD and the subsequent development of BOS, and suggest that this association may be mediated by receptor/TGF-β biology.
Collapse
Affiliation(s)
- Ariss DerHovanessian
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, California
| | - S. Samuel Weigt
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, California
| | - Vyacheslav Palchevskiy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, California
| | - Michael Y. Shino
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, California
| | - David M. Sayah
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, California
| | - Aric L. Gregson
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, California
| | - Paul W. Noble
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles California
| | - Scott M. Palmer
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University, Durham, North Carolina
| | - Michael C. Fishbein
- Depatment of Pathology and Laboratory Medicine, University of California, Los Angeles, California
| | - Bernard M. Kubak
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, California
| | - Abbas Ardehali
- Division of Cardiothoracic Surgery, Department of Surgery, University of California, Los Angeles, California
| | - David J. Ross
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, California
| | - Rajan Saggar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, California
| | - Joseph P. Lynch
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, California
| | - Robert M. Elashoff
- Department of Biomathematics, University of California, Los Angeles, California
| | - John A. Belperio
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, Los Angeles, California
| |
Collapse
|
21
|
Tu T, Calabro SR, Lee A, Maczurek AE, Budzinska MA, Warner FJ, McLennan SV, Shackel NA. Hepatocytes in liver injury: Victim, bystander, or accomplice in progressive fibrosis? J Gastroenterol Hepatol 2015; 30:1696-704. [PMID: 26239824 DOI: 10.1111/jgh.13065] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/26/2015] [Indexed: 12/11/2022]
Abstract
Chronic liver disease causes significant morbidity and mortality through progressive fibrosis, cirrhosis, and liver cancer. The classical theory of fibrogenesis has hepatic stellate cells (HSCs) as the principal and only significant source of abnormal extracellular matrix (ECM). Further, HSCs have the major role in abnormal ECM turnover. It is the death of hepatocytes, as the initial target of injury, that initiates a sequence of events including the recruitment of inflammatory cells and activation of HSCs. Following this initial response, the ongoing insult to hepatocytes is regarded as perpetuating injury, but otherwise, hepatocytes are regarded as "victims" and "bystanders" in progressive fibrosis. Recent developments, however, challenge this view and suggest the concept of the hepatocyte being an active participant in liver injury. It is clear now that hepatocytes undergo phenotypic changes, adapt to injury, and react to the altered microenvironment. In this review, we describe studies showing that hepatocytes contribute to progressive fibrosis by direct manipulation of the surrounding ECM and through signaling to effector cells, particularly HSCs and intrahepatic immune cells. Together, these findings suggest an active "accomplice" role for the hepatocyte in progressive liver fibrosis and highlight novel pathways that could be targeted for development of future anti-fibrotic therapies.
Collapse
Affiliation(s)
- Thomas Tu
- Liver Injury and Cancer, Centenary Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Sarah R Calabro
- Liver Injury and Cancer, Centenary Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Aimei Lee
- Liver Injury and Cancer, Centenary Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Annette E Maczurek
- Liver Injury and Cancer, Centenary Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Magdalena A Budzinska
- Liver Injury and Cancer, Centenary Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Fiona J Warner
- Liver Injury and Cancer, Centenary Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Susan V McLennan
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Nicholas A Shackel
- Liver Injury and Cancer, Centenary Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,A. W. Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
22
|
Roth KJ, Copple BL. Role of Hypoxia-Inducible Factors in the Development of Liver Fibrosis. Cell Mol Gastroenterol Hepatol 2015; 1:589-597. [PMID: 28210703 PMCID: PMC5301877 DOI: 10.1016/j.jcmgh.2015.09.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 09/16/2015] [Indexed: 02/08/2023]
Abstract
Liver fibrosis remains a significant clinical problem in the United States and throughout the world. Although important advances in the understanding of this disease have been made, no effective pharmacologic agents have been developed that directly prevent or reverse the fibrotic process. Many of the successes in liver fibrosis treatment have been targeted toward treating the cause of fibrosis, such as the development of new antivirals that eradicate hepatitis virus. For many patients, however, this is not feasible, so a liver transplant remains the only viable option. Thus, there is a critical need to identify new therapeutic targets that will slow or reverse the progression of fibrosis in such patients. Research over the last 16 years has identified hypoxia-inducible factors (HIFs) as key transcription factors that drive many aspects of liver fibrosis, making them potential targets of therapy. In this review, we discuss the latest work on HIFs and liver fibrosis, including the cell-specific functions of these transcription factors in the development of liver fibrosis.
Collapse
Key Words
- BDL, bile duct ligation
- CCl4, carbon tetrachloride
- Ccr, C-C chemokine receptor
- FGF, fibroblast growth factor
- HGF, hepatocyte growth factor
- HIFs, hypoxia-inducible factors
- HSC, hepatic stellate cell
- Hepatic Stellate Cells
- Hypoxia-Inducible Factors
- Jmjd, Jumonji domain-containing
- Kupffer Cells
- Liver Fibrosis
- PAI-1, plasminogen activator inhibitor-1
- PDGF, platelet-derived growth factor
- Rgs, regulator of G-protein signaling
- TGF-β, transforming growth factor β
- VEGF, vascular endothelial growth factor
- α-SMA, α-smooth muscle actin
Collapse
Affiliation(s)
| | - Bryan L. Copple
- Correspondence Address correspondence to: Bryan L. Copple, PhD, Department of Pharmacology and Toxicology, Michigan State University, 1355 Bogue Street, B403 Life Sciences Building, East Lansing, Michigan 48824.Department of Pharmacology and ToxicologyMichigan State University1355 Bogue Street, B403 Life Sciences BuildingEast LansingMichigan 48824
| |
Collapse
|
23
|
Karcioglu SS, Palabiyik SS, Bayir Y, Karakus E, Mercantepe T, Halici Z, Albayrak A. The Role of RAAS Inhibition by Aliskiren on Paracetamol-Induced Hepatotoxicity Model in Rats. J Cell Biochem 2015; 117:638-46. [DOI: 10.1002/jcb.25313] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 08/13/2015] [Indexed: 12/19/2022]
Affiliation(s)
| | - Saziye Sezin Palabiyik
- Department of Pharmaceutical Toxicology; Faculty of Pharmacy; Ataturk University; Erzurum Turkey
| | - Yasin Bayir
- Department of Biochemistry; Faculty of Pharmacy; Ataturk University; Erzurum Turkey
| | - Emre Karakus
- Department of Pharmacology and Toxicology; Faculty of Veterinary Medicine; Ataturk University; Erzurum Turkey
| | - Tolga Mercantepe
- Department of Histology and Embryology, Faculty of Medicine; Recep Tayyip Erdogan University; Rize Turkey
| | - Zekai Halici
- Department of Pharmacology; Faculty of Medicine; Ataturk University; Erzurum Turkey
| | - Abdulmecit Albayrak
- Department of Pharmacology; Faculty of Medicine; Ataturk University; Erzurum Turkey
| |
Collapse
|
24
|
Human Keratoconus Cell Contractility is Mediated by Transforming Growth Factor-Beta Isoforms. J Funct Biomater 2015; 6:422-38. [PMID: 26096146 PMCID: PMC4493522 DOI: 10.3390/jfb6020422] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 05/29/2015] [Accepted: 06/10/2015] [Indexed: 01/05/2023] Open
Abstract
Keratoconus (KC) is a progressive disease linked to defects in the structural components of the corneal stroma. The extracellular matrix (ECM) is secreted and assembled by corneal keratocytes and regulated by transforming growth factor-β (TGF-β). We have previously identified alterations in the TGF-β pathway in human keratoconus cells (HKCs) compared to normal corneal fibroblasts (HCFs). In our current study, we seeded HKCs and HCFs in 3D-collagen gels to identify variations in contractility, and expression of matrix metalloproteases (MMPs) by HKCs in response the TGF-β isoforms. HKCs showed delayed contractility with decreased Collagen I:Collagen V ratios. TGF-β1 significantly increased ECM contraction, Collagen I, and Collagen V expression by HKCs. We also found that HKCs have significantly decreased Collagen I:Collagen III ratios suggesting a potential link to altered collagen isoform expression in KC. Our findings show that HKCs have significant variations in collagen secretion in a 3D collagen gel and have delayed contraction of the matrix compared to HCFs. For the first time, we utilize a collagen gel model to characterize the contractility and MMP expression by HKCs that may contribute to the pathobiology of KC.
Collapse
|
25
|
Liang B, Guo XL, Jin J, Ma YC, Feng ZQ. Glycyrrhizic acid inhibits apoptosis and fibrosis in carbon-tetrachloride-induced rat liver injury. World J Gastroenterol 2015; 21:5271-5280. [PMID: 25954100 PMCID: PMC4419067 DOI: 10.3748/wjg.v21.i17.5271] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 01/27/2015] [Accepted: 02/11/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate anti-apoptotic effects of glycyrrhizic acid (GA) against fibrosis in carbon tetrachloride (CCl4)-induced liver injury and its contributing factors.
METHODS: Liver fibrosis was induced by administration of CCl4 for 8 wk. Pathological changes in the liver of rats were examined by hematoxylin-eosin staining. Collagen fibers were detected by Sirius red staining. Hepatocyte apoptosis was determined by TUNEL assay and the expression levels of cleaved caspase-3, Bax, α-SMA, connective tissue growth factor (CTGF), matrix metalloproteinase (MMP) 2 and MMP9 proteins were evaluated by western blot analysis, and α-SMA mRNA, collagen type I and III mRNA were estimated by real-time PCR.
RESULTS: Treatment with GA significantly improved the pathological changes in the liver and markedly decreased the positive area of Sirius red compared with rats in the CCl4-treated group. TUNEL assay showed that GA significantly reduced the number of TUNEL-positive cells compared with the CCl4-treated group. The expression levels of cleaved caspase-3, Bax, α-SMA, CTGF, MMP2 and MMP9 proteins, and α-SMA mRNA, collagen type I and III mRNA were also significantly reduced by GA compared with the CCl4-treated group (P < 0.05).
CONCLUSION: GA treatment can ameliorate CCl4-induced liver fibrosis by inhibiting hepatocyte apoptosis and hepatic stellate cell activation.
Collapse
|
26
|
Chen Y, Williams V, Filippova M, Filippov V, Duerksen-Hughes P. Viral carcinogenesis: factors inducing DNA damage and virus integration. Cancers (Basel) 2014; 6:2155-86. [PMID: 25340830 PMCID: PMC4276961 DOI: 10.3390/cancers6042155] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/03/2014] [Accepted: 10/09/2014] [Indexed: 12/13/2022] Open
Abstract
Viruses are the causative agents of 10%-15% of human cancers worldwide. The most common outcome for virus-induced reprogramming is genomic instability, including accumulation of mutations, aberrations and DNA damage. Although each virus has its own specific mechanism for promoting carcinogenesis, the majority of DNA oncogenic viruses encode oncogenes that transform infected cells, frequently by targeting p53 and pRB. In addition, integration of viral DNA into the human genome can also play an important role in promoting tumor development for several viruses, including HBV and HPV. Because viral integration requires the breakage of both the viral and the host DNA, the integration rate is believed to be linked to the levels of DNA damage. DNA damage can be caused by both endogenous and exogenous factors, including inflammation induced by either the virus itself or by co-infections with other agents, environmental agents and other factors. Typically, cancer develops years to decades following the initial infection. A better understanding of virus-mediated carcinogenesis, the networking of pathways involved in transformation and the relevant risk factors, particularly in those cases where tumorigenesis proceeds by way of virus integration, will help to suggest prophylactic and therapeutic strategies to reduce the risk of virus-mediated cancer.
Collapse
Affiliation(s)
- Yan Chen
- Department of Basic Science, Loma Linda University, Loma Linda, CA 92354, USA.
| | - Vonetta Williams
- Department of Basic Science, Loma Linda University, Loma Linda, CA 92354, USA.
| | - Maria Filippova
- Department of Basic Science, Loma Linda University, Loma Linda, CA 92354, USA.
| | - Valery Filippov
- Department of Basic Science, Loma Linda University, Loma Linda, CA 92354, USA.
| | | |
Collapse
|
27
|
Xu Q, Sakai K, Suzuki Y, Tambo C, Sakai Y, Matsumoto K. Suppression of fibrogenic gene expression and liver fibrosis using a synthetic prostacyclin agonist. Biomed Res 2014; 34:241-50. [PMID: 24190236 DOI: 10.2220/biomedres.34.241] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Chronic injury and inflammation in the liver are associated with the development of liver fibrosis. Expressions of transforming growth factor-β1 (TGF-β1) and hepatocyte growth factor (HGF) participate in the development and suppression, respectively, of liver fibrosis. Here, we investigated the effect of ONO-1301, a synthetic prostaglandin I2/IP receptor agonist, on liver fibrosis and on changes in the hepatic expressions of genes that regulate the progression of fibrosis in mice. Liver fibrosis was caused by the repetitive administration of CCl4 for 12 weeks, with ONO-1301 being administered during the last 4 weeks. The expressions of fibrogenic genes: TGF-β1, connective tissue growth factor, α-smooth muscle actin, type-I collagen, and type-III collagen were upregulated by chronic liver injury, which was associated with the expansion of myofibroblasts and the development of liver fibrosis. Treatment with ONO-1301 increased hepatic HGF mRNA expression, but decreased the expressions of TGF-β1, connective tissue growth factor, α-smooth muscle actin, and type-I and type-III collagen, which was associated with the suppression of myofibroblast expansion and liver fibrosis. Neutralizing antibody for HGF significantly attenuated the suppressive action of ONO-1301 on liver fibrosis and fibrogenic gene expressions. The therapeutic action of ONO-1301 on liver fibrosis may have occurred partly through HGF-mediated pathways.
Collapse
Affiliation(s)
- Qing Xu
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | | | | | | | | | | |
Collapse
|
28
|
Gene expression profiling and secretome analysis differentiate adult-derived human liver stem/progenitor cells and human hepatic stellate cells. PLoS One 2014; 9:e86137. [PMID: 24516514 PMCID: PMC3906387 DOI: 10.1371/journal.pone.0086137] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Accepted: 12/04/2013] [Indexed: 12/20/2022] Open
Abstract
Adult-derived human liver stem/progenitor cells (ADHLSC) are obtained after primary culture of the liver parenchymal fraction. The cells are of fibroblastic morphology and exhibit a hepato-mesenchymal phenotype. Hepatic stellate cells (HSC) derived from the liver non-parenchymal fraction, present a comparable morphology as ADHLSC. Because both ADHLSC and HSC are described as liver stem/progenitor cells, we strived to extensively compare both cell populations at different levels and to propose tools demonstrating their singularity. ADHLSC and HSC were isolated from the liver of four different donors, expanded in vitro and followed from passage 5 until passage 11. Cell characterization was performed using immunocytochemistry, western blotting, flow cytometry, and gene microarray analyses. The secretion profile of the cells was evaluated using Elisa and multiplex Luminex assays. Both cell types expressed α-smooth muscle actin, vimentin, fibronectin, CD73 and CD90 in accordance with their mesenchymal origin. Microarray analysis revealed significant differences in gene expression profiles. HSC present high expression levels of neuronal markers as well as cytokeratins. Such differences were confirmed using immunocytochemistry and western blotting assays. Furthermore, both cell types displayed distinct secretion profiles as ADHLSC highly secreted cytokines of therapeutic and immuno-modulatory importance, like HGF, interferon-γ and IL-10. Our study demonstrates that ADHLSC and HSC are distinct liver fibroblastic cell populations exhibiting significant different expression and secretion profiles.
Collapse
|
29
|
White JF, Werkmeister JA, Bisucci T, Darby IA, Ramshaw JAM. Temporal variation in the deposition of different types of collagen within a porous biomaterial implant. J Biomed Mater Res A 2013; 102:3550-5. [DOI: 10.1002/jbm.a.35027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 10/31/2013] [Indexed: 01/24/2023]
Affiliation(s)
- Jacinta F. White
- CSIRO Materials Science and Engineering; Bayview Avenue Clayton Victoria 3169 Australia
| | - Jerome A. Werkmeister
- CSIRO Materials Science and Engineering; Bayview Avenue Clayton Victoria 3169 Australia
| | - Teresa Bisucci
- School of Medical Sciences; RMIT University; Bundoora Victoria 3083 Australia
| | - Ian A. Darby
- School of Medical Sciences; RMIT University; Bundoora Victoria 3083 Australia
| | - John A. M. Ramshaw
- CSIRO Materials Science and Engineering; Bayview Avenue Clayton Victoria 3169 Australia
| |
Collapse
|
30
|
Lee J, French B, Morgan T, French SW. The liver is populated by a broad spectrum of markers for macrophages. In alcoholic hepatitis the macrophages are M1 and M2. Exp Mol Pathol 2013; 96:118-25. [PMID: 24145004 PMCID: PMC3944995 DOI: 10.1016/j.yexmp.2013.09.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 09/27/2013] [Indexed: 12/19/2022]
Abstract
Background Liver cell injury in alcoholic hepatitis (AH) is in part, due to macrophage generated proinflammatory cytokines i.e., M1, M2a, M2b, and M2c might be involved in ALD. The T cell response to chemokines and cytokines differs not only when M1 and M2 macrophages are compared but even when individual M2 subtypes are profiled. Purpose In AH, M1 monocytes in the blood show increased sensitivity in the TNF-α response to LPS. Immunohistochemistry (IHC) studies showed that the liver sinusoids in ALD are abundantly populated by CD163 expressing type 2 macrophages. In this report, we profile many of the molecules associated with M1 and M2 macrophages in livers with AH using IHC. Methods Using immunofluorescent antibody-labeling, we profiled the receptors, cytokines and chemokines observed in M1, M2a, M2b, and M2c macrophages in liver biopsies from patients with AH. Results The increased CD 163 expression found in previous studies was confirmed as well an additional macrophage phenotypic marker CD206, suggesting that AH pathogenesis at least partially involves M2a and M2c macrophages. TGF-β was found to be robustly over expressed by liver sinusoidal macrophages. Macrophage expression of the phenotypic markers TLR-2, TLR-4 and TLR-8 – found in both M1 and M2 macrophages – as well as the chemokines CCL-1 and CCL-18 was found. However, IRF-4, which is related to IL-4 production and M2a polarization as well as the cytokines CCL-1 and Il-1β and the chemokine CXCL-1 were also observed, suggesting that M2a and M2b also play a role in AH pathogenesis. Conclusion Livers with AH show robust macrophage over expression of TGF-β, a growth factor more commonly associated with M2 type macrophages and mostly known for its fibrogenetic properties. However, our immunoprofiling of macrophage over expression also shows that AH is driven by receptors, interferons, and cytokines that are commonly associated not just with M2 macrophages, but with M1 as well. Thus, a complex interplay between different types of macrophages expressing a diverse array of molecules and receptors is involved in AH.
Collapse
Affiliation(s)
- James Lee
- Harbor-UCLA Medical Center Department of Pathology, Torrance, CA 90502, USA
| | - B French
- Harbor-UCLA Medical Center Department of Pathology, Torrance, CA 90502, USA
| | - T Morgan
- Veteran's Administration Long Beach California, Department of Medicine, USA
| | - Samuel W French
- Harbor-UCLA Medical Center Department of Pathology, Torrance, CA 90502, USA
| |
Collapse
|
31
|
Perepelyuk M, Terajima M, Wang AY, Georges PC, Janmey PA, Yamauchi M, Wells RG. Hepatic stellate cells and portal fibroblasts are the major cellular sources of collagens and lysyl oxidases in normal liver and early after injury. Am J Physiol Gastrointest Liver Physiol 2013; 304:G605-14. [PMID: 23328207 PMCID: PMC3602686 DOI: 10.1152/ajpgi.00222.2012] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Liver fibrosis is characterized by excessive deposition of extracellular matrix proteins by myofibroblasts derived from hepatic stellate cells and portal fibroblasts. Activation of these precursors to myofibroblasts requires matrix stiffness, which results in part from increased collagen cross-linking mediated by lysyl oxidase (LOX) family proteins. The aims of this study were to characterize the mechanical changes of early fibrosis, to identify the cells responsible for LOX production in early injury, and to determine which cells in normal liver produce collagens and elastins, which serve as substrates for LOXs early after injury. Hepatocytes and liver nonparenchymal cells were isolated from normal and early-injured liver and examined immediately for expression of LOXs and matrix proteins. We found that stellate cells and portal fibroblasts were the major cellular sources of fibrillar collagens and LOXs in normal liver and early after injury (1 day after bile duct ligation and 2 and 7 days after CCl(4) injury). Activity assays using stellate cells and portal fibroblasts in culture demonstrated significant increases in LOX family enzymatic activity as cells became myofibroblastic. LOX family-mediated deoxypyridinoline and pyridinoline cross-links increased after CCl(4)-mediated injury. There was a significant association between liver stiffness (as quantified by the shear storage modulus G') and deoxypyridinoline levels; increased deoxypyridinoline levels were also coincident with significantly increased elastic resistance to large strain deformations, consistent with increased cross-linking of the extracellular matrix. These data suggest a model in which the liver is primed to respond quickly to injury, activating potential mechanical feed-forward mechanisms.
Collapse
Affiliation(s)
- Maryna Perepelyuk
- 1Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania;
| | | | - Andrew Y. Wang
- 5University of Virginia Health System, Charlottesville, Virginia; and
| | - Penelope C. Georges
- 4Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania;
| | - Paul A. Janmey
- 2Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; ,4Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania;
| | - Mitsuo Yamauchi
- 6North Carolina Oral Health Institute, Durham, North Carolina
| | - Rebecca G. Wells
- 1Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; ,3Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania;
| |
Collapse
|
32
|
Anti-inflammatory effects of Stephania tetrandra S. Moore on interleukin-6 production and experimental inflammatory disease models. Mediators Inflamm 2012; 5:280-91. [PMID: 18475741 PMCID: PMC2365809 DOI: 10.1155/s0962935196000415] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Deregulation of interleukin-6 (IL-6) expression caused the synthesis and release of many inflammatory mediators. It is involved in chronic inflammation, autoimmune diseases, and malignancy. Stephania tetrandra S. Moore is a Chinese medicinal herb which has been used traditionary as a remedy for neuralgia and arthritis in China. To investigate the anti-inflammatory effects of S. tetrandra S. Moore in vitro and in vivo, its effects on the production of IL-6 and inflammatory mediators were analysed. When human monocytes/macrophages stimulated with silica were treated with 0.1–10 μg/ml S. tetranda S. Moore, the production of IL-6 was inhibited up to 50%. At these concentrations, it had no cytotoxicity effect on these cells. It also suppressed the production of IL-6 by alveolar macrophages stimulated with silica. In addition, it inhibited the release of superoxide anion and hydrogen peroxide from human monocytes/macrophages. To assess the anti-fibrosis effects of S. tetrandra S. Moore, its effects on in vivo experimental inflammatory models were evaluated. In the experimental silicosis model, IL-6 activities in the sera and in the culture supernatants of pulmonary fibroblasts were also inhibited by it. In vitro and in vivo treatment of S. tetrandra S. Moore reduced collagen production by rat lung fibroblasts and lung tissue. Also, S. tetrandra S. Moore reduced the levels of serum GOT and GPT in the rat cirrhosis model induced by CCL4, and it was effective in reducing hepatic fibrosis and nodular formation. Taken together, these data indicate that it has a potent anti-inflammatory and antifibrosis effect by reducing IL-6 production.
Collapse
|
33
|
Oliveira FDS, Silveira TRD, Matte U. Relationship between serum transforming growth factor ß1 and liver collagen content in rats treated with carbon tetrachloride. ARQUIVOS DE GASTROENTEROLOGIA 2012; 49:232-4. [DOI: 10.1590/s0004-28032012000300013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 06/15/2012] [Indexed: 11/22/2022]
Abstract
CONTEXT: Transforming Growth Factor ß1 (TGFß1) plays a fundamental role in fibrogenesis, although its importance as a biomarker of liver disease is still matter of debate. OBJECTIVE: Quantify serum TGFß1 and its association to liver collagen content in rats exposed to Carbon Tetrachloride (CCl4). METHODS: Rats were submitted to a fibrosis model using CCl4 and sacrificed after 6, 10, 12 and 16 weeks of treatment. Serum levels of TGFß1 were measured by ELISA and collagen content was defined by morphometric analysis. RESULTS: Serum levels of TGFß1 increased between 6 and 10 weeks, whereas collagen density increased between 12 and 16 weeks. A negative correlation was observed between liver collagen deposition and serum levels of TGFß1 (r = -0. 48; P<0. 05). CONCLUSION: Serum levels of TGFß1 were inversely proportional to collagen intensity in cirrhotic livers of rats exposed to CCl4, thus suggesting a limited use as biomarker in advanced liver disease.
Collapse
Affiliation(s)
| | | | - Ursula Matte
- Hospital de Clínicas de Porto Alegre; Hospital de Clínicas de Porto Alegre, Brasil
| |
Collapse
|
34
|
Harn HJ, Lin SZ, Hung SH, Subeq YM, Li YS, Syu WS, Ding DC, Lee RP, Hsieh DK, Lin PC, Chiou TW. Adipose-derived stem cells can abrogate chemical-induced liver fibrosis and facilitate recovery of liver function. Cell Transplant 2012; 21:2753-64. [PMID: 22776464 DOI: 10.3727/096368912x652959] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Adipose-derived stem cells (ADSCs) are easy to harvest and have the ability for self-renewal and to differentiate into various cell types, including those of the hepatic lineage. Studies on the use of ADSCs for liver transplantation are, however, limited. The objective of this study was to investigate the feasibility of using human ADSCs and to better understand their mechanism of action for the repair of liver damage in a thioacetamide (TAA)-induced model of chronic liver damage in the rat. To induce liver damage, 200 mg/kg TAA was injected intraperitoneally into Wistar rats every 3 days for 60 days. For cell therapy, 1 × 10(6) human ADSCs suspended in 300 μl of phosphate-buffered saline were transplanted into each experimental rat by direct liver injection. Immunohistochemistry showed that the transplanted ADSCs differentiated into albumin- and α-fetoprotein-secreting liver-like cells 1 week after transplantation. In addition, liver function recovered significantly, as determined by biochemical analyses that analyzed total bilirubin, prothrombin time, and albumin levels. The Metavir score, derived from histopathological analysis, also showed a significant decrease in liver fibrosis and inflammatory activity after ADSC transplantation. Finally, we found a reduction in the expression of α-smooth muscle actin, a marker of hepatic stellate cells, which produce collagen fiber, and an increase in the expression of matrix metalloproteinase-9, which degrades collagen fiber, after ADSC transplantation. These findings are consistent with abrogation of liver fibrosis in the ADSC therapy group. Consequently, these results suggest that ADSC transplantation may facilitate recovery from chronic liver damage and thus may have clinical applications.
Collapse
Affiliation(s)
- Horng-Jyh Harn
- Department of Pathology, China Medical University Hospital, Taichung, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Qiang G, Zhang L, Yang X, Xuan Q, Shi L, Zhang H, Chen B, Li X, Zu M, Zhou D, Guo J, Yang H, Du G. Effect of valsartan on the pathological progression of hepatic fibrosis in rats with type 2 diabetes. Eur J Pharmacol 2012; 685:156-64. [PMID: 22546234 DOI: 10.1016/j.ejphar.2012.04.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 04/04/2012] [Accepted: 04/05/2012] [Indexed: 12/17/2022]
Abstract
Currently there is no effective treatment for nonalcoholic fatty liver disease (NAFLD), especially hepatic fibrosis induced by type 2 diabetes. Valsartan maybe has beneficial effect on the liver disease. The aim of the present study was to investigate the effect of valsartan on the pathological progression of hepatic fibrosis in rats with type 2 diabetes. An animal model of hepatic fibrosis with type 2 diabetes was developed using a high-sucrose, high-fat diet and low-dose streptozotocin. Valsartan (15 mg/kg/day, i.g.) was orally administered for four months. The livers were removed to make hematoxylin-eosin (HE) staining and Picric acid-Sirius red staining, and immunohistochemistry staining of α-smooth-muscle-actin (α-SMA), transforming growth factor β1 (TGF-β1), tumor necrosis factor (TNF-α) and monocyte chemotactic protein-1 (MCP-1). Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining was performed to detect hepatocyte apoptosis. The liver mitochondria were isolated to measure the mitochondrial respiratory function. The results showed that valsartan significantly alleviated the lesion of hepatic steatosis and hepatic fibrosis by HE staining and Picric acid-Sirius red staining. Immunohistochemical staining suggested that the expression of α-SMA, TGF-β1, TNF-α and MCP-1 in liver tissue of diabetic rats was markedly reduced by valsartan. TUNEL staining showed that there were fewer TUNEL-positive apoptotic hepatocytes in valsartan group. In addition, valsartan restored the injured hepatic mitochondrial respiratory function. The findings demonstrated that valsartan prevented the pathological progression of hepatic fibrosis in type 2 diabetic rats, correlated with reducing α-SMA, TGF-β1, TNF-α and MCP-1 expression, also anti-apoptosis and mitochondria-protective potential.
Collapse
Affiliation(s)
- Guifen Qiang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Chen ZCC, Ekaputra AK, Gauthaman K, Adaikan PG, Yu H, Hutmacher DW. In vitro and in vivo analysis of co-electrospun scaffolds made of medical grade poly(ε-caprolactone) and porcine collagen. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2012; 19:693-707. [DOI: 10.1163/156856208784089580] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Z. C. C. Chen
- a Department of Obstetrics and Gynecology, National University of Singapore, Singapore
| | - A. K. Ekaputra
- b Graduate Program in Bioengineering, Faculty of Medicine, National University of Singapore, Singapore
| | - K. Gauthaman
- c Department of Obstetrics and Gynecology, National University of Singapore, Singapore
| | - P. G. Adaikan
- d Department of Obstetrics and Gynecology, National University of Singapore, Singapore
| | - H. Yu
- e Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute of Bioengineering and Nanotechnology, A*STAR, Singapore
| | - D. W. Hutmacher
- f Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, QLD 4059, Australia
| |
Collapse
|
37
|
Bin WT, Ma LM, Xu Q, Shi XL. Embryonic hepatocyte transplantation for hepatic cirrhosis: Efficacy and mechanism of action. World J Gastroenterol 2012; 18:309-22. [PMID: 22294837 PMCID: PMC3261526 DOI: 10.3748/wjg.v18.i4.309] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 07/14/2011] [Accepted: 07/21/2011] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the efficacy and mechanism of action of allogeneic embryonic hepatocyte transplantation for the treatment of hepatic cirrhosis.
METHODS: Rat embryonic hepatocytes were characterized by examining cell markers. Wistar rats with CCl4-induced cirrhosis were randomly divided into two groups: a model group receiving continuous CCl4, and a cell transplantation group receiving continuous CCl4 and transplanted with embryonic fluorescent-labeled hepatocytes. In addition, a normal control group was composed of healthy rats. All rats were sacrificed after 2 wk following the initiation of the cell transplant. Ultrasound, pathological analyses and serum biochemical tests were used to evaluate the efficacy of embryonic hepatocyte transplantation. To analyze the recovery status of cirrhotic hepatocytes and the signaling pathways influenced by embryonic hepatocyte transplantation, real-time polymerase chain reaction was performed to examine the mRNA expression of stellate activation-associated protein (STAP), c-myb, α smooth muscle actin (α-SMA) and endothelin-1 (ET-1). Western blotting and immunohistochemistry were employed to detect α-SMA and ET-1 protein expression in hepatic tissues.
RESULTS: Gross morphological, ultrasound and histopathological examinations, serum biochemical tests and radioimmunoassays demonstrated that hepatic cirrhosis was successfully established in the Wistar rats. Stem cell factor receptor (c-kit), hepatocyte growth factor receptor (c-Met), Nestin, α fetal protein, albumin and cytokeratin19 markers were observed in the rat embryonic hepatocytes. Following embryonic hepatocyte transplantation, there was a significant reversal in the gross appearance, ultrasound findings, histopathological properties, and serum biochemical parameters of the rat liver. In addition, after the activation of hepatic stellate cells and STAP signaling, α-SMA, c-myb and ET-1 mRNA levels became significantly lower than in the untreated cirrhotic group (P < 0.05). These levels, however, were not statistically different from those of the normal healthy group. Immunohistochemical staining and Western blot analyses revealed that α-SMA and ET-1 protein expression levels in the transplantation group were significantly lower than in the untreated cirrhotic group, but being not statistically different from the normal group.
CONCLUSION: Transplantation of embryonic hepatocytes in rats has therapeutic effects on cirrhosis. The described treatment may significantly reduce the expression of STAP and ET-1.
Collapse
|
38
|
Copple BL, Kaska S, Wentling C. Hypoxia-inducible factor activation in myeloid cells contributes to the development of liver fibrosis in cholestatic mice. J Pharmacol Exp Ther 2012; 341:307-16. [PMID: 22271822 DOI: 10.1124/jpet.111.189340] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Macrophages play an integral role in the development of liver fibrosis by releasing mediators, such as platelet-derived growth factor-B (PDGF-B) and transforming growth factor-β1, which stimulate hepatic stellate cell proliferation, chemotaxis, and collagen production. However, the mechanism by which chronic liver injury stimulates macrophages to release these mediators is not completely understood. We tested the hypothesis that chronic liver injury activates hypoxia-inducible factor (HIF) transcription factors in macrophages that regulate the production of mediators that promote fibrosis. To test this hypothesis, Cre/lox technology was used to generate myeloid cell-specific HIF-1α or HIF-1β knockout mice. When these mice were subjected to bile duct ligation (BDL), levels of α-smooth muscle actin and type I collagen in the liver were reduced compared with those of mice with normal levels of HIFs. The deficiency of HIFs in macrophages did not affect liver injury or inflammation after BDL but reduced PDGF-B mRNA and protein, suggesting that HIF activation in macrophages may promote fibrosis by regulating the production of PDGF-B. Consistent with a role for HIFs in liver fibrosis in cholestatic liver disease, nuclear HIF-1α protein was present in macrophages, hepatocytes, and fibroblasts in the livers from patients with primary biliary cirrhosis and primary sclerosing cholangitis. These studies demonstrate that HIFs are important regulators of profibrotic mediator production by macrophages during the development of liver fibrosis and suggest that HIFs may be a novel therapeutic target for the treatment of chronic liver disease in patients.
Collapse
Affiliation(s)
- Bryan L Copple
- Department of Pharmacology and Toxicology, Michigan State University, B403 Life Sciences Building, East Lansing, MI 48823, USA.
| | | | | |
Collapse
|
39
|
Chuang HY, Ng LT, Lin LT, Chang JS, Chen JY, Lin TC, Lin CC. Hydrolysable tannins of tropical almond show antifibrotic effects in TGF-β1-induced hepatic stellate cells. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2011; 91:2777-2784. [PMID: 21725979 DOI: 10.1002/jsfa.4521] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 04/28/2011] [Accepted: 05/16/2011] [Indexed: 05/31/2023]
Abstract
BACKGROUND Persistent activation of hepatic stellate cells (HSC-T6) has been known to cause liver fibrosis. In this study, our objective was to investigate the effects of chebulagic acid and chebulinic acid, two hydrolysable tannins of tropical almond (Terminalia chebula) fruits, on collagen synthesis and signal transduction in transforming growth factor-β1-stimulated HSC-T6 cells. The expression of Smad2, Smad3, Smad4, collagen I(α1)/III, and plasminogen activator inhibitor 1 (PAI-1) mRNAs was determined by reverse-transcription polymerase chain reaction and their protein levels were assessed by western blotting. RESULTS Results showed that chebulagic acid and chebulinic acid at 20 µmol L(-1) exhibited cytotoxic and anti-proliferative effects on HSC-T6 cells. They also significantly decreased the expression of Smd2, Smad3 and Smad4, and the synthesis of collagen, procollagen I (α1) and III, as well as suppressing the activation of PAI-1; these events consequently facilitated the resolution of fibrosis. CONCLUSION These results indicate that both chebulagic acid and chebulinic acid possess antifibrotic activity, and their mechanism of action could be through the inhibition of the Smad pathway.
Collapse
Affiliation(s)
- Hsin-Ying Chuang
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Taiwan
| | | | | | | | | | | | | |
Collapse
|
40
|
Rath T, Roderfeld M, Güler C, Wenzel C, Graf J, Beitinger F, Roeb E, Zachoval R. YKL-40 and transient elastography, a powerful team to assess hepatic fibrosis. Scand J Gastroenterol 2011; 46:1369-80. [PMID: 21905976 DOI: 10.3109/00365521.2011.613949] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Transient elastography (TE) is a non-invasive and accurate method for the diagnosis of severe hepatic fibrosis and cirrhosis (F = 3 and F = 4). However, the assessment of significant fibrosis (F = 2) by TE is impaired due to a high variation in the diagnostic accuracy. Within this study, we aim to compare the diagnostic value of TE and experimental biomarkers of liver fibrosis. MATERIAL AND METHODS A total of 55 patients with chronic liver disease of different etiologies were included in the study. Among them, patients with HCV infection represented the largest cohort (n = 25). Liver fibrosis was evaluated according to the Desmet/Scheuer score. All patients received TE. Serum concentrations of YKL-40, hyaluronic acid (HA), Laminin, C-terminal procollagen I peptide, MMP-9, TIMP-1, TIMP-2 and MMP-9/TIMP-1 complex were determined by ELISA. RESULTS In the total patient population, areas under the receiver operator characteristic curve (AUROC) for TE were 0.798 (F ≥ 2), 0.880 (F ≥ 3) and 1 (F = 4). Among the serum markers, highest diagnostic accuracies were calculated for YKL-40 for F ≥ 2 (0.792) and F ≥ 3 (0.914) and for YKL-40 and HA for F = 4 (both 0.936). In the subgroup of HCV patients, the following AUROCs for TE were calculated: 0.802 (F ≥ 2), 0.798 (F ≥ 3) and 0.998 (F = 4). YKL-40 exhibited the highest diagnostic accuracy of all biomarkers in the HCV population (0.880, 0.854 and 0.986, respectively). CONCLUSIONS YKL-40 is a powerful fibrosis marker with high diagnostic accuracy, in particular in HCV-associated liver disease. Its determination may confirm and improve the diagnostic accuracy of TE especially in early stages of liver fibrosis.
Collapse
Affiliation(s)
- Timo Rath
- Department of Internal Medicine, Division of Gastroenterology, Justus-Liebig-University Giessen, Giessen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Transforming growth factor-β (TGF-β) is a central regulator in chronic liver disease contributing to all stages of disease progression from initial liver injury through inflammation and fibrosis to cirrhosis and hepatocellular carcinoma. Liver-damage-induced levels of active TGF-β enhance hepatocyte destruction and mediate hepatic stellate cell and fibroblast activation resulting in a wound-healing response, including myofibroblast generation and extracellular matrix deposition. Being recognised as a major profibrogenic cytokine, the targeting of the TGF-β signalling pathway has been explored with respect to the inhibition of liver disease progression. Whereas interference with TGF-β signalling in various short-term animal models has provided promising results, liver disease progression in humans is a process of decades with different phases in which TGF-β or its targeting might have both beneficial and adverse outcomes. Based on recent literature, we summarise the cell-type-directed double-edged role of TGF-β in various liver disease stages. We emphasise that, in order to achieve therapeutic effects, we need to target TGF-β signalling in the right cell type at the right time.
Collapse
|
42
|
Segovia-Silvestre T, Reichenbach V, Fernández-Varo G, Vassiliadis E, Barascuk N, Morales-Ruiz M, Karsdal MA, Jiménez W. Circulating CO3-610, a degradation product of collagen III, closely reflects liver collagen and portal pressure in rats with fibrosis. FIBROGENESIS & TISSUE REPAIR 2011; 4:19. [PMID: 21813019 PMCID: PMC3170588 DOI: 10.1186/1755-1536-4-19] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Accepted: 08/03/2011] [Indexed: 12/19/2022]
Abstract
Background Hepatic fibrosis is characterized by intense tissue remodeling, mainly driven by matrix metalloproteinases. We previously identified CO3-610, a type III collagen neoepitope generated by matrix metalloproteinase (MMP)-9, and tested its performance as a fibrosis marker in rats with bile-duct ligation. In this study, we assessed whether CO3-610 could be used as a surrogate biomarker of liver fibrosis and portal hypertension in carbon tetrachloride-induced experimental fibrosis. Results For this study, 68 Wistar rats were used. Serum CO3-610 was measured by ELISA. Liver fibrosis was quantified by Sirius red staining. Serum hyaluronic acid (HA) was measured with a binding-protein assay. Gene expression of collagens I and III, Mmp2 and Mmp9, and tissue inhibitors of matrix metalloproteinase 1 (Timp1) and 2(Timp2) was quantified by PCR. Hemodynamic measurements were taken in a subgroup of animals. A close direct relationship was found between serum CO3-610 and hepatic collagen content (r = 0.78; P < 0.001), superior to that found for serum HA (r = 0.49; P < 0.05). CO3-610 levels in rats with severe fibrosis (43.5 ± 3.3 ng/mL, P < 0.001) and cirrhosis (60.6 ± 4.3 ng/mL, P < 0.001) were significantly higher than those in control animals (26.6 ± 1.3 ng/mL). Importantly, a highly significant relationship was found between serum CO3-610 and portal hypertension (r = 0.84; P < 0.001). Liver Mmp9 expression increased significantly in fibrotic animals but decreased to control levels in cirrhotic ones. Conclusions Circulating CO3-610 behaves as a reliable indicator of hepatic remodeling and portal hypertension in experimental fibrosis. This peptide could ultimately be a useful marker for the management of liver disease in patients.
Collapse
|
43
|
Abstract
Progression of fibrosis involves interstitial hypercellularity, matrix accumulation, and atrophy of epithelial structures, resulting in loss of normal function and ultimately organ failure. There is common agreement that the fibroblast/myofibroblast is the cell type most responsible for interstitial matrix accumulation and consequent structural deformations associated with fibrosis. During wound healing and progressive fibrotic events, fibroblasts transform into myofibroblasts acquiring smooth muscle features, most notably the expression of alpha-smooth muscle actin and synthesis of mesenchymal cell-related matrix proteins. In renal disease, glomerular mesangial cells also acquire a myofibroblast phenotype and synthesize the same matrix proteins. The origin of interstitial myofibroblasts during fibrosis is a matter of debate, where the cells are proposed to derive from resident fibroblasts, pericytes, perivascular adventitial, epithelial, and/or endothelial sources. Regardless of the origin of the cells, transforming growth factor-beta1 (TGF-β1) is the principal growth factor responsible for myofibroblast differentiation to a profibrotic phenotype and exerts its effects via Smad signaling pathways involving mitogen-activated protein kinase and Akt/protein kinase B. Additionally, reactive oxygen species (ROS) have important roles in progression of fibrosis. ROS are derived from a variety of enzyme sources, of which the nicotinamide adenine dinucleotide phosphate (NAD(P)H) oxidase family has been identified as a major source of superoxide and hydrogen peroxide generation in the cardiovasculature and kidney during health and disease. Recent evidence indicates that the NAD(P)H oxidase homolog Nox4 is most accountable for ROS-induced fibroblast and mesangial cell activation, where it has an essential role in TGF-β1 signaling of fibroblast activation and differentiation into a profibrotic myofibroblast phenotype and matrix production. Information on the role of ROS in mesangial cell and fibroblast signaling is incomplete, and further research on myofibroblast differentiation during fibrosis is warranted.
Collapse
|
44
|
Liu H, Wang X, Wang C, Yuan X, Bao H, Fu X, Chu Y. Molecular cloning, in vitro expression and bioactivity of rabbit transforming growth factor-beta receptor type II (rTGF-βRII). Vet Immunol Immunopathol 2010; 140:55-62. [PMID: 21167609 DOI: 10.1016/j.vetimm.2010.11.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Revised: 11/17/2010] [Accepted: 11/22/2010] [Indexed: 01/25/2023]
Abstract
Transforming growth factor-beta receptor II (TGF-βRII) is an attractive target for anti-scarring therapy in wound healing because it attenuates excessive TGF-β which has pleiotropic effects on the immune system. In the present study, the cDNA of rabbit TGF-βRII (rTGF-βRII) was amplified from rabbit peripheral blood by RT-PCR. The open reading frame of rTGF-βRII encodes a protein consisting of 567 amino acids, which contains a predicted transmembrane domain and a Serine/Threonine protein kinase domain similar to other identified mammalian TGF-βRIIs. The amino acid sequences of the biologically active, soluble rTGF-βRII and mouse, rat, human and chicken counterparts are 81%, 81%, 89% and 61%, respectively, identical. Recombinant soluble rTGF-βRII (rsTGF-βRII) fused with His tag was efficiently expressed in Escherichia coli BL21 (DE3) expression host strain. This fusion protein's molecular weight of ∼ 19 kDa was identified by SDS-PAGE and Western blotting. In vitro, purified rsTGF-βRII was able to inhibit the proliferation of keloid rabbit fibroblasts and decrease the level of collagen. These findings indicate that rTGF-βRII plays an important role in inhibiting the proliferation of keloid rabbit fibroblasts and provides the basis for investigations on the role of TGF-βRII in this important domestic species.
Collapse
Affiliation(s)
- Haifeng Liu
- Heilongjiang Province Key Laboratory for Anti-fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang 157011, PR China
| | | | | | | | | | | | | |
Collapse
|
45
|
|
46
|
Panakanti R, Pratap A, Yang N, Jackson JS, Mahato RI. Triplex forming oligonucleotides against type α1(I) collagen attenuates liver fibrosis induced by bile duct ligation. Biochem Pharmacol 2010; 80:1718-26. [PMID: 20816672 DOI: 10.1016/j.bcp.2010.08.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 08/08/2010] [Accepted: 08/25/2010] [Indexed: 12/20/2022]
Abstract
Liver fibrosis is a consequence of chronic liver disorders which lead to the accumulation of extracellular matrix (ECM). Particularly, there is an increased accumulation of collagen in the fibrotic liver. We have therefore used a triplex forming oligonucleotide (TFO) against the type α1(I) collagen and evaluated, whether it can attenuate liver fibrosis induced by common bile duct ligation (CBDL) in rats. There was a significant decrease in hydroxyproline levels and Masson's trichrome staining for collagen in TFO-treated CBDL groups compared to non-treated CBDL group. There was over expression of type α1(I) collagen, α-smooth muscle actin (α-SMA) and TGF-β1 expression in the CBDL group compared to TFO-treated CBDL group. Also, the serum alanine transaminase (ALT) and aspartate transaminase (AST) concentrations were less in the TFO treated group compared to non-treated CBDL group. There was also less neutrophils accumulation in TFO treated CBDL group assayed by myeloperoxidase (MPO) assay. These results suggests that TFO can be used to downregulate type 1 collagen gene expression and can alleviate liver fibrosis induced by common bile duct ligation.
Collapse
Affiliation(s)
- Ravikiran Panakanti
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | | | | | | | | |
Collapse
|
47
|
Marinho CC, Bretas T, Voieta I, Queiroz LCD, Ruiz-Guevara R, Teixeira AL, Antunes CM, Prata A, Lambertucci JR. Serum hyaluronan and collagen IV as non-invasive markers of liver fibrosis in patients from an endemic area for schistosomiasis mansoni: a field-based study in Brazil. Mem Inst Oswaldo Cruz 2010; 105:471-8. [DOI: 10.1590/s0074-02762010000400020] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2009] [Accepted: 09/04/2009] [Indexed: 12/27/2022] Open
|
48
|
Zakaria S, Youssef M, Moussa M, Akl M, El-Ahwany E, El-Raziky M, Mostafa O, Helmy AH, El-Hindawi A. Value of α-smooth muscle actin and glial fibrillary acidic protein in predicting early hepatic fibrosis in chronic hepatitis C virus infection. Arch Med Sci 2010; 6:356-65. [PMID: 22371771 PMCID: PMC3282512 DOI: 10.5114/aoms.2010.14255] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Revised: 03/12/2009] [Accepted: 04/10/2009] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION α-Smooth muscle actin (α-SMA)-positive hepatic stellate cells (HSCs) are pericytes responsible for fibrosis in chronic liver injury. The glial fibrillary acidic protein (GFAP), commonly expressed by astrocytes in the central nervous system, is expressed in vivo in the liver in a subpopulation of quiescent stellate cells. The reports concerning GFAP expression in human liver are still conflicting. The aim of the study is investigation the utility of GFAP compared to α-SMA as an indicator of early activated HSCs, in predicting fibrosis in chronic hepatitis C (CHC) patients. MATERIAL AND METHODS With immunohistochemistry and a semi-quantitative scoring system, the expressions of α-SMA and GFAP on HSCs in liver biopsies from patients with pure CHC (n = 34), hepatitis C virus-induced cirrhosis (n = 24), mixed CHC/schistosomiasis (n = 11) and normal controls (n = 10) were analysed. RESULTS The immunoreactivity of α-SMA and GFAP in perisinusoidal, periportal and pericentral areas was assessed. α-Smooth muscle actin and GFAP-positive HSCs were significantly increased in all diseased groups compared with normal controls. In pure CHC with or without cirrhosis, perisinusoidal α-SMA-positive HSCs were predominant in relation to GFAP-positive cells. On the other hand, GFAP-positive cells were predominant in the group of schistosomiasis as compared with the other diseased groups. It was noticed that expression of GFAP on perisinusoidal HSCs in CHC patients sequentially decreased with the progression of fibrosis. CONCLUSIONS Glial fibrillary acidic protein could represent a more useful marker than α-SMA of early activation of HSCs in CHC patients and seems to be an early indicator of hepatic fibrogenesis.
Collapse
Affiliation(s)
- Sohier Zakaria
- Department of Tropical Medicine, Cairo University, Cairo, Egypt
| | - Magdy Youssef
- Department of Hepato-Gastroenterology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Mona Moussa
- Department of Pathology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Maha Akl
- Department of Pathology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Eman El-Ahwany
- Department of Immunology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Maysa El-Raziky
- Department of Tropical Medicine, Cairo University, Cairo, Egypt
| | - Omaima Mostafa
- Department of Hepato-Gastroenterology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Ahmed-Hazem Helmy
- Department of Surgery, Theodor Bilharz Research Institute, Giza, Egypt
| | | |
Collapse
|
49
|
Copple BL, Bai S, Moon JOK. Hypoxia-inducible factor-dependent production of profibrotic mediators by hypoxic Kupffer cells. Hepatol Res 2010; 40:530-9. [PMID: 20412331 PMCID: PMC2886188 DOI: 10.1111/j.1872-034x.2010.00635.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
AIM Liver fibrosis develops when chronic liver injury stimulates cells in the liver to produce mediators that activate hepatic stellate cells and stimulate them to secrete collagen. Recent studies suggest that the hypoxia-regulated transcription factor, hypoxia-inducible factor-1alpha, is essential for upregulation of profibrotic mediators, such as platelet-derived growth factor, in the liver during the development of liver fibrosis. What remains unknown, however, is the cell type-specific regulation of profibrotic mediators by hypoxia-inducible factors. Accordingly, in the present study the hypothesis tested was that hypoxia-inducible factors regulate production of profibrotic mediators by hypoxic Kupffer cells. METHODS Kupffer cells were isolated from control mice and hypoxia-inducible factor-1beta-deficient mice and exposed to room air or 1% oxygen (i.e. hypoxia). Levels of profibrotic mediators were quantified by real-time polymerase chain reaction. RESULTS Exposure of Kupffer cells isolated from control mice to 1% oxygen activated hypoxia-inducible factor-1alpha, and increased mRNA levels of platelet-derived growth factor-B, vascular endothelial growth factor, angiopoietin-1 and monocyte chemotactic protein-1. Upregulation of all of these mediators by hypoxia was prevented in Kupffer cells isolated from hypoxia-inducible factor-1beta-deficient mice. CONCLUSION RESULTS from these studies suggest that hypoxia-inducible factors are critical regulators of profibrotic mediator production by hypoxic Kupffer cells.
Collapse
Affiliation(s)
- Bryan L. Copple
- Department of Pharmacology, Toxicology, and Experimental Therapeutics University of Kansas Medical Center, Kansas City, KS 66160
| | - Shan Bai
- Department of Pharmacology, Toxicology, and Experimental Therapeutics University of Kansas Medical Center, Kansas City, KS 66160
| | - Jeon-OK Moon
- Department of Pharmacology, Toxicology, and Experimental Therapeutics University of Kansas Medical Center, Kansas City, KS 66160,Department of Pharmacy, Pusan National University, Busan 609-735, South Korea
| |
Collapse
|
50
|
Allman M, Gaskin L, Rivera CA. CCl4-induced hepatic injury in mice fed a Western diet is associated with blunted healing. J Gastroenterol Hepatol 2010; 25:635-43. [PMID: 20370733 PMCID: PMC2953717 DOI: 10.1111/j.1440-1746.2009.06112.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Feeding a Western diet (WD) enriched in saturated fat protects against chronic alcoholic hepatitis. However, saturated fat induces lipotoxicity in cultured hepatocytes. The purpose of the present study was to elucidate the influence of WD on acute hepatic injury and healing. METHODS Male C57BL/6 mice were fed a purified control diet (CD) or WD enriched in palmitate and cholesterol. After 3 weeks, carbon tetrachloride (CCl(4)) was administered (0.1 microL/g, intraperitoneally). Hepatic inflammation and proliferation were assessed by immunostaining for neutrophils and intracellular adhesion molecule-1, and Ki67, respectively. Cytokine expression was analyzed by real-time polymerase chain reaction. Protein levels of peroxisome proliferator-activated receptor-gamma (PPAR-gamma) were assessed by western blotting. RESULTS Feeding a WD resulted in markedly greater histological evidence of necrosis and enhanced alanine aminotransferase activity (188 +/- 6.2 U/L) compared to CD-fed mice (99.1 +/- 6.3 U/L) by day 2 post-CCl(4). In contrast, WD blunted leukocyte accumulation in necrotic areas and the expression of cytokines (tumor necrosis factor-alpha and interleukin-6) involved in tissue regeneration. Diminished repair was further indexed by lower collagen-alphaI and Ki67 expression in the mice fed a WD. Finally, feeding a WD, as well as the treatment of cultured hepatocytes with palmitic acid, upregulated the expression of PPAR-gamma, which has been previously shown to prevent hepatic repair following CCl(4) exposure. CONCLUSIONS These findings suggest that impaired healing in WD-fed mice blunted recovery from acute injury and underscored the complex relationship between diet and hepatic injury.
Collapse
Affiliation(s)
| | | | - Chantal A. Rivera
- Send all correspondence to: Chantal A. Rivera, Ph.D., LSU Health Sciences Center, Dept. of Molecular and Cellular Physiology, 1501 Kings Hwy, Shreveport, LA 71130, Phone: 011 318-675-8140, Fax: 318-675-4217,
| |
Collapse
|