1
|
Papierska K, Judasz E, Tonińska W, Kubicki M, Krajka-Kuźniak V. Modulatory Effects of Chalcone Thio-Derivatives on NF-κB and STAT3 Signaling Pathways in Hepatocellular Carcinoma Cells: A Study on Selected Active Compounds. Int J Mol Sci 2024; 25:10739. [PMID: 39409068 PMCID: PMC11476945 DOI: 10.3390/ijms251910739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/29/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
Our previous studies demonstrated the modulatory effects of new synthetic thio-chalcone derivatives in dishes on the Nrf2, NF-κB, and STAT3 signaling pathways in colon cancer cells. This study aimed to evaluate the effect of four selected active chalcone thio-derivatives on the NF-κB and STAT3 signaling pathways involved in inflammatory processes and cell proliferation in human liver cancer cells. Cell survival was assessed for cancer (HepG2) and normal (THLE-2) cell lines. Activation of NF-κB and STAT3 signaling pathways and the expression of proteins controlled by these pathways were estimated by Western blot, and qRT-PCR assessed the expression of NF-κB and STAT3 target genes. We also evaluated the impact on the selected kinases responsible for the phosphorylation of the studied transcription factors by MagneticBead-Based Multiplex Immunoassay. Among the thio-derivatives tested, especially derivatives 1 and 5, there was an impact on cell viability, cell cycle, apoptosis, and activation of NF-κB and STAT3 pathways in hepatocellular carcinoma (HCC), which confirms the possibilities of using them in combinatorial molecular targeted therapy of HCC. The tested synthetic thio-chalcones exhibit anticancer activity by initiating proapoptotic processes in HCC while showing low toxicity to non-cancerous cells. These findings confirm the possibility of using chalcone thio-derivatives in molecularly targeted combination therapy for HCC.
Collapse
Affiliation(s)
- Katarzyna Papierska
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland; (E.J.); (W.T.); (V.K.-K.)
| | - Eliza Judasz
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland; (E.J.); (W.T.); (V.K.-K.)
| | - Wiktoria Tonińska
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland; (E.J.); (W.T.); (V.K.-K.)
| | - Maciej Kubicki
- Faculty of Chemistry, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 8, 61-712 Poznań, Poland;
| | - Violetta Krajka-Kuźniak
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland; (E.J.); (W.T.); (V.K.-K.)
| |
Collapse
|
2
|
Yuan M, Shi H, Wang B, Cai J, Yu W, Wang W, Qian Q, Wang Y, Zhou X, Liu J. Targeting SOCS2 alleviates myocardial fibrosis by reducing nuclear translocation of β-catenin. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119804. [PMID: 39084528 DOI: 10.1016/j.bbamcr.2024.119804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/25/2024] [Accepted: 07/20/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Myocardial fibrosis is an important pathological feature of dilated cardiomyopathy (DCM). The roles of SOCS2 in fibrosis of different organs are controversial. Herein, we investigated the function and potential mechanism of SOCS2 in myocardial fibrosis. METHODS Bioinformatics, immunohistochemistry (IHC), immunofluorescence (IF), western blot (WB), real-time fluorescence quantitative PCR (qPCR), rat primary myocardial fibroblasts (rCFs) culture, doxorubicin (DOX) induced mouse dilated cardiomyopathy (DCM) model, and in vivo adeno-associated virus (AAV) infection were used to explore the role of SOCS2 in DCM. RESULTS Bioinformatics analysis showed that SOCS2 was positively correlated with fibrosis related factors. SOCS2 was significantly upregulated in patients and mice with DCM. In vivo experiments showed that targeted inhibition of cardiac SOCS2 could improve mouse cardiac function and alleviate myocardial fibrosis. Further research demonstrated that SOCS2 promoted the transformation of myofibroblasts. Knockdown of SOCS2 reduced the nuclear localization of β-catenin, which inhibited the fibrogenic effect of Wnt/β-catenin pathway. In addition, bioinformatics analysis suggested that lymphoid enhancer binding factor 1 (LEF1) was significantly positively correlated with SOCS2. Finally, dual luciferase assays demonstrated that LEF1 could bind to the promoter region of SOCS2, thereby mediating its transcriptional activation. CONCLUSION SOCS2 could activate the Wnt/β-catenin by regulating the nuclear translocation of β-catenin, which induces the transcriptional activation of SOCS2. Overall, these results indicated a positive feedback activation phenomenon between SOCS2, β-catenin and LEF1 in DCM. These results suggested that inhibition of SOCS2 could effectively alleviate the progression of myocardial fibrosis and improve cardiac function.
Collapse
Affiliation(s)
- Ming Yuan
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China
| | - Hongjie Shi
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China
| | - Bin Wang
- Department of Cardiovascular Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Jie Cai
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China
| | - Wenjun Yu
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China
| | - Wei Wang
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China
| | - Qiaofeng Qian
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China
| | - Yumou Wang
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China
| | - Xianwu Zhou
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China.
| | - Jinping Liu
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China.
| |
Collapse
|
3
|
Sayed Amr K, Mohamed Ezzat W, Ibrahim Saleh A, Heiba A, Amin H, Refaat Kamel R, Eltaweel N, Henery H, Omaia A, Ibrahim Siddik R, Abdelghany Abdelazeem Elhosary Y. Measuring the differential expression of the major hypermethylated tumor suppressor genes in tissues of primary hepatocellular carcinoma. J Genet Eng Biotechnol 2024; 22:100394. [PMID: 39179317 PMCID: PMC11214395 DOI: 10.1016/j.jgeb.2024.100394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2024]
Abstract
BACKGROUND Hepatocarcinogenesis is a multifactorial process that arises from a integration of genetic and epigenetic anomalies leading to abnormal gene expression and function. It is difficult to characterize HCC with a single biomarker. Our study aimed at detecting the expression of a panel of 8 methylated genes (SOCS1, APC, Gadd45b, CDKN1B, P15, PAX6, STAT1 and MSH2) as regulatory factors among Egyptian patients with HCC. METHODS This study was conducted on HCC tissue samples of 30 Egyptian patients in comparison with their non-cancerous adjacent cirrhotic tissue as a control. Tissue samples were obtained from patients who have undergone living donor liver transplantation (LDLT) or liver resection at El Sahel Teaching Hospital (Cairo, Egypt). A special Custom designed PCR Arrays was used to analyze the expression profiles of chosen methylated genes associated with HCC. RESULTS Expression of SOCS1, APC, Gadd45b, CDKN1B, P15, PAX6, STAT1 and MSH2 were lower in the HCC tissue compared to the cirrhotic tissue (pvalue = 0.015, 0.081, 0.004, 0.027, 0.211, 0.015, 0.025 and 0.0001 respectively). 5 genes (SOCS1, APC, GAdd45b, CDKN1B, and MSH2) showed the ability to be used as diagnostic biomarkers for HCC with high sensitivity and specificity values at cut off values: 1.05, 1.17, 0.995, 0.546, and 0.125 respectively. As for the other 3 genes (P15, PAX6, STAT1), PAX6 gene has the highest sensitivity at a cut off value of 0.3364. A significant negative correlation was shown between alpha fetoprotein (AFP) and 5 of the studied genes (SOCS1, APC, Gadd45b, STAT1, and MSH2). CONCLUSIONS Expression of the selected hypermethylated genes (SOCS1, APC, Gadd45b, CDKN1B, P15, PAX6, STAT1 and MSH2) in HCC tissue samples was lower than adjacent tissue. Their role should be further studied to solve the mystery that surrounds the pathogenesis of HCC.
Collapse
Affiliation(s)
- Khalda Sayed Amr
- Medical Molecular Genetics Dept, Human Genetics and Genome Research Institute, National Research Center, 33 El Buhouth St, Dokki, Cairo 12622, Egypt
| | - Wafaa Mohamed Ezzat
- Internal Medicine Dept, Medical Research Institute, National Research Center, 33 El Buhouth St, Dokki, Cairo 12622, Egypt
| | - Ahmed Ibrahim Saleh
- Internal Medicine Dept, Medical Research Institute, National Research Center, 33 El Buhouth St, Dokki, Cairo 12622, Egypt.
| | - Ahmed Heiba
- Internal Medicine Dept, Medical Research Institute, National Research Center, 33 El Buhouth St, Dokki, Cairo 12622, Egypt
| | - Hend Amin
- Internal Medicine Dept, Medical Research Institute, National Research Center, 33 El Buhouth St, Dokki, Cairo 12622, Egypt
| | - Refaat Refaat Kamel
- Surgery Dept, Faculty Of Medicine, Ain Shams University, 38 Abbassia, Cairo 1181, Egypt
| | - Noha Eltaweel
- Medical Molecular Genetics Dept, Human Genetics and Genome Research Institute, National Research Center, 33 El Buhouth St, Dokki, Cairo 12622, Egypt
| | - Hoda Henery
- Surgery Dept, Elsahel Teaching Hospital, 2 Youssef Karam, Borham, El Sahel, Cairo 11697, Egypt
| | - Amr Omaia
- Internal Medicine Dept, Medical Research Institute, National Research Center, 33 El Buhouth St, Dokki, Cairo 12622, Egypt
| | - Reham Ibrahim Siddik
- Internal Medicine Dept, Medical Research Institute, National Research Center, 33 El Buhouth St, Dokki, Cairo 12622, Egypt
| | | |
Collapse
|
4
|
Sande-Melon M, Bergemann D, Fernández-Lajarín M, González-Rosa JM, Cox AG. Development of a hepatic cryoinjury model to study liver regeneration. Development 2024; 151:dev203124. [PMID: 38975841 PMCID: PMC11318111 DOI: 10.1242/dev.203124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/20/2024] [Indexed: 07/09/2024]
Abstract
The liver is a remarkable organ that can regenerate in response to injury. Depending on the extent of injury, the liver can undergo compensatory hyperplasia or fibrosis. Despite decades of research, the molecular mechanisms underlying these processes are poorly understood. Here, we developed a new model to study liver regeneration based on cryoinjury. To visualise liver regeneration at cellular resolution, we adapted the CUBIC tissue-clearing approach. Hepatic cryoinjury induced a localised necrotic and apoptotic lesion characterised by inflammation and infiltration of innate immune cells. After this initial phase, we observed fibrosis, which resolved as regeneration re-established homeostasis in 30 days. Importantly, this approach enables the comparison of healthy and injured parenchyma within an individual animal, providing unique advantages to previous models. In summary, the hepatic cryoinjury model provides a fast and reproducible method for studying the cellular and molecular pathways underpinning fibrosis and liver regeneration.
Collapse
Affiliation(s)
- Marcos Sande-Melon
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - David Bergemann
- Cardiovascular Research Centre, Massachusetts General Hospital Research Institute, Charlestown Navy Yard Campus, 149, 13th Street, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Miriam Fernández-Lajarín
- Cardiovascular Research Centre, Massachusetts General Hospital Research Institute, Charlestown Navy Yard Campus, 149, 13th Street, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA 02467, USA
| | - Juan Manuel González-Rosa
- Cardiovascular Research Centre, Massachusetts General Hospital Research Institute, Charlestown Navy Yard Campus, 149, 13th Street, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA 02467, USA
| | - Andrew G. Cox
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria 3000, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, Victoria 3000, Australia
| |
Collapse
|
5
|
Sande-Melon M, Bergemann D, Fernández-Lajarín M, González-Rosa JM, Cox AG. Development of a hepatic cryoinjury model to study liver regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.24.550437. [PMID: 38948752 PMCID: PMC11212901 DOI: 10.1101/2023.07.24.550437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The liver is a remarkable organ that can regenerate in response to injury. Depending on the extent of injury, the liver can undergo compensatory hyperplasia or fibrosis. Despite decades of research, the molecular mechanisms underlying these processes are poorly understood. Here, we developed a new model to study liver regeneration based on cryoinjury. To visualise liver regeneration at cellular resolution, we adapted the CUBIC tissue-clearing approach. Hepatic cryoinjury induced a localised necrotic and apoptotic lesion characterised by inflammation and infiltration of innate immune cells. Following this initial phase, we observed fibrosis, which resolved as regeneration re-established homeostasis in 30 days. Importantly, this approach enables the comparison of healthy and injured parenchyma with an individual animal, providing unique advantages to previous models. In summary, the hepatic cryoinjury model provides a fast and reproducible method for studying the cellular and molecular pathways underpinning fibrosis and liver regeneration.
Collapse
Affiliation(s)
- Marcos Sande-Melon
- Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, 3000, Australia
| | - David Bergemann
- Cardiovascular Research Centre, Massachusetts General Hospital Research Institute, Charlestown Navy Yard Campus, 149, 13 Street, 02129 MA, USA
- Harvard Medical School
| | - Miriam Fernández-Lajarín
- Cardiovascular Research Centre, Massachusetts General Hospital Research Institute, Charlestown Navy Yard Campus, 149, 13 Street, 02129 MA, USA
- Harvard Medical School
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA 02467
| | - Juan Manuel González-Rosa
- Cardiovascular Research Centre, Massachusetts General Hospital Research Institute, Charlestown Navy Yard Campus, 149, 13 Street, 02129 MA, USA
- Harvard Medical School
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA 02467
| | - Andrew G. Cox
- Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, 3000, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, Victoria, 3000, Australia
| |
Collapse
|
6
|
Mokhtarpour K, Akbarzadehmoallemkolaei M, Rezaei N. A viral attack on brain tumors: the potential of oncolytic virus therapy. J Neurovirol 2024; 30:229-250. [PMID: 38806994 DOI: 10.1007/s13365-024-01209-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/30/2024]
Abstract
Managing malignant brain tumors remains a significant therapeutic hurdle that necessitates further research to comprehend their treatment potential fully. Oncolytic viruses (OVs) offer many opportunities for predicting and combating tumors through several mechanisms, with both preclinical and clinical studies demonstrating potential. OV therapy has emerged as a potent and effective method with a dual mechanism. Developing innovative and effective strategies for virus transduction, coupled with immune checkpoint inhibitors or chemotherapy drugs, strengthens this new technique. Furthermore, the discovery and creation of new OVs that can seamlessly integrate gene therapy strategies, such as cytotoxic, anti-angiogenic, and immunostimulatory, are promising advancements. This review presents an overview of the latest advancements in OVs transduction for brain cancer, focusing on the safety and effectiveness of G207, G47Δ, M032, rQNestin34.5v.2, C134, DNX-2401, Ad-TD-nsIL12, NSC-CRAd-S-p7, TG6002, and PVSRIPO. These are evaluated in both preclinical and clinical models of various brain tumors.
Collapse
Affiliation(s)
- Kasra Mokhtarpour
- Animal Model Integrated Network (AMIN), Universal Scientific Education and Research Network (USERN), Tehran, 1419733151, Iran
| | - Milad Akbarzadehmoallemkolaei
- Animal Model Integrated Network (AMIN), Universal Scientific Education and Research Network (USERN), Tehran, 1419733151, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Gharib St, Keshavarz Blvd, Tehran, 1419733151, Iran
| | - Nima Rezaei
- Animal Model Integrated Network (AMIN), Universal Scientific Education and Research Network (USERN), Tehran, 1419733151, Iran.
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Gharib St, Keshavarz Blvd, Tehran, 1419733151, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, 1417653761, Iran.
| |
Collapse
|
7
|
Ilangumaran S, Gui Y, Shukla A, Ramanathan S. SOCS1 expression in cancer cells: potential roles in promoting antitumor immunity. Front Immunol 2024; 15:1362224. [PMID: 38415248 PMCID: PMC10897024 DOI: 10.3389/fimmu.2024.1362224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 01/31/2024] [Indexed: 02/29/2024] Open
Abstract
Suppressor of cytokine signaling 1 (SOCS1) is a potent regulator immune cell responses and a proven tumor suppressor. Inhibition of SOCS1 in T cells can boost antitumor immunity, whereas its loss in tumor cells increases tumor aggressivity. Investigations into the tumor suppression mechanisms so far focused on tumor cell-intrinsic functions of SOCS1. However, it is possible that SOCS1 expression in tumor cells also regulate antitumor immune responses in a cell-extrinsic manner via direct and indirect mechanisms. Here, we discuss the evidence supporting the latter, and its implications for antitumor immunity.
Collapse
Affiliation(s)
- Subburaj Ilangumaran
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | | | | |
Collapse
|
8
|
Kandhi R, Menendez A, Ramanathan S, Ilangumaran S. Regulation of High-fat Diet-induced Liver Fibrosis by SOCS1 Expression in Hepatic Stellate Cells. J Clin Exp Hepatol 2024; 14:101280. [PMID: 38076369 PMCID: PMC10709186 DOI: 10.1016/j.jceh.2023.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/03/2023] [Indexed: 10/10/2024] Open
Abstract
Background Hepatic stellate cells (HSC) are the key mediators of fibrosis development in non-alcoholic fatty liver disease (NAFLD). Hepatic inflammation induced by high-fat diet activates HSCs, which differentiate to myofibroblasts and produce extracellular fibrillar matrix. HSC activation during hepatic fibrogenesis is modulated by cytokines and growth factors produced by stressed hepatocytes and macrophages. SOCS1 is a negative feedback regulator of certain cytokines and growth factors implicated in liver fibrosis. Aim The goal of this study was to understand the regulatory functions of SOCS1 in HSCs during NAFLD-induced liver fibrosis. Methodology Mice lacking SOCS1 specifically in HSCs (Socs1ΔHSC) and control Socs1-floxed (Socs1fl/fl) mice were fed choline-deficient L-amino acid-defined high-fat diet (CDA-HFD) or normal control diet for 14 weeks. Body weight gain was regularly monitored. Serum alanine aminotransferase levels and liver weight were assessed at the endpoint. Fibrosis development was evaluated by Sirius red staining and hydroxyproline content, and myofibroblast differentiation by immunohistochemistry. Expression of genes encoding pro-fibrogenic factors, cytokines, growth factors and chemokines, and the phenotype and numbers of intrahepatic leukocytes were evaluated. Results Socs1ΔHSC mice showed increased liver/body weight ratio and displayed increased collagen deposition and myofibroblast differentiation. Induction of Acta2, Col1a1, Pdgfb, IL1b and Ccl2 genes was significantly elevated in Socs1ΔHSC mice compared to Socs1fl/fl controls fed CDA-HFD. Tgfb gene induction was comparable between the two groups, however, Socs1ΔHSC livers displayed increased SMAD3 phosphorylation. The fibrotic livers of Socs1ΔHSC mice showed increased inflammatory cell infiltration, and flow cytometry analysis revealed elevated numbers of myeloid cells, granulocytes and myeloid-derived dendritic cells. Socs1ΔHSC livers harbored increased numbers of Ly6ChiCCR2+ pro-inflammatory macrophages, largely comprised of Ly6ChiCCR2+CX3CR1+ cells, suggesting impaired transition to anti-inflammatory macrophages. Conclusion Our findings show that SOCS1 exerts non-redundant regulatory functions in HSCs that are critical for attenuating high-fat diet-induced inflammatory response and liver fibrosis development.
Collapse
Affiliation(s)
- Rajani Kandhi
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Alfredo Menendez
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Sheela Ramanathan
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Subburaj Ilangumaran
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| |
Collapse
|
9
|
Bai J, Chen S. LncRNA CASC9 enhances the stability of SOCS-1 by combining with FUS to alleviate sepsis-induced liver injury. Cytokine 2023; 171:156346. [PMID: 37751673 DOI: 10.1016/j.cyto.2023.156346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 07/25/2023] [Accepted: 08/21/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND Liver injury plays a major role in the development of sepsis. Liver damage after sepsis is an independent risk factor for multiple organ failure and death. Cancer susceptibility candidate 9 (CASC9) exerts a protective effect on sepsis-induced acute lung injury (ALI). However, the role and underlying mechanism haven't been fully evaluated. METHODS Animal and cell models of sepsis were established in vivo and in vitro experiments. The histological and apoptosis analyses of liver tissues were tested by hematoxylin-eosin (HE) staining and terminal dUTP nick end labeling (TUNEL) assay, respectively. Serum levels of inflammatory cytokines were detected via using an enzyme-linked immunosorbent assay (ELISA). The expressions of CASC9, suppressor of cytokine signaling (SOCS)-1, Bcl-2, Bax, Bad, and caspase3 were measured by reverse-transcription quantitative polymerase chain reaction (RT-qPCR) and western blotting. Cell counting kit-8 (CCK-8) and flow cytometry were applied to examine cell viability and apoptosis, respectively. RNA immunoprecipitation (RIP) and RNA-pull down assay were used to verify the binding relationships among CASC9, SOCS-1 and FUS. RESULTS CASC9 and SOCS-1 were lowly expressed in animal and cell models of sepsis liver injury. CASC9 or SOCS-1 overexpression could inhibit cell apoptosis upon lipopolysaccharide (LPS) induction. Meanwhile, the serum levels of tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6 and IL-8 were reduced by CASC9 or SOCS-1 overexpression in LPS-induced LO2 cells. Mechanistically, CASC9 interacted with fused in sarcoma (FUS) to stabilize the mRNA of SOCS-1. SOCS-1 silencing antagonized the effects of CASC9 on improving sepsis liver injury. CONCLUSION CASC9 overexpression ameliorated the sepsis-induced liver injury, and the probable underlying mechanism may be that CASC9 stabilized the SOCS-1 mRNA by interacting with FUS.
Collapse
Affiliation(s)
- Jinquan Bai
- The Affiliated Nanhua Hospital, Department of Critical Care Medicine, Hengyang Medical School, University of South China, Hengyang 421002, Hunan Province, PR China
| | - Shiyi Chen
- The Affiliated Nanhua Hospital, Department of Hepatobiliary Surgery, Hengyang Medical School, University of South China, Hengyang 421002, Hunan Province, PR China.
| |
Collapse
|
10
|
Kandhi R, Yeganeh M, Yoshimura A, Menendez A, Ramanathan S, Ilangumaran S. Hepatic stellate cell-intrinsic role of SOCS1 in controlling hepatic fibrogenic response and the pro-inflammatory macrophage compartment during liver fibrosis. Front Immunol 2023; 14:1259246. [PMID: 37860002 PMCID: PMC10582746 DOI: 10.3389/fimmu.2023.1259246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023] Open
Abstract
Introduction Hepatic stellate cells (HSC) become activated, differentiate to myofibroblasts and produce extracellular fibrillar matrix during liver fibrosis. The hepatic fibrogenic response is orchestrated by reciprocal interactions between HSCs and macrophages and their secreted products. SOCS1 can regulate several cytokines and growth factors implicated in liver fibrosis. Here we investigated the role of SOCS1 in regulating HSC activation. Methods Mice lacking SOCS1 in HSCs (Socs1ΔHSC) were generated by crossing Socs1fl/fl and LratCre mice. Liver fibrosis was induced by carbon tetrachloride and evaluated by Sirius red staining, hydroxyproline content and immunostaining of myofibroblasts. Gene expression of pro-fibrogenic factors, cytokines, growth factors and chemokines were quantified by RT-qPCR. The phenotype and the numbers of intrahepatic leukocyte subsets were studied by flow cytometry. The impact of fibrosis on the development of diethyl nitrosamine-induced hepatocellular carcinoma was evaluated. Results Socs1ΔHSC mice developed more severe liver fibrosis than control Socs1fl/fl mice that was characterized by increased collagen deposition and myofibroblast differentiation. Socs1ΔHSC mice showed a significant increase in the expression of smooth muscle actin, collagens, matrix metalloproteases, cytokines, growth factors and chemokines in the liver following fibrosis induction. The fibrotic livers of Socs1ΔHSC mice displayed heightened inflammatory cell infiltration with increased proportion and numbers of Ly6ChiCCR2+ pro-inflammatory macrophages. This macrophage population contained elevated numbers of CCR2+CX3CR1+ cells, suggesting impaired transition towards restorative macrophages. Fibrosis induction following exposure to diethyl nitrosamine resulted in more numerous and larger liver tumor nodules in Socs1ΔHSC mice than in Socs1fl/fl mice. Discussion Our findings indicate that (i) SOCS1 expression in HSCs is a critical to control liver fibrosis and development of hepatocaellular carcinoma, and (ii) attenuation of HSC activation by SOCS1 regulates pro-inflammatory macrophage recruitment and differentiation during liver fibrosis.
Collapse
Affiliation(s)
- Rajani Kandhi
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Mehdi Yeganeh
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Alfredo Menendez
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Sheela Ramanathan
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Subburaj Ilangumaran
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
11
|
Wu YY, Xu YM, Lau ATY. Epigenetic effects of herbal medicine. Clin Epigenetics 2023; 15:85. [PMID: 37179342 PMCID: PMC10183144 DOI: 10.1186/s13148-023-01481-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 04/08/2023] [Indexed: 05/15/2023] Open
Abstract
Epigenetic memory is essential for life that governs the predefined functional features of cells. Recent evidence has indicated that the epigenetic modification provides a potential link to gene expression changes that may be involved in the development of various chronic diseases, and targeting the epigenome becomes a plausible method for treating diseases. Traditional herbal medicine has gradually entered the vision of researchers due to its low toxicity and its effectiveness in treating diseases. As a matter of fact, researchers found that the possessed epigenetic modification capacity of herbal medicine had the ability to combat the progression of the disease, such as various types of cancer, diabetes, inflammation, amnesia, liver fibrosis, asthma, and hypertension-induced renal injury. Studies on the epigenetic effects of herbal medicine will provide valuable insights into the molecular mechanisms of human diseases, which may lead to new therapeutic approaches and diagnoses. Thus, this review summarized the impact of herbal medicine and its bioactive components on disease epigenome as examples of how utilization of epigenetic plasticity could be useful as the basis for the future development of targeted therapies in chronic diseases.
Collapse
Affiliation(s)
- Yu-Yao Wu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
| | - Yan-Ming Xu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
| | - Andy T Y Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China.
| |
Collapse
|
12
|
Jiao Y, Yan Z, Yang A. The Roles of Innate Lymphoid Cells in the Gastric Mucosal Immunology and Oncogenesis of Gastric Cancer. Int J Mol Sci 2023; 24:ijms24076652. [PMID: 37047625 PMCID: PMC10095467 DOI: 10.3390/ijms24076652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/25/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
Innate lymphoid cells (ILCs) are a group of innate immune cells that have garnered considerable attention due to their critical roles in regulating immunity and tissue homeostasis. They are particularly abundant in the gastrointestinal tract, where they have been shown to interact with commensal bacteria, pathogens, and other components of the local microenvironment to influence host immune responses to infection and oncogenesis. Their tissue-residency properties enable gastric ILCs a localized and rapid response to alert and stress, which indicates their key potential in regulating immunosurveillance. In this review, we discuss the current understanding of the role of ILCs in the gastric mucosa, with a focus on their interactions with the gastric microbiota and Helicobacter pylori and their contributions to tissue homeostasis and inflammation. We also highlight recent findings on the involvement of ILCs in the pathogenesis of gastric cancer and the implications of targeting ILCs as a therapeutic approach. Overall, this review provides an overview of the diverse functions of ILCs in gastric mucosa and highlights their potential as targets for future therapies for gastric cancer.
Collapse
Affiliation(s)
- Yuhao Jiao
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Zhiyu Yan
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
- 4 + 4 M.D. Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Aiming Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
13
|
SOCS1 Deficiency Promotes Hepatocellular Carcinoma via SOCS3-Dependent CDKN1A Induction and NRF2 Activation. Cancers (Basel) 2023; 15:cancers15030905. [PMID: 36765862 PMCID: PMC9913612 DOI: 10.3390/cancers15030905] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
SOCS1 deficiency, which increases susceptibility to hepatocellular carcinoma (HCC), promotes CDKN1A expression in the liver. High CDKN1A expression correlates with disease severity in many cancers. Here, we demonstrate a crucial pathogenic role of CDKN1A in diethyl nitrosamine (DEN)-induced HCC in SOCS1-deficient mice. Mechanistic studies on DEN-induced genotoxic response revealed that SOCS1-deficient hepatocytes upregulate SOCS3 expression, SOCS3 promotes p53 activation, and Cdkn1a induction that were abolished by deleting either Socs3 or Tp53. Previous reports implicate CDKN1A in promoting oxidative stress response mediated by NRF2, which is required for DEN-induced hepatocarcinogenesis. We show increased induction of NRF2 and its target genes in SOCS1-deficient livers following DEN treatment that was abrogated by the deletion of either Cdkn1a or Socs3. Loss of SOCS3 in SOCS1-deficient mice reduced the growth of DEN-induced HCC without affecting tumor incidence. In the TCGA-LIHC dataset, the SOCS1-low/SOCS3-high subgroup displayed increased CDKN1A expression, enrichment of NRF2 transcriptional signature, faster disease progression, and poor prognosis. Overall, our findings show that SOCS1 deficiency in hepatocytes promotes compensatory SOCS3 expression, p53 activation, CDKN1A induction, and NRF2 activation, which can facilitate cellular adaptation to oxidative stress and promote neoplastic growth. Thus, the NRF2 pathway represents a potential therapeutic target in SOCS1-low/SOCS3-high HCC cases.
Collapse
|
14
|
Li D, Guo X, Zhao W, Jingyu J, Xia C, Yu G. Genome-wide DNA methylation dynamics in carbon tetrachloride-induced mice liver fibrosis. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:85-92. [PMID: 36594057 PMCID: PMC9790058 DOI: 10.22038/ijbms.2022.66256.14555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 11/22/2022] [Indexed: 01/04/2023]
Abstract
Objectives Many persistent harmful stimuli can result in chronic liver diseases, which lead to about 2 million deaths per year in the whole world. Liver fibrosis was found to exist in all kinds of chronic liver diseases. Many studies suggested that DNA methylation was associated with the pathogenesis of liver fibrosis. This study aimed to quantitatively detect DNA methylation changes in the whole genome in fibrotic liver tissues of mice. Materials and Methods Liver fibrosis was induced by intraperitoneal injection of carbon tetrachloride (CCl4) for 4 weeks. A genome-wide methylome analysis was performed using 850K BeadChips assays. The methylation status of 27 CpG dinucleotides located in 3 genes was detected by pyrosequencing to confirm chip data accuracy, and mRNA expressions of these 3 genes were examined by RT-qPCR methods. Results A total of 130,068 differentially methylated sites (DMS, 58,474 hypermethylated, and 71,594 hypomethylated) between fibrotic liver tissues and control mice liver tissues were identified by the 850k BeadChips array. Consistency between pyrosequencing data and 850k BeadChips array data was observed (R=0.928; P<0.01). Apoptosis, positive regulation of transcription of Notch receptor target, and negative regulation of p38MAPK signal cascade activities were significantly enriched in the Gene Ontology (GO) analyses. Cholesterol metabolism, bile secretion, and more biosynthesis and metabolism pathways were enriched in KEGG pathway analyses. Ten key genes were identified by the Cytoscape plugin cytoHubba. Conclusion 7850 genes were found to have methylation change in fibrotic liver tissues of mice, which facilitates future research for clinical application.
Collapse
Affiliation(s)
- Deming Li
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), College of Life Science, Henan Normal University, Xinxiang, Henan, China,These authors contributed eqully to this work
| | - Xiaoshu Guo
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), College of Life Science, Henan Normal University, Xinxiang, Henan, China,Department of Physiology, Changzhi Medical College, Shanxi, China,These authors contributed eqully to this work
| | - Wenyu Zhao
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Jingyu Jingyu
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Cong Xia
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Guoying Yu
- State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), College of Life Science, Henan Normal University, Xinxiang, Henan, China,Corresponding author: Guoying Yu. State Key Laboratory Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), College of Life Science, Henan Normal University, Xinxiang, Henan, China. Tel: +86-03733326340; Fax: +86-0373 3326524;
| |
Collapse
|
15
|
Edwards JS, Delabat SA, Badilla AD, DiCaprio RC, Hyun J, Burgess RA, Silva T, Dykxhoorn DM, Chen SX, Wang L, Ishida Y, Saito T, Thomas E. Downregulation of SOCS1 increases interferon-induced ISGylation during differentiation of induced-pluripotent stem cells to hepatocytes. JHEP REPORTS : INNOVATION IN HEPATOLOGY 2022; 4:100592. [PMID: 36439639 PMCID: PMC9685392 DOI: 10.1016/j.jhepr.2022.100592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 12/24/2022]
Abstract
Background & Aims Increased expression of IFN-stimulated gene 15 (ISG15) and subsequently increased ISGylation are key factors in the host response to viral infection. In this study, we sought to characterize the expression of ISG15, ISGylation, and associated enzymes at each stage of differentiation from induced pluripotent stem cells (iPSCs) to hepatocytes. Methods To study the regulation of ISGylation, we utilized patient samples and in vitro cell culture models including iPSCs, hepatocytes-like cells, immortalized cell lines, and primary human hepatocytes. Protein/mRNA expression were measured following treatment with poly(I:C), IFNα and HCV infection. Results When compared to HLCs, we observed several novel aspects of the ISGylation pathway in iPSCs. These include a lower baseline expression of the ISGylation-activating enzyme, UBE1L, a lack of IFN-induced expression of the ISGylation-conjugation enzyme UBE2L6, an attenuated activation of the transcription factor STAT1 and constitutive expression of SOCS1. ISGylation was observed in iPSCs following downregulation of SOCS1, which facilitated STAT1 activation and subsequently increased expression of UBE2L6. Intriguingly, HCV permissive transformed hepatoma cell lines demonstrated higher intrinsic expression of SOCS1 and weaker ISGylation following IFN treatment. SOCS1 downregulation in HCV-infected Huh 7.5.1 cells led to increased ISGylation. Conclusions Herein, we show that high basal levels of SOCS1 inhibit STAT1 activation and subsequently IFN-induced UBE2L6 and ISGylation in iPSCs. Furthermore, as iPSCs differentiate into hepatocytes, epigenetic mechanisms regulate ISGylation by modifying UBE1L and SOCS1 expression levels. Overall, this study demonstrates that the development of cell-intrinsic innate immunity during the differentiation of iPSCs to hepatocytes provides insight into cell type-specific regulation of host defense responses and related oncogenic processes. Impact and implications To elucidate the mechanism underlying regulation of ISGylation, a key process in the innate immune response, we studied changes in ISGylation-associated genes at the different stages of differentiation from iPSCs to hepatocytes. We found that high basal levels of SOCS1 inhibit STAT1 activation and subsequently IFN-induced UBE2L6 and ISGylation in iPSCs. Importantly, epigenetic regulation of SOCS1 and subsequently ISGylation may be important factors in the development of cell type-specific host defense responses in hepatocytes that should be considered when studying chronic infections and oncogenic processes in the liver.
Collapse
Key Words
- AFP, alpha-fetoprotein
- ALB, albumin
- Antiviral Response
- Epigenetic Regulation
- FOXA2, forkhead Box A2
- HB, hepatoblast
- HCC, hepatocellular carcinoma
- HCV
- HLC, hepatocyte-like cell
- Hepatocellular Carcinoma
- Host Defense
- IFN, interferon
- IRF3, interferon regulatory factor 3
- ISG, interferon-stimulated gene
- ISG15
- Innate Immunity
- JAK, Janus kinase
- Liver Cancer
- OCT4, octamer-binding transcription factor 4
- PHHs, primary human hepatocytes
- RIG-I, retinoic acid-inducible gene I
- RLR, RIGI-like receptor
- RNAseq, RNA sequencing
- SOCS1
- SOCS1, suppressor of cytokine signaling 1
- STAT1
- STAT1, signal transducer and activator of transcription 1
- TLR, toll-like receptor
- UBE1L/UBA7, ubiquitin-activating enzyme E1
- USP18, deconjugation enzyme ubiquitin specific peptidase 18
- UbcH8/UBE2L6, ubiquitin-conjugating enzyme E2 L6
- iPSC, induced-pluripotent stem cell
- pSTAT1, phosphorylated STAT1
Collapse
Affiliation(s)
- Jasmine S. Edwards
- University of Miami Miller School of Medicine Department of Microbiology and Immunology, USA
| | | | - Alejandro D. Badilla
- University of Miami Miller School of Medicine Department of Microbiology and Immunology, USA
| | - Robert C. DiCaprio
- University of Miami Miller School of Medicine Department of Pathology, USA
| | - Jinhee Hyun
- University of Miami Miller School of Medicine Department of Pathology, USA
| | - Robert A. Burgess
- University of Miami Miller School of Medicine Department of Pathology, USA
| | - Tiago Silva
- University of Miami Department of Public Health Sciences, USA
| | - Derek M. Dykxhoorn
- University of Miami Miller School of Medicine Department of Human Genetics, USA
| | - Steven Xi Chen
- University of Miami Department of Public Health Sciences, USA
| | - Lily Wang
- University of Miami Department of Public Health Sciences, USA
| | - Yuji Ishida
- Department of Medicine, Division of Gastrointestinal and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Research & Development Department, PhoenixBio, Co., Ltd, Higashi-Hiroshima, Hiroshima, Japan
| | - Takeshi Saito
- Department of Medicine, Division of Gastrointestinal and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,USC Research Center for Liver Diseases, Los Angeles, California, USA
| | - Emmanuel Thomas
- University of Miami Miller School of Medicine Department of Microbiology and Immunology, USA,University of Miami Miller School of Medicine Department of Pathology, USA,Corresponding author. Address: 1550 NW 10th Avenue, Papanicolaou Building Room 109, Miami, FL 33136, United States; Tel.: (305) 243-2895.
| |
Collapse
|
16
|
Kang I, Kim JA, Kim J, Lee JH, Kim MJ, Ahn JK. Hepatitis B virus X protein promotes epithelial-mesenchymal transition of hepatocellular carcinoma cells by regulating SOCS1. BMB Rep 2022. [PMID: 35168698 PMCID: PMC9152579 DOI: 10.5483/bmbrep.2022.55.5.157] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Hepatocellular carcinoma (HCC), a primary type of liver cancer, is one of the leading causes of cancer related deaths worldwide. HCC patients have poor prognosis due to intrahepatic and extrahepatic metastasis. Hepatitis B virus (HBV) infection is one of the major causes of various liver diseases including HCC. Among HBV gene products, HBV X protein (HBx) plays an important role in the development and metastasis of HCC. However, the mechanism of HCC metastasis induced by HBx has not been elucidated yet. In this study, for the first time, we report that HBx interacts with the suppressor of cytokine signaling 1 (SOCS1) which negatively controls NF-κB by degrading p65, a subunit of NF-κB. NF-κB activates the transcription of factors associated with epithelial-mesenchymal transition (EMT), a crucial cellular process associated with invasiveness and migration of cancer cells. Here, we report that HBx physically binds to SOCS1, subsequently prevents the ubiquitination of p65, activates the transcription of EMT transcription factors and enhance cell migration and invasiveness, suggesting a new mechanism of HBV-associated HCC metastasis.
Collapse
Affiliation(s)
- Inho Kang
- Department of Microbiology & Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 34134, Korea
| | - Ji Ae Kim
- Department of Microbiology & Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 34134, Korea
| | - Jinchul Kim
- Department of Microbiology & Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 34134, Korea
| | - Ju Hyeon Lee
- Department of Microbiology & Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 34134, Korea
| | - Mi-jee Kim
- Department of Microbiology & Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 34134, Korea
| | - Jeong Keun Ahn
- Department of Microbiology & Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
17
|
Masuzaki R, Kanda T, Sasaki R, Matsumoto N, Nirei K, Ogawa M, Karp SJ, Moriyama M, Kogure H. Suppressors of Cytokine Signaling and Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:cancers14102549. [PMID: 35626153 PMCID: PMC9139988 DOI: 10.3390/cancers14102549] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 05/21/2022] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Hepatocellular carcinoma (HCC) is a common malignancy worldwide. The HCC generally develops in the liver of patients already suffering from chronic liver disease. There have been significant advances in both the curative and palliative treatment of HCC. Although liver resection is a curative treatment for HCC, its indication is often limited due to an impaired liver function reservoir. There is still a need to understand how to control liver regeneration after resection and find better cancer immunotherapy and anticancer drugs for advanced HCC. Suppressors of cytokine signaling (SOCS) negatively regulate cytokine signaling related to cell proliferation, differentiation, and immune response; therefore, SOCS are thought to play an important role in HCC development and liver regeneration. Abstract Cytokines are secreted soluble glycoproteins that regulate cellular growth, proliferation, and differentiation. Suppressors of cytokine signaling (SOCS) proteins negatively regulate cytokine signaling and form a classical negative feedback loop in the signaling pathways. There are eight members of the SOCS family. The SOCS proteins are all comprised of a loosely conserved N-terminal domain, a central Src homology 2 (SH2) domain, and a highly conserved SOCS box at the C-terminus. The role of SOCS proteins has been implicated in the regulation of cytokines and growth factors in liver diseases. The SOCS1 and SOCS3 proteins are involved in immune response and inhibit protective interferon signaling in viral hepatitis. A decreased expression of SOCS3 is associated with advanced stage and poor prognosis of patients with hepatocellular carcinoma (HCC). DNA methylations of SOCS1 and SOCS3 are found in HCC. Precise regulation of liver regeneration is influenced by stimulatory and inhibitory factors after partial hepatectomy (PH), in particular, SOCS2 and SOCS3 are induced at an early time point after PH. Evidence supporting the important role of SOCS signaling during liver regeneration also supports a role of SOCS signaling in HCC. Immuno-oncology drugs are now the first-line therapy for advanced HCC. The SOCS can be potential targets for HCC in terms of cell proliferation, cell differentiation, and immune response. In this literature review, we summarize recent findings of the SOCS family proteins related to HCC and liver diseases.
Collapse
Affiliation(s)
- Ryota Masuzaki
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Itabashi, Tokyo 173-8610, Japan; (T.K.); (R.S.); (N.M.); (K.N.); (M.O.); (M.M.); (H.K.)
- Correspondence: ; Tel.: +81-3-3972-8111
| | - Tatsuo Kanda
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Itabashi, Tokyo 173-8610, Japan; (T.K.); (R.S.); (N.M.); (K.N.); (M.O.); (M.M.); (H.K.)
| | - Reina Sasaki
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Itabashi, Tokyo 173-8610, Japan; (T.K.); (R.S.); (N.M.); (K.N.); (M.O.); (M.M.); (H.K.)
| | - Naoki Matsumoto
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Itabashi, Tokyo 173-8610, Japan; (T.K.); (R.S.); (N.M.); (K.N.); (M.O.); (M.M.); (H.K.)
| | - Kazushige Nirei
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Itabashi, Tokyo 173-8610, Japan; (T.K.); (R.S.); (N.M.); (K.N.); (M.O.); (M.M.); (H.K.)
| | - Masahiro Ogawa
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Itabashi, Tokyo 173-8610, Japan; (T.K.); (R.S.); (N.M.); (K.N.); (M.O.); (M.M.); (H.K.)
| | - Seth J. Karp
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Mitsuhiko Moriyama
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Itabashi, Tokyo 173-8610, Japan; (T.K.); (R.S.); (N.M.); (K.N.); (M.O.); (M.M.); (H.K.)
| | - Hirofumi Kogure
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Itabashi, Tokyo 173-8610, Japan; (T.K.); (R.S.); (N.M.); (K.N.); (M.O.); (M.M.); (H.K.)
| |
Collapse
|
18
|
Li L, Zhang J, Peng H, Jiang X, Liu Z, Tian H, Hou S, Xie X, Peng Q, Zhou T. Knockdown of miR-92a suppresses the stemness of colorectal cancer cells via mediating SOCS3. Bioengineered 2022; 13:5613-5624. [PMID: 35184640 PMCID: PMC8974062 DOI: 10.1080/21655979.2021.2022267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
MiRNAs (microRNAs) participate in colorectal cancer (CRC) progression and act as potential biomarkers for CRC prognosis. In this study, we investigated the mechanisms of microRNA-92a (miR-92a) in CRC. Expressions of miR-92a and SOCS3 (Suppressor Of Cytokine Signaling 3) were investigated by qRT-PCR in CRC cell lines and 30 cases of CRC. The self-renewal capacity and proliferation of CRC stem cells were estimated by the sphere formation assay, EdU staining, and Flow cytometry analysis. Moreover, the interplay between miR-92a and SOCS3 in CRC cells was validated by luciferase reporter experiments. MiR-92a was found to be remarkably increased while SOCS3 was significantly downregulated in CRC tissues. Inhibition of miR-92a or SOCS3 attenuated the sphere formation capacity, decreased expressions of stemness-related proteins, and inhibited the proliferation of cancer stem-like cells. Knockdown of SOCS3 reversed the repressive impacts of miR-92a inhibitors on self-renewal and growth of CRC cancer stem cells. This study suggested that miR-92a functions as an oncogene of CRC through mediating the stemness of colorectal cancer cells by directly binding and repressing SOCS3.
Collapse
Affiliation(s)
- Lifa Li
- Department of Gastrointestinal Surgery II, Hepatobiliary and Pancreatic Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jingxiao Zhang
- Department of Medica, The Second Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hong Peng
- Department of Anorectal Surgery, Nanchong Central Hospital, the Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xianhong Jiang
- Department of Gastrointestinal Surgery II, Hepatobiliary and Pancreatic Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Zuoliang Liu
- Department of Gastrointestinal Surgery II, Hepatobiliary and Pancreatic Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hongpeng Tian
- Department of Gastrointestinal Surgery II, Hepatobiliary and Pancreatic Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Songlin Hou
- Department of Gastrointestinal Surgery II, Hepatobiliary and Pancreatic Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xingjiang Xie
- Department of Gastrointestinal Surgery II, Hepatobiliary and Pancreatic Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Qiang Peng
- Department of Gastrointestinal Surgery II, Hepatobiliary and Pancreatic Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Tong Zhou
- Department of Gastrointestinal Surgery II, The Second Affiliated Hospital of North Sichuan Medical College, Sichuan, China
- Department of Gastrointestinal Surgery II, Affiliated Hospital of North Sichuan Medical College, Sichuan, China
| |
Collapse
|
19
|
OUP accepted manuscript. Hum Reprod 2022; 37:1489-1504. [DOI: 10.1093/humrep/deac098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/13/2022] [Indexed: 11/14/2022] Open
|
20
|
Zhu H, Zhao H, Xu S, Zhang Y, Ding Y, Li J, Huang C, Ma T. Sennoside A alleviates inflammatory responses by inhibiting the hypermethylation of SOCS1 in CCl 4-induced liver fibrosis. Pharmacol Res 2021; 174:105926. [PMID: 34619344 DOI: 10.1016/j.phrs.2021.105926] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 02/07/2023]
Abstract
Liver fibrosis is the consequence of chronic liver injury and is a major challenge to global health. However, successful therapy for liver fibrosis is still lacking. Sennoside A (SA), a commonly used clinical stimulant laxative, is reported to improve hepatic disease, but the underlying mechanisms remain largely elusive. Here, we show for the first time that SA enhanced suppressor of cytokine signaling 1 (SOCS1) expression in a DNA methyltransferase 1 (DNMT1)-dependent manner and thereby attenuated liver fibrosis. Consistently, SA inhibited the expression of the liver fibrogenesis markers α-smooth muscle actin (α-SMA) and type I collagen alpha-1 (Col1α1) and suppressed inflammatory responses in vivo and in vitro. Coculture experiments with macrophages/hepatic stellate cells (HSCs) revealed that SA suppressed HSC proliferation by downregulating proinflammatory cytokines in macrophages. Mechanically, SA promoted the aberrant expression of SOCS1 in liver fibrosis. However, blocking SOCS1 expression weakened the inhibitory effect of SA on HSC proliferation, indicating that SOCS1 may play an important role in mediating the antifibrotic effect of SA. Furthermore, SA inhibited DNMT1-mediated SOCS1 and reduced HSC proliferation by inhibiting inflammatory responses in carbon tetrachloride (CCl4) -induced liver fibrosis.
Collapse
Affiliation(s)
- Hong Zhu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Huizi Zhao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Songbing Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yuan Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yuhao Ding
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Taotao Ma
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
21
|
Sun H, Li Y, Quan X, Chen N, Jin X, Jin W, Jin Y, Shen X. PIAS3/SOCS1-STAT3 axis responses to oxidative stress in hepatocellular cancer cells. Am J Transl Res 2021; 13:12395-12409. [PMID: 34956461 PMCID: PMC8661178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/23/2021] [Indexed: 06/14/2023]
Abstract
The participation of STAT3 and its upstream inhibitors, PIAS3 and SOCS1, in the oxidative response of hepatocellular carcinoma (HCC) cells was uncertain. Here, the expression of PIAS3 and SOCS1 in HCC tissues and cell lines was explored, and we sought to determine whether oxidative stress epigenetically regulated PIAS3 and SOCS1 expression and STAT3 activation in HCC cells. The expression of PIAS3 and SOCS1 was markedly decreased in HCC cell lines and tissues compared to normal hepatic cells and tissues. In HCC patients, low PIAS3 and SOCS1 expression were associated with poor survival. Oxidative stress induced by H2O2 in HepG2 cells was indicated by low antioxidant levels and high protein carbonyl content. Moreover, oxidative stress in HepG2 cells contributed to reduced proliferation but increased apoptosis, migration, and invasion capacity, which might be counteracted by antioxidants, such as tocopheryl acetate (TA). PIAS3 and SOCS1 expression was markedly decreased, while STAT3 was activated in HepG2 cells in response to H2O2 exposure. Co-treatment with antioxidant TA effectively increased the expression of PIAS3 and SOCS1, but it dephosphorylated STAT3 in H2O2-treated cells. PIAS1 or SOCS1 overexpression in HepG2 cells after H2O2 treatment restored cell viability and anti-oxidative responses and decreased apoptosis, migration, and invasion ability, and dephosphorylated STAT3 levels. Co-administration of the STAT3 activator, colivelin, partially abolished the effect of PIAS3 and SOCS1 overexpression in these processes. Therefore, oxidative stress in HCC cells may improve their migration and reduce proliferation through STAT3 activation through the repression of PIAS3 and SOCS1 expression.
Collapse
Affiliation(s)
- Honghua Sun
- Department of Oncology, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Yanglong Li
- Department of Oncology, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Xianglan Quan
- Department of Oncology, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Ning Chen
- Department of Infection Disease, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Xinglin Jin
- Department of General Surgert, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Wenbiao Jin
- Department of Oncology, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Yongmin Jin
- Department of Oncology, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Xionghu Shen
- Department of Oncology, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| |
Collapse
|
22
|
Stevenson AW, Melton PE, Moses EK, Wallace HJ, Wood FM, Rea S, Danielsen PL, Alghamdi M, Hortin N, Borowczyk J, Deng Z, Manzur M, Fear MW. A methylome and transcriptome analysis of normal human scar cells reveals a role for FOXF2 in scar maintenance. J Invest Dermatol 2021; 142:1489-1498.e12. [PMID: 34687743 DOI: 10.1016/j.jid.2021.08.445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 07/22/2021] [Accepted: 08/01/2021] [Indexed: 12/12/2022]
Abstract
Scar is maintained for life and increases in size during periods of growth such as puberty. Epigenetic changes in fibroblasts after injury may underpin the maintenance and growth of scar. Here, we, combined methylome and transcriptome data from normotrophic mature scar and contralateral uninjured normal skin fibroblasts to identify potential regulators of scar maintenance. 219 significantly differentially expressed and 1199 significantly differentially methylated promoters were identified, of which there were 12 genes both significantly differentially methylated and expressed. Of these the two transcription factors, Forkhead Box F2 (FOXF2) and Mohawk Homeobox (MKX) were selected for further analysis. Immunocytochemistry and qPCR suggested FOXF2 but not MKX had elevated expression in scar fibroblasts. Using RNASeq, FOXF2 knockdown was shown to significantly reduce expression of extracellular matrix related genes, whilst MKX did not appear to affect similar pathways. Finally, FOXF2 knockdown was also shown to significantly decrease collagen I production in scar and keloid fibroblasts. This study provides insights into the maintenance of normotrophic scar, suggesting FOXF2 is an important regulator of this process. Targeting genes responsible for maintenance of scar phenotype may ameliorate scar appearance and improve patient outcomes in the future.
Collapse
Affiliation(s)
- Andrew W Stevenson
- Burn Injury Research Unit, School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Australia.
| | - Phillip E Melton
- School of Population and Global Health, The University of Western Australia, Perth, Australia; School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Perth, Australia; Menzies Research Institute, University of Tasmania, Hobart, Tasmania, Australia
| | - Eric K Moses
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Perth, Australia; Menzies Research Institute, University of Tasmania, Hobart, Tasmania, Australia; School of Biomedical Sciences, University of Western Australia, Crawley, Western Australia, Australia
| | - Hilary J Wallace
- Burn Injury Research Unit, School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Australia; School of Medicine, The University of Notre Dame Australia, Fremantle, Australia
| | - Fiona M Wood
- Burn Injury Research Unit, School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Australia; Burns Service of Western Australia, Perth Children's Hospital and Fiona Stanley Hospital, Perth, Australia
| | - Suzanne Rea
- Burns Service of Western Australia, Perth Children's Hospital and Fiona Stanley Hospital, Perth, Australia
| | - Patricia L Danielsen
- Department of Dermatology and Copenhagen Wound Healing Center, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mansour Alghamdi
- Department of Anatomy, College of Medicine, King Khalid University, Abha, Saudi Arabia; Genomics and Personalised Medicine Unit, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Nicole Hortin
- Burn Injury Research Unit, School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Australia
| | - Julia Borowczyk
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Zhenjun Deng
- Burn Injury Research Unit, School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Australia
| | - Mitali Manzur
- Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, Nedlands, Australia
| | - Mark W Fear
- Burn Injury Research Unit, School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Australia
| |
Collapse
|
23
|
Xue T, Qiu X, Liu H, Gan C, Tan Z, Xie Y, Wang Y, Ye T. Epigenetic regulation in fibrosis progress. Pharmacol Res 2021; 173:105910. [PMID: 34562602 DOI: 10.1016/j.phrs.2021.105910] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 08/23/2021] [Accepted: 09/15/2021] [Indexed: 02/08/2023]
Abstract
Fibrosis, a common process of chronic inflammatory diseases, is defined as a repair response disorder when organs undergo continuous damage, ultimately leading to scar formation and functional failure. Around the world, fibrotic diseases cause high mortality, unfortunately, with limited treatment means in clinical practice. With the development and application of deep sequencing technology, comprehensively exploring the epigenetic mechanism in fibrosis has been allowed. Extensive remodeling of epigenetics controlling various cells phenotype and molecular mechanisms involved in fibrogenesis was subsequently verified. In this review, we summarize the regulatory mechanisms of DNA methylation, histone modification, noncoding RNAs (ncRNAs) and N6-methyladenosine (m6A) modification in organ fibrosis, focusing on heart, liver, lung and kidney. Additionally, we emphasize the diversity of epigenetics in the cellular and molecular mechanisms related to fibrosis. Finally, the potential and prospect of targeted therapy for fibrosis based on epigenetic is discussed.
Collapse
Affiliation(s)
- Taixiong Xue
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xingyu Qiu
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongyao Liu
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Cailing Gan
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zui Tan
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuting Xie
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuxi Wang
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China; Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.
| | - Tinghong Ye
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
24
|
Yoshimura A, Ito M, Mise-Omata S, Ando M. SOCS: negative regulators of cytokine signaling for immune tolerance. Int Immunol 2021; 33:711-716. [PMID: 34415326 DOI: 10.1093/intimm/dxab055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/18/2021] [Indexed: 11/14/2022] Open
Abstract
Cytokines are important intercellular communication tools for immunity. Many cytokines promote gene transcription and proliferation through the JAK/STAT (Janus kinase / signal transducers and activators of transcription) and the Ras/ERK (GDP/GTP-binding rat sarcoma protein / extracellular signal-regulated kinase) pathways, and these signaling pathways are tightly regulated. The SOCS (suppressor of cytokine signaling) family are representative negative regulators of JAK/STAT-mediated cytokine signaling and regulate the differentiation and function of T cells, thus being involved in immune tolerance. Human genetic analysis has shown that SOCS family members are strongly associated with autoimmune diseases, allergy and tumorigenesis. SOCS family proteins also function as immune-checkpoint molecules that contribute to the unresponsiveness of T cells to cytokines.
Collapse
Affiliation(s)
- Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinanomachi, Shinjyuku-ku, Tokyo, Japan
| | - Minako Ito
- Medical Institute of Bioregulation Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan
| | - Setsuko Mise-Omata
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinanomachi, Shinjyuku-ku, Tokyo, Japan
| | - Makoto Ando
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinanomachi, Shinjyuku-ku, Tokyo, Japan
| |
Collapse
|
25
|
Prêle CM, Iosifidis T, McAnulty RJ, Pearce DR, Badrian B, Miles T, Jamieson SE, Ernst M, Thompson PJ, Laurent GJ, Knight DA, Mutsaers SE. Reduced SOCS1 Expression in Lung Fibroblasts from Patients with IPF Is Not Mediated by Promoter Methylation or Mir155. Biomedicines 2021; 9:biomedicines9050498. [PMID: 33946612 PMCID: PMC8147237 DOI: 10.3390/biomedicines9050498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/12/2021] [Accepted: 04/23/2021] [Indexed: 01/16/2023] Open
Abstract
The interleukin (IL)-6 family of cytokines and exaggerated signal transducer and activator of transcription (STAT)3 signaling is implicated in idiopathic pulmonary fibrosis (IPF) pathogenesis, but the mechanisms regulating STAT3 expression and function are unknown. Suppressor of cytokine signaling (SOCS)1 and SOCS3 block STAT3, and low SOCS1 levels have been reported in IPF fibroblasts and shown to facilitate collagen production. Fibroblasts and lung tissue from IPF patients and controls were used to examine the mechanisms underlying SOCS1 down-regulation in IPF. A significant reduction in basal SOCS1 mRNA in IPF fibroblasts was confirmed. However, there was no difference in the kinetics of activation, and methylation of SOCS1 in control and IPF lung fibroblasts was low and unaffected by 5′-aza-2′-deoxycytidine’ treatment. SOCS1 is a target of microRNA-155 and although microRNA-155 levels were increased in IPF tissue, they were reduced in IPF fibroblasts. Therefore, SOCS1 is not regulated by SOCS1 gene methylation or microRNA155 in these cells. In conclusion, we confirmed that IPF fibroblasts had lower levels of SOCS1 mRNA compared with control fibroblasts, but we were unable to determine the mechanism. Furthermore, although SOCS1 may be important in the fibrotic process, we were unable to find a significant role for SOCS1 in regulating fibroblast function.
Collapse
Affiliation(s)
- Cecilia M. Prêle
- Institute for Respiratory Health, Nedland, WA 6009, Australia; (C.M.P.); (T.I.); (B.B.); (T.M.); (P.J.T.); (G.J.L.)
- Centre for Respiratory Health and Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, University of Western Australia, Nedland, WA 6009, Australia
| | - Thomas Iosifidis
- Institute for Respiratory Health, Nedland, WA 6009, Australia; (C.M.P.); (T.I.); (B.B.); (T.M.); (P.J.T.); (G.J.L.)
| | - Robin J. McAnulty
- Centre for Inflammation and Tissue Repair, Rayne Institute, Department of Medicine, University College London, London WC1E 6JJ, UK; (R.J.M.); (D.R.P.)
| | - David R. Pearce
- Centre for Inflammation and Tissue Repair, Rayne Institute, Department of Medicine, University College London, London WC1E 6JJ, UK; (R.J.M.); (D.R.P.)
| | - Bahareh Badrian
- Institute for Respiratory Health, Nedland, WA 6009, Australia; (C.M.P.); (T.I.); (B.B.); (T.M.); (P.J.T.); (G.J.L.)
| | - Tylah Miles
- Institute for Respiratory Health, Nedland, WA 6009, Australia; (C.M.P.); (T.I.); (B.B.); (T.M.); (P.J.T.); (G.J.L.)
| | - Sarra E. Jamieson
- Telethon Kids Institute and Centre for Child Health Research, University of Western Australia, Nedlands, WA 6009, Australia;
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
| | - Philip J. Thompson
- Institute for Respiratory Health, Nedland, WA 6009, Australia; (C.M.P.); (T.I.); (B.B.); (T.M.); (P.J.T.); (G.J.L.)
| | - Geoffrey J. Laurent
- Institute for Respiratory Health, Nedland, WA 6009, Australia; (C.M.P.); (T.I.); (B.B.); (T.M.); (P.J.T.); (G.J.L.)
- Centre for Respiratory Health and Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, University of Western Australia, Nedland, WA 6009, Australia
| | - Darryl A. Knight
- Faculty of Medicine, University of British Columbia (UBC), Vancouver, BC V6Z 1Y5, Canada;
| | - Steven E. Mutsaers
- Institute for Respiratory Health, Nedland, WA 6009, Australia; (C.M.P.); (T.I.); (B.B.); (T.M.); (P.J.T.); (G.J.L.)
- Centre for Respiratory Health and Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, University of Western Australia, Nedland, WA 6009, Australia
- Correspondence: ; Tel.: +61-(0)8-6151-0891; Fax: +61-(0)8-6151-1027
| |
Collapse
|
26
|
Ye W, Siwko S, Tsai RYL. Sex and Race-Related DNA Methylation Changes in Hepatocellular Carcinoma. Int J Mol Sci 2021; 22:ijms22083820. [PMID: 33917049 PMCID: PMC8067720 DOI: 10.3390/ijms22083820] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 12/11/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer and fourth leading cause of cancer-related death worldwide. The number of HCC cases continues to rise despite advances in screening and therapeutic inventions. More importantly, HCC poses two major health disparity issues. First, HCC occurs more commonly in men than women. Second, with the global increase in non-alcoholic fatty liver diseases (NAFLD), it has also become evident that HCC is more prevalent in some races and/or ethnic groups compared to others, depending on its predisposing etiology. Most studies on HCC in the past have been focused on genetic factors as the driving force for HCC development, and the results revealed that genetic mutations associated with HCC are often heterogeneous and involve multiple pathogenic pathways. An emerging new research field is epigenetics, in which gene expression is modified without altering DNA sequences. In this article, we focus on reviewing current knowledge on HCC-related DNA methylation changes that show disparities among different sexes or different racial/ethnic groups, in an effort to establish a point of departure for resolving the broader issue of health disparities in gastrointestinal malignancies using cutting-edge epigenetic approaches.
Collapse
|
27
|
Karki P, Cha B, Zhang CO, Li Y, Ke Y, Promnares K, Kaibuchi K, Yoshimura A, Birukov KG, Birukova AA. Microtubule-dependent mechanism of anti-inflammatory effect of SOCS1 in endothelial dysfunction and lung injury. FASEB J 2021; 35:e21388. [PMID: 33724556 PMCID: PMC10069762 DOI: 10.1096/fj.202001477rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 12/21/2020] [Accepted: 01/07/2021] [Indexed: 12/15/2022]
Abstract
Suppressors of cytokine signaling (SOCS) provide negative regulation of inflammatory reaction. The role and precise cellular mechanisms of SOCS1 in control of endothelial dysfunction and barrier compromise associated with acute lung injury remain unexplored. Our results show that siRNA-mediated SOCS1 knockdown augmented lipopolysaccharide (LPS)-induced pulmonary endothelial cell (EC) permeability and enhanced inflammatory response. Consistent with in vitro data, EC-specific SOCS1 knockout mice developed more severe lung vascular leak and accumulation of inflammatory cells in bronchoalveolar lavage fluid. SOCS1 overexpression exhibited protective effects against LPS-induced endothelial permeability and inflammation, which were dependent on microtubule (MT) integrity. Biochemical and image analysis of unstimulated EC showed SOCS1 association with the MT, while challenge with LPS or MT depolymerizing agent colchicine impaired this association. SOCS1 directly interacted with N2 domains of MT-associated proteins CLIP-170 and CLASP2. Furthermore, N-terminal region of SOCS1 was indispensable for these interactions and SOCS1-ΔN mutant lacking N-terminal 59 amino acids failed to rescue LPS-induced endothelial dysfunction. Depletion of endogenous CLIP-170 or CLASP2 abolished SOCS1 interaction with Toll-like receptor-4 and Janus kinase-2 leading to impairment of SOCS1 inhibitory effects on LPS-induced inflammation. Altogether, these findings suggest that endothelial barrier protective and anti-inflammatory effects of SOCS1 are critically dependent on its targeting to the MT.
Collapse
Affiliation(s)
- Pratap Karki
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Boyoung Cha
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Chen-Ou Zhang
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yue Li
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kamoltip Promnares
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University, Nagoya, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University, Tokyo, Japan
| | - Konstantin G Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Anna A Birukova
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
28
|
YOSHIMURA A, AKI D, ITO M. SOCS, SPRED, and NR4a: Negative regulators of cytokine signaling and transcription in immune tolerance. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2021; 97:277-291. [PMID: 34121041 PMCID: PMC8403526 DOI: 10.2183/pjab.97.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Cytokines are important intercellular communication tools for immunity. Most cytokines utilize the JAK-STAT and Ras-ERK pathways to promote gene transcription and proliferation; however, this signaling is tightly regulated. The suppressor of cytokine signaling (SOCS) family and SPRED family are a representative negative regulators of the JAK-STAT pathway and the Ras-ERK pathway, respectively. The SOCS family regulates the differentiation and function of CD4+ T cells, CD8+ T cells, and regulatory T cells, and is involved in immune tolerance, anergy, and exhaustion. SPRED family proteins have been shown to inactivate Ras by recruiting the Ras-GTPase neurofibromatosis type 1 (NF1) protein. Human genetic analysis has shown that SOCS family members are strongly associated with autoimmune diseases, allergies, and tumorigenesis, and SPRED1 is involved in NF1-like syndromes and tumors. We also identified the NR4a family of nuclear receptors as a key transcription factor for immune tolerance that suppresses cytokine expression and induces various immuno-regulatory molecules including SOCS1.
Collapse
Affiliation(s)
- Akihiko YOSHIMURA
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- Correspondence should be addressed: A. Yoshimura, Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan (e-mail: )
| | - Daisuke AKI
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Minako ITO
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
29
|
Khan MGM, Ghosh A, Variya B, Santharam MA, Ihsan AU, Ramanathan S, Ilangumaran S. Prognostic significance of SOCS1 and SOCS3 tumor suppressors and oncogenic signaling pathway genes in hepatocellular carcinoma. BMC Cancer 2020; 20:774. [PMID: 32807134 PMCID: PMC7433106 DOI: 10.1186/s12885-020-07285-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022] Open
Abstract
Background SOCS1 and SOCS3 genes are considered tumor suppressors in hepatocellular carcinoma (HCC) due to frequent epigenetic repression. Consistent with this notion, mice lacking SOCS1 or SOCS3 show increased susceptibility to diethylnitrosamine (DEN)-induced HCC. As SOCS1 and SOCS3 are important regulators of cytokine and growth factor signaling, their loss could activate oncogenic signaling pathways. Therefore, we examined the correlation between SOCS1/SOCS3 and key oncogenic signaling pathway genes as well as their prognostic significance in HCC. Methods The Cancer Genome Atlas dataset on HCC comprising clinical and transcriptomic data was retrieved from the cBioportal platform. The correlation between the expression of SOCS1 or SOCS3 and oncogenic pathway genes was evaluated using the GraphPad PRISM software. The inversely correlated genes were assessed for their impact on patient survival using the UALCAN platform and their expression quantified in the regenerating livers and DEN-induced HCC tissues of mice lacking Socs1 or Socs3. Finally, the Cox proportional hazards model was used to evaluate the predictive potential of SOCS1 and SOCS3 when combined with the genes of select oncogenic signaling pathways. Results SOCS1 expression was comparable between HCC and adjacent normal tissues, yet higher SOCS1 expression predicted favorable prognosis. In contrast, SOCS3 expression was significantly low in HCC, yet it lacked predictive potential. The correlation between SOCS1 or SOCS3 expression and key genes of the cell cycle, receptor tyrosine kinase, growth factor and MAPK signaling pathways were mostly positive than negative. Among the negatively correlated genes, only a few showed elevated expression in HCC and predicted survival. Many PI3K pathway genes showed mutual exclusivity with SOCS1 and/or SOCS3 and displayed independent predictive ability. Among genes that negatively correlated with SOCS1 and/or SOCS3, only CDK2 and AURKA showed corresponding modulations in the regenerating livers and DEN-induced tumors of hepatocyte-specific Socs1 or Socs3 deficient mice and predicted patient survival. The Cox proportional hazards model identified the combinations of SOCS1 or SOCS3 with CXCL8 and DAB2 as highly predictive. Conclusions SOCS1 expression in HCC has an independent prognostic value whereas SOCS3 expression does not. The predictive potential of SOCS1 expression is increased when combined with other oncogenic signaling pathway genes.
Collapse
Affiliation(s)
- Md Gulam Musawwir Khan
- Immunology graduate program, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Amit Ghosh
- Cell biology graduate program, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Bhavesh Variya
- Cell biology graduate program, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Madanraj Appiya Santharam
- Cell biology graduate program, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Awais Ullah Ihsan
- Cell biology graduate program, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Sheela Ramanathan
- Immunology graduate program, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada.,Cell biology graduate program, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada.,CRCHUS, Sherbrooke, Québec, J1H 5N4, Canada
| | - Subburaj Ilangumaran
- Immunology graduate program, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada. .,Cell biology graduate program, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada. .,CRCHUS, Sherbrooke, Québec, J1H 5N4, Canada.
| |
Collapse
|
30
|
Thaventhiran JED, Lango Allen H, Burren OS, Rae W, Greene D, Staples E, Zhang Z, Farmery JHR, Simeoni I, Rivers E, Maimaris J, Penkett CJ, Stephens J, Deevi SVV, Sanchis-Juan A, Gleadall NS, Thomas MJ, Sargur RB, Gordins P, Baxendale HE, Brown M, Tuijnenburg P, Worth A, Hanson S, Linger RJ, Buckland MS, Rayner-Matthews PJ, Gilmour KC, Samarghitean C, Seneviratne SL, Sansom DM, Lynch AG, Megy K, Ellinghaus E, Ellinghaus D, Jorgensen SF, Karlsen TH, Stirrups KE, Cutler AJ, Kumararatne DS, Chandra A, Edgar JDM, Herwadkar A, Cooper N, Grigoriadou S, Huissoon AP, Goddard S, Jolles S, Schuetz C, Boschann F, Lyons PA, Hurles ME, Savic S, Burns SO, Kuijpers TW, Turro E, Ouwehand WH, Thrasher AJ, Smith KGC. Whole-genome sequencing of a sporadic primary immunodeficiency cohort. Nature 2020; 583:90-95. [PMID: 32499645 PMCID: PMC7334047 DOI: 10.1038/s41586-020-2265-1] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 02/26/2020] [Indexed: 12/19/2022]
Abstract
Primary immunodeficiency (PID) is characterized by recurrent and often life-threatening infections, autoimmunity and cancer, and it poses major diagnostic and therapeutic challenges. Although the most severe forms of PID are identified in early childhood, most patients present in adulthood, typically with no apparent family history and a variable clinical phenotype of widespread immune dysregulation: about 25% of patients have autoimmune disease, allergy is prevalent and up to 10% develop lymphoid malignancies1-3. Consequently, in sporadic (or non-familial) PID genetic diagnosis is difficult and the role of genetics is not well defined. Here we address these challenges by performing whole-genome sequencing in a large PID cohort of 1,318 participants. An analysis of the coding regions of the genome in 886 index cases of PID found that disease-causing mutations in known genes that are implicated in monogenic PID occurred in 10.3% of these patients, and a Bayesian approach (BeviMed4) identified multiple new candidate PID-associated genes, including IVNS1ABP. We also examined the noncoding genome, and found deletions in regulatory regions that contribute to disease causation. In addition, we used a genome-wide association study to identify loci that are associated with PID, and found evidence for the colocalization of-and interplay between-novel high-penetrance monogenic variants and common variants (at the PTPN2 and SOCS1 loci). This begins to explain the contribution of common variants to the variable penetrance and phenotypic complexity that are observed in PID. Thus, using a cohort-based whole-genome-sequencing approach in the diagnosis of PID can increase diagnostic yield and further our understanding of the key pathways that influence immune responsiveness in humans.
Collapse
Affiliation(s)
- James E D Thaventhiran
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK.
- Medical Research Council Toxicology Unit, School of Biological Sciences, University of Cambridge, Cambridge, UK.
| | - Hana Lango Allen
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- Medical Research Council Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Oliver S Burren
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - William Rae
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Daniel Greene
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- Medical Research Council Biostatistics Unit, Cambridge Institute of Public Health, Cambridge Biomedical Campus, Cambridge, UK
| | - Emily Staples
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Zinan Zhang
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology and Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - James H R Farmery
- Medical Research Council Biostatistics Unit, Cambridge Institute of Public Health, Cambridge Biomedical Campus, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Ilenia Simeoni
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Elizabeth Rivers
- UCL Great Ormond Street Institute of Child Health, London, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Jesmeen Maimaris
- UCL Great Ormond Street Institute of Child Health, London, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Christopher J Penkett
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Jonathan Stephens
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Sri V V Deevi
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Alba Sanchis-Juan
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Nicholas S Gleadall
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Moira J Thomas
- Department of Immunology, Queen Elizabeth University Hospital, Glasgow, UK
- Gartnavel General Hospital, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Ravishankar B Sargur
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Pavels Gordins
- East Yorkshire Regional Adult Immunology and Allergy Unit, Hull Royal Infirmary, Hull and East Yorkshire Hospitals NHS Trust, Hull, UK
| | - Helen E Baxendale
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Royal Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - Matthew Brown
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Paul Tuijnenburg
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam University Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Austen Worth
- UCL Great Ormond Street Institute of Child Health, London, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Steven Hanson
- Institute of Immunity and Transplantation, University College London, London, UK
- Department of Immunology, Royal Free London NHS Foundation Trust, London, UK
| | - Rachel J Linger
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Matthew S Buckland
- Institute of Immunity and Transplantation, University College London, London, UK
- Department of Immunology, Royal Free London NHS Foundation Trust, London, UK
| | - Paula J Rayner-Matthews
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Kimberly C Gilmour
- UCL Great Ormond Street Institute of Child Health, London, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Crina Samarghitean
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Suranjith L Seneviratne
- Institute of Immunity and Transplantation, University College London, London, UK
- Department of Immunology, Royal Free London NHS Foundation Trust, London, UK
| | - David M Sansom
- Institute of Immunity and Transplantation, University College London, London, UK
- Department of Immunology, Royal Free London NHS Foundation Trust, London, UK
| | - Andy G Lynch
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- School of Mathematics and Statistics, University of St Andrews, St Andrews, UK
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Karyn Megy
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Eva Ellinghaus
- K.G. Jebsen Inflammation Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo University Hospital, Oslo, Norway
| | - David Ellinghaus
- Department of Transplantation, Institute of Clinical Medicine, University of Oslo, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Silje F Jorgensen
- Section of Clinical Immunology and Infectious Diseases, Department of Rheumatology, Dermatology and Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Oslo, Norway
| | - Tom H Karlsen
- K.G. Jebsen Inflammation Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo University Hospital, Oslo, Norway
| | - Kathleen E Stirrups
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Antony J Cutler
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Dinakantha S Kumararatne
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Department of Clinical Biochemistry and Immunology, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Anita Chandra
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Department of Clinical Biochemistry and Immunology, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - J David M Edgar
- St James's Hospital, Dublin, Ireland
- Trinity College Dublin, Dublin, Ireland
| | | | - Nichola Cooper
- Department of Medicine, Imperial College London, London, UK
| | | | - Aarnoud P Huissoon
- West Midlands Immunodeficiency Centre, University Hospitals Birmingham, Birmingham, UK
- Birmingham Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Sarah Goddard
- University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, UK
| | - Stephen Jolles
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, UK
| | - Catharina Schuetz
- Department of Pediatric Immunology, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Felix Boschann
- Institute of Medical Genetics and Human Genetics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Paul A Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Matthew E Hurles
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Sinisa Savic
- Department of Clinical Immunology and Allergy, St James's University Hospital, Leeds, UK
- The NIHR Leeds Biomedical Research Centre, Leeds, UK
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, UK
| | - Siobhan O Burns
- Institute of Immunity and Transplantation, University College London, London, UK
- Department of Immunology, Royal Free London NHS Foundation Trust, London, UK
| | - Taco W Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam University Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
- Department of Blood Cell Research, Sanquin, Amsterdam, The Netherlands
| | - Ernest Turro
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- Medical Research Council Biostatistics Unit, Cambridge Institute of Public Health, Cambridge Biomedical Campus, Cambridge, UK
| | - Willem H Ouwehand
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Adrian J Thrasher
- UCL Great Ormond Street Institute of Child Health, London, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK.
| |
Collapse
|
31
|
Mafanda EK, Kandhi R, Bobbala D, Khan MGM, Nandi M, Menendez A, Ramanathan S, Ilangumaran S. Essential role of suppressor of cytokine signaling 1 (SOCS1) in hepatocytes and macrophages in the regulation of liver fibrosis. Cytokine 2019; 124:154501. [DOI: 10.1016/j.cyto.2018.07.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 07/26/2018] [Accepted: 07/28/2018] [Indexed: 12/31/2022]
|
32
|
Yamaguchi T, Matsuzaki J, Katsuda T, Saito Y, Saito H, Ochiya T. Generation of functional human hepatocytes in vitro: current status and future prospects. Inflamm Regen 2019; 39:13. [PMID: 31308858 PMCID: PMC6604181 DOI: 10.1186/s41232-019-0102-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 05/22/2019] [Indexed: 12/17/2022] Open
Abstract
Liver and hepatocyte transplantation are the only effective therapies for late-stage liver diseases, in which the liver loses its regenerative capacity. However, there is a shortage of donors. As a potential alternative approach, functional hepatocytes were recently generated from various cell sources. Analysis of drug metabolism in the human liver is important for drug development. Consequently, cells that metabolize drugs similar to human primary hepatocytes are required. This review discusses the current challenges and future perspectives concerning hepatocytes and hepatic progenitor cells that have been reprogrammed from various cell types, focusing on their functions in transplantation models and their ability to metabolize drugs.
Collapse
Affiliation(s)
- Tomoko Yamaguchi
- 1Division of Pharmacotherapeutics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512 Japan.,2Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045 Japan
| | - Juntaro Matsuzaki
- 2Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045 Japan.,3Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Takeshi Katsuda
- 2Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045 Japan
| | - Yoshimasa Saito
- 1Division of Pharmacotherapeutics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512 Japan
| | - Hidetsugu Saito
- 1Division of Pharmacotherapeutics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512 Japan
| | - Takahiro Ochiya
- 2Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045 Japan.,4Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402 Japan
| |
Collapse
|
33
|
Chen XF, Wu J, Zhang YD, Zhang CX, Chen XT, Zhao W, Chen TX. Role of SOCS3 in enhanced acute-phase protein genes by neonatal macrophages in response to IL-6. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2019; 54:206-212. [PMID: 31204209 DOI: 10.1016/j.jmii.2019.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 04/18/2019] [Accepted: 05/15/2019] [Indexed: 01/16/2023]
Abstract
BACKGROUND Interleukin 6 (IL-6) induce the inflammatory response directly related with the morbidity and mortality of neonatal. Here we aimed to explore the mechanism of IL-6 in neonatal inflammatory response by studying the IL-6/STAT3 signaling pathway. METHODS Cord blood samples from health term neonatal and peripheral venous blood from health volunteers were collected. The monocytes of adults and cord blood were isolated and induced into macrophages. Then the macrophages were pretreated with or without MG132 before IL-6 stimulation. Proteins were analyzed by Western blot, mRNA by real time PCR and membrane molecule by flow cytometry. RESULTS The acute phase protein gene expression in neonatal macrophages after stimulated with IL-6 were higher than that in adult. Significantly enhanced phosphorylation of STAT3 was seen in neonatal macrophages. Both mRNA and protein expression of SOCS3 in neonatal macrophages were lower than that in adult. After pretreated with MG132, the expression of SOCS3 protein was increased which lead to attenuate the STAT3 phosphorylation and APP gene expression. CONCLUSION Neonatal exhibit an enhanced expression of downstream target genes and IL-6/STAT3 signal pathway which is related with the diminished SOCS3. This provides a new sight into inflammatory responses in neonatal.
Collapse
Affiliation(s)
- Xia-Fang Chen
- Department of Rheumatology/Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China; Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jing Wu
- Department of Rheumatology/Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China; Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yi-Dan Zhang
- Department of Internal Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Chen-Xing Zhang
- Department of Rheumatology/Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China; Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xu-Ting Chen
- Department of Rheumatology/Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China; Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Wei Zhao
- Division of Allergy and Immunology, Department of Pediatrics, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Tong-Xin Chen
- Department of Rheumatology/Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China; Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
34
|
Khan MGM, Ghosh A, Variya B, Santharam MA, Kandhi R, Ramanathan S, Ilangumaran S. Hepatocyte growth control by SOCS1 and SOCS3. Cytokine 2019; 121:154733. [PMID: 31154249 DOI: 10.1016/j.cyto.2019.154733] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 05/18/2019] [Accepted: 05/21/2019] [Indexed: 02/06/2023]
Abstract
The extraordinary capacity of the liver to regenerate following injury is dependent on coordinated and regulated actions of cytokines and growth factors. Whereas hepatocyte growth factor (HGF) and epidermal growth factor (EGF) are direct mitogens to hepatocytes, inflammatory cytokines such as TNFα and IL-6 also play essential roles in the liver regeneration process. These cytokines and growth factors activate different signaling pathways in a sequential manner to elicit hepatocyte proliferation. The kinetics and magnitude of these hepatocyte-activating stimuli are tightly regulated to ensure restoration of a functional liver mass without causing uncontrolled cell proliferation. Hepatocyte proliferation can become deregulated under conditions of chronic inflammation, leading to accumulation of genetic aberrations and eventual neoplastic transformation. Among the control mechanisms that regulate hepatocyte proliferation, negative feedback inhibition by the 'suppressor of cytokine signaling (SOCS)' family proteins SOCS1 and SOCS3 play crucial roles in attenuating cytokine and growth factor signaling. Loss of SOCS1 or SOCS3 in the mouse liver increases the rate of liver regeneration and renders hepatocytes susceptible to neoplastic transformation. The frequent epigenetic repression of the SOCS1 and SOCS3 genes in hepatocellular carcinoma has stimulated research in understanding the growth regulatory mechanisms of SOCS1 and SOCS3 in hepatocytes. Whereas SOCS3 is implicated in regulating JAK-STAT signaling induced by IL-6 and attenuating EGFR signaling, SOCS1 is crucial for the regulation of HGF signaling. These two proteins also module the functions of certain key proteins that control the cell cycle. In this review, we discuss the current understanding of the functions of SOCS1 and SOCS3 in controlling hepatocyte proliferation, and its implications to liver health and disease.
Collapse
Affiliation(s)
- Md Gulam Musawwir Khan
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke J1H 5N4, Québec, Canada
| | - Amit Ghosh
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke J1H 5N4, Québec, Canada
| | - Bhavesh Variya
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke J1H 5N4, Québec, Canada
| | - Madanraj Appiya Santharam
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke J1H 5N4, Québec, Canada
| | - Rajani Kandhi
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke J1H 5N4, Québec, Canada
| | - Sheela Ramanathan
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke J1H 5N4, Québec, Canada
| | - Subburaj Ilangumaran
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke J1H 5N4, Québec, Canada.
| |
Collapse
|
35
|
Reactive Oxygen Species Drive Epigenetic Changes in Radiation-Induced Fibrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4278658. [PMID: 30881591 PMCID: PMC6381575 DOI: 10.1155/2019/4278658] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/06/2018] [Accepted: 12/12/2018] [Indexed: 12/14/2022]
Abstract
Radiation-induced fibrosis (RIF) develops months to years after initial radiation exposure. RIF occurs when normal fibroblasts differentiate into myofibroblasts and lay down aberrant amounts of extracellular matrix proteins. One of the main drivers for developing RIF is reactive oxygen species (ROS) generated immediately after radiation exposure. Generation of ROS is known to induce epigenetic changes and cause differentiation of fibroblasts to myofibroblasts. Several antioxidant compounds have been shown to prevent radiation-induced epigenetic changes and the development of RIF. Therefore, reviewing the ROS-linked epigenetic changes in irradiated fibroblast cells is essential to understand the development and prevention of RIF.
Collapse
|
36
|
High Risk of Hepatocellular Carcinoma Development in Fibrotic Liver: Role of the Hippo-YAP/TAZ Signaling Pathway. Int J Mol Sci 2019; 20:ijms20030581. [PMID: 30700007 PMCID: PMC6387126 DOI: 10.3390/ijms20030581] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 01/24/2019] [Accepted: 01/28/2019] [Indexed: 02/07/2023] Open
Abstract
Liver cancer is the fourth leading cause of cancer-related death globally, accounting for approximately 800,000 deaths annually. Hepatocellular carcinoma (HCC) is the most common type of liver cancer, making up about 80% of cases. Liver fibrosis and its end-stage disease, cirrhosis, are major risk factors for HCC. A fibrotic liver typically shows persistent hepatocyte death and compensatory regeneration, chronic inflammation, and an increase in reactive oxygen species, which collaboratively create a tumor-promoting microenvironment via inducing genetic alterations and chromosomal instability, and activating various oncogenic molecular signaling pathways. In this article, we review recent advances in fields of liver fibrosis and carcinogenesis, and consider several molecular signaling pathways that promote hepato-carcinogenesis under the microenvironment of liver fibrosis. In particular, we pay attention to emerging roles of the Hippo-YAP/TAZ signaling pathway in stromal activation, hepatic fibrosis, and liver cancer.
Collapse
|
37
|
Abstract
Interferon-gamma (IFNG) has long been implicated as a central orchestrator of antitumor immune responses in the elimination stage of the immunoediting paradigm. However, mounting evidence suggests that IFNG may also have important and significant protumor roles to play in the equilibrium and escape phases through its regulatory effects on immunoevasive functions that promote tumorigenesis. These seemingly contradictory effects of IFNG undoubtedly play profound roles in not only the activation of inflammatory response to cancer but also in the determination of its outcome. In the face of the recent explosion of anticancer immunotherapeutic strategies in the clinic, it is critical that a complete understanding is achieved of the underpinnings of the mechanisms that determine the two faces of IFNG signaling in cancer. Here, the current state of this dichotomy is reviewed.
Collapse
Affiliation(s)
- M Raza Zaidi
- Fels Institute for Cancer Research & Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
38
|
Jamhiri I, Shahin K, Khodabandeh Z, Kalantar K, Sarvari J, Atapour A, Mina F, Ahmadnejad A, Hosseini SY. Recombinant NS3 Protein Induced Expression of Immune Modulatory Elements in Hepatic Stellate Cells During Its Fibrotic Activity. Viral Immunol 2018; 31:575-582. [PMID: 30281404 DOI: 10.1089/vim.2018.0018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
There is a growing body of studies that show the important role of NS3 protein from hepatitis C virus in fibrosis. However, mechanisms of the effects of this protein on immune modulation of stellate cells remain to be investigated. Herein, the effect of NS3 protein on the expression level of suppressor of cytokine signaling (SOCS)1/3 and interleukin-24 (IL-24)-related genes was investigated in hepatic stellate cell (HSC), LX-2. Recombinant NS3 protein was added to LX-2 HSC culture. Leptin and standard medium treatments were also included in experiments as positive and negative controls, respectively. Total RNA was extracted from each well at 6, 12, and 24 h after NS3 addition. The expression levels of the fibrotic (transforming growth factor beta 1 [TGF-β], alpha-smooth muscle actin [α-SMA], and COL1A1), inflammatory (IL-6 and IL-24), IL-20R, IL-22R, and immunosuppressive genes (SOCS1 and SOCS3) were evaluated by real-time polymerase chain reaction (PCR). Recombinant NS3 protein induced activated phenotypes of LX-2 with a significant increase in the expression level of α-SMA COL1A1 (p < 0.0001) and TGF-β. Moreover, this exposure led to a meaningful elevation in the expression of IL-6. Furthermore, compared with leptin (control), after the stellate cell treatment with NS3, SOCS1 and SOCS3 gene expression induced at a comparable level. Compared with the control sample, the NS3 protein significantly increased the expression level of IL-24 and its related receptors, IL-20R and IL-22R. This study not only confirmed the previously proved inflammatory and fibrotic effect of this protein but also indicated that high expression levels of SOCS1, SOCS3, and IL-24 have a significant effect on HSC activation. Therefore, these two molecules can be used as a potential therapeutic target candidate.
Collapse
Affiliation(s)
- Iman Jamhiri
- 1 Stem Cell Technology Research Center, Shiraz University of Medical Sciences , Shiraz, Iran
| | - Khashayar Shahin
- 2 Department of Biology, Faculty of Sciences, University of Isfahan , Isfahan, Iran
| | - Zahra Khodabandeh
- 1 Stem Cell Technology Research Center, Shiraz University of Medical Sciences , Shiraz, Iran
| | - Kurosh Kalantar
- 3 Department of Immunology, School of Medicine, Shiraz University of Medical Sciences , Shiraz, Iran
| | - Jamal Sarvari
- 4 Gastroenterohepatology Research Center, Shiraz University of Medical Sciences , Shiraz, Iran .,5 Department of Bacteriology and Virology, Shiraz University of Medical Sciences , Shiraz, Iran
| | - Amir Atapour
- 6 Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences , Shiraz, Iran
| | - Fatemeh Mina
- 5 Department of Bacteriology and Virology, Shiraz University of Medical Sciences , Shiraz, Iran
| | - Asma Ahmadnejad
- 5 Department of Bacteriology and Virology, Shiraz University of Medical Sciences , Shiraz, Iran
| | - Seyed Younes Hosseini
- 1 Stem Cell Technology Research Center, Shiraz University of Medical Sciences , Shiraz, Iran .,5 Department of Bacteriology and Virology, Shiraz University of Medical Sciences , Shiraz, Iran
| |
Collapse
|
39
|
Yoshimura A, Ito M, Chikuma S, Akanuma T, Nakatsukasa H. Negative Regulation of Cytokine Signaling in Immunity. Cold Spring Harb Perspect Biol 2018; 10:a028571. [PMID: 28716890 PMCID: PMC6028070 DOI: 10.1101/cshperspect.a028571] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cytokines are key modulators of immunity. Most cytokines use the Janus kinase and signal transducers and activators of transcription (JAK-STAT) pathway to promote gene transcriptional regulation, but their signals must be attenuated by multiple mechanisms. These include the suppressors of cytokine signaling (SOCS) family of proteins, which represent a main negative regulation mechanism for the JAK-STAT pathway. Cytokine-inducible Src homology 2 (SH2)-containing protein (CIS), SOCS1, and SOCS3 proteins regulate cytokine signals that control the polarization of CD4+ T cells and the maturation of CD8+ T cells. SOCS proteins also regulate innate immune cells and are involved in tumorigenesis. This review summarizes recent progress on CIS, SOCS1, and SOCS3 in T cells and tumor immunity.
Collapse
Affiliation(s)
- Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Minako Ito
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shunsuke Chikuma
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takashi Akanuma
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hiroko Nakatsukasa
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
40
|
Teng C, Chang H, Tsai H, Hsieh W, Kuo Y, Su I, Lin Y. Liver regeneration accelerates hepatitis B virus-related tumorigenesis of hepatocellular carcinoma. Mol Oncol 2018; 12:1175-1187. [PMID: 29729074 PMCID: PMC6026873 DOI: 10.1002/1878-0261.12318] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/16/2018] [Accepted: 04/23/2018] [Indexed: 12/15/2022] Open
Abstract
Although partial hepatectomy (PH) to remove tumors provides a potential cure of hepatocellular carcinoma (HCC), long-term survival of hepatitis B virus (HBV)-related HCC patients after PH remains a big challenge. Early recurrence within 2 years post-PH is associated with the dissemination of primary HCC. However, late recurrence after 2 years post-PH is supposed due to the de novo or a secondary tumor. Since PH initiates liver regeneration (LR), we hypothesize that LR may accelerate tumorigenesis through activation of pre-existing precancerous lesions in the remaining liver. In this study, we explored the potential role of several LR-related factors in the de novo recurrence in a HBV X protein (HBx) transgenic mouse model receiving PH to mimic human HCC development. Following PH, we observed that tumor development was significantly accelerated from 16.9 to 10.4 months in HBx transgenic mice. The expression of suppressor of cytokine signaling (SOCS) family proteins was remarkably suppressed in livers of HBx transgenic relative to non-transgenic mice from early to late stages after PH as compared with non-PH mice. The expression of transforming growth factor-β (TGF-β)/Smad pathway, hepatocyte growth factor (HGF), Myc, signal transducer and activator of transcription 3 (STAT3), and β-Catenin also showed a significant difference between livers of HBx transgenic and non-transgenic mice at variable time points after PH in comparison with non-PH mice. Taken together, our results provide an explanation for the high de novo recurrence of HBV-related HCC after PH, probably through induction of the sequential changes of LR-related SOCS family proteins, growth factors, and transcription factors, which may promote growth on the precancerous remnant liver.
Collapse
Affiliation(s)
- Chiao‐Fang Teng
- Graduate Institute of Biomedical SciencesChina Medical UniversityTaichungTaiwan
- Organ Transplantation CenterChina Medical University HospitalTaichungTaiwan
| | - Hong‐Yi Chang
- Department of BiotechnologySouthern Taiwan University of Science and TechnologyTainanTaiwan
| | - Hung‐Wen Tsai
- Department of PathologyNational Cheng Kung University HospitalTainanTaiwan
- Institute of Clinical MedicineCollege of MedicineNational Cheng Kung UniversityTainanTaiwan
| | - Wen‐Chuan Hsieh
- National Institute of Infectious Diseases and VaccinologyNational Health Research InstitutesTainanTaiwan
| | - Yu‐Hao Kuo
- Organ Transplantation CenterChina Medical University HospitalTaichungTaiwan
| | - Ih‐Jen Su
- Department of BiotechnologySouthern Taiwan University of Science and TechnologyTainanTaiwan
- Department of PathologyNational Cheng Kung University HospitalTainanTaiwan
- National Institute of Infectious Diseases and VaccinologyNational Health Research InstitutesTainanTaiwan
| | - Yih‐Jyh Lin
- Division of General and Transplant SurgeryDepartment of SurgeryNational Cheng Kung University HospitalTainanTaiwan
- Department of SurgeryCollege of MedicineNational Cheng Kung UniversityTainanTaiwan
- Liver Cancer Collaborative Oncology GroupNational Cheng Kung University HospitalTainanTaiwan
| |
Collapse
|
41
|
Kot M, Daujat-Chavanieu M. Altered cytokine profile under control of the serotonergic system determines the regulation of CYP2C11 and CYP3A isoforms. Food Chem Toxicol 2018; 116:369-378. [PMID: 29698782 DOI: 10.1016/j.fct.2018.04.051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/12/2018] [Accepted: 04/21/2018] [Indexed: 01/16/2023]
Abstract
The aim of this study is to assess a potential mechanism by which the serotonergic system can control the expression and activity of cytochrome (CYP) 2C11 and CYP3A isoforms during liver insufficiency. A rat model of diethylnitrosamine (DEN)-induced liver insufficiency was developed by administering 50 mg/kg of DEN twice a week for 7 weeks. Dysfunction of the serotonergic system was evoked by feeding the rats with a tryptophan-free diet for three weeks. Dysfunction of the serotonergic system during liver insufficiency decreased the level of proinflammatory cytokines (TGF-β and IL-1β) and increased the level of an anti-inflammatory cytokine (IL-4). Simultaneously, activation of the repressive mechanism IL-4/JAK1/STAT6/SOCS1 of the JAK2/STAT5b-mediated signal transduction pathway and the pERK1/2/GR/STAT6 signal transduction pathway resulted in the suppression of the CYP2C11 and CYP3A isoforms. Moreover, dysfunction of the serotonergic system during liver insufficiency equalized the level of testosterone to the basal level, did not change the steady state of the corticosterone level and significantly enhanced the reduced level of growth hormone. An altered cytokine profile under control of the serotonergic system determines the regulation of CYP2C11 and CYP3A isoforms during liver insufficiency through mechanisms based on posttranscriptional and posttranslational processes.
Collapse
Affiliation(s)
- Marta Kot
- Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Smętna Street 12, Poland.
| | - Martine Daujat-Chavanieu
- Institute for Regenerative Medicine and Biotherapy, Univ Montpellier, Inserm U1183, CHU Montpellier, Montpellier, France
| |
Collapse
|
42
|
Alterations in the p53-SOCS2 axis contribute to tumor growth in colon cancer. Exp Mol Med 2018; 50:1-10. [PMID: 29622769 PMCID: PMC5940812 DOI: 10.1038/s12276-017-0001-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 10/07/2017] [Accepted: 10/11/2017] [Indexed: 12/28/2022] Open
Abstract
Altered expression of suppressor of cytokine signaling (SOCS) is found in various tumors. However, regulation of SOCS2 by upstream molecules has yet to be clearly elucidated, particularly in tumor cells. SCOCS2 expression was examined in tumor cells transfected with an inducible p53 expression system. The impact of SOCS2 on cell proliferation was measured with in vitro assays. Inhibition of tumorigenicity by SOCS2 knockdown was assessed via a mouse model. Expression profiles were compared and genes differentially expressed were identified using four types of p53-null cells (Saos, HLK3, PC3, and H1299) and the same cells stably expressing p53. Twelve kinds of target genes were simultaneously upregulated or downregulated by p53 in three or more sets of p53-null cells. SOCS2 expression was reciprocally inhibited by inducible p53 expression in p53-null cells, even colon cancer cells. SOCS2 promoter activity was inhibited by wild type but not mutant p53. SOCS2 knockdown inhibited tumor growth in vitro and in an animal xenograph model. SOCS2 overexpression was detected in a murine model of azoxymethane/dextran sulfate sodium-induced colitis-associated colon cancer compared to mock-treated controls. SOCS2 expression was heterogeneously upregulated in some human colon cancers. Thus, SOCS2 was upregulated by p53 dysfunction and seemed to be associated with the tumorigenic potential of colon cancer. Insights into a signaling protein’s role in cell growth could inform new therapeutic strategies for treating colon cancer. SOCS-2 acts as an ‘off switch’ for cell signaling pathways. It has been identified as possibly protective against many cancers, although some cancers are associated with elevated SOCS-2 levels. Researchers led by Daeghon Kim at Chonbuk National University Hospital in South Korea have now shown that the effects of SOCS-2 are apparently dependent on how much of it is present. Moderate levels of SOCS-2 can suppress growth in colon cancer cells, but Kim’s team showed that excessive SOCS-2 has the opposite effect, promoting proliferation. The researchers also identified a gene commonly mutated in cancer cells that can drive overproduction of SOCS-2. Drugs that inhibit SOCS-2 or block its production may therefore offer useful treatments for colorectal cancer.
Collapse
|
43
|
Cordero-Espinoza L, Huch M. The balancing act of the liver: tissue regeneration versus fibrosis. J Clin Invest 2018; 128:85-96. [PMID: 29293095 DOI: 10.1172/jci93562] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Epithelial cell loss alters a tissue's optimal function and awakens evolutionarily adapted healing mechanisms to reestablish homeostasis. Although adult mammalian organs have a limited regeneration potential, the liver stands out as one remarkable exception. Following injury, the liver mounts a dynamic multicellular response wherein stromal cells are activated in situ and/or recruited from the bloodstream, the extracellular matrix (ECM) is remodeled, and epithelial cells expand to replenish their lost numbers. Chronic damage makes this response persistent instead of transient, tipping the system into an abnormal steady state known as fibrosis, in which ECM accumulates excessively and tissue function degenerates. Here we explore the cellular and molecular switches that balance hepatic regeneration and fibrosis, with a focus on uncovering avenues of disease modeling and therapeutic intervention.
Collapse
|
44
|
Mojic M, Takeda K, Hayakawa Y. The Dark Side of IFN-γ: Its Role in Promoting Cancer Immunoevasion. Int J Mol Sci 2017; 19:E89. [PMID: 29283429 PMCID: PMC5796039 DOI: 10.3390/ijms19010089] [Citation(s) in RCA: 218] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 12/26/2017] [Accepted: 12/26/2017] [Indexed: 02/07/2023] Open
Abstract
Interferon-γ (IFN-γ) is a pleiotropic cytokine that has long been praised as an important effector molecule of anti-tumor immunity, capable of suppressing tumor growth through various mechanisms. On the contrary to such a bright side of IFN-γ, it has also been involved in promoting an outgrowth of tumor cells with immunoevasive phenotype suggesting an existence of a dark "tumor-promoting" side effect of IFN-γ. In this review, we will summarize this multi-functional role of IFN-γ in tumor context, how it promotes changes in tumor phenotype towards increased fitness for growth in immunocompetent host. Furthermore, we summarize how IFN-γ is involved in homeostatic or cancer-triggered mechanisms to establish an immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Marija Mojic
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama, Sugitani 2630, Toyama 930-0194, Japan.
| | - Kazuyoshi Takeda
- Division of Cell Biology, Biomedical Research Center, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo 113-8421, Japan.
- Department of Biofunctional Microbiota, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Yoshihiro Hayakawa
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama, Sugitani 2630, Toyama 930-0194, Japan.
| |
Collapse
|
45
|
Planello AC, Singhania R, Kron KJ, Bailey SD, Roulois D, Lupien M, Line SRP, de Souza AP, De Carvalho DD. Pre-neoplastic epigenetic disruption of transcriptional enhancers in chronic inflammation. Oncotarget 2017; 7:15772-86. [PMID: 26908456 PMCID: PMC4941276 DOI: 10.18632/oncotarget.7513] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 02/09/2016] [Indexed: 02/06/2023] Open
Abstract
Chronic periodontitis (CP) is a chronic inflammatory disease independently associated with higher incidence of oral cavity squamous cell carcinoma (OSCC). However, the molecular mechanism responsible for this increased incidence is unknown. Here we profiled the DNA methylome of CP patients and healthy controls and compared to a large set of OSCC samples from TCGA. We observed a significant overlap between the altered DNA methylation patterns in CP and in OSCC, suggesting an emergence of a pre-neoplastic epigenome in CP. Remarkably, the hypermethylated CpGs in CP were significantly enriched for enhancer elements. This aberrant enhancer methylation is functional and able to disrupt enhancer activity by preventing the binding of chromatin looping factors. This study provides new insights on the molecular mechanisms linking chronic inflammation and tumor predisposition, highlighting the role of epigenetic disruption of transcriptional enhancers.
Collapse
Affiliation(s)
- Aline C Planello
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Department of Morphology, Piracicaba Dental School, University of Campinas, Piracicaba, SP, Brazil
| | - Rajat Singhania
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ken J Kron
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Swneke D Bailey
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - David Roulois
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Sérgio R Peres Line
- Department of Morphology, Piracicaba Dental School, University of Campinas, Piracicaba, SP, Brazil
| | - Ana Paula de Souza
- Department of Morphology, Piracicaba Dental School, University of Campinas, Piracicaba, SP, Brazil
| | - Daniel D De Carvalho
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
46
|
Wang H, Wang J, Xia Y. Defective Suppressor of Cytokine Signaling 1 Signaling Contributes to the Pathogenesis of Systemic Lupus Erythematosus. Front Immunol 2017; 8:1292. [PMID: 29085365 PMCID: PMC5650678 DOI: 10.3389/fimmu.2017.01292] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 09/26/2017] [Indexed: 12/19/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease involving injuries in multiple organs and systems. Exaggerated inflammatory responses are characterized as end-organ damage in patients with SLE. Although the explicit pathogenesis of SLE remains unclear, increasing evidence suggests that dysregulation of cytokine signals contributes to the progression of SLE through the Janus kinase/signal transducer and activator of transcription (STAT) signaling pathway. Activated STAT proteins translocate to the cell nucleus and induce transcription of target genes, which regulate downstream cytokine production and inflammatory cell infiltration. The suppressor of cytokine signaling 1 (SOCS1) is considered as a classical inhibitor of cytokine signaling. Recent studies have demonstrated that SOCS1 expression is decreased in patients with SLE and in murine lupus models, and this negatively correlates with the magnitude of inflammation. Dysregulation of SOCS1 signals participates in various pathological processes of SLE such as hematologic abnormalities and autoantibody generation. Lupus nephritis is one of the most serious complications of SLE, and it correlates with suppressed SOCS1 signals in renal tissues. Moreover, SOCS1 insufficiency affects the function of several other organs, including skin, central nervous system, liver, and lungs. Therefore, SOCS1 aberrancy contributes to the development of both systemic and local inflammation in SLE patients. In this review, we discuss recent studies regarding the roles of SOCS1 in the pathogenesis of SLE and its therapeutic implications.
Collapse
Affiliation(s)
- Huixia Wang
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Jiaxing Wang
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
47
|
Ge J, Huang Z, Liu H, Chen J, Xie Z, Chen Z, Peng J, Sun J, Hou J, Zhang X. Lower Expression of MicroRNA-155 Contributes to Dysfunction of Natural Killer Cells in Patients with Chronic Hepatitis B. Front Immunol 2017; 8:1173. [PMID: 29018442 PMCID: PMC5614978 DOI: 10.3389/fimmu.2017.01173] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 09/05/2017] [Indexed: 12/23/2022] Open
Abstract
MicroRNAs have been reported to be regulated in different ways in a variety of liver diseases. As a key modulator of cellular function in both innate and adaptive immunity, the role of miR-155 in chronic hepatitis B virus infection remains largely unknown. Here, we investigated the expression and function of miR-155 in chronic hepatitis B (CHB) patients. It was found that miR-155 expression in peripheral blood mononuclear cells (PBMCs) was lower in CHB patients than healthy controls (HC). Among CHB infection, immune-active (IA) patients with abnormal alanine aminotransferase (ALT) levels had relatively higher miR-155 expression in PBMCs and serum than immune-tolerant carriers, but were comparable to inactive carriers. Moreover, there was a positive correlation between miR-155 expression and ALT levels in CHB patients. Particularly, miR-155 expression in natural killer (NK) cells was significantly downregulated in IA patients compared with HC. Inversely, suppressor of cytokine signaling 1 (SOCS1), a target of miR-155, was upregulated in NK cells of IA patients. Overexpression of miR-155 in NK cells from IA patients led to a decrease in SOCS1 expression and an increase of IFN-γ production. Finally, accompanied by the normalization of ALT, miR-155 expression in PBMCs gradually decreased during telbivudine or peg-IFN-α-2a therapy. Interestingly, higher miR-155 expression at baseline was associated with better response to telbivudine therapy, but not peg-IFN-α-2a. In conclusion, our data suggested that miR-155 downregulation in NK cells of IA patients impaired IFN-γ production by targeting SOCS1, which may contribute to immune dysfunction during CHB infection. Additionally, baseline miR-155 expression could predict the treatment response to telbivudine therapy.
Collapse
Affiliation(s)
- Jun Ge
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zuxiong Huang
- Department of Hepatology, Mengchao Haptobiliary Hospital of Fujian Medical University, Fuzhou, China.,Department of Hepatology, Affiliated Infectious Disease Hospital of Fujian Medical University, Fuzhou, China
| | - Hongyan Liu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiehua Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhanglian Xie
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zide Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jie Peng
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Sun
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinlin Hou
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoyong Zhang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
48
|
Piñeros Alvarez AR, Glosson-Byers N, Brandt S, Wang S, Wong H, Sturgeon S, McCarthy BP, Territo PR, Alves-Filho JC, Serezani CH. SOCS1 is a negative regulator of metabolic reprogramming during sepsis. JCI Insight 2017; 2:92530. [PMID: 28679957 DOI: 10.1172/jci.insight.92530] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 05/25/2017] [Indexed: 01/01/2023] Open
Abstract
Sepsis can induce an overwhelming systemic inflammatory response, resulting in organ damage and death. Suppressor of cytokine signaling 1 (SOCS1) negatively regulates signaling by cytokine receptors and Toll-like receptors (TLRs). However, the cellular targets and molecular mechanisms for SOCS1 activity during polymicrobial sepsis are unknown. To address this, we utilized a cecal ligation and puncture (CLP) model for sepsis; C57BL/6 mice subjected to CLP were then treated with a peptide (iKIR) that binds the SOCS1 kinase inhibitory region (KIR) and blocks its activity. Treatment with iKIR increased CLP-induced mortality, bacterial burden, and inflammatory cytokine production. Myeloid cell-specific SOCS1 deletion (Socs1Δmyel) mice were also more susceptible to sepsis, demonstrating increased mortality, higher bacterial loads, and elevated inflammatory cytokines, compared with Socs1fl littermate controls. These effects were accompanied by macrophage metabolic reprograming, as evidenced by increased lactic acid production and elevated expression of the glycolytic enzymes hexokinase, lactate dehydrogenase A, and glucose transporter 1 in septic Socs1Δmyel mice. Upregulation was dependent on the STAT3/HIF-1α/glycolysis axis, and blocking glycolysis ameliorated increased susceptibility to sepsis in iKIR-treated CLP mice. These results reveal a role of SOCS1 as a regulator of metabolic reprograming that prevents overwhelming inflammatory response and organ damage during sepsis.
Collapse
Affiliation(s)
- Annie Rocio Piñeros Alvarez
- Department Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Nicole Glosson-Byers
- Department Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana.,Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Stephanie Brandt
- Department Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana.,Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Soujuan Wang
- Department Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana.,Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Hector Wong
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center and Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio, USA
| | - Sarah Sturgeon
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Brian Paul McCarthy
- Center for In Vivo Imaging, Department Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Paul R Territo
- Center for In Vivo Imaging, Department Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jose Carlos Alves-Filho
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - C Henrique Serezani
- Department Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana.,Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
49
|
Ilangumaran S, Bobbala D, Ramanathan S. SOCS1: Regulator of T Cells in Autoimmunity and Cancer. Curr Top Microbiol Immunol 2017; 410:159-189. [PMID: 28900678 DOI: 10.1007/82_2017_63] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
SOCS1 is a negative feedback regulator of cytokine and growth factor receptor signaling, and plays an indispensable role in attenuating interferon gamma signaling. Studies on SOCS1-deficient mice have established a crucial role for SOCS1 in regulating CD8+ T cell homeostasis. In the thymus, SOCS1 prevents thymocytes that had failed positive selection from surviving and expanding, ensures negative selection and prevents inappropriate developmental skewing toward the CD8 lineage. In the periphery, SOCS1 not only controls production of T cell stimulatory cytokines but also attenuates the sensitivity of CD8+ T cells to synergistic cytokine stimulation and antigen non-specific activation. As cytokine stimulation of CD8+ T lymphocytes increases their sensitivity to low affinity TCR ligands, SOCS1 likely contributes to peripheral T cell tolerance by putting brakes on aberrant T cell activation driven by inflammatory cytokines. In addition, SOCS1 is critical to maintain the stability of T regulatory cells and control their plasticity to become pathogenic Th17 and Th1 cells under the harmful influence of inflammatory cytokines. SOCS1 also regulates T cell activation by dendritic cells via modulating their generation, maturation, antigen presentation, costimulatory signaling, and cytokine production. The above control mechanisms of SOCS1 on T cells, T regulatory cells and dendritic cells collectively contribute to immunological tolerance and prevent autoimmune manifestation. On other hand, silencing SOCS1 in dendritic cells or CD8+ T cells stimulates efficient antitumor immunity. Thus, even though SOCS1 is not a cell surface checkpoint inhibitor, its regulatory functions on T cell responses qualify SOCS1as a "non-classical" checkpoint blocker. SOCS1 also functions as a tumor suppressor in cancer cells by regulating oncogenic signal transduction pathways. The loss of SOCS1 expression observed in many tumors may have an impact on classical checkpoint pathways. The potential to exploit SOCS1 to treat inflammatory/autoimmune diseases and elicit antitumor immunity is discussed.
Collapse
Affiliation(s)
- Subburaj Ilangumaran
- Immunology Division, Faculty of Medicine and Health Sciences, Department of Pediatrics, Université de Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada.
| | - Diwakar Bobbala
- Immunology Division, Faculty of Medicine and Health Sciences, Department of Pediatrics, Université de Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Sheela Ramanathan
- Immunology Division, Faculty of Medicine and Health Sciences, Department of Pediatrics, Université de Sherbrooke, 3001 North 12th avenue, Sherbrooke, QC, J1H 5N4, Canada
| |
Collapse
|
50
|
Bian F, Jiang H, Man M, Mai K, Zhou H, Xu W, He G. Dietary gossypol suppressed postprandial TOR signaling and elevated ER stress pathways in turbot (Scophthalmus maximus L.). Am J Physiol Endocrinol Metab 2017; 312:E37-E47. [PMID: 27894064 DOI: 10.1152/ajpendo.00285.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 01/17/2023]
Abstract
Gossypol is known to be a polyphenolic compound toxic to animals. However, its molecular targets are far from fully characterized. To evaluate the physiological and molecular effects of gossypol, we chose turbot (Scophthalmus maximus L.), a carnivorous fish, as our model species. Juvenile turbots (7.83 ± 0.02 g) were fed diets containing gradient levels of gossypol at 0 (G0), 600 (G1), and 1,200 (G2) mg/kg diets for 11 wk. After the feeding trial, fish growth, body protein, and fat contents were significantly reduced in the G2 group compared with those of the G0 group (P < 0.05). Gossypol had little impact on digestive enzyme activities and intestine morphology. However, gossypol caused liver fibrosis and stimulated chemokine and proinflammatory cytokine secretions. More importantly, gossypol suppressed target of rapamycin (TOR) signaling and induced endoplasmic reticulum (ER) stress pathway in both the feeding experiment and cell cultures. Our results demonstrated that gossypol inhibited TOR signaling and elevated ER stress pathways both in vivo and in vitro, thus providing new mechanism of action of gossypol in nutritional physiology.
Collapse
Affiliation(s)
- Fuyun Bian
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, Ocean University of China, Qingdao, China
| | - Haowen Jiang
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, Ocean University of China, Qingdao, China
| | - Mingsan Man
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, Ocean University of China, Qingdao, China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, Ocean University of China, Qingdao, China
| | - Huihui Zhou
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, Ocean University of China, Qingdao, China
| | - Wei Xu
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, Ocean University of China, Qingdao, China
| | - Gen He
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, Ocean University of China, Qingdao, China
| |
Collapse
|