1
|
Jiang H, Fang Y, Wang Y, Li T, Lin H, Lin J, Pan T, Liu Q, Lv J, Chen D, Chen Y. FGF4 improves hepatocytes ferroptosis in autoimmune hepatitis mice via activation of CISD3. Int Immunopharmacol 2023; 116:109762. [PMID: 36702076 DOI: 10.1016/j.intimp.2023.109762] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/14/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023]
Abstract
Autoimmune hepatitis (AIH) is increasingly affecting human health but pharmacotherapies remain to be identified. Growing evidence reveals that ferroptosis, a newly recognized form of programmed cell death, is critical for AIH. However, the exact mechanisms of the ferroptotic cascade remain elusive. Data in this study showed that ferroptosis aggravation was associated with protectively-elevated fibroblast growth factor 4 (FGF4) expression in Concanavalin A (ConA)-induced AIH liver injury, with these effects being effectively reversed by Ferrostatin-1 (Fer-1). Moreover, hepatic Fgf4 depletion was more susceptible to lipid peroxidation and iron accumulation, as well as hepatic lesion and inflammation caused by ConA administration. Conversely, treatment with non-mitogenic recombinant FGF4 (rFGF4) mitigated liver damage and hepatocellular ferroptosis while being accompanied by the upregulation of CDGSH iron-sulfur domain-containing protein 3 (CISD3) in vivo and in vitro. Furthermore, CISD3 overexpression exhibited stronger resistance to ferroptosis while CISD3 knockdown reduced ferroptotic biomarkers cystine/glutamate transporter (xCT) and glutathione peroxidase 4(GPX4) in rFGF4-treated Erastin-induced AML12 cells. In addition, rFGF4 significantly enhanced the levels of heme oxygenase 1 (HO-1) and nuclear factor erythroid 2-related factor 2 (Nrf2) in ConA-induced AIH mice. Overall, this study showed that FGF4 can act as a phylactic role in AIH progression, with rFGF4 treatment inhibiting ferroptosis of hepatocytes by increasing CISD3 levels and activating Nrf2/HO-1 signaling.
Collapse
Affiliation(s)
- Huimian Jiang
- Department of Infectious Diseases, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou 325035, China
| | - Yan Fang
- Department of Infectious Diseases, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou 325035, China
| | - Yuxin Wang
- Department of Infectious Diseases, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou 325035, China
| | - Ting Li
- Department of Infectious Diseases, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou 325035, China
| | - Hongwei Lin
- Department of Infectious Diseases, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou 325035, China
| | - Jing Lin
- Department of Infectious Diseases, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou 325035, China
| | - Tongtong Pan
- Department of Infectious Diseases, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou 325035, China
| | - Qingxiu Liu
- Department of Infectious Diseases, the People's Hospital of Lishui, Lishui 323000, China
| | - Jiaojian Lv
- Department of Infectious Diseases, the People's Hospital of Lishui, Lishui 323000, China
| | - Dazhi Chen
- Department of Clinical Medicine, Hangzhou Medical College, Hangzhou 310053, China.
| | - Yongping Chen
- Department of Infectious Diseases, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
2
|
Mountford S, Effenberger M, Noll-Puchta H, Griessmair L, Ringleb A, Haas S, Denk G, Reiter FP, Mayr D, Dinarello CA, Tilg H, Bufler P. Modulation of Liver Inflammation and Fibrosis by Interleukin-37. Front Immunol 2021; 12:603649. [PMID: 33746950 PMCID: PMC7970756 DOI: 10.3389/fimmu.2021.603649] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/08/2021] [Indexed: 12/20/2022] Open
Abstract
Background and Aims: Chronic inflammation induces liver fibrosis, cirrhosis and potentially liver cancer. Kupffer cells modulate hepatic stellate cells by secreting immunologically active proteins as TGF-β. TGF-β promotes liver fibrosis via the activation of Sma- and Mad-related protein 3. IL-37 broadly suppresses innate and adaptive immune responses. Intracellular IL-37 interacts with Smad3. We hypothesize that IL-37 downregulates the activation of hepatic Kupffer and stellate cells and interferes with the TGF-β signaling cascade to modulate liver fibrogenesis. Methods: The role of IL-37 on liver inflammation and fibrogenesis was assessed in three mouse models as well as isolated Kupffer- and stellate cells. Serum IL-37 was tested by ELISA in a clinical cohort and correlated with liver disease severity. Results: Transgene expression of IL-37 in mice extends survival, reduces hepatic damage, expression of early markers of fibrosis and histologically assessed liver fibrosis after bile duct ligation. IL-37tg mice were protected against CCl4-induced liver inflammation. Colitis-associated liver inflammation and fibrosis was less severe in IL-10 knockout IL-37tg mice. Spontaneous and LPS/TGF-β-induced cytokine release and profibrogenic gene expression was lower in HSC and KC isolated from IL-37tg mice and IL-37 overexpressing, IL-1β stimulated human LX-2 stellate cells. However, administration of recombinant human IL-37 did not modulate fibrosis pathways after BDL in mice, LX2 cells or murine HSCs. In a large clinical cohort, we observed a positive correlation of serum IL-37 levels with disease severity in liver cirrhosis. Conclusions: Predominantly intracellular IL-37 downregulates liver inflammation and fibrosis. The correlation of serum IL-37 with disease severity in cirrhosis suggests its potential as a novel target modulating the course of liver fibrosis.
Collapse
Affiliation(s)
- Steffeni Mountford
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Maria Effenberger
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Heidi Noll-Puchta
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Lucas Griessmair
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Andrea Ringleb
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sonja Haas
- RNA Biology, Ethris GmbH, Planegg, Germany
| | - Gerald Denk
- Department of Medicine II, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Florian P. Reiter
- Department of Medicine II, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Doris Mayr
- Department of Pathology, Institute of Pathology, Ludwig-Maximilians-University, Munich, Germany
| | - Charles A. Dinarello
- Department of Medicine and Immunology, University of Colorado Denver, Aurora, CO, United States
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Philip Bufler
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
3
|
Peng X, Pan X, Tan J, Li Y, Li M. Protective effect of interleukin-36 receptor antagonist on liver injury induced by concanavalin A in mice. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:623-628. [PMID: 32742600 PMCID: PMC7374990 DOI: 10.22038/ijbms.2020.35614.8492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Objective(s): Interleukin-36 receptor antagonist (IL-36Ra) is a new member of the IL-1 family that exhibits anti-inflammatory activity in a variety of inflammatory and immune diseases. Our purpose was to determine the effect of IL-36Ra on liver injury in a mouse hepatitis model induced by concanavalin A (ConA). Materials and Methods: Mice were treated with IL-36Ra DNA or pcDNA3.1 control plasmid using a hydrodynamic gene delivery approach. Results: Our data reveal that treatment with IL-36Ra decreased liver inflammation and serum level of aminotransferases. Furthermore, IL-36Ra reduced ConA-induced pro-inflammatory cytokines (interferon-γ, tumor necrosis factor-α, and IL-17A) production when compared to control plasmid. Conclusion: Our results demonstrated that IL-36Ra is a critical protector against ConA-induced liver injury.
Collapse
Affiliation(s)
- Xiao Peng
- Department of Immunology, Medical School of Ningbo University, Ningbo 315211, China
| | - Xiuhe Pan
- Department of Immunology, Medical School of Ningbo University, Ningbo 315211, China
| | - Jun Tan
- Department of Hepatology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo 315010, China
| | - Yan Li
- Department of Immunology, Medical School of Ningbo University, Ningbo 315211, China
| | - Mingcai Li
- Department of Immunology, Medical School of Ningbo University, Ningbo 315211, China
| |
Collapse
|
4
|
Noh JR, Kim JH, Na SY, Lee IB, Seo YJ, Choi JH, Seo Y, Lee TG, Choi HS, Kim YH, Lee CH. Hepatocyte CREBH deficiency aggravates inflammatory liver injury following chemokine-dependent neutrophil infiltration through upregulation of NF-κB p65 in mice. Arch Toxicol 2019; 94:509-522. [DOI: 10.1007/s00204-019-02633-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/26/2019] [Indexed: 12/31/2022]
|
5
|
Hines IN, Kremer M, Moore SM, Wheeler MD. Impaired T cell-mediated hepatitis in peroxisome proliferator activated receptor alpha (PPARα)-deficient mice. Biol Res 2018; 51:5. [PMID: 29448959 PMCID: PMC5815252 DOI: 10.1186/s40659-018-0153-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 02/06/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Peroxisome proliferator activated receptor alpha (PPARα), a regulator of enzymes involved in β oxidation, has been reported to influence lymphocyte activation. The purpose of this study was to determine whether PPARα plays a role in T cell-mediated hepatitis induced by Concanavalin A (ConA). METHODS Wild type (wt) or PPARα-deficient (PPARα-/-) mice were treated with ConA (15 mg/kg) by intravenous injection 0, 10 or 24 h prior to sacrifice and serum and tissue collection for analysis of tissue injury, cytokine response, T cell activation and characterization. RESULTS Ten and 24 h following ConA administration, wt mice had significant liver injury as demonstrated by serum transaminase levels, inflammatory cell infiltrate, hepatocyte apoptosis, and expression of several cytokines including interleukin 4 (IL4) and interferon gamma (IFNγ). In contrast, PPARα-/- mice were protected from ConA-induced liver injury with significant reductions in serum enzyme release, greatly reduced inflammatory cell infiltrate, hepatocellular apoptosis, and IFNγ expression, despite having similar levels of hepatic T cell activation and IL4 expression. This resistance to liver injury was correlated with reduced numbers of hepatic natural killer T (NKT) cells and their in vivo responsiveness to alpha-galactosylceramide. Interestingly, adoptive transfer of either wt or PPARα-/- splenocytes reconstituted ConA liver injury and cytokine production in lymphocyte-deficient, severe combined immunodeficient mice implicating PPARα within the liver, possibly through support of IL15 expression and/or suppression of IL12 production and not the lymphocyte as the key regulator of T cell activity and ConA-induced liver injury. CONCLUSION Taken together, these data suggest that PPARα within the liver plays an important role in ConA-mediated liver injury through regulation of NKT cell recruitment and/or survival.
Collapse
Affiliation(s)
- Ian N. Hines
- Department of Nutrition Science, College of Allied Health Sciences, East Carolina University, Health Sciences Bldg. Room 4165F, Greenville, NC 27834 USA
- Department of Medicine, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Michael Kremer
- Department of General Surgery, University of Ulm, Ulm, Germany
- Department of Medicine, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Sherri M. Moore
- Department of Nutrition Science, College of Allied Health Sciences, East Carolina University, Health Sciences Bldg. Room 4165F, Greenville, NC 27834 USA
| | - Michael D. Wheeler
- Department of Nutrition Science, College of Allied Health Sciences, East Carolina University, Health Sciences Bldg. Room 4165F, Greenville, NC 27834 USA
- Department of Medicine, University of North Carolina, Chapel Hill, NC 27599 USA
| |
Collapse
|
6
|
Noh JR, Kim YH, Kim DK, Hwang JH, Kim KS, Choi DH, Lee SJ, Lee HG, Lee TG, Weng HL, Dooley S, Choi HS, Lee CH. Small Heterodimer Partner Deficiency Increases Inflammatory Liver Injury Through C-X-C motif chemokine ligand 2-Driven Neutrophil Recruitment in Mice. Toxicol Sci 2018; 163:254-264. [DOI: 10.1093/toxsci/kfy030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Affiliation(s)
- Jung-Ran Noh
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, South Korea
| | - Yong-Hoon Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, South Korea
- University of Science and Technology (UST), Daejeon 34113, South Korea
| | - Don-Kyu Kim
- National Creative Research Initiatives Center for Nuclear Receptor Signals and Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, South Korea
| | - Jung Hwan Hwang
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, South Korea
- University of Science and Technology (UST), Daejeon 34113, South Korea
| | - Kyoung-Shim Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, South Korea
- University of Science and Technology (UST), Daejeon 34113, South Korea
| | - Dong-Hee Choi
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, South Korea
| | - Seon-Jin Lee
- University of Science and Technology (UST), Daejeon 34113, South Korea
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Hee Gu Lee
- University of Science and Technology (UST), Daejeon 34113, South Korea
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Tae Geol Lee
- Center for Nano Bio Measurement, Korea Research Institute of Standard and Science, Yuseong-gu, Daejeon 34113, South Korea
| | - Hong-Lei Weng
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Steven Dooley
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Hueng-Sik Choi
- National Creative Research Initiatives Center for Nuclear Receptor Signals and Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, South Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, South Korea
- University of Science and Technology (UST), Daejeon 34113, South Korea
| |
Collapse
|
7
|
Li X, Yao QY, Liu HC, Jin QW, Xu BL, Zhang SC, Tu CT. Placental growth factor silencing ameliorates liver fibrosis and angiogenesis and inhibits activation of hepatic stellate cells in a murine model of chronic liver disease. J Cell Mol Med 2017; 21:2370-2385. [PMID: 28378526 PMCID: PMC5618674 DOI: 10.1111/jcmm.13158] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/13/2017] [Indexed: 01/18/2023] Open
Abstract
Placental growth factor (PlGF) is a member of the vascular endothelial growth factor (VEGF) family and is involved in pathological angiogenesis associated with chronic liver diseases. However, the precise mechanisms underlying PlGF signalling contributing to liver fibrosis and angiogenesis remain largely unexplored. This study aimed to assess the effect of reducing PlGF expression using small interfering RNA (siRNA) on experimental liver fibrosis and angiogenesis, and to elucidate the underlying molecular mechanisms. Fibrosis was induced in mice by carbon tetrachloride (CCl4) for 8 weeks, and mice were treated with PlGF siRNA or non‐targeting control siRNA starting two weeks after initiating CCl4 injections. The results showed that PlGF was highly expressed in cirrhotic human and mice livers; which mainly distributed in activated hepatic stellate cells (HSCs). PlGF silencing robustly reduced liver inflammation, fibrosis, intrahepatic macrophage recruitment, and inhibited the activation of HSCs in vivo. Moreover, PlGF siRNA‐treated fibrotic mice showed diminished hepatic microvessel density and angiogenic factors, such as hypoxia‐inducible factor‐1α (HIF‐1α), VEGF and VEGF receptor‐1. Moreover, down‐regulation of PlGF with siRNA in HSCs inhibited the activation and proliferation of HSCs. Mechanistically, overexpression of PlGF in activated HSCs was induced by hypoxia dependent on HIF‐1α, and PlGF induces HSC activation and proliferation via activation the phosphatidylinositol 3‐kinase (PI3K)/Akt signalling pathways. These findings indicate that PlGF plays an important role in liver fibrosis‐associated angiogenesis and that blockage of PlGF could be an effective strategy for chronic liver disease.
Collapse
Affiliation(s)
- Xi Li
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qun-Yan Yao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University and Shanghai Institute of Liver Diseases, Shanghai, China
| | - Hong-Chun Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University and Shanghai Institute of Liver Diseases, Shanghai, China
| | - Qian-Wen Jin
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University and Shanghai Institute of Liver Diseases, Shanghai, China
| | - Bei-Li Xu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University and Shanghai Institute of Liver Diseases, Shanghai, China
| | - Shun-Cai Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University and Shanghai Institute of Liver Diseases, Shanghai, China
| | - Chuan-Tao Tu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University and Shanghai Institute of Liver Diseases, Shanghai, China
| |
Collapse
|
8
|
Xiang M, Liu T, Tan W, Ren H, Li H, Liu J, Cao H, Cheng Q, Liu X, Zhu H, Tuo Y, Wang J, Zhang Y. Effects of kinsenoside, a potential immunosuppressive drug for autoimmune hepatitis, on dendritic cells/CD8 + T cells communication in mice. Hepatology 2016; 64:2135-2150. [PMID: 27639182 DOI: 10.1002/hep.28825] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 07/23/2016] [Accepted: 08/09/2016] [Indexed: 12/11/2022]
Abstract
UNLABELLED The central purpose of this study was to investigate therapeutic effects of the botanical derivative, kinsenoside (KD), in experimental autoimmune hepatitis (AIH). Treatment with KD substantially reduced hepatic histopathological damage, induced by lymphocyte infiltration and proinflammatory cytokines, in concanavalin A-induced T-cell-mediated hepatitis, and in dendritic cells (DCs) loaded with hepatocellular carcinoma cells (DC/Hepa1-6) induced murine AIH. Interactions between immune cells after KD treatment in AIH were detected by anti-CD8 antibody blocking, CD8+ T cell sorting, and vaccinated mice with KD-pretreated DCs in a DC/Hepa1-6 model. These results showed that KD inhibited the elevated expressions of CD86 and major histocompatibility complex II, densities of chemokine receptor C-C chemokine receptor type 7, and extensive migration to lymph nodes, and increased the programmed death ligand 1 level of DCs, followed by suppressing CD8+ T cells, characterized as low differentiation and cytotoxicity, and eliciting cytokines balance. Furthermore, biochemical analysis, two-dimensional fingerprint screen and three-dimensional molecular docking results showed that KD bound to the vascular endothelial growth factor receptor 2 (VEGFR2) kinase domain, which inhibited the metabolism-related phosphatidylinositol 3 kinase/protein kinase B (PI3K-AKT) pathway in DCs and DC-modulated CD8+ T cells to lower the mitochondrial membrane potential and glucose/lipid utilization ratio in both cells. KD reversed activation of the PI3K-AKT pathway by 740 Y-P (PI3K agonist), thereby impeding the translocation and dimerization of signal transducer and activators of transcription (STAT) 3 and synergistically blocking the inflammation-related Janus kinase (JAK) 2/STAT3 pathway in DCs and DC-modulated T cells. CONCLUSION KD treatment elicits immunosuppression against autoimmune liver injury by targeting VEGFR2, followed by diminishing the cross-talk of metabolism-related PI3K-AKT and inflammation-related JAK2-STAT3 pathways, and thereby disrupts DC-induced cross-priming of CD8+ T cell responses. (Hepatology 2016;64:2135-2150).
Collapse
Affiliation(s)
- Ming Xiang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Liu
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wanyue Tan
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyu Ren
- Department of Digestive Disease, Affiliated Xiehe Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Li
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junjun Liu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Cao
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Cheng
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiulan Liu
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hucheng Zhu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yali Tuo
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianping Wang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yonghui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Kremer M, Son G, Zhang K, Moore SM, Norris A, Manzini G, Wheeler MD, Hines IN. Smad3 signaling in the regenerating liver: implications for the regulation of IL-6 expression. Transpl Int 2014; 27:748-58. [PMID: 24649805 DOI: 10.1111/tri.12322] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 11/24/2013] [Accepted: 03/16/2014] [Indexed: 12/12/2022]
Abstract
Liver regeneration is vital for graft survival and adequate organ function. Smad activation regulates hepatocyte proliferation and macrophage function. The aim of the current study was to evaluate the impact of Smad3 signaling during liver regeneration in the mouse. Male C57Bl/6 wild-type (wt) mice or mice deficient in Smad3 (Smad3(-/-) ) were subjected to a 70% partial hepatectomy (pHx) or sham surgery and sacrificed 24, 42, or 48 h later. Tissue was analyzed for TGF-β signaling, the mitogenic cytokine response [i.e., tumor necrosis factor alpha, TNF-α; interleukin (IL)-6], and liver regeneration. Partial hepatectomy stimulated a strong regenerative response measured by proliferating cell nuclear antigen-positive hepatocytes 42 and 48 h post-pHx in conjunction with an increased expression of IL-6, TNF-α, and Smad2/3 phosphorylation 24 h post-pHx in both hepatocytes and nonparenchymal cells. Surprisingly, Smad3 deficiency led to reduced hepatocyte proliferation 42 h post-pHx which recovered by 48 h, a process that correlated with and was preceded by significant reductions in IL-6 expression and signal transducer and activator of transcription 3 phosphorylation, and cyclin D1 induction 24 h post-pHx. Loss of Smad3 signaling suppresses the expression of key mitogenic cytokines and delays hepatocellular regeneration. Therapies directed at finely regulating Smad3 activation early within the regenerating liver may prove useful in promoting liver cell proliferation and restoration of liver mass.
Collapse
Affiliation(s)
- Michael Kremer
- Department of General Surgery, University of Ulm, Ulm, Germany; Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina, Chapel Hill, NC, USA
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Zhou YC, Chen S, Cao JJ, Chen SY, Xie YF, Niu QX. Adenovirus-mediated viral interleukin-10 gene transfer prevents concanavalin A-induced liver injury. Dig Liver Dis 2012; 44:398-405. [PMID: 22209949 DOI: 10.1016/j.dld.2011.11.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 11/14/2011] [Accepted: 11/25/2011] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIM Liver injury is closely associated with immune inflammation. Lacking immunostimulatory functions, viral interleukin-10 (vIL-10), a cellular IL-10 homologue, has been an attractive molecule for immunomodulatory therapy. We aimed to reveal a protective effect of the gene transfer of an adenoviral vector encoding vIL-10 on liver injury induced by concanavalin A. METHODS C57BL/6J mice were intravenously injected with adenoviral vector encoding vIL-10 before concanavalin A challenge. Liver injury was assessed. Interferon-γ and interleukin-4 levels were measured by ELISA. The activation of splenic and hepatic immune cells was analysed using an MTT assay. RESULTS Adenoviral vector encoding vIL-10 pretreatment significantly decreased concanavalin A-mediated elevations in serum alanine aminotransaminase and aspartate aminotransaminase activity, and necrotic area in liver tissues. The protective effect of adenoviral vector encoding vIL-10 was attributed to its inhibition of T cell activation, and production of interferon-γ and interleukin-4 by the immune cells. Recombinant mouse IL-10, a high homologous cytokine to vIL-10, effectively downregulated interferon-γ and interleukin-4 release by hepatic mononuclear cells. CONCLUSION Adenovirus vector-mediated vIL-10 gene transfer can prevent concanavalin A-induced hepatic injury, minimise pro-inflammatory cytokine release, and inhibit the activation of T lymphocytes.
Collapse
Affiliation(s)
- Yan-Chun Zhou
- Institute of Inflammation and Immune Diseases, Shantou University Medical College, Shantou, Guangdong, China
| | | | | | | | | | | |
Collapse
|
11
|
Tu CT, Han B, Yao QY, Zhang YA, Liu HC, Zhang SC. Curcumin attenuates Concanavalin A-induced liver injury in mice by inhibition of Toll-like receptor (TLR) 2, TLR4 and TLR9 expression. Int Immunopharmacol 2011; 12:151-7. [PMID: 22138522 DOI: 10.1016/j.intimp.2011.11.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2011] [Revised: 11/14/2011] [Accepted: 11/15/2011] [Indexed: 12/31/2022]
Abstract
Curcumin has antiviral, antioxidant, and anti-inflammatory properties. However, the hepatoprotective effects and molecular mechanisms of curcumin on acute liver injury have not been carefully examined. The aims of this study were to examine the anti-inflammatory effect of curcumin on Concanavalin A (Con A) induced hepatitis, and to elucidate its underlying molecular mechanisms in mice. Mice received curcumin (200 mg/kg body weight) by gavage before Con A intravenous administration. We found that curcumin pretreatment was able to significantly reduce the elevated plasma aminotransferase levels and liver necrosis in Con A-induced hepatitis. Also, curcumin pretreatment reduced intrahepatic expression of genes encoding pro-inflammatory molecules such as tumor necrosis factor α (TNF-α) and interferon γ (IFN-γ) as compared with the vehicle controls, but augmented anti-inflammatory cytokine interleukin 10 (IL-10) by enzyme linked immunosorbent assay (ELISA). Furthermore, the expression levels of Toll-like receptor (TLR) 2, TLR4 and TLR9 mRNA or protein in liver tissues were significantly lowered by curcumin treatment. Curcumin pretreatment did not affect hepatic Kupffer cell numbers after Con A injection. These results suggest that curcumin pretreatment protects against T cell-mediated hepatitis in mice. The beneficial effect of curcumin may be partly mediated by inhibiting the expression levels of TLR2, TLR4 and TLR9 in the liver.
Collapse
|
12
|
Hines IN, Hartwell HJ, Feng Y, Theve EJ, Hall GA, Hashway S, Connolly J, Fecteau M, Fox JG, Rogers AB. Insulin resistance and metabolic hepatocarcinogenesis with parent-of-origin effects in A×B mice. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:2855-65. [PMID: 21967816 DOI: 10.1016/j.ajpath.2011.08.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 06/24/2011] [Accepted: 08/16/2011] [Indexed: 01/01/2023]
Abstract
Insulin resistance is a defining feature of metabolic syndrome and type 2 diabetes mellitus but also may occur independently of these conditions. Nonalcoholic fatty liver disease (NAFLD), the hepatic manifestation of these disorders, increases the risk of hepatocellular carcinoma (HCC). However, mechanisms linking hyperinsulinemia to NAFLD and HCC require clarification. We describe a novel model of primary insulin resistance and HCC with strong parent-of-origin effects. Male AB6F1 (A/JCr dam × C57BL/6 sire) but not B6AF1 (B6 dam × A/J sire) mice developed spontaneous insulin resistance, NAFLD, and HCC without obesity or diabetes. A survey of mitochondrial, imprinted, and sex-linked traits revealed modest associations with X-linked genes. However, a diet-induced obesity study, including B6.A chromosome substitution-strain (consomic) mice, showed no segregation by sex chromosome. Thus, parent-of-origin effects were specified within the autosomal genome. Next, we interrogated mechanisms of insulin-associated hepatocarcinogenesis. Steatotic hepatocytes exhibited adipogenic transition characterized by vacuolar metaplasia and up-regulation of vimentin, adipsin, fatty acid translocase (CD36), peroxisome proliferator-activated receptor-γ, and related products. This profile was largely recapitulated in insulin-supplemented primary mouse hepatocyte cultures. Importantly, pyruvate kinase M2, a fetal anabolic enzyme implicated in the Warburg effect, was activated by insulin in vivo and in vitro. Thus, our study reveals parent-of-origin effects in heritable insulin resistance, implicating adipogenic transition with acquired anabolic metabolism in the progression from NAFLD to HCC.
Collapse
Affiliation(s)
- Ian N Hines
- Department of Nutrition Sciences, East Carolina University, Greenville, North Carolina, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
Emerging evidence suggests a strong interaction between the gut microbiota and health and disease. The interactions of the gut microbiota and the liver have only recently been investigated in detail. Receiving approximately 70% of its blood supply from the intestinal venous outflow, the liver represents the first line of defense against gut-derived antigens and is equipped with a broad array of immune cells (i.e., macrophages, lymphocytes, natural killer cells, and dendritic cells) to accomplish this function. In the setting of tissue injury, whereby the liver is otherwise damaged (e.g., viral infection, toxin exposure, ischemic tissue damage, etc.), these same immune cell populations and their interactions with the infiltrating gut bacteria likely contribute to and promote these pathologies. The following paper will highlight recent studies investigating the relationship between the gut microbiota, liver biology, and pathobiology. Defining these connections will likely provide new targets for therapy or prevention of a wide variety of acute and chronic liver pathologies.
Collapse
|
14
|
Abstract
The structural organization of research facilities within a surgical university center should aim at strengthening the department's research output and likewise provide opportunities for the scientific education of academic surgeons. We suggest a model in which several independent research groups within a surgical department engage in research projects covering various aspects of surgically relevant basic, translational or clinical research. In order to enhance the translational aspects of surgical research, a permanent link needs to be established between the department's scientific research projects and its chief interests in clinical patient care. Importantly, a focus needs to be placed on obtaining evidence-based data to judge the efficacy of novel diagnostic and treatment concepts. Integration of modern technologies from the fields of physics, computer science and molecular medicine into surgical research necessitates cooperation with external research facilities, which can be strengthened by coordinated support programs offered by research funding institutions.
Collapse
|
15
|
Shi G, Zhang Z, Feng D, Xu Y, Lu Y, Wang J, Jiang J, Zhang Z, Li X, Ning G. Selection of reference genes for quantitative real-time reverse transcription-polymerase chain reaction in concanavalin A-induced hepatitis model. Anal Biochem 2010; 401:81-90. [DOI: 10.1016/j.ab.2010.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 02/04/2010] [Accepted: 02/06/2010] [Indexed: 11/16/2022]
|
16
|
Fujii Y, Kawamura H, Kawamura T, Kanda Y, Matsumoto H, Kobayashi T, Yamamoto T, Aoyama T, Abo T. Co-appearance of autoantibody-producing B220(low) B cells with NKT cells in the course of hepatic injury. Cell Immunol 2010; 260:105-12. [PMID: 19857863 DOI: 10.1016/j.cellimm.2009.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Revised: 09/24/2009] [Accepted: 09/25/2009] [Indexed: 11/19/2022]
Abstract
Severe hepatic injury is induced by Concanavalin A (Con A) administration in mice, the major effector cells being CD4(+) T cells, NKT cells and macrophages. Since autologous lymphocyte subsets are associated with tissue damage, Con A-induced hepatic injury is considered to be autoimmune hepatitis. However, it has remained to be investigated how autoantibodies and B-1 cells are responsible for this phenomenon. In this study, it was demonstrated that autoantibodies which were detected using Hep-2 cells in immunofluorescence tests and using double-strand (ds) DNA in the ELISA method, appeared after Con A administration (a peak at day 14). Moreover, autoantibody-producing B220(low) cells (i.e., B-1 cells) also appeared at this time. Purified B220(low) cells were found to have a potential to produce autoantibodies. These results suggest that Con A-induced hepatic injury indeed includes the mechanism of autoimmune hepatitis.
Collapse
Affiliation(s)
- Yuki Fujii
- Department of Immunology, Niigata University School of Medicine, Niigata 951-8510, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Kremer M, Thomas E, Milton RJ, Perry AW, van Rooijen N, Wheeler MD, Zacks S, Fried M, Rippe RA, Hines IN. Kupffer cell and interleukin-12-dependent loss of natural killer T cells in hepatosteatosis. Hepatology 2010; 51:130-41. [PMID: 20034047 PMCID: PMC3761962 DOI: 10.1002/hep.23292] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
UNLABELLED Hepatosteatosis is associated with increased expression of tumor necrosis factor alpha (TNF-alpha) and interleukin (IL)-12, major T helper (Th) 1 cytokines, and reduced hepatic natural killer T (NKT) cell numbers. The relationship between lipid accumulation, cytokine expression, and hepatic NKT cells is not known. This study was conducted to assess the role of IL-12 in the development of hepatic steatosis and its potential impact on liver NKT cells. Male C57Bl/6 wildtype (WT) and IL-12-deficient (IL-12(-/-)) mice were fed a choline-deficient diet (CDD) for 0, 10, or 20 weeks. CDD led to marked hepatosteatosis, reduced hepatic but not splenic NKT cell numbers and function, and increased hepatic expression of the T(h)1-type cytokines IL-12, interferon gamma (IFN-gamma), and TNF-alpha in WT mice. The absence of IL-12 resulted in similar CDD-induced hepatosteatosis, but preserved hepatic NKT cells and significantly reduced hepatic IFN-gamma and TNF-alpha expression. Treatment of CDD-fed mice with lipopolysaccharide led to a significant increase in hepatic IL-12 expression, and Kupffer cell (KC) depletion reduced liver IL-12 expression and restored NKT cells in CDD-induced fatty liver. Interestingly, KCs from CDD-fed mice failed to produce increased quantities of IL-12 upon activation in vitro when compared to similarly treated KCs from control fed mice, suggesting that secondary factors in vivo promote heightened IL-12 production. Finally, human livers with severe steatosis showed a substantial decrease in NKT cells. CONCLUSION Hepatosteatosis reduces the numbers of hepatic NKT cells in a KC-and IL-12-dependent manner. Our results suggest a pivotal and multifunctional role of KC-derived IL-12 in the altered immune response in steatotic liver, a process that is likely active within human nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Michael Kremer
- Center for Alcohol Sudies, University of North Carolina at Chapel Hill, NC 27599
,Department of Surgery, University of Heidelberg, Germany
| | - Emmanuel Thomas
- Department of Medicine, University of North Carolina at Chapel Hill, NC 27599
| | - Richard J. Milton
- Center for Alcohol Sudies, University of North Carolina at Chapel Hill, NC 27599
| | - Ashley W. Perry
- Center for Alcohol Sudies, University of North Carolina at Chapel Hill, NC 27599
| | - Nico van Rooijen
- Department of Cell Biology, Free University, Amsterdam, Netherlands
| | - Michael D. Wheeler
- Center for Alcohol Sudies, University of North Carolina at Chapel Hill, NC 27599
| | - Steven Zacks
- Department of Medicine, University of North Carolina at Chapel Hill, NC 27599
| | | | - Richard A. Rippe
- Department of Medicine, University of North Carolina at Chapel Hill, NC 27599
| | - Ian N. Hines
- Center for Alcohol Sudies, University of North Carolina at Chapel Hill, NC 27599
,Department of Medicine, University of North Carolina at Chapel Hill, NC 27599
| |
Collapse
|
18
|
Wang ZL, Wu XH, Song LF, Wang YS, Hu XH, Luo YF, Chen ZZ, Ke J, Peng XD, He CM, Zhang W, Chen LJ, Wei YQ. Phosphoinositide 3-kinase gamma inhibitor ameliorates concanavalin A-induced hepatic injury in mice. Biochem Biophys Res Commun 2009; 386:569-74. [PMID: 19538942 DOI: 10.1016/j.bbrc.2009.06.060] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Accepted: 06/11/2009] [Indexed: 02/05/2023]
Abstract
A pivotal role of phosphoinositide 3-kinase-gamma (PI3Kgamma) in inflammatory cell activation and recruitment makes it an attractive target for immunomodulatory therapy. In present study we investigated the therapeutic efficiency of AS605240, a selective PI3Kgamma inhibitor, on hepatitis and liver fibrosis in murine models induced by concanavalin A (ConA). Orally administration of AS605240 significantly improved survival, decreased the serum levels of alanine aminotransaminase (ALT), prevented inflammatory infiltration to liver in ConA-induced hepatitis. TNF-alpha and IFN-gamma at protein levels in serum and mRNA levels in liver were markedly reduced. Downregulated phospho-Akt level of inflammatory cells infiltrating the liver by AS605240 treatment was detected by immunohistochemistry analysis in liver and further confirmed by Western blotting analysis in splenocytes. In ConA-induced chronic liver fibrosis model, accumulation of smooth-muscle actin (SMA)-expressing cells was partially inhibited by AS605240 treatment. These observations suggest that AS605240 might be of therapeutic value for the treatment of ConA-induced hepatic injury.
Collapse
Affiliation(s)
- Zhen-ling Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Osawa Y, Seki E, Brenner DA. Apoptosis in Liver Injury and Liver Diseases. ESSENTIALS OF APOPTOSIS 2009:547-564. [DOI: 10.1007/978-1-60327-381-7_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
20
|
Jeong DH, Goo MJ, Hong IH, Yang HJ, Ki MR, DO SH, Ha JH, Lee SS, Park JK, Jeong KS. Inhibition of radiation-induced apoptosis via overexpression of SMP30 in Smad3-knockout mice liver. JOURNAL OF RADIATION RESEARCH 2008; 49:653-660. [PMID: 19001783 DOI: 10.1269/jrr.08042] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Apoptosis occurs early after irradiation and may be a good indicator of radiation damages. Since elevated levels of TGF-beta are associated with radiation-induced inflammation, the null mice of Smad3, a key downstream mediator of TGF-beta, show accelerated healing of irradiated injury. In order to evaluate resistance to radiation-induced liver injuries in Smad3-null mice, we determined the occurrence of apoptosis and the expression of senescence marker protein-30 (SMP30), as an anti-apoptotic marker, after irradiation to the liver. The livers of Smad3-mutant mice were exposed to local irradiation of 15 gray, from a (60)Co-gamma radiation. One week after irradiation, in Smad3-KO mice, radiation-induced apoptosis was at lower levels compared to those of irradiated WT mice. These findings were well matched with the expression of CYP2E1, which plays a role in hepatic injuries produced by oxidative stress. In addition, antioxidant related protein, the SMP30 levels were reduced by gamma irradiation in both groups. Interestingly, the increased expression of SMP30 expression in Smad3-KO mice liver was preserved at a higher level than that of the WT mice after irradiation. Therefore, these results suggest that the interruption of TGF-beta signaling by deletion of Smad3 brings about inhibition of hepatic apoptosis after ionizing irradiation. Moreover, the protective effect to ionizing radiation might be in correlation with the overexpression of SMP30 in the Smad3-null mice, which may act as an anti-apoptotic signaling molecule. The alteration of SMP30 by interruption of Smad3 might be a useful therapeutic target and diagnostic marker for radiation-induced liver damages.
Collapse
Affiliation(s)
- Da-Hee Jeong
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Samon JB, Champhekar A, Minter LM, Telfer JC, Miele L, Fauq A, Das P, Golde TE, Osborne BA. Notch1 and TGFbeta1 cooperatively regulate Foxp3 expression and the maintenance of peripheral regulatory T cells. Blood 2008; 112:1813-21. [PMID: 18550850 PMCID: PMC2518888 DOI: 10.1182/blood-2008-03-144980] [Citation(s) in RCA: 156] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2008] [Accepted: 05/23/2008] [Indexed: 12/11/2022] Open
Abstract
Notch and its ligands have been implicated in the regulation and differentiation of various CD4(+) T-helper cells. Regulatory T cells (T(regs)), which express the transcription factor Foxp3, suppress aberrant immune responses that are typically associated with autoimmunity or excessive inflammation. Previous studies have shown that transforming growth factor beta (TGFbeta1) induces Foxp3 expression and a regulatory phenotype in peripheral T cells. Here, we show that pharmacologic inhibition of Notch signaling using gamma-secretase inhibitor (GSI) treatment blocks (1) TGFbeta1-induced Foxp3 expression, (2) the up-regulation of Foxp3-target genes, and (3) the ability to suppress naive T-cell proliferation. In addition, the binding of Notch1, CSL, and Smad to conserved binding sites in the foxp3 promoter can be inhibited by treatment with GSI. Finally, in vivo administration of GSI results in reduced Foxp3 expression and development of symptoms consistent with autoimmune hepatitis, a disease previously found to result from dysregulation of TGFbeta signaling and regulatory T cells. Together, these findings indicate that the Notch and TGFbeta signaling pathways cooperatively regulate Foxp3 expression and regulatory T-cell maintenance both in vitro and in vivo.
Collapse
MESH Headings
- Alanine/analogs & derivatives
- Alanine/pharmacology
- Amyloid Precursor Protein Secretases/antagonists & inhibitors
- Animals
- Azepines/pharmacology
- Base Sequence
- Cell Differentiation
- Cell Proliferation/drug effects
- DNA Primers/genetics
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/physiology
- Gene Expression/drug effects
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Muscle Proteins/metabolism
- Protease Inhibitors/pharmacology
- Receptor, Notch1/antagonists & inhibitors
- Receptor, Notch1/deficiency
- Receptor, Notch1/genetics
- Receptor, Notch1/physiology
- Signal Transduction/drug effects
- Smad Proteins/metabolism
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/physiology
- Transforming Growth Factor beta1/pharmacology
- Transforming Growth Factor beta1/physiology
Collapse
Affiliation(s)
- Jeremy B Samon
- Program in Animal Biotechnology and Biomedical Science, University of Massachusetts Amherst, MA 01003, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Autoinflammatory liver disease represents an important aspect of global hepatological practice. The three principal disease divisions recognized are autoimmune hepatitis, primary sclerosing cholangitis and primary biliary cirrhosis. Largely, but not exclusively, these diseases are considered to be autoimmune in origin. Increased recognition of outlier and overlap syndromes, changes in presentation and natural history, as well as the increased awareness of IgG4-associated sclerosing cholangitis, all highlight the limitations of the classic terminology. New insights continue to improve the care given to patients, and have arisen from carefully conducted clinical studies, therapeutic trials, as well as genetic and laboratory investigations. The challenges remain to treat patients before liver injury becomes permanent and to prevent the development of organ failure.
Collapse
Affiliation(s)
- Teru Kumagi
- Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|