101
|
Cho K, Kim S, Choi SH. Suppressor of cytokine signaling 2 is induced in Huntington's disease and involved in autophagy. Biochem Biophys Res Commun 2021; 559:21-27. [PMID: 33933990 DOI: 10.1016/j.bbrc.2021.04.089] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 04/20/2021] [Indexed: 11/15/2022]
Abstract
Suppressor of cytokine signaling (SOCS) proteins are primarily feedback inhibitors of cytokine signaling. The two conserved domains of SOCS proteins have distinct functions. Src homology 2 (SH2) domain inhibits cytokine receptor, while SOCS box acts as an E3 ubiquitin ligase. SOCS2, a cytokine signaling suppressor, has been primarily implicated in regulating inflammatory conditions in neuronal diseases. However, SOCS proteins have been suggested to play diverse roles in healthy and diseased nervous system including neurodegenerative disorders. In this study, SOCS2 was found to be upregulated in Huntington's disease and was substantially induced in extended polyglutamine (polyQ)-expressing striatal cells. The induced level was augmented under aging conditions. In extended polyQ-expressing cells, downregulated SOCS2 improved autophagic dysfunction rather than altered inflammatory conditions. Overall, we suggest that SOCS2 involves in regulating autophagy by functioning as an E3 ligase in extended polyQ conditions, and consequently regulates cell damage and cell death type.
Collapse
Affiliation(s)
- KyoungJoo Cho
- Department of Life Science, Kyonggi University, Suwon, South Korea.
| | - Sejeong Kim
- College of Korean Medicine, Sangji University, Wonju, South Korea; Department of Cognitive Science, Yonsei University, Seoul, South Korea
| | - Seung Ho Choi
- Department of Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea; Samsung Biomedical Research Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul, South Korea
| |
Collapse
|
102
|
Sugawara S, El-Diwany R, Cohen LK, Rousseau KE, Williams CYK, Veenhuis RT, Mehta SH, Blankson JN, Thomas DL, Cox AL, Balagopal A. People with HIV-1 demonstrate type 1 interferon refractoriness associated with upregulated USP18. J Virol 2021; 95:JVI.01777-20. [PMID: 33658340 PMCID: PMC8139647 DOI: 10.1128/jvi.01777-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 02/19/2021] [Indexed: 01/04/2023] Open
Abstract
HIV-1 infection persists in humans despite expression of antiviral type 1 interferons (IFN). Even exogenous administration of IFNα only marginally reduces HIV-1 abundance, raising the hypothesis that people living with HIV-1 (PLWH) are refractory to type 1 IFN. We demonstrated type 1 IFN refractoriness in CD4+ and CD8+ T cells isolated from HIV-1 infected persons by detecting diminished STAT1 phosphorylation (pSTAT1) and interferon-stimulated gene (ISG) induction upon type 1 IFN stimulation compared to healthy controls. Importantly, HIV-1 infected people who were virologically suppressed with antiretrovirals also showed type 1 IFN refractoriness. We found that USP18 levels were elevated in people with refractory pSTAT1 and ISG induction and confirmed this finding ex vivo in CD4+ T cells from another cohort of HIV-HCV coinfected persons who received exogenous pegylated interferon-α2b in a clinical trial. We used a cell culture model to recapitulate type 1 IFN refractoriness in uninfected CD4+ T cells that were conditioned with media from HIV-1 inoculated PBMCs, inhibiting de novo infection with antiretroviral agents. In this model, RNA interference against USP18 partly restored type 1 IFN responses in CD4+ T cells. We found evidence of type 1 IFN refractoriness in PLWH irrespective of virologic suppression that was associated with upregulated USP18, a process that might be therapeutically targeted to improve endogenous control of infection.ImportancePeople living with HIV-1 (PLWH) have elevated constitutive expression of type 1 interferons (IFN). However, it is unclear whether this impacts downstream innate immune responses. We identified refractory responses to type 1 IFN stimulation in T cells from PLWH, independent of antiretroviral treatment. Type 1 IFN refractoriness was linked to elevated USP18 levels in the same cells. Moreover, we found that USP18 levels predicted the anti-HIV-1 effect of type 1 IFN-based therapy on PLWH. In vitro, we demonstrated that refractory type 1 IFN responses were transferrable to HIV-1 uninfected target CD4+ T cells, and this phenomenon was mediated by type 1 IFN from HIV-1 infected cells. Type 1 IFN responses were partially restored by USP18 knockdown. Our findings illuminate a new mechanism by which HIV-1 contributes to innate immune dysfunction in PLWH, through the continuous production of type 1 IFN that induces a refractory state of responsiveness.
Collapse
Affiliation(s)
- Sho Sugawara
- Department of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ramy El-Diwany
- Department of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Laura K Cohen
- Department of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kimberly E Rousseau
- Department of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Rebecca T Veenhuis
- Department of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shruti H Mehta
- Department of Epidemiology, Johns Hopkins University School of Public Health, Baltimore, Maryland, USA
| | - Joel N Blankson
- Department of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David L Thomas
- Department of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrea L Cox
- Department of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ashwin Balagopal
- Department of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
103
|
Xie J, Wang M, Cheng A, Jia R, Zhu D, Liu M, Chen S, Zhao X, Yang Q, Wu Y, Zhang S, Luo Q, Wang Y, Xu Z, Chen Z, Zhu L, Liu Y, Yu Y, Zhang L, Chen X. The role of SOCS proteins in the development of virus- induced hepatocellular carcinoma. Virol J 2021; 18:74. [PMID: 33849568 PMCID: PMC8045357 DOI: 10.1186/s12985-021-01544-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 04/03/2021] [Indexed: 01/08/2023] Open
Abstract
Background Liver cancer has become one of the most common cancers and has a high mortality rate. Hepatocellular carcinoma is one of the most common liver cancers, and its occurrence and development process are associated with chronic hepatitis B virus (HBV) and hepatitis C virus (HCV) infections. Main body The serious consequences of chronic hepatitis virus infections are related to the viral invasion strategy. Furthermore, the viral escape mechanism has evolved during long-term struggles with the host. Studies have increasingly shown that suppressor of cytokine signaling (SOCS) proteins participate in the viral escape process. SOCS proteins play an important role in regulating cytokine signaling, particularly the Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathway. Cytokines stimulate the expression of SOCS proteins, in turn, SOCS proteins inhibit cytokine signaling by blocking the JAK-STAT signaling pathway, thereby achieving homeostasis. By utilizing SOCS proteins, chronic hepatitis virus infection may destroy the host’s antiviral responses to achieve persistent infection. Conclusions This review provides recent knowledge regarding the role of SOCS proteins during chronic hepatitis virus infection and provides some new ideas for the future treatment of chronic hepatitis.
Collapse
Affiliation(s)
- Jinyan Xie
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| | - Mingshu Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| | - Anchun Cheng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China. .,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China. .,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.
| | - Renyong Jia
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| | - Dekang Zhu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| | - Mafeng Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| | - Shun Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| | - XinXin Zhao
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| | - Qiao Yang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| | - Ying Wu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| | - Shaqiu Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| | - Qihui Luo
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| | - Yin Wang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| | - Zhiwen Xu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| | - Zhengli Chen
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| | - Ling Zhu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| | - Yunya Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| | - Yanling Yu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| | - Ling Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| | - Xiaoyue Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, 611130, Sichuan, People's Republic of China
| |
Collapse
|
104
|
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: 1.7] [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
|
105
|
Winslow S, Odqvist L, Diver S, Riise R, Abdillahi S, Wingren C, Lindmark H, Wellner A, Lundin S, Yrlid L, Ax E, Djukanovic R, Sridhar S, Higham A, Singh D, Southworth T, Brightling CE, Olsson HK, Jevnikar Z. Multi-omics links IL-6 trans-signalling with neutrophil extracellular trap formation and Haemophilus infection in COPD. Eur Respir J 2021; 58:13993003.03312-2020. [PMID: 33766947 DOI: 10.1183/13993003.03312-2020] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 03/04/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND IL-6 trans-signalling (IL-6TS) is emerging as a pathogenic mechanism in chronic respiratory diseases, however the drivers of IL-6TS in the airways and the phenotypic characteristic of patients with increased IL-6TS pathway activation remain poorly understood. OBJECTIVE Our aim was to identify and characterize COPD patients with increased airway IL-6TS and to elucidate the biological drivers of IL-6TS pathway activation. METHODS We used an IL-6TS-specific sputum biomarker profile (sIL-6R, IL-6, IL-1β, IL-8, MIP-1β) to stratify sputum data from patients with COPD (n=74; BEAT-COPD) by hierarchical clustering. The IL-6TS signature was related to clinical characteristics and sputum microbiome profiles. The induction of neutrophil extracellular trap formation (NETosis) and IL-6TS by Haemophilus influenzae were studied in human neutrophils. RESULTS Hierarchical clustering revealed an IL-6TS-high subset (n=24) of COPD patients, which shared phenotypic traits with an IL-6TS-high subset previously identified in asthma. The subset was characterized by increased sputum cell counts (p=0.0001), persistent sputum neutrophilia (p=0.0004), reduced quality of life (CRQ total score; p=0.008), and increased levels of pro-inflammatory mediators and MMPs in sputum. IL-6TS-high COPD patients showed an increase in Proteobacteria, with Haemophilus as the dominating genus. NETosis induced by H. influenzae was identified as a potential mechanism for increased soluble IL-6 receptor (sIL-6R) levels. This was supported by a significant positive correlation between sIL-6R and NETosis markers in bronchoalveolar lavage fluid from COPD patients. CONCLUSION IL-6TS pathway activation due to chronic colonization with Haemophilus may be an important disease driver in a subset of COPD patients.
Collapse
Affiliation(s)
- Sofia Winslow
- Translational Science and Experimental Medicine, Early Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Lina Odqvist
- Bioscience COPD/IPF, Early Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Sarah Diver
- Department of Respiratory Science, Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester and University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Rebecca Riise
- Bioscience COPD/IPF, Early Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Suado Abdillahi
- Bioscience COPD/IPF, Early Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Cecilia Wingren
- Bioscience COPD/IPF, Early Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Helena Lindmark
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Annika Wellner
- Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Sofia Lundin
- Translational Science and Experimental Medicine, Early Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Linda Yrlid
- Bioscience COPD/IPF, Early Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Elisabeth Ax
- Translational Science and Experimental Medicine, Early Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.,Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ratko Djukanovic
- NIHR Southampton Respiratory Biomedical Research Unit, Clinical and Experimental Sciences and Human Development and Health, University of Southampton, Southampton, UK
| | - Sriram Sridhar
- Oncology Bioinformatics, Translational Science, Early Oncology, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Andrew Higham
- The University of Manchester Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and University Hospital of South Manchester, NHS Foundation Trust, Manchester, UK
| | - Dave Singh
- The University of Manchester Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and University Hospital of South Manchester, NHS Foundation Trust, Manchester, UK
| | - Thomas Southworth
- The University of Manchester Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and University Hospital of South Manchester, NHS Foundation Trust, Manchester, UK
| | - Christopher E Brightling
- Department of Respiratory Science, Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester and University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Henric K Olsson
- Translational Science and Experimental Medicine, Early Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Zala Jevnikar
- Translational Science and Experimental Medicine, Early Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
106
|
Abolfathi H, Sheikhpour M, Shahraeini SS, Khatami S, Nojoumi SA. Studies in lung cancer cytokine proteomics: a review. Expert Rev Proteomics 2021; 18:49-64. [PMID: 33612047 DOI: 10.1080/14789450.2021.1892491] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Proteins are molecules that have role in the progression of the diseases. Proteomics is a tool that can play an effective role in identifying diagnostic and therapeutic biomarkers for lung cancer. Cytokines are proteins that play a decisive role in activating body's immune system in lung cancer. They can increase the growth of the tumor (oncogenic cytokines) or limit tumor growth (anti-tumor cytokines) by regulating related signaling pathways such as proliferation, growth, metastasis, and apoptosis. AREAS COVERED In the present study, a total of 223 papers including 196 research papers and 27 review papers, extracted from PubMed and Scopus and published from 1997 to present, are reviewed. The most important involved-cytokines in lung cancer including TNF-α, IFN- γ, TGF-β, VEGF and interleukins such as IL-6, IL-17, IL-8, IL-10, IL-22, IL-1β and IL-18 are introduced. Also, the pathological and biological role of such cytokines in cancer signaling pathways is explained. EXPERT OPINION In lung cancer, the cytokine expression changes under the physiological conditions of the immune system, and inflammatory cytokines are associated with the progression of lung cancer. Therefore, the cytokine expression profile can be used in the diagnosis, prognosis, prediction of therapeutic responses, and survival of patients with lung cancer.
Collapse
Affiliation(s)
- Hanie Abolfathi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Mojgan Sheikhpour
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Sadegh Shahraeini
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Shohreh Khatami
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Ali Nojoumi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
107
|
Aydemir MN, Aydemir HB, Korkmaz EM, Budak M, Cekin N, Pinarbasi E. Computationally predicted SARS-COV-2 encoded microRNAs target NFKB, JAK/STAT and TGFB signaling pathways. GENE REPORTS 2021; 22:101012. [PMID: 33398248 PMCID: PMC7773562 DOI: 10.1016/j.genrep.2020.101012] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 11/27/2020] [Accepted: 12/13/2020] [Indexed: 12/13/2022]
Abstract
Recently an outbreak that emerged in Wuhan, China in December 2019, spread to the whole world in a short time and killed >1,410,000 people. It was determined that a new type of beta coronavirus called severe acute respiratory disease coronavirus type 2 (SARS-CoV-2) was causative agent of this outbreak and the disease caused by the virus was named as coronavirus disease 19 (COVID19). Despite the information obtained from the viral genome structure, many aspects of the virus-host interactions during infection is still unknown. In this study we aimed to identify SARS-CoV-2 encoded microRNAs and their cellular targets. We applied a computational method to predict miRNAs encoded by SARS-CoV-2 along with their putative targets in humans. Targets of predicted miRNAs were clustered into groups based on their biological processes, molecular function, and cellular compartments using GO and PANTHER. By using KEGG pathway enrichment analysis top pathways were identified. Finally, we have constructed an integrative pathway network analysis with target genes. We identified 40 SARS-CoV-2 miRNAs and their regulated targets. Our analysis showed that targeted genes including NFKB1, NFKBIE, JAK1-2, STAT3-4, STAT5B, STAT6, SOCS1-6, IL2, IL8, IL10, IL17, TGFBR1-2, SMAD2-4, HDAC1-6 and JARID1A-C, JARID2 play important roles in NFKB, JAK/STAT and TGFB signaling pathways as well as cells' epigenetic regulation pathways. Our results may help to understand virus-host interaction and the role of viral miRNAs during SARS-CoV-2 infection. As there is no current drug and effective treatment available for COVID19, it may also help to develop new treatment strategies.
Collapse
Key Words
- ACE-2, angiotensin-converting enzyme 2
- AKT1, AKT serine/threonine kinase 1
- BCL2, BCL2 apoptosis regulator
- CDK1, cyclin dependent kinase 1
- CDKL2, cyclin dependent kinase like 2
- COVID19, new type corona virus disease
- CTNNB1, catenin beta 1
- CXCL1, C-X-C motif chemokine ligand 1
- CXCL10, C-X-C motif chemokine ligand 10
- CXCL11, C-X-C motif chemokine ligand 11
- CXCL16, C-X-C motif chemokine ligand 16
- CXCL9, C-X-C motif chemokine ligand 9
- E2F1, E2F transcription factor 1
- EIF4A1, eukaryotic translation initiation factor 4A1
- GRB2, growth factor receptor bound protein 2
- HDAC1, histone deacetylase 1
- HDAC2, histone deacetylase 2
- HDAC3, histone deacetylase 3
- HIF1A, hypoxia inducible factor 1 subunit alpha
- ICTV, International Committee on Taxonomy of Viruses
- IFNGR2, interferon gamma receptor 2
- IKBKE, inhibitor of nuclear factor kappa B kinase subunit epsilon
- IL10, interleukin 10
- IL13, interleukin 13
- IL15, interleukin 15
- IL16, interleukin 16
- IL17A, interleukin 17 A
- IL2, interleukin 2
- IL21, interleukin 21
- IL22, interleukin 22
- IL24, interleukin 24
- IL25, interleukin 25
- IL33, interleukin 33
- IL5, interleukin 5
- IL7, interleukin 7
- IL8, interleukin 8
- JAK/STAT
- JAK1, Janus kinase 1
- JAK2, Janus kinase 2
- JARID1A, lysine demethylase 5A
- JARID1B, lysine demethylase 5B
- JARID1C, lysine demethylase 5C
- JARID2, Jumonji and AT-rich interaction domain containing 2
- KEGG, Kyoto Encyclopedia of Genes and Genomes
- MAPK1, mitogen-activated protein kinase 1
- MAPK3, mitogen-activated protein kinase 3
- MAPK4, mitogen-activated protein kinase 4
- MAPK6, mitogen-activated protein kinase 6
- MAPK7, mitogen-activated protein kinase 7
- NFKB
- NFKB1, nuclear factor kappa B subunit 1
- NFKBIE, NFKB inhibitor epsilon
- NOS3, nitric oxide synthase 3
- PANTHER, protein analysis through evolutionary relationships
- PIK3CA, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha
- PTEN, phosphatase and tensin homolog
- RB1, RB transcriptional corepressor 1
- RHOA, ras homolog family member A
- SARS-CoV-2
- SARS-CoV-2, severe acute respiratory disease coronavirus type 2
- SMAD2, SMAD family member 2
- SMAD3, SMAD family member 3
- SMAD4, SMAD family member 4
- SOCS1, suppressor of cytokine signaling 1
- SOCS3, suppressor of cytokine signaling 3
- SOCS4, suppressor of cytokine signaling 4
- SOCS5, suppressor of cytokine signaling 5
- SOCS6, suppressor of cytokine signaling 6
- SOS1, SOS Ras/Rac guanine nucleotide exchange factor 1
- SP1, Sp1 transcription factor
- STAT3, signal transducer and activator of transcription 3
- STAT4, signal transducer and activator of transcription 4
- STAT5B, signal transducer and activator of transcription 5B
- STAT6, signal transducer and activator of transcription 6
- SUMO1, small ubiquitin like modifier 1
- SUMO2, small ubiquitin like modifier 2
- TBP, TATA-box binding protein
- TGFB
- TGFBR1, transforming growth factor beta receptor 1
- TGFBR2, transforming growth factor beta receptor 2
- TMPRSS11A, transmembrane serine protease 11A
- TMPRSS4, transmembrane serine protease 4
- TNFRSF21, TNF receptor superfamily member 21
- WHO, World Health Organization
- miRNA
Collapse
Affiliation(s)
- Merve Nur Aydemir
- Department of Molecular Biology and Genetics, Faculty of Science, Sivas Cumhuriyet University, Sivas, Turkey
| | - Habes Bilal Aydemir
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Gaziosmanpaşa University, Tokat, Turkey
| | - Ertan Mahir Korkmaz
- Department of Molecular Biology and Genetics, Faculty of Science, Sivas Cumhuriyet University, Sivas, Turkey
| | - Mahir Budak
- Department of Molecular Biology and Genetics, Faculty of Science, Sivas Cumhuriyet University, Sivas, Turkey
| | - Nilgun Cekin
- Sivas Cumhuriyet University, Faculty of Medicine, Department of Medical Biology, 58140 Sivas, Turkey
| | - Ergun Pinarbasi
- Sivas Cumhuriyet University, Faculty of Medicine, Department of Medical Biology, 58140 Sivas, Turkey
| |
Collapse
|
108
|
Ruan Z, Chen G, Xie T, Mo G, Wang G, Luo W, Li H, Shi M, Liu WS, Zhang X. Cytokine inducible SH2-containing protein potentiate J subgroup avian leukosis virus replication and suppress antiviral responses in DF-1 chicken fibroblast cells. Virus Res 2021; 296:198344. [PMID: 33636239 DOI: 10.1016/j.virusres.2021.198344] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 12/26/2022]
Abstract
Cytokine-inducible Srchomology2 (SH2)-containing protein (CIS) belongs to the suppressors of cytokine signaling (SOCS) protein family function as a negative feedback loop inhibiting cytokine signal transduction. J subgroup avian leukosis virus (ALV-J), a commonly-seen avian virus with a feature of immunosuppression, poses an unmeasurable threat to the poultry industry across the world. However, commercial medicines or vaccines are still no available for this virus. This study aims to evaluate the potential effect of chicken CIS in antiviral response and its role on ALV-J replication. The results showed that ALV-J strain SCAU-HN06 infection induced CIS expression in DF-1 cells, which was derived from chicken embryo free of endogenous avian sarcoma-leukosis virus (ASLV) like sequences. By overexpressing CIS, the expression of chicken type I interferon (IFN-I) and interferon-stimulated genes (ISGs; PKR, ZAP, CH25H, CCL4, IFIT5, and ISG12) were both suppressed. Meanwhile, data showed that CIS overexpression also increased viral yield. Interestingly, knockdown of CIS enhanced induction of IFN-I and ISGs and inhibited viral replication. Collectively, we proved that modulation of CIS expression not only affected SCAU-HN06 replication in vitro but also altered the expression of IFN-I and ISGs that act as an essential part of antiviral innate immune system. Our data provide a potential target for developing antiviral agents for ALV-J.
Collapse
Affiliation(s)
- Zhuohao Ruan
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, China; College of Marine Sciences, South China Agricultural University, Guangzhou, China.
| | - Genghua Chen
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, China.
| | - Tingting Xie
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, China.
| | - Guodong Mo
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, China.
| | - Guiyan Wang
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, China.
| | - Wen Luo
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, China.
| | - Hongmei Li
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, China.
| | - Meiqing Shi
- Division of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MA, 20742, USA.
| | - Wen-Sheng Liu
- College of Marine Sciences, South China Agricultural University, Guangzhou, China; Guangdong Province Engineering Research Centre of Aquatic Immunization and Aquaculture Health Techniques, South China Agricultural University, Guangzhou, China.
| | - Xiquan Zhang
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, 510642, China.
| |
Collapse
|
109
|
Mizoguchi I, Ohashi M, Hasegawa H, Chiba Y, Orii N, Inoue S, Kawana C, Xu M, Sudo K, Fujita K, Kuroda M, Hashimoto SI, Matsushima K, Yoshimoto T. EBV-induced gene 3 augments IL-23Rα protein expression through a chaperone calnexin. J Clin Invest 2021; 130:6124-6140. [PMID: 32809973 DOI: 10.1172/jci122732] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/05/2020] [Indexed: 12/27/2022] Open
Abstract
Epstein-Barr virus-induced gene 3 (EBI3) is a subunit common to IL-27, IL-35, and IL-39. Here, we explore an intracellular role of EBI3 that is independent of its function in cytokines. EBI3-deficient naive CD4+ T cells had reduced IFN-γ production and failed to induce T cell-dependent colitis in mice. Similarly reduced IFN-γ production was observed in vitro in EBI3-deficient CD4+ T cells differentiated under pathogenic Th17 polarizing conditions with IL-23. This is because the induction of expression of one of the IL-23 receptor (IL-23R) subunits, IL-23Rα, but not another IL-23R subunit, IL-12Rβ1, was selectively decreased at the protein level, but not the mRNA level. EBI3 augmented IL-23Rα expression via binding to the chaperone molecule calnexin and to IL-23Rα in a peptide-dependent manner, but not a glycan-dependent manner. Indeed, EBI3 failed to augment IL-23Rα expression in the absence of endogenous calnexin. Moreover, EBI3 poorly augmented the expression of G149R, an IL-23Rα variant that protects against the development of human colitis, because binding of EBI3 to the variant was reduced. Taken together with the result that EBI3 expression is inducible in T cells, the present results suggest that EBI3 plays a critical role in augmenting IL-23Rα protein expression via calnexin under inflammatory conditions.
Collapse
Affiliation(s)
- Izuru Mizoguchi
- Department of Immunoregulation, Institute of Medical Science
| | - Mio Ohashi
- Department of Immunoregulation, Institute of Medical Science
| | | | - Yukino Chiba
- Department of Immunoregulation, Institute of Medical Science
| | - Naoko Orii
- Department of Immunoregulation, Institute of Medical Science
| | - Shinya Inoue
- Department of Immunoregulation, Institute of Medical Science
| | - Chiaki Kawana
- Department of Immunoregulation, Institute of Medical Science
| | - Mingli Xu
- Department of Immunoregulation, Institute of Medical Science
| | | | - Koji Fujita
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| | - Masahiko Kuroda
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| | - Shin-Ichi Hashimoto
- Department of Laboratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa, Japan
| | - Kouji Matsushima
- Department of Molecular Preventive Medicine, School of Medicine, University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
110
|
Arima M, Fujii Y, Sonoda KH. Translational Research in Retinopathy of Prematurity: From Bedside to Bench and Back Again. J Clin Med 2021; 10:331. [PMID: 33477419 PMCID: PMC7830975 DOI: 10.3390/jcm10020331] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/09/2021] [Accepted: 01/15/2021] [Indexed: 12/11/2022] Open
Abstract
Retinopathy of prematurity (ROP), a vascular proliferative disease affecting preterm infants, is a leading cause of childhood blindness. Various studies have investigated the pathogenesis of ROP. Clinical experience indicates that oxygen levels are strongly correlated with ROP development, which led to the development of oxygen-induced retinopathy (OIR) as an animal model of ROP. OIR has been used extensively to investigate the molecular mechanisms underlying ROP and to evaluate the efficacy of new drug candidates. Large clinical trials have demonstrated the efficacy of anti-vascular endothelial growth factor (VEGF) agents to treat ROP, and anti-VEGF therapy is presently becoming the first-line treatment worldwide. Anti-VEGF therapy has advantages over conventional treatments, including being minimally invasive with a low risk of refractive error. However, long-term safety concerns and the risk of late recurrence limit this treatment. There is an unmet medical need for novel ROP therapies, which need to be addressed by safe and minimally invasive therapies. The recent progress in biotechnology has contributed greatly to translational research. In this review, we outline how basic ROP research has evolved with clinical experience and the subsequent emergence of new drugs. We discuss previous and ongoing trials and present the candidate molecules expected to become novel targets.
Collapse
Affiliation(s)
- Mitsuru Arima
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 8128582, Japan; (Y.F.); (K.-H.S.)
- Center for Clinical and Translational Research, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka 8128582, Japan
| | - Yuya Fujii
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 8128582, Japan; (Y.F.); (K.-H.S.)
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 8128582, Japan; (Y.F.); (K.-H.S.)
| |
Collapse
|
111
|
Westerberg J, Tideholm E, Piersiala K, Drakskog C, Kumlien Georén S, Mäki-Torkko E, Cardell LO. JAK/STAT Dysregulation With SOCS1 Overexpression in Acquired Cholesteatoma-Adjacent Mucosa. Otol Neurotol 2021; 42:e94-e100. [PMID: 33201080 DOI: 10.1097/mao.0000000000002850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
IMPORTANCE Surgery remains the gold standard in cholesteatoma treatment. However, the rate of recurrence is significant and the development of new nonsurgical treatment alternatives is warranted. One of the possible molecular pathways to target is the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway. OBJECTIVE To investigate the JAK/STAT pathway in the middle ear mucosa in patients with acquired cholesteatoma compared with middle ear mucosa from healthy controls. DESIGN Case-control study. SETTING Linköping University Hospital, Sweden, and Karolinska Institutet, Stockholm, Sweden. Sampling period: February 2011 to December 2016. PARTICIPANTS Middle ear mucosa from 26 patients with acquired cholesteatoma undergoing tympanoplasty and mastoidectomy, and 27 healthy controls undergoing translabyrinthine surgery for vestibular schwannoma or cochlear implantation was investigated. MAIN OUTCOMES/MEASURES The expression of Interleukin-7 receptor alpha, JAK1, JAK2, JAK3, STAT5A, STAT5B, and suppressor of cytokine signaling-1 (SOCS1) were quantified using quantitative polymerase chain reaction. In addition, expression level of cyclin D2, transforming growth factor beta 1, thymic stromal lymphopoietin, CD3, and CD19 was evaluated. RESULTS In cholesteatoma-adjacent mucosa, SOCS1 was significantly upregulated (p= 0.0003) compared with healthy controls, whereas STAT5B was significantly downregulated (p = 0.0006). The expression of JAK1, JAK2, JAK3, and STAT5A did not differ significantly between groups. CONCLUSIONS AND RELEVANCE To the best of our knowledge, this is the first article reporting dysregulation of the JAK/STAT pathway in cholesteatoma-adjacent mucosa. The main finding is that important players of the aforementioned pathway are significantly altered, namely SOCS1 is upregulated and STAT5B is downregulated compared with healthy controls.
Collapse
Affiliation(s)
- Johanna Westerberg
- Department of Biomedical and Clinical Sciences, Division of Sensory Organs and Communication, Linköping University, Region Östergötland, Sweden
| | - Ellen Tideholm
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm
| | - Krzysztof Piersiala
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm
- Department of ENT Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Cecilia Drakskog
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm
| | - Susanna Kumlien Georén
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm
| | - Elina Mäki-Torkko
- Department of Biomedical and Clinical Sciences, Division of Sensory Organs and Communication, Linköping University, Region Östergötland, Sweden
- Audiological Research Center, Faculty of Medicine and Health, Örebro university, Sweden
| | - Lars Olaf Cardell
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm
- Department of ENT Diseases, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
112
|
Park JM, An JM, Han YM, Surh YJ, Hwang SJ, Kim SJ, Hahm KB. Walnut polyphenol extracts inhibit Helicobacter pylori-induced STAT3 Tyr705 phosphorylation through activation of PPAR-γ and SOCS1 induction. J Clin Biochem Nutr 2021. [DOI: 10.3164/jcbn.20-105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
| | - Jeong Min An
- CHA Cancer Preventive Research Center, CHA Bio Complex, CHA University
| | | | | | | | | | - Ki Baik Hahm
- CHA Cancer Preventive Research Center, CHA Bio Complex, CHA University
- Medpacto Research Institute, Medpacto
| |
Collapse
|
113
|
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: 5] [Impact Index Per Article: 1.7] [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
|
114
|
Karki P, Ke Y, Zhang CO, Li Y, Tian Y, Son S, Yoshimura A, Kaibuchi K, Birukov KG, Birukova AA. SOCS3-microtubule interaction via CLIP-170 and CLASP2 is critical for modulation of endothelial inflammation and lung injury. J Biol Chem 2021; 296:100239. [PMID: 33372035 PMCID: PMC7949054 DOI: 10.1074/jbc.ra120.014232] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 11/23/2020] [Accepted: 12/28/2020] [Indexed: 12/12/2022] Open
Abstract
Proinflammatory cytokines such as IL-6 induce endothelial cell (EC) barrier disruption and trigger an inflammatory response in part by activating the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway. The protein suppressor of cytokine signaling-3 (SOCS3) is a negative regulator of JAK-STAT, but its role in modulation of lung EC barrier dysfunction caused by bacterial pathogens has not been investigated. Using human lung ECs and EC-specific SOCS3 knockout mice, we tested the hypothesis that SOCS3 confers microtubule (MT)-mediated protection against endothelial dysfunction. SOCS3 knockdown in cultured ECs or EC-specific SOCS3 knockout in mice resulted in exacerbated lung injury characterized by increased permeability and inflammation in response to IL-6 or heat-killed Staphylococcus aureus (HKSA). Ectopic expression of SOCS3 attenuated HKSA-induced EC dysfunction, and this effect required assembled MTs. SOCS3 was enriched in the MT fractions, and treatment with HKSA disrupted SOCS3-MT association. We discovered that-in addition to its known partners gp130 and JAK2-SOCS3 interacts with MT plus-end binding proteins CLIP-170 and CLASP2 via its N-terminal domain. The resulting SOCS3-CLIP-170/CLASP2 complex was essential for maximal SOCS3 anti-inflammatory effects. Both IL-6 and HKSA promoted MT disassembly and disrupted SOCS3 interaction with CLIP-170 and CLASP2. Moreover, knockdown of CLIP-170 or CLASP2 impaired SOCS3-JAK2 interaction and abolished the anti-inflammatory effects of SOCS3. Together, these findings demonstrate for the first time an interaction between SOCS3 and CLIP-170/CLASP2 and reveal that this interaction is essential to the protective effects of SOCS3 in lung endothelium.
Collapse
Affiliation(s)
- Pratap Karki
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Chen-Ou Zhang
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yue Li
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yufeng Tian
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Sophia Son
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University, Tokyo, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University, Nagoya, Japan
| | - Konstantin G Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Anna A Birukova
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
115
|
Zhu J, Wang J, Huang J, Du W, He Y, Pan H, Luo J. MicroRNA-140-5p regulates the proliferation, apoptosis and inflammation of RA FLSs by repressing STAT3. Exp Ther Med 2020; 21:171. [PMID: 33456538 PMCID: PMC7792473 DOI: 10.3892/etm.2020.9602] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 04/09/2020] [Indexed: 12/15/2022] Open
Abstract
Ectopic expression of microRNA (miRNA) in rheumatoid arthritis (RA) fibroblast-like synoviocyte (RA FLS) is associated with the development of rheumatoid arthritis. The present study aimed to evaluate the effects of miRNA-140-5p (miR-140) on the properties of RA FLSs. It was found that miR-140 expression was decreased in 33 RA patients and extracted RA FLS samples, when compared to the corresponding healthy controls. Abnormally increased miR-140 expression in RA FLSs attenuated cell proliferation and increased cell apoptosis. Additionally, reduced pro-inflammatory cytokine production was observed in RA FLSs transfected with a miR-140 precursor. Furthermore, the 3'-UTR of the signal transducer and activator of transcription (STAT) 3 gene was identified as a target of miR-140. Notably, restoration of STAT3 expression rescued the regulatory effect of miR-140 on the proliferation, apoptosis and inflammatory cytokine production of RA FLSs. Therefore, the current findings indicated that miR-140 is a crucial modulator of both proliferation and apoptosis, shedding light on the etiology behind RA FLS viability, which is modulated by an interplay between miR-140 and STAT3 in the context of RA.
Collapse
Affiliation(s)
- Jiehua Zhu
- Department of Laboratory Medicine, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jianglin Wang
- School of Laboratory Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jialin Huang
- School of Laboratory Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Wensheng Du
- Department of Laboratory Medicine, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Yingzhong He
- Department of Laboratory Medicine, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Hongfei Pan
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Junmin Luo
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
116
|
Hajj GNM, Nunes PBC, Roffe M. Genome-wide translation patterns in gliomas: An integrative view. Cell Signal 2020; 79:109883. [PMID: 33321181 DOI: 10.1016/j.cellsig.2020.109883] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/01/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Gliomas are the most frequent tumors of the central nervous system (CNS) and include the highly malignant glioblastoma (GBM). Characteristically, gliomas have translational control deregulation related to overactivation of signaling pathways such as PI3K/AKT/mTORC1 and Ras/ERK1/2. Thus, mRNA translation appears to play a dominant role in glioma gene expression patterns. The, analysis of genome-wide translated transcripts, together known as the translatome, may reveal important information for understanding gene expression patterns in gliomas. This review provides a brief overview of translational control mechanisms altered in gliomas with a focus on the current knowledge related to the translatomes of glioma cells and murine glioma models. We present an integrative meta-analysis of selected glioma translatome data with the aim of identifying recurrent patterns of gene expression preferentially regulated at the level of translation and obtaining clues regarding the pathological significance of these alterations. Re-analysis of several translatome datasets was performed to compare the translatomes of glioma models with those of their non-tumor counterparts and to document glioma cell responses to radiotherapy and MNK modulation. The role of recurrently altered genes in the context of translational control and tumorigenesis are discussed.
Collapse
Affiliation(s)
- Glaucia Noeli Maroso Hajj
- International Research Institute, A.C.Camargo Cancer Center, Rua Taguá, 440, São Paulo ZIP Code: 01508-010, Brazil; National Institute of Oncogenomics and Innovation, Brazil.
| | - Paula Borzino Cordeiro Nunes
- International Research Institute, A.C.Camargo Cancer Center, Rua Taguá, 440, São Paulo ZIP Code: 01508-010, Brazil
| | - Martin Roffe
- International Research Institute, A.C.Camargo Cancer Center, Rua Taguá, 440, São Paulo ZIP Code: 01508-010, Brazil; National Institute of Oncogenomics and Innovation, Brazil.
| |
Collapse
|
117
|
Cai C, Hu Z, Yu X. Accelerator or Brake: Immune Regulators in Malaria. Front Cell Infect Microbiol 2020; 10:610121. [PMID: 33363057 PMCID: PMC7758250 DOI: 10.3389/fcimb.2020.610121] [Citation(s) in RCA: 4] [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: 09/25/2020] [Accepted: 11/09/2020] [Indexed: 12/15/2022] Open
Abstract
Malaria is a life-threatening infectious disease, affecting over 250 million individuals worldwide each year, eradicating malaria has been one of the greatest challenges to public health for a century. Growing resistance to anti-parasitic therapies and lack of effective vaccines are major contributing factors in controlling this disease. However, the incomplete understanding of parasite interactions with host anti-malaria immunity hinders vaccine development efforts to date. Recent studies have been unveiling the complexity of immune responses and regulators against Plasmodium infection. Here, we summarize our current understanding of host immune responses against Plasmodium-derived components infection and mainly focus on the various regulatory mechanisms mediated by recent identified immune regulators orchestrating anti-malaria immunity.
Collapse
Affiliation(s)
- Chunmei Cai
- Research Center for High Altitude Medicine, School of Medical, Qinghai University, Xining, China
- Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province, Qinghai University, Xining, China
| | - Zhiqiang Hu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiao Yu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Lab of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| |
Collapse
|
118
|
Martinez-Fabregas J, Wang L, Pohler E, Cozzani A, Wilmes S, Kazemian M, Mitra S, Moraga I. CDK8 Fine-Tunes IL-6 Transcriptional Activities by Limiting STAT3 Resident Time at the Gene Loci. Cell Rep 2020; 33:108545. [PMID: 33357429 PMCID: PMC7773550 DOI: 10.1016/j.celrep.2020.108545] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/28/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023] Open
Abstract
Cytokines are highly pleiotropic ligands that regulate the immune response. Here, using interleukin-6 (IL-6) as a model system, we perform detailed phosphoproteomic and transcriptomic studies in human CD4+ T helper 1 (Th-1) cells to address the molecular bases defining cytokine functional pleiotropy. We identify CDK8 as a negative regulator of STAT3 transcriptional activities, which interacts with STAT3 upon IL-6 stimulation. Inhibition of CDK8 activity, using specific small molecule inhibitors, reduces the IL-6-induced phosphoproteome by 23% in Th-1 cells, including STAT3 S727 phosphorylation. STAT3 binding to target DNA sites in the genome is increased upon CDK8 inhibition, which results in a concomitant increase in STAT3-mediated transcriptional activity. Importantly, inhibition of CDK8 activity under Th-17 polarizing conditions results in an enhancement of Th-17 differentiation. Our results support a model where CDK8 regulates STAT3 transcriptional processivity by modulation of its gene loci resident time, critically contributing to diversification of IL-6 responses. CDK8 regulates IL-6-mediated STAT3 S727 phosphorylation in primary human T cells CDK8 controls STAT3 activity by limiting its resident time at gene loci CDK8 inhibition increases IL-6-mediated Th17 differentiation
Collapse
Affiliation(s)
| | - Luopin Wang
- Department of Computer Science, Purdue University, West Lafayette, IN, USA
| | - Elizabeth Pohler
- Division of Cell Signaling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Adeline Cozzani
- Université de Lille, INSERM UMR1277 CNRS UMR9020-CANTHER and Institut pour la Recherche sur le Cancer de Lille (IRCL), Lille, France
| | - Stephan Wilmes
- Division of Cell Signaling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Majid Kazemian
- Department of Computer Science, Purdue University, West Lafayette, IN, USA; Department of Biochemistry, Purdue University, West Lafayette, IN, USA.
| | - Suman Mitra
- Université de Lille, INSERM UMR1277 CNRS UMR9020-CANTHER and Institut pour la Recherche sur le Cancer de Lille (IRCL), Lille, France.
| | - Ignacio Moraga
- Division of Cell Signaling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK.
| |
Collapse
|
119
|
Du J, Liao W, Liu W, Deb DK, He L, Hsu PJ, Nguyen T, Zhang L, Bissonnette M, He C, Li YC. N 6-Adenosine Methylation of Socs1 mRNA Is Required to Sustain the Negative Feedback Control of Macrophage Activation. Dev Cell 2020; 55:737-753.e7. [PMID: 33220174 DOI: 10.1016/j.devcel.2020.10.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/12/2020] [Accepted: 10/29/2020] [Indexed: 12/16/2022]
Abstract
Bacterial infection triggers a cytokine storm that needs to be resolved to maintain the host's wellbeing. Here, we report that ablation of m6A methyltransferase subunit METTL14 in myeloid cells exacerbates macrophage responses to acute bacterial infection in mice, leading to high mortality due to sustained production of pro-inflammatory cytokines. METTL14 depletion blunts Socs1 m6A methylation and reduces YTHDF1 binding to the m6A sites, which diminishes SOCS1 induction leading to the overactivation of TLR4/NF-κB signaling. Forced expression of SOCS1 in macrophages depleted of METTL14 or YTHDF1 rescues the hyper-responsive phenotype of these macrophages in vitro and in vivo. We further show that LPS treatment induces Socs1 m6A methylation and sustains SOCS1 induction by promoting Fto mRNA degradation, and forced FTO expression in macrophages mimics the phenotype of METTL14-depleted macrophages. We conclude that m6A methylation-mediated SOCS1 induction is required to maintain the negative feedback control of macrophage activation in response to bacterial infection.
Collapse
Affiliation(s)
- Jie Du
- Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, IL, USA; Institute of Biomedical Research, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wang Liao
- Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, IL, USA; Department of Cardiology, Hainan General Hospital, Hainan Clinical Research Institute, Haikou, Hainan, China
| | - Weicheng Liu
- Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, IL, USA
| | - Dilip K Deb
- Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, IL, USA
| | - Lei He
- Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, IL, USA
| | - Phillip J Hsu
- Departments of Chemistry, Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| | - Tivoli Nguyen
- Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, IL, USA
| | - Linda Zhang
- Departments of Chemistry, Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| | - Marc Bissonnette
- Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, IL, USA
| | - Chuan He
- Departments of Chemistry, Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Yan Chun Li
- Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
120
|
Strous GJ, Almeida ADS, Putters J, Schantl J, Sedek M, Slotman JA, Nespital T, Hassink GC, Mol JA. Growth Hormone Receptor Regulation in Cancer and Chronic Diseases. Front Endocrinol (Lausanne) 2020; 11:597573. [PMID: 33312162 PMCID: PMC7708378 DOI: 10.3389/fendo.2020.597573] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022] Open
Abstract
The GHR signaling pathway plays important roles in growth, metabolism, cell cycle control, immunity, homeostatic processes, and chemoresistance via both the JAK/STAT and the SRC pathways. Dysregulation of GHR signaling is associated with various diseases and chronic conditions such as acromegaly, cancer, aging, metabolic disease, fibroses, inflammation and autoimmunity. Numerous studies entailing the GHR signaling pathway have been conducted for various cancers. Diverse factors mediate the up- or down-regulation of GHR signaling through post-translational modifications. Of the numerous modifications, ubiquitination and deubiquitination are prominent events. Ubiquitination by E3 ligase attaches ubiquitins to target proteins and induces proteasomal degradation or starts the sequence of events that leads to endocytosis and lysosomal degradation. In this review, we discuss the role of first line effectors that act directly on the GHR at the cell surface including ADAM17, JAK2, SRC family member Lyn, Ubc13/CHIP, proteasome, βTrCP, CK2, STAT5b, and SOCS2. Activity of all, except JAK2, Lyn and STAT5b, counteract GHR signaling. Loss of their function increases the GH-induced signaling in favor of aging and certain chronic diseases, exemplified by increased lung cancer risk in case of a mutation in the SOCS2-GHR interaction site. Insight in their roles in GHR signaling can be applied for cancer and other therapeutic strategies.
Collapse
Affiliation(s)
- Ger J. Strous
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
- BIMINI Biotech B.V., Leiden, Netherlands
| | - Ana Da Silva Almeida
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Joyce Putters
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Julia Schantl
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Magdalena Sedek
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Johan A. Slotman
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Tobias Nespital
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Gerco C. Hassink
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Jan A. Mol
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
121
|
Park JM, An JM, Han YM, Surh YJ, Hwang SJ, Kim SJ, Hahm KB. Walnut polyphenol extracts inhibit Helicobacter pylori-induced STAT3 Tyr705 phosphorylation through activation of PPAR-γ and SOCS1 induction. J Clin Biochem Nutr 2020; 67:248-256. [PMID: 33293765 PMCID: PMC7705089 DOI: 10.3164/jcbn.20-89] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 06/13/2020] [Indexed: 12/14/2022] Open
Abstract
The health beneficial effects of walnut plentiful of n-3 polyunsaturated fatty acid had been attributed to its anti-inflammatory and anti-oxidative properties against various clinical diseases. Since we have published Fat-1 transgenic mice overexpressing 3-desaturase significantly mitigated Helicobacter pylori (H. pylori)-associated gastric pathologies including rejuvenation of chronic atrophic gastritis and prevention of gastric cancer, in this study, we have explored the underlying molecular mechanisms of walnut against H. pylori infection. Fresh walnut polyphenol extracts (WPE) were found to suppress the phosphorylation and nuclear translocation of signal transducer and activator of transcription 3 (STAT3) induced by H. pylori infection in RGM-1 gastric mucosal cells. Notably, H. pylori infection significantly decreased suppressor of cytokine signaling 1 (SOCS1), but WPE induced expression of SOCS1, by which the suppressive effect of walnut extracts on STAT3Tyr705 phosphorylation was not seen in SOCS1 KO cells. WPE induced significantly increased nuclear translocation nuclear translocation of PPAR-γ in RGM1 cells, by which PPAR-γ KO inhibited transcription of SOCS1 and suppressive effect of WPE on p-STAT3Tyr705 was not seen. WPE inhibited the expression of c-Myc and IL-6/IL-6R signaling, which was attenuated in the RGM1 cells harboring SOCS1 specific siRNA. Conclusively, WPE inhibits H. pylori-induced STAT3 phosphorylation in a PPAR-γ and SOCS1-dependent manner.
Collapse
Affiliation(s)
- Jong Min Park
- College of Oriental Medicine, Daejeon University, Daehak-ro 62, Dong-gu, Daejeon, 34520, Korea
| | - Jeong Min An
- CHA Cancer Preventive Research Center, CHA Bio Complex, 330 Pangyo-dong, Bundang-gu, Seongnam, 13497, Korea
| | - Young Min Han
- Western Seoul Center, Korea Basic Science Institute, University-Industry Cooperate Building, 150 Bugahyeon-ro, Seodaemun-gu, Seoul, 03759, Korea
| | - Young Joon Surh
- College of Pharmacy Seoul National University, Seoul, 08826, Korea
| | - Sun Jin Hwang
- Medpacto Research Institute, Medpacto Inc., 92, Myeongdal-ro, Seocho-gu, Seoul, 06668, Korea
| | - Seong Jin Kim
- Medpacto Research Institute, Medpacto Inc., 92, Myeongdal-ro, Seocho-gu, Seoul, 06668, Korea
| | - Ki Baik Hahm
- CHA Cancer Preventive Research Center, CHA Bio Complex, 330 Pangyo-dong, Bundang-gu, Seongnam, 13497, Korea.,Medpacto Research Institute, Medpacto Inc., 92, Myeongdal-ro, Seocho-gu, Seoul, 06668, Korea
| |
Collapse
|
122
|
Huang S, Liu K, Cheng A, Wang M, Cui M, Huang J, Zhu D, Chen S, Liu M, Zhao X, Wu Y, Yang Q, Zhang S, Ou X, Mao S, Gao Q, Yu Y, Tian B, Liu Y, Zhang L, Yin Z, Jing B, Chen X, Jia R. SOCS Proteins Participate in the Regulation of Innate Immune Response Caused by Viruses. Front Immunol 2020; 11:558341. [PMID: 33072096 PMCID: PMC7544739 DOI: 10.3389/fimmu.2020.558341] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 08/24/2020] [Indexed: 12/17/2022] Open
Abstract
The host immune system has multiple innate immune receptors that can identify, distinguish and react to viral infections. In innate immune response, the host recognizes pathogen-associated molecular patterns (PAMP) in nucleic acids or viral proteins through pathogen recognition receptors (PRRs), especially toll-like receptors (TLRs) and induces immune cells or infected cells to produce type I Interferons (IFN-I) and pro-inflammatory cytokines, thus when the virus invades the host, innate immunity is the earliest immune mechanism. Besides, cytokine-mediated cell communication is necessary for the proper regulation of immune responses. Therefore, the appropriate activation of innate immunity is necessary for the normal life activities of cells. The suppressor of the cytokine signaling proteins (SOCS) family is one of the main regulators of the innate immune response induced by microbial pathogens. They mainly participate in the negative feedback regulation of cytokine signal transduction through Janus kinase signal transducer and transcriptional activator (JAK/STAT) and other signal pathways. Taken together, this paper reviews the SOCS proteins structures and the function of each domain, as well as the latest knowledge of the role of SOCS proteins in innate immune caused by viral infections and the mechanisms by which SOCS proteins assist viruses to escape host innate immunity. Finally, we discuss potential values of these proteins in future targeted therapies.
Collapse
Affiliation(s)
- Shanzhi Huang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ke Liu
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Anchun Cheng
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Mingshu Wang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Min Cui
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Juan Huang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Dekang Zhu
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Shun Chen
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Mafeng Liu
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xinxin Zhao
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yin Wu
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qiao Yang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Shaqiu Zhang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xumin Ou
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Sai Mao
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qun Gao
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yanling Yu
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Bin Tian
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yunya Liu
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ling Zhang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhongqiong Yin
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Bo Jing
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xiaoyue Chen
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Renyong Jia
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
123
|
Lawson KA, Sousa CM, Zhang X, Kim E, Akthar R, Caumanns JJ, Yao Y, Mikolajewicz N, Ross C, Brown KR, Zid AA, Fan ZP, Hui S, Krall JA, Simons DM, Slater CJ, De Jesus V, Tang L, Singh R, Goldford JE, Martin S, Huang Q, Francis EA, Habsid A, Climie R, Tieu D, Wei J, Li R, Tong AHY, Aregger M, Chan KS, Han H, Wang X, Mero P, Brumell JH, Finelli A, Ailles L, Bader G, Smolen GA, Kingsbury GA, Hart T, Kung C, Moffat J. Functional genomic landscape of cancer-intrinsic evasion of killing by T cells. Nature 2020; 586:120-126. [PMID: 32968282 DOI: 10.1038/s41586-020-2746-2] [Citation(s) in RCA: 239] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 06/30/2020] [Indexed: 01/05/2023]
Abstract
The genetic circuits that allow cancer cells to evade destruction by the host immune system remain poorly understood1-3. Here, to identify a phenotypically robust core set of genes and pathways that enable cancer cells to evade killing mediated by cytotoxic T lymphocytes (CTLs), we performed genome-wide CRISPR screens across a panel of genetically diverse mouse cancer cell lines that were cultured in the presence of CTLs. We identify a core set of 182 genes across these mouse cancer models, the individual perturbation of which increases either the sensitivity or the resistance of cancer cells to CTL-mediated toxicity. Systematic exploration of our dataset using genetic co-similarity reveals the hierarchical and coordinated manner in which genes and pathways act in cancer cells to orchestrate their evasion of CTLs, and shows that discrete functional modules that control the interferon response and tumour necrosis factor (TNF)-induced cytotoxicity are dominant sub-phenotypes. Our data establish a central role for genes that were previously identified as negative regulators of the type-II interferon response (for example, Ptpn2, Socs1 and Adar1) in mediating CTL evasion, and show that the lipid-droplet-related gene Fitm2 is required for maintaining cell fitness after exposure to interferon-γ (IFNγ). In addition, we identify the autophagy pathway as a conserved mediator of the evasion of CTLs by cancer cells, and show that this pathway is required to resist cytotoxicity induced by the cytokines IFNγ and TNF. Through the mapping of cytokine- and CTL-based genetic interactions, together with in vivo CRISPR screens, we show how the pleiotropic effects of autophagy control cancer-cell-intrinsic evasion of killing by CTLs and we highlight the importance of these effects within the tumour microenvironment. Collectively, these data expand our knowledge of the genetic circuits that are involved in the evasion of the immune system by cancer cells, and highlight genetic interactions that contribute to phenotypes associated with escape from killing by CTLs.
Collapse
Affiliation(s)
- Keith A Lawson
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.,Division of Urology, Department of Surgery, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | | | - Xiaoyu Zhang
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Eiru Kim
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rummy Akthar
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | - Yuxi Yao
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | - Catherine Ross
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Kevin R Brown
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Abdelrahman Abou Zid
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada.,Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | | | - Shirley Hui
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | - Lujia Tang
- Agios Pharmaceuticals, Cambridge, MA, USA
| | | | | | | | - Qian Huang
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | | | - Andrea Habsid
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Ryan Climie
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - David Tieu
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Jiarun Wei
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Ren Li
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Amy Hin Yan Tong
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Michael Aregger
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Katherine S Chan
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Hong Han
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Xiaowei Wang
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Patricia Mero
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - John H Brumell
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.,Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Antonio Finelli
- Division of Urology, Department of Surgery, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Laurie Ailles
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Gary Bader
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Gromoslaw A Smolen
- Agios Pharmaceuticals, Cambridge, MA, USA.,Celsius Therapeutics, Cambridge, MA, USA
| | | | - Traver Hart
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Jason Moffat
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada. .,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada. .,Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
124
|
Huo R, Chu Q, Zhao X, Liu X, Xu T. Molecular evolution and functional characterization of SOCS3a and SOCS3b in miiuy croaker (Miichthys miiuy). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 110:103723. [PMID: 32387555 DOI: 10.1016/j.dci.2020.103723] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 04/26/2020] [Accepted: 04/26/2020] [Indexed: 06/11/2023]
Abstract
The suppressor of cytokine signaling 3 (SOCS3), as a negative regulator in inferferon (IFN) signaling pathways in mammals, has a vital role in immune systems. However, studies on the function of SOCS3 in lower vertebrates are limited. In this study, we identified SOCS3a and fish-specific SOCS3b gene in miiuy croaker. Sequence analysis results showed that SOCS3a and SOCS3b were evolutionarily conservative in fish. Expression analysis indicated that miiuy croaker SOCS3a and SOCS3b (mmSOCS3a and mmSOCS3b) were expressed in all of the tested miiuy croaker tissues, thus revealing the potential ability to perceive poly (I:C) stimulation. Further functional experiments showed that mmSOCS3a and mmSOCS3b could inhibit the IFNγ- and IFNα-induced ISRE reporter activation, respectively. Accordingly, the investigation of mmSOCS3a and mmSOCS3b can provide insights into fish SOCS3 and a basis for future research on the SOCS family of fish immune systems.
Collapse
Affiliation(s)
- Ruixuan Huo
- Laboratory of Fish Biogenetics & Immune Evolution, College of Marine Science, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Qing Chu
- Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266200, China; Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Xueyan Zhao
- Laboratory of Fish Biogenetics & Immune Evolution, College of Marine Science, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Xuezhu Liu
- Laboratory of Fish Biogenetics & Immune Evolution, College of Marine Science, Zhejiang Ocean University, Zhoushan, 316022, China.
| | - Tianjun Xu
- Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266200, China; Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China.
| |
Collapse
|
125
|
Cordes F, Foell D, Ding JN, Varga G, Bettenworth D. Differential regulation of JAK/STAT-signaling in patients with ulcerative colitis and Crohn’s disease. World J Gastroenterol 2020; 26:4055-4075. [PMID: 32821070 PMCID: PMC7403801 DOI: 10.3748/wjg.v26.i28.4055] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/24/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
In 2018, the pan-Janus kinase (JAK) inhibitor tofacitinib was launched for the treatment of ulcerative colitis (UC). Although tofacitinib has proven efficacious in patients with active UC, it failed in patients with Crohn’s disease (CD). This finding strongly hints at a different contribution of JAK signaling in both entities. Here, we review the current knowledge on the interplay between the JAK/signal transducer and activator of transcription (STAT) pathway and inflammatory bowel diseases (IBD). In particular, we provide a detailed overview of the differences and similarities of JAK/STAT-signaling in UC and CD, highlight the impact of the JAK/STAT pathway in experimental colitis models and summarize the published evidence on JAK/STAT-signaling in immune cells of IBD as well as the genetic association between the JAK/STAT pathway and IBD. Finally, we describe novel treatment strategies targeting JAK/STAT inhibition in UC and CD and comment on the limitations and challenges of the new drug class.
Collapse
Affiliation(s)
- Friederike Cordes
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster D-48149, Germany
| | - Dirk Foell
- Department of Pediatric Rheumatology and Immunology, University Children’s Hospital Münster, Münster D-48149, Germany
| | - John Nik Ding
- Department of Gastroenterology, St. Vincent’s Hospital, Melbourne 3002, Australia
- Department of Medicine, University of Melbourne, East Melbourne 3002, Australia
| | - Georg Varga
- Department of Pediatric Rheumatology and Immunology, University Children’s Hospital Münster, Münster D-48149, Germany
| | - Dominik Bettenworth
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster D-48149, Germany
| |
Collapse
|
126
|
Metcalfe RD, Putoczki TL, Griffin MDW. Structural Understanding of Interleukin 6 Family Cytokine Signaling and Targeted Therapies: Focus on Interleukin 11. Front Immunol 2020; 11:1424. [PMID: 32765502 PMCID: PMC7378365 DOI: 10.3389/fimmu.2020.01424] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022] Open
Abstract
Cytokines are small signaling proteins that have central roles in inflammation and cell survival. In the half-century since the discovery of the first cytokines, the interferons, over fifty cytokines have been identified. Amongst these is interleukin (IL)-6, the first and prototypical member of the IL-6 family of cytokines, nearly all of which utilize the common signaling receptor, gp130. In the last decade, there have been numerous advances in our understanding of the structural mechanisms of IL-6 family signaling, particularly for IL-6 itself. However, our understanding of the detailed structural mechanisms underlying signaling by most IL-6 family members remains limited. With the emergence of new roles for IL-6 family cytokines in disease and, in particular, roles of IL-11 in cardiovascular disease, lung disease, and cancer, there is an emerging need to develop therapeutics that can progress to clinical use. Here we outline our current knowledge of the structural mechanism of signaling by the IL-6 family of cytokines. We discuss how this knowledge allows us to understand the mechanism of action of currently available inhibitors targeting IL-6 family cytokine signaling, and most importantly how it allows for improved opportunities to pharmacologically disrupt cytokine signaling. We focus specifically on the need to develop and understand inhibitors that disrupt IL-11 signaling.
Collapse
Affiliation(s)
- Riley D Metcalfe
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Technology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Tracy L Putoczki
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Michael D W Griffin
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Technology Institute, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
127
|
Yoshizumi T, Kubo A, Murata H, Shinonaga M, Kanno H. BC-Box Motif in SOCS6 Induces Differentiation of Epidermal Stem Cells into GABAnergic Neurons. Int J Mol Sci 2020; 21:ijms21144947. [PMID: 32668737 PMCID: PMC7403999 DOI: 10.3390/ijms21144947] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/09/2020] [Accepted: 07/11/2020] [Indexed: 12/11/2022] Open
Abstract
The BC-box motif in suppressor of cytokine signaling 6 (SOCS6) promotes the neuronal differentiation of somatic stem cells, including epidermal stem cells. SOCS6 protein belongs to the group of SOCS proteins and inhibits cytokine signaling. Here we showed that epidermal stem cells were induced to differentiate into GABAnergic neurons by the intracellular delivery of a peptide composed of the amino-acid sequences encoded by the BC-box motif in SOCS6 protein. The BC-box motif (SLQYLCRFVI) in SOCS6 corresponded to the binding site of elongin BC. GABAnergic differentiation mediated by the BC-box motif in SOCS6 protein was caused by ubiquitination of JAK2 and inhibition of the JAK2-STAT3 pathway. Furthermore, GABAnergic neuron-like cells generated from epidermal stem cells were transplanted into the brain of a rodent ischemic model. Then, we demonstrated that these transplanted cells were GAD positive and that the cognitive function of the ischemic model rodents with the transplanted cells was improved. This study could contribute to not only elucidating the mechanism of GABAnergic neuronal differentiation but also to neuronal regenerative medicine utilizing GABAnergic neurons.
Collapse
Affiliation(s)
- Tetsuya Yoshizumi
- Department of Neurosurgery, International University of Health and Welfare Atami Hospital, Atami 413-0012, Japan; (T.Y.); (M.S.)
| | - Atsuhiko Kubo
- Nerve Care Clinic, Yokosuka 238-0012, Japan;
- Department of Neurosurgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan;
| | - Hidetoshi Murata
- Department of Neurosurgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan;
| | - Masamichi Shinonaga
- Department of Neurosurgery, International University of Health and Welfare Atami Hospital, Atami 413-0012, Japan; (T.Y.); (M.S.)
| | - Hiroshi Kanno
- Department of Neurosurgery, International University of Health and Welfare Atami Hospital, Atami 413-0012, Japan; (T.Y.); (M.S.)
- Department of Neurosurgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan;
- Correspondence: ; Tel.: +81-557-81-9171; Fax: +81-557-83-6632
| |
Collapse
|
128
|
Ye M, Joosse ME, Liu L, Sun Y, Dong Y, Cai C, Song Z, Zhang J, Brant SR, Lazarev M, Li X. Deletion of IL-6 Exacerbates Colitis and Induces Systemic Inflammation in IL-10-Deficient Mice. J Crohns Colitis 2020; 14:831-840. [PMID: 31679013 PMCID: PMC7346894 DOI: 10.1093/ecco-jcc/jjz176] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Interleukin 6 [IL-6] or its receptor is currently a candidate for targeted biological therapy of inflammatory bowel disease [IBD]. Thus, a comprehensive understanding of the consequences of blocking IL-6 is imperative. We investigated this by evaluating the effects of IL-6 deletion on the spontaneous colitis of IL-10-deficient mice [IL-10-/-]. METHODS IL-6/IL-10 double-deficient mice [IL-6-/-/IL-10-/-] were generated and analysed for intestinal inflammation, general phenotypes and molecular/biochemical changes in the colonic mucosa compared with wild-type and IL-10-/- mice. RESULTS Unexpectedly, the IL-6-/-/IL-10-/- mice showed more pronounced gut inflammation and earlier disease onset than IL-10-/- mice, both locally [colon and small bowel] and systemically [splenomegaly, ulcerative dermatitis, leukocytosis, neutrophilia and monocytosis]. IL-6-/-/IL-10-/- mice exhibited elevations of multiple cytokines [IL-1β, IL-4, IL-12, TNFα] and chemokines [MCP-1 and MIG], but not IFN-γ [Th1], IL-17A and IL-17G [Th17], or IL-22 [Th22]. FOXP3 and TGF-β, two key factors for regulatory T [Treg] cell differentiation, were significantly down-regulated in the colonic mucosa, but not in the thymus or mesenteric lymph nodes, of IL-6-/-/IL-10-/- mice. CTLA-4 was diminished while iNOS was up-regulated in the colonic mucosa of IL-6-/-/IL-10-/- mice. CONCLUSION In IL-10-/- mice, complete IL-6 blockade significantly aggravates gut inflammation, at least in part by suppressing Treg/CTLA-4 and promoting the IL-1β/Th2 pathway. In addition, the double mutant exhibits signs of severe systemic inflammation. Our data define a new function of IL-6 and suggest that caution should be exercised when targeting IL-6 in IBD patients, particularly those with defects in IL-10 signalling.
Collapse
Affiliation(s)
- Mei Ye
- Division of Gastroenterology, Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, USA,Department of Gastroenterology, Zhongnan Hospital, Wuhan University, Hubei Province, China
| | - Maria E Joosse
- Division of Gastroenterology, Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Ling Liu
- Division of Gastroenterology, Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, USA,Department of Gastroenterology, West China Hospital, Sichuan University, Sichuan Province, China
| | - Yu Sun
- Division of Gastroenterology, Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Ying Dong
- Division of Gastroenterology, Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Changchun Cai
- Division of Gastroenterology, Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Zhenmei Song
- Division of Gastroenterology, Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Jennifer Zhang
- Division of Gastroenterology, Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Steven R Brant
- Division of Gastroenterology, Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Mark Lazarev
- Division of Gastroenterology, Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Xuhang Li
- Division of Gastroenterology, Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, USA,Corresponding author: Xuhang Li, PhD, Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, 720 Rutland Ave., Ross 918, Baltimore, MD 21205, USA.
| |
Collapse
|
129
|
MacLeod G, Bozek DA, Rajakulendran N, Monteiro V, Ahmadi M, Steinhart Z, Kushida MM, Yu H, Coutinho FJ, Cavalli FMG, Restall I, Hao X, Hart T, Luchman HA, Weiss S, Dirks PB, Angers S. Genome-Wide CRISPR-Cas9 Screens Expose Genetic Vulnerabilities and Mechanisms of Temozolomide Sensitivity in Glioblastoma Stem Cells. Cell Rep 2020; 27:971-986.e9. [PMID: 30995489 DOI: 10.1016/j.celrep.2019.03.047] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 12/19/2018] [Accepted: 03/13/2019] [Indexed: 01/14/2023] Open
Abstract
Glioblastoma therapies have remained elusive due to limitations in understanding mechanisms of growth and survival of the tumorigenic population. Using CRISPR-Cas9 approaches in patient-derived GBM stem cells (GSCs) to interrogate function of the coding genome, we identify actionable pathways responsible for growth, which reveal the gene-essential circuitry of GBM stemness and proliferation. In particular, we characterize members of the SOX transcription factor family, SOCS3, USP8, and DOT1L, and protein ufmylation as important for GSC growth. Additionally, we reveal mechanisms of temozolomide resistance that could lead to combination strategies. By reaching beyond static genome analysis of bulk tumors, with a genome-wide functional approach, we reveal genetic dependencies within a broad range of biological processes to provide increased understanding of GBM growth and treatment resistance.
Collapse
Affiliation(s)
- Graham MacLeod
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Danielle A Bozek
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Vernon Monteiro
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Moloud Ahmadi
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Zachary Steinhart
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Michelle M Kushida
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Helen Yu
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Fiona J Coutinho
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Florence M G Cavalli
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ian Restall
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Xiaoguang Hao
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Traver Hart
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - H Artee Luchman
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Samuel Weiss
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Peter B Dirks
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, Department of Laboratory Medicine and Pathobiology, Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Division of Neurosurgery, The Hospital for Sick Children, Toronto, ON, Canada.
| | - Stephane Angers
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada; Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
130
|
Aziz SGG, Aziz SGG, Khabbazi A, Alipour S. The methylation status of TNF-α and SOCS3 promoters and the regulation of these gene expressions in patients with Behçet's disease. Biomarkers 2020; 25:384-390. [PMID: 32475174 DOI: 10.1080/1354750x.2020.1754912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Introduction: The aim of this study was to evaluate the methylation status of TNF-α and SOCS3 promoters in patients with BD and compare them with a healthy group.Method: This was a case-control study, in which 47 subjects with BD and 61 individuals as the control participated. Blood samples were collected from all the participants. Then, PBMCs were isolated using the Ficoll method and methylation of considered sites was investigated using the qMS-PCR technique after DNA extraction by the rapid genomic DNA extraction method and its analysis with Nano-drop.Results: The methylation and expression of TNF-α showed that the methylation level significantly declined in the patient in comparison with the healthy (p < 0.05). Moreover, the results on the mean expression showed that it significantly increased in the patient group, as compared with the healthy group (p < 0.05). In addition, the expression of the SOCS3 gene was not significantly different between the patients and healthy subjects while the level of SOCS3 methylation was significantly higher in the patient group than that in the healthy group (p < 0.05).Discussion: The present study revealed that the gene expression of TNF-alpha increased in BD patients, suggesting that TNF-alpha likely has a role in the pathogenesis of BD.
Collapse
Affiliation(s)
| | - Sara Gholizadeh-Ghaleh Aziz
- Department of Food Science and Technology, College of Agriculture, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Alireza Khabbazi
- Connective Tissue Disease, Tabriz University of Medical Science, Tabriz, Iran
| | - Shahriar Alipour
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
131
|
Liu H, Tang F, Su J, Ma J, Qin Y, Ji L, Geng H, Wang S, Zhang P, Liu J, Cui S, Ge RL, Li Z. EPAS1 regulates proliferation of erythroblasts in chronic mountain sickness. Blood Cells Mol Dis 2020; 84:102446. [PMID: 32470757 DOI: 10.1016/j.bcmd.2020.102446] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 01/13/2023]
Abstract
Excessive erythrocytosis (EE) is a characteristic of chronic mountain sickness (CMS). Currently, the pathogenesis of CMS remains unclear. This study was intended to investigate the role of EPAS1 in the proliferation of erythroblasts in CMS. Changes of HIF-1α and EPAS1/HIF-2α in the bone marrow erythroblasts of 21 patients with CMS and 14 control subjects residing at the same altitudes were determined by RT-qPCR and western blotting. We also developed a lentiviral vector, Lv-EPAS1/sh-EPAS1, to over-express/silence EPAS1 in K562 cells. Cells cycle and proliferation were detected by flow cytometry. Transcriptome analyses were carried out on Illumina. CMS patients showed a higher expression of EPAS1/HIF-2α in the bone marrow erythroblasts than those of controls. Variations in EPAS1 expression in CMS patients were positively correlated with RBC levels, and negatively correlated with SaO2. Over-expressing of EPAS1 in K562 cells accelerated the erythroid cells cycle progression and promoted the erythroid cells proliferation-and vice versa. Transcriptome data indicated that proliferation-related DEGs were significantly enriched in EPAS1 overexpression/silencing K562 cells. Our results suggest that EPAS1 might participate in the pathogenesis of EE by regulating the proliferation of erythroblasts.
Collapse
Affiliation(s)
- Huihui Liu
- Research Center for High Altitude Medicine, Qinghai University, Xining, China; Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, Xining, China; Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Xining, China; Department of Rheumatology, Affiliated Hospital of Qinghai University, Xining, China
| | - Feng Tang
- Research Center for High Altitude Medicine, Qinghai University, Xining, China; Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, Xining, China; Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Xining, China
| | - Juan Su
- Department of Rheumatology, Affiliated Hospital of Qinghai University, Xining, China
| | - Jie Ma
- Department of Hematology, Affiliated Hospital of Qinghai University, Xining, China
| | - Yajing Qin
- Department of Rheumatology, Affiliated Hospital of Qinghai University, Xining, China
| | - Linhua Ji
- Department of Hematology, Affiliated Hospital of Qinghai University, Xining, China
| | - Hui Geng
- Department of Rheumatology, Affiliated Hospital of Qinghai University, Xining, China
| | - Shengyan Wang
- Research Center for High Altitude Medicine, Qinghai University, Xining, China; Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, Xining, China; Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Xining, China
| | - Peili Zhang
- Research Center for High Altitude Medicine, Qinghai University, Xining, China; Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, Xining, China; Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Xining, China
| | - Junli Liu
- Research Center for High Altitude Medicine, Qinghai University, Xining, China; Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, Xining, China; Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Xining, China
| | - Sen Cui
- Department of Hematology, Affiliated Hospital of Qinghai University, Xining, China
| | - Ri-Li Ge
- Research Center for High Altitude Medicine, Qinghai University, Xining, China; Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, Xining, China; Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Xining, China
| | - Zhanquan Li
- Department of Rheumatology, Affiliated Hospital of Qinghai University, Xining, China.
| |
Collapse
|
132
|
Delen E, Doğanlar O, Delen Ö, Doğanlar ZB, Kılınçer C. The Role of JAK-STAT Signaling Activation in Hypertrophied Ligamentum Flavum. World Neurosurg 2020; 137:e506-e516. [PMID: 32059970 DOI: 10.1016/j.wneu.2020.02.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Although previous studies have reported the expression of JAK1, STAT3, and phosphorylated STAT3 in hypertrophied ligamentum flavum (LF), the role of the Janus kinase-signal transducer and activator of transcription (JAK/STAT) signaling pathway in hypertrophied LF has not been fully elucidated. The aim of this study was to identify the important JAK/STAT gene expression patterns of the 3 main receptors involved in this pathway: interferon (IFN)-γ receptor (IFN-γR), IFN-α receptor (IFNAR), and interleukin (IL)-6 receptor (IL-6R). METHODS The human LF specimens were obtained from 28 patients who underwent lumbar spine surgery for either degenerative lumbar canal stenosis (DLCS) (n = 28) or lumbar disc herniation (LDH) (n = 20). In this design, patients with LDH served as the control group. The degree of fibrosis was demonstrated by Masson's trichrome staining. The location and expression profiling of the JAK/STAT pathway were analyzed by quantitative real-time polymerase chain reaction and Western blotting. The thickness of the LF was measured with axial T1-weighted magnetic resonance imaging. RESULTS The most severe fibrotic changes were on the dorsal side of the LF. IL-6 and IFN-I expression levels were significantly increased on the dorsal side of the LF. While expression levels of IL-6R and IFNAR on the dural and dorsal side were significantly higher in the DLCS samples, IFN-γR and endothelial epidermal growth factor receptor in LF samples showed a significant increase only on the dorsal side. JAK/STAT genes were significantly expressed, especially on the dorsal side. CONCLUSIONS Our data suggest that IFNAR- and IL-6R-dependent JAK/STAT signaling pathways may be significant targets in drug development strategies for the treatment of LF hypertrophy.
Collapse
Affiliation(s)
- Emre Delen
- Department of Neurosurgery, Trakya University School of Medicine, Edirne, Turkey.
| | - Oğuzhan Doğanlar
- Department of Medical Biology, Trakya University School of Medicine, Edirne, Turkey
| | - Özlem Delen
- Department of Histology and Embryology, Trakya University School of Medicine, Edirne, Turkey
| | - Zeynep Banu Doğanlar
- Department of Medical Biology, Trakya University School of Medicine, Edirne, Turkey
| | - Cumhur Kılınçer
- Department of Neurosurgery, Trakya University School of Medicine, Edirne, Turkey
| |
Collapse
|
133
|
SOCS3 Attenuates GM-CSF/IFN-γ-Mediated Inflammation During Spontaneous Spinal Cord Regeneration. Neurosci Bull 2020; 36:778-792. [PMID: 32306216 PMCID: PMC7340708 DOI: 10.1007/s12264-020-00493-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 12/06/2019] [Indexed: 12/13/2022] Open
Abstract
SOCS3, a feedback inhibitor of the JAK/STAT signal pathway, negatively regulates axonal regrowth and inflammation in the central nervous system (CNS). Here, we demonstrated a distinct role of SOCS3 in the injured spinal cord of the gecko following tail amputation. Severing the gecko spinal cord did not evoke an inflammatory cascade except for an injury-stimulated elevation of the granulocyte/macrophage colony-stimulating factor (GM-CSF) and interferon gamma (IFN-γ) cytokines. Simultaneously, the expression of SOCS3 was upregulated in microglia, and unexpectedly not in neurons. Enforced expression of SOCS3 was sufficient to suppress the GM-CSF/IFN-γ-driven inflammatory responses through its KIR domain by attenuating the activities of JAK1 and JAK2. SOCS3 was also linked to GM-CSF/IFN-γ-induced cross-tolerance. Transfection of adenovirus overexpressing SOCS3 in the injured cord resulted in a significant decrease of inflammatory cytokines. These results reveal a distinct role of SOCS3 in the regenerating spinal cord, and provide new hints for CNS repair in mammals.
Collapse
|
134
|
Cai M, Shi Y, Zheng T, Hu S, Du K, Ren A, Jia X, Chen S, Wang J, Lai S. Mammary epithelial cell derived exosomal MiR-221 mediates M1 macrophage polarization via SOCS1/STATs to promote inflammatory response. Int Immunopharmacol 2020; 83:106493. [PMID: 32289739 DOI: 10.1016/j.intimp.2020.106493] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/31/2020] [Accepted: 04/06/2020] [Indexed: 01/10/2023]
Abstract
Lactational mastitis seriously alters the normal physiological function of mammary gland and activates the innate immune. Mammary epithelial cells (MECs) secret cytokines and regulate the function of immune system. However, the mechanism MECs mediated crosstalk with immune cells, such as macrophages, during mastitis is unclear. In this study, mouse mammary epithelial cells (HC11), treated with Lipoteichoic acid (LTA), and macrophages (RAW264.7) were used to mimic intercellular communication. Our results showed that exosomal miR-221 level was up-regulated and reached the peak at 12 h after infected by LTA. The expression of miR-211, CD11b protein and TNF-α mRNA were upregulated and the expression of CD206 protein and Arg-1 mRNA were inhibited in RAW264.7 treated with exosomes. In addition, miR-221 mimics and inhibitors enhanced and depressed HC11-derived exosomal miR-221 level, respectively. After treatment of Exo(mimic) in RAW264.7, the expression of CD11b protein and TNF-α mRNA were up-regulated, the expression of CD206 and Arg-1 mRNA were down-regulated. Additionally, Exo(inhibitor) enhanced CD206 protein and Arg-1 mRNA levels and inhibited CD11b protein and TNF-α mRNA levels. Furthermore, SOCS1 was identified to be a target gene of miR-221 by using Luciferase assays. And western blot assays showed that the expression of p-STAT1 and p-STAT3 were elevated and repressed, respectively. Taken together, we suggest that exosomal miR-221 promotes polarization of M1 macrophages via SOCS1, STAT1 and STAT3. And we reveal a novel crosstalk signaling pathway between mammary epithelial cells and macrophages in the process of inflammation.
Collapse
Affiliation(s)
- Mingcheng Cai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; College of Landscape Architecture and Life Science/Institute of Special Plants, Chongqing University of Arts and Sciences, Yongchuan, Chongqing 402160, China
| | - Yu Shi
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Tianhao Zheng
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Kun Du
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Anyong Ren
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Xianbo Jia
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.
| | - Shiyi Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Jie Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Songjia Lai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.
| |
Collapse
|
135
|
Stephens LM, Varga SM. Function and Modulation of Type I Interferons during Respiratory Syncytial Virus Infection. Vaccines (Basel) 2020; 8:vaccines8020177. [PMID: 32290326 PMCID: PMC7349809 DOI: 10.3390/vaccines8020177] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/04/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory infections in infants and young children, accounting for an estimated 3 million hospitalizations annually worldwide. Despite the major health burden, there is currently no licensed RSV vaccine. RSV is recognized by a range of cellular receptors including both toll-like receptors (TLR) and retinoic acid-inducible gene-I-like receptors (RIG-I). This interaction initiates signaling through mitochondrial antiviral signaling (MAVS) and interferon regulatory factor (IRF) proteins, resulting in the induction of type I interferons (IFN). Early viral control is mediated by either IFN-α or IFN-β signaling through the IFN receptor (IFNAR), inducing the production of antiviral interferon-stimulating genes (ISGs). Type I IFNs also initiate the early production of proinflammatory cytokines including interleukin 6 (IL-6), tumor necrosis factor (TNF), and IFN-γ. Type I IFN levels correlate with age, and inadequate production may be a critical factor in facilitating the increased RSV disease severity observed in infants. Here, we review the current literature on the function of type I IFNs in RSV pathogenesis, as well as their involvement in the differential immune responses observed in infants and adults.
Collapse
Affiliation(s)
- Laura M. Stephens
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA;
| | - Steven M. Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA;
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
- Correspondence: ; Tel.: +1-319-335-7784
| |
Collapse
|
136
|
Xu Y, Wu Y, Xiong Y, Tao J, Pan T, Tan S, Gao G, Chen Y, Abbas N, Getachew A, Zhuang Y, You K, Yang F, Li YX. Ascorbate protects liver from metabolic disorder through inhibition of lipogenesis and suppressor of cytokine signaling 3 (SOCS3). Nutr Metab (Lond) 2020; 17:17. [PMID: 32158492 PMCID: PMC7057613 DOI: 10.1186/s12986-020-0431-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 01/20/2020] [Indexed: 12/28/2022] Open
Abstract
Background Fatty liver is a reversible status, but also an origin stage to develop to other metabolic syndromes, such as diabetes and heart disease that threatens public health worldwide. Ascorbate deficiency is reported to be correlated with increasing risks for metabolic syndromes, but whether ascorbate has a therapeutic effect is unknown. Here, we investigated if ascorbate treatment alone could work on protecting from the development of steatosis and mechanisms beyond. Methods Guinea pigs were fed with a chow diet or a high palm oil diet (HPD) respectively. HPD induced animals were administered different concentrations of ascorbate in different time intervals through water. Besides, hepatocyte-like cells derived from human embryonic stem cells and HepG2 cells were treated with palmitic acid (PA) to induce lipid accumulation for molecular mechanism study. Results We find that ascorbate rescues HPD and PA induced steatosis and insulin tolerance in vivo and in vitro. We demonstrate that ascorbate changes cellular lipid profiles via inhibits lipogenesis, and inhibits the expression of SOCS3 via STAT3, thus enhances insulin signal transduction. Overexpression of SOCS3 abolishes the ascorbate rescue effects on insulin signal and lipid accumulation in hepatic cells. Conclusions Ascorbate ameliorates hepatic steatosis and improves insulin sensitivity through inhibiting lipogenesis and SOCS3.
Collapse
Affiliation(s)
- Yingying Xu
- 1Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, 510000 China.,2Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou, 510000 China
| | - Yuhang Wu
- 1Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, 510000 China.,2Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou, 510000 China
| | - Yue Xiong
- 1Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, 510000 China.,2Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou, 510000 China.,3University of Chinese Academy of Sciences, Beijing, 100000 China
| | - Jiawang Tao
- 1Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, 510000 China.,2Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou, 510000 China.,3University of Chinese Academy of Sciences, Beijing, 100000 China
| | - Tingcai Pan
- 1Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, 510000 China.,2Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou, 510000 China.,3University of Chinese Academy of Sciences, Beijing, 100000 China
| | - Shenglin Tan
- 1Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, 510000 China.,2Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou, 510000 China.,3University of Chinese Academy of Sciences, Beijing, 100000 China
| | - Ge Gao
- 1Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, 510000 China.,2Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou, 510000 China
| | - Yan Chen
- 1Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, 510000 China.,2Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou, 510000 China.,4Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, GIBH, CAS, Guangzhou, 510000 China.,5Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
| | - Nasir Abbas
- 1Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, 510000 China.,2Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou, 510000 China.,3University of Chinese Academy of Sciences, Beijing, 100000 China
| | - Anteneh Getachew
- 1Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, 510000 China.,2Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou, 510000 China.,3University of Chinese Academy of Sciences, Beijing, 100000 China
| | - Yuanqi Zhuang
- 1Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, 510000 China.,2Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou, 510000 China
| | - Kai You
- 1Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, 510000 China.,2Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou, 510000 China
| | - Fan Yang
- 1Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, 510000 China.,2Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou, 510000 China.,4Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, GIBH, CAS, Guangzhou, 510000 China.,5Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
| | - Yin-Xiong Li
- 1Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, 510000 China.,2Guangdong Provincial Key Laboratory of Biocomputing, GIBH, CAS, Guangzhou, 510000 China.,4Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, GIBH, CAS, Guangzhou, 510000 China.,5Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
| |
Collapse
|
137
|
Abbas MN, Kausar S, Zhao E, Cui H. Suppressors of cytokine signaling proteins as modulators of development and innate immunity of insects. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 104:103561. [PMID: 31785267 DOI: 10.1016/j.dci.2019.103561] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 06/10/2023]
Abstract
The suppressors of cytokine signaling (SOCS) are a family of intracellular molecules. Many members of this family have been reported to be involved in various physiological processes in invertebrates and vertebrates (e.g., developmental process and immune response). The functions of SOCS molecules seem to remain conserved in animals throughout evolutionary history. The members of the SOCS family play vital roles in the physiological processes by regulating the extent and duration of signaling activities of both Janus Kinase-Signal Transducer and Activators of Transcription (JAK-STAT) and epidermal growth factor receptor (EGFR) pathways in vivo. So far, in different insect species, a variable number of SOCS and SOCS box domain-containing proteins have been identified. These proteins are categorized into different types based on their sequence diversification, leading to an alteration in structure and regulatory function. The biological roles of the many SOCS proteins have been established as a negative or positive regulator of the signaling pathways, as mentioned earlier. Here, we discussed the existing knowledge on the SOCS proteins and their involvement in different biological functions in insects, and future perspectives to further elucidate their physiological roles.
Collapse
Affiliation(s)
- Muhammad Nadeem Abbas
- State Key Laboratory of Silkworm Genome Biology, College of Biotechnology, Southwest University, Chongqing, 400715, China; Key Laboratory of Sericulture Biology and Genetic Breeding, Ministry of Agricultural and Rural Affairs, Southwest University, Chongqing, 400715, China; Medical Research Institute, Southwest University, Chongqing, 400715, China.
| | - Saima Kausar
- State Key Laboratory of Silkworm Genome Biology, College of Biotechnology, Southwest University, Chongqing, 400715, China; Key Laboratory of Sericulture Biology and Genetic Breeding, Ministry of Agricultural and Rural Affairs, Southwest University, Chongqing, 400715, China; Medical Research Institute, Southwest University, Chongqing, 400715, China.
| | - Erhu Zhao
- State Key Laboratory of Silkworm Genome Biology, College of Biotechnology, Southwest University, Chongqing, 400715, China; Key Laboratory of Sericulture Biology and Genetic Breeding, Ministry of Agricultural and Rural Affairs, Southwest University, Chongqing, 400715, China; Medical Research Institute, Southwest University, Chongqing, 400715, China.
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, College of Biotechnology, Southwest University, Chongqing, 400715, China; Key Laboratory of Sericulture Biology and Genetic Breeding, Ministry of Agricultural and Rural Affairs, Southwest University, Chongqing, 400715, China; Medical Research Institute, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
138
|
Arshad S, Naveed M, Ullia M, Javed K, Butt A, Khawar M, Amjad F. Targeting STAT-3 signaling pathway in cancer for development of novel drugs: Advancements and challenges. Genet Mol Biol 2020; 43:e20180160. [PMID: 32167126 PMCID: PMC7198026 DOI: 10.1590/1678-4685-gmb-2018-0160] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 10/20/2018] [Indexed: 12/25/2022] Open
Abstract
Signal transducers and activators of transcription 3 (STAT-3) is a transcription
factor that regulates the gene expression of several target genes. These factors
are activated by the binding of cytokines and growth factors with STAT-3
specific receptors on cell membrane. Few years ago, STAT-3 was considered an
acute phase response element having several cellular functions such as
inflammation, cell survival, invasion, metastasis and proliferation, genetic
alteration, and angiogenesis. STAT-3 is activated by several types of
inflammatory cytokines, carcinogens, viruses, growth factors, and oncogenes.
Thus, the STAT3 pathway is a potential target for cancer therapeutics. Abnormal
STAT-3 activity in tumor development and cellular transformation can be targeted
by several genomic and pharmacological methodologies. An extensive review of the
literature has been conducted to emphasize the role of STAT-3 as a unique cancer
drug target. This review article discusses in detail the wide range of STAT-3
inhibitors that show antitumor effects both in vitro and
in vivo. Thus, targeting constitutive STAT-3 signaling is a
remarkable therapeutic methodology for tumor progression. Finally, current
limitations, trials and future perspectives of STAT-3 inhibitors are also
critically discussed.
Collapse
Affiliation(s)
- Sundas Arshad
- University of Lahore, Department of Allied Health Sciences, Gujrat Campus, Pakistan
| | - Muhammad Naveed
- University of Central Punjab, Faculty of life sciences, Department of Biotechnology, Lahore, Pakistan
| | - Mahad Ullia
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Khadija Javed
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Ayesha Butt
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Masooma Khawar
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Fazeeha Amjad
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| |
Collapse
|
139
|
Antihyperglycemic Effects and Mode of Actions of Musa paradisiaca Leaf and Fruit Peel Hydroethanolic Extracts in Nicotinamide/Streptozotocin-Induced Diabetic Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:9276343. [PMID: 32047529 PMCID: PMC7007756 DOI: 10.1155/2020/9276343] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 12/27/2019] [Indexed: 02/06/2023]
Abstract
The present study aimed to evaluate the antihyperglycemic effects of Musa paradisiaca (M. paradisiaca) leaf and fruit peel hydroethanolic extracts and to suggest their probable mode of actions in nicotinamide (NA)/streptozotocin (STZ)-induced diabetic rats. The leaf and fruit peel hydroethanolic extracts were analyzed by GC-MS that indicated the presence of phytol, octadecatrienoic acid, hexadecanoic acid, and octadecadienoic acid as major components in the leaf extract and vitamin E, octadecenamide, β-sitosterol, and stigmasterol as major phytochemicals in the fruit peel extract. Diabetes mellitus was induced by a single intraperitoneal injection of STZ (60 mg/kg body weight) dissolved in citrate buffer (pH 4.5), 15 minutes after intraperitoneal injection of NA (120 mg/kg body weight). The NA/STZ-induced diabetic rats were, respectively, treated with M. paradisiaca leaf and fruit peel hydroethanolic extracts at a dose of 100 mg/kg body weight/day by oral administration for 28 days. The treatment of NA/STZ-induced diabetic rats with leaf and fruit peel extracts significantly improved the impaired oral glucose tolerance and significantly increased the lowered serum insulin and C-peptide levels. The HOMA-IR (as the index of insulin resistance) and QUICKI (as a marker for insulin sensitivity), as well as HOMA-β cell function were significantly alleviated as a result of treatment of diabetic rats with leaf and fruit peel extracts. In association, the elevated serum-free fatty acids, TNF-α, and IL-6 levels were significantly decreased. In addition, the suppressed adipose tissue PPARγ, GLUT4, adiponectin, and insulin receptor β-subunit mRNA expressions were upregulated while the elevated adipose tissue resistin expression was downregulated in diabetic rats as a result of treatment with the leaf and peel extract. Based on these results, it can be concluded that M. paradisiaca leaf and fruit peel hydroethanolic extracts have antihyperglycemic effects which may be mediated via their insulinotropic and insulin-sensitizing effects.
Collapse
|
140
|
Xin P, Xu X, Deng C, Liu S, Wang Y, Zhou X, Ma H, Wei D, Sun S. The role of JAK/STAT signaling pathway and its inhibitors in diseases. Int Immunopharmacol 2020; 80:106210. [PMID: 31972425 DOI: 10.1016/j.intimp.2020.106210] [Citation(s) in RCA: 466] [Impact Index Per Article: 116.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/19/2019] [Accepted: 01/08/2020] [Indexed: 02/09/2023]
Abstract
The JAK/STAT signaling pathway is an universally expressed intracellular signal transduction pathway and involved in many crucial biological processes, including cell proliferation, differentiation, apoptosis, and immune regulation. It provides a direct mechanism for extracellular factors-regulated gene expression. Current researches on this pathway have been focusing on the inflammatory and neoplastic diseases and related drug. The mechanism of JAK/STAT signaling is relatively simple. However, the biological consequences of the pathway are complicated due to its crosstalk with other signaling pathways. In addition, there is increasing evidence indicates that the persistent activation of JAK/STAT signaling pathway is closely related to many immune and inflammatory diseases, yet the specific mechanism remains unclear. Therefore, it is necessary to study the detailed mechanisms of JAK/STAT signaling in disease formation to provide critical reference for clinical treatments of the diseases. In this review, we focus on the structure of JAKs and STATs, the JAK/STAT signaling pathway and its negative regulators, the associated diseases, and the JAK inhibitors for the clinical therapy.
Collapse
Affiliation(s)
- Ping Xin
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Xiaoyun Xu
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Chengjie Deng
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Shuang Liu
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Youzhi Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xuegang Zhou
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Hongxing Ma
- Clinical Laboratory Department, Najing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Najing 211200, China
| | - Donghua Wei
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Shiqin Sun
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China.
| |
Collapse
|
141
|
Wolde M, Laan LC, Medhin G, Gadissa E, Berhe N, Tsegaye A. Human Monocytes/Macrophage Inflammatory Cytokine Changes Following in vivo and in vitro Schistomam manoni Infection. J Inflamm Res 2020; 13:35-43. [PMID: 32021377 PMCID: PMC6970607 DOI: 10.2147/jir.s233381] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/06/2019] [Indexed: 11/23/2022] Open
Abstract
Introduction Epidemiological and animal studies indicate that helminth infections have positive effects due to their potential to protect against autoimmune diseases. Here, we aim to assess the effect of S. mansoni infection on immune modulation of human monocytes and their potential protection against autoimmune disease development both in vivo and in vitro. Materials and Methods Monocytes were isolated from helminth-infected Ethiopians (MHIE), and from Dutch healthy volunteers (MHV). The MHV were stimulated in vitro with S. mansoni soluble egg antigens (SEA) or soluble worm antigens (SWA). In addition, phenotypical changes were studied directly, as well as after culturing for 6 days in the presence of human serum to obtain macrophages. Q-PCR, flow cytometry, multiplex bead immunoassay, and live-cell imaging were employed during analysis. Results MHIE showed elevated transcripts of SOCS-1 and TNF-α compared to MHV. Similarly, MHV that were stimulated with SEA demonstrated enhanced levels of SOCS-1, IL-10, and IL-12 mRNA, compared to control MHV. Remarkably, the SEA-treated monocytes showed a much higher motility than control monocytes, a hallmark of a patrolling phenotype. Furthermore, in vitro cultured macrophages that were stimulated by SEA exhibited enhanced mRNA levels of SOCS-1, IL-10, TNF-α, IL-12 and TGF-β, compared to control macrophages. Conclusion Macrophages from MHIE as well as SEA-treated MHV show an intermediate activation phenotype with both pro-inflammatory and anti-inflammatory characteristics in vitro. The observed pro-inflammatory properties might reflect a recent response of the cells due to contact with a pathogen, whereas the anti-inflammatory properties might contribute to helminth-induced protection against inflammatory diseases. Large-scale study is recommended to consolidate the findings of the present study. ![]()
Point your SmartPhone at the code above. If you have a QR code reader the video abstract will appear. Or use: https://youtu.be/SYOVExqwTRU
Collapse
Affiliation(s)
- Mistire Wolde
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia.,Department of Medical Laboratory Sciences, College of Health Science, Addis Ababa University, Addis Ababa, Ethiopia
| | - Lisa C Laan
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, the Netherlands
| | - Girmay Medhin
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| | | | - Nega Berhe
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia.,Oslo University Hospital-Ulleval, Centre for Imported and Tropical Diseases, Oslo, Norway
| | - Aster Tsegaye
- Department of Medical Laboratory Sciences, College of Health Science, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
142
|
Fruit and vegetable intake modifies the associations between suppressor of cytokine signaling 3 genetic variants and type 2 diabetes. Eur J Nutr 2020; 59:3441-3449. [PMID: 31927672 DOI: 10.1007/s00394-020-02178-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 01/03/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE Type 2 diabetes is a complex disease determined by variable genes and environmental factors. The study was designed to investigate the effect of interactions of four polymorphisms of suppressor of cytokine signaling 3 (SOCS3) with fruit and vegetable (F&V) intake on type 2 diabetes in a rural population of China. METHODS A total of 4411 participants from the rural areas of Henan, China were included in the study. Multivariate logistic regression and restricted cubic splines were used to estimate the associations between polymorphisms and risk allele score of SOCS3 and type 2 diabetes in different groups. Haplotype analysis was conducted to examine the effects of linkage inheritance at these four loci on type 2 diabetes. RESULTS Three of the four polymorphisms showed significant associations with type 2 diabetes in the less F&V intake group after adjusting the covariates, the odds ratios (ORs) and corresponding 95% confidence intervals (95% CIs) were 1.24 (1.08-1.41) for rs4969168, 1.16 (1.02-1.32) for rs9892622, and 1.21 (1.06-1.39) for rs9914220. No significant association was detected in the more F&V intake group. The obvious dose-response relationship between the risk allele score and type 2 diabetes was also noted only in the less F&V intake group. CONCLUSIONS Variants of SOCS3 gene were associated with type 2 diabetes and the associations could be modified by the F&V intake.
Collapse
|
143
|
Lee J, Cho CW, Jang M, Lim TG, Lee E, Hong HD, Rhee Y, Lee Y. Immunostimulatory activities of a high molecular weight fraction of Cynanchum auriculatum royle ex wight root obtained by ultrafiltration. Pharmacogn Mag 2020. [DOI: 10.4103/pm.pm_449_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
144
|
Qiu M, Mo L, Li J, Liang H, Zhu W, Zheng X, Duan X, Xu W. Effects of miR-150-5p on the growth and SOCS1 expression of rheumatoid arthritis synovial fibroblasts. Clin Rheumatol 2019; 39:909-917. [PMID: 31879859 DOI: 10.1007/s10067-019-04894-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 10/19/2019] [Accepted: 12/12/2019] [Indexed: 01/10/2023]
Abstract
OBJECTIVE miR-150-5p has been implicated in the regulation and onset of immune diseases. We investigated the effects of miR-150-5p on the functions of RA synovial fibroblasts (RASFs). METHOD The binding site between suppressor of cytokine signaling 1 (SOCS1) and miR-150-5p was analyzed using European Bioinformatics Institute database, and the 3' UTR of SOCS1 mRNA, including the binding site, was amplified and ligated to the 3'-end of LUC2 gene in the pmirGL0 dual-luciferase vector. The pmirGL0 vector and corresponding mimics were subsequently co-transfected into 293T cells to compare the relative fluorescence intensity of LUC2 between the miR-150-5p mimics and the negative control (NC) mimics groups. Further, the RASF cell line MH7A was transfected with miR-150-5p or NC mimics and subjected to flow cytometric analysis, cell counting kit-8 assay, western blot analysis, qPCR, and enzyme-linked immunosorbent (ELISA) assay 48 h after transfection. RESULTS miR-150-5p mimics resulted in a lower cell apoptotic rate and proportion of cells in the S phase. Using a dual-luciferase reporter gene assay, we then found that SOCS1 is a potential target of miR-150-5p. Compared with NC mimics, miR-150-5p mimics significantly decreased the protein and mRNA expression levels of SOCS1. ELISA assay showed that miR-150-5p mimics increased interleukin-6 level in the cell culture medium but did not influence tumor necrosis factor-alpha levels. CONCLUSIONS Overall, the growth-promoting effect of miR-150-5p on MH7A cells may be attributed to the miR-150-5p-induced degradation of SOCS1 mRNA, suggesting a potential therapeutic target for RA.Key Points• SOCS1 is a potential target of miR-150-5p.• miR-150-5p promoted the growth of RASF cell line MH7A.• miR-150-5p increased the secretion of IL-6 but did not significantly affect TNF-α levels in MH7A cells.
Collapse
Affiliation(s)
- Mingliang Qiu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- Department of Rheumatology, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China
| | - Lisha Mo
- Department of Rheumatology, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China
| | - Juxiang Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Hua Liang
- Department of Clinical Laboratory, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China
| | - Weina Zhu
- Department of Pediatrics, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China
| | - Xiangjuan Zheng
- Department of Chemistry, Nanchang University, Nanchang, 330031, China
| | - Xinwang Duan
- Department of Rheumatology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Weidong Xu
- Department of Rheumatology, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China.
| |
Collapse
|
145
|
Alluri K, Nair KPM, Ghosh S. Differential expression of zinc transporters in functionally contrasting tissues involved in zinc homeostasis. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2019; 39:615-629. [PMID: 31852371 DOI: 10.1080/15257770.2019.1670838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Zinc homeostasis is maintained by 24 tissue-specific zinc transporters which include ZnTs (ZnT1-10), ZIPs (ZIP1-14), in addition to metallothionein (MT). Current study aimed the role of zinc transporters in maintaining the basal levels of zinc in functionally contrasting tissue specific THP-1 (monocyte), RD (muscle), and Saos-2 (bone) cells. Zinc transporters expression was assessed by qRT-PCR. The mRNA levels of ZnTs (ZnT5-7 & ZnT9), ZIPs (ZIP6-10, ZIP13-14), and MT were significantly (p < 0.05) higher in Saos-2 compared to THP-1 and RD. The present study suggests that distinct expression pattern of zinc transporters and metallothionein might be responsible for the differential zinc assimilation.
Collapse
Affiliation(s)
- Kiran Alluri
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Krishna Pillay Madhavan Nair
- Micronutrient Research Group, ICMR- National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Sudip Ghosh
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| |
Collapse
|
146
|
Val CH, de Oliveira MC, Lacerda DR, Barroso A, Batista NV, Menezes-Garcia Z, de Assis DRR, Cramer AT, Brant F, Teixeira MM, Glória Souza D, Ferreira AM, Machado FS. SOCS2 modulates adipose tissue inflammation and expansion in mice. J Nutr Biochem 2019; 76:108304. [PMID: 31816561 DOI: 10.1016/j.jnutbio.2019.108304] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 11/06/2019] [Accepted: 11/12/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Obesity is usually triggered by a nutrient overload that favors adipocyte hypertrophy and increases the number of pro-inflammatory cells and mediators into adipose tissue. These mediators may be regulated by suppressors of cytokine signaling (SOCS), such as SOCS2, which is involved in the regulation of the inflammatory response of many diseases, but its role in obesity is not yet known. We aimed to investigate the role of SOCS2 in metabolic and inflammatory dysfunction induced by a high-refined carbohydrate-containing diet (HC). MATERIAL AND METHODS Male C57BL/6 wild type (WT) and SOCS2 deficient (SOCS2-/-) mice were fed chow or an HC diet for 8 weeks. RESULTS In general, SOCS2 deficient mice, independent of the diet, showed higher adipose tissue mass compared with their WT counterparts that were associated with decreased lipogenesis rate in adipose tissue, lipolysis in adipocyte culture and energy expenditure. An anti-inflammatory profile was observed in adipose tissue of SOCS2-/- by reduced secretion of cytokines, such as TNF and IL-6, and increased M2-like macrophages and regulatory T cells compared with WT mice. Also, SOCS2 deficiency reduced the differentiation/expansion of pro-inflammatory cells in the spleen but increased Th2 and Treg cells compared with their WT counterparts. CONCLUSION The SOCS2 protein is an important modulator of obesity that regulates the metabolic pathways related to adipocyte size. Additionally, SOCS2 is an inflammatory regulator that appears to be essential for controlling the release of cytokines and the differentiation/recruitment of cells into adipose tissue during the development of obesity.
Collapse
Affiliation(s)
- Cynthia Honorato Val
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Brazil
| | | | | | - Andreia Barroso
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Brazil
| | | | | | | | | | - Fátima Brant
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Brazil
| | | | | | | | - Fabiana Simão Machado
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Brazil.
| |
Collapse
|
147
|
De Feudis M, Walker GE, Genoni G, Manfredi M, Agosti E, Giordano M, Caputo M, Di Trapani L, Marengo E, Aimaretti G, Filigheddu N, Bellone S, Bona G, Prodam F. Identification of Haptoglobin as a Readout of rhGH Therapy in GH Deficiency. J Clin Endocrinol Metab 2019; 104:5263-5273. [PMID: 31215990 DOI: 10.1210/jc.2019-00562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/13/2019] [Indexed: 02/12/2023]
Abstract
BACKGROUND GH deficiency (GHD) is characterized by a cluster of cardiovascular risk factors and subtle inflammation. We aimed to demonstrate, through a proteomic approach, molecules directly modulated by GHD and involved in the inflammatory state. METHODS Ten children with isolated GHD were studied before and after 1 year of treatment with rhGH and compared with 14 matched controls. A two-dimensional electrophoresis plasma proteomics analysis was performed at baseline and after GH treatment to identify the top molecules modulated by GH. In vitro studies on human hepatoma (HepG2) cells were performed to validate the data. RESULTS Twelve of 20 proteomic spots were predicted to be isoforms α and β of haptoglobin (Hp) and confirmed by liquid chromatography tandem mass spectrometry and Western immunoblot analyses. Hp levels were higher in patients with GHD than controls at baseline (P < 0.001) and were reduced following GH treatment (P < 0.01). In HepG2 cells, both GH and IGF-1 were able to downregulate IL-6-induced Hp secretion. Moreover, Hp secretion was restored in pegvisomant-treated HepG2 cells. CONCLUSIONS Hp is a molecule acting in the inflammatory state of GHD and a possible biomarker for GH treatment. Nevertheless, the contribution of other factors and the molecular pathways involved in the GH downregulation of Hp remain to be clearly defined.
Collapse
Affiliation(s)
- Marilisa De Feudis
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | | | - Giulia Genoni
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Marcello Manfredi
- Interdisciplinary Research Center of Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
- Innovative Solutions and Advanced LED Imaging Techniques s.r.l., Spin-off of Department of Sciences and Technological Innovation, University of Piemonte Orientale, Alessandria, Italy
| | - Emanuela Agosti
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Mara Giordano
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Marina Caputo
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Luisa Di Trapani
- Clinical Biochemistry, Maggiore della Carità Hospital, Novara, Italy
| | - Emilio Marengo
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, Alessandria, Italy
| | - Gianluca Aimaretti
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Nicoletta Filigheddu
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Simonetta Bellone
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Gianni Bona
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Flavia Prodam
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
- Interdisciplinary Research Center of Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
148
|
Chen Y, Vandereyken M, Newton IP, Moraga I, Näthke IS, Swamy M. Loss of adenomatous polyposis coli function renders intestinal epithelial cells resistant to the cytokine IL-22. PLoS Biol 2019; 17:e3000540. [PMID: 31770366 PMCID: PMC6903767 DOI: 10.1371/journal.pbio.3000540] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 12/10/2019] [Accepted: 11/07/2019] [Indexed: 12/26/2022] Open
Abstract
Interleukin-22 (IL-22) is a critical immune defence cytokine that maintains intestinal homeostasis and promotes wound healing and tissue regeneration, which can support the growth of colorectal tumours. Mutations in the adenomatous polyposis coli gene (Apc) are a major driver of familial colorectal cancers (CRCs). How IL-22 contributes to APC-mediated tumorigenesis is poorly understood. To investigate IL-22 signalling in wild-type (WT) and APC-mutant cells, we performed RNA sequencing (RNAseq) of IL-22-treated murine small intestinal epithelial organoids. In WT epithelia, antimicrobial defence and cellular stress response pathways were most strongly induced by IL-22. Surprisingly, although IL-22 activates signal transducer and activator of transcription 3 (STAT3) in APC-mutant cells, STAT3 target genes were not induced. Our analyses revealed that ApcMin/Min cells are resistant to IL-22 due to reduced expression of the IL-22 receptor, and increased expression of inhibitors of STAT3, particularly histone deacetylases (HDACs). We further show that IL-22 increases DNA damage and genomic instability, which can accelerate cellular transition from heterozygosity (ApcMin/+) to homozygosity (ApcMin/Min) to drive tumour formation. Our data reveal an unexpected role for IL-22 in promoting early tumorigenesis while excluding a function for IL-22 in transformed epithelial cells.
Collapse
Affiliation(s)
- Yu Chen
- Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
- MRC Protein Phosphorylation and Ubiquitylation Unit (PPU), School of Life Sciences, University of Dundee, Dundee, United Kingdom
- Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Maud Vandereyken
- MRC Protein Phosphorylation and Ubiquitylation Unit (PPU), School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Ian P. Newton
- Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Ignacio Moraga
- Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Inke S. Näthke
- Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Mahima Swamy
- MRC Protein Phosphorylation and Ubiquitylation Unit (PPU), School of Life Sciences, University of Dundee, Dundee, United Kingdom
- Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
149
|
Estrada-Reyes ZM, Tsukahara Y, Amadeu RR, Goetsch AL, Gipson TA, Sahlu T, Puchala R, Wang Z, Hart SP, Mateescu RG. Signatures of selection for resistance to Haemonchus contortus in sheep and goats. BMC Genomics 2019; 20:735. [PMID: 31615414 PMCID: PMC6792194 DOI: 10.1186/s12864-019-6150-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 09/29/2019] [Indexed: 11/20/2022] Open
Abstract
Background Gastrointestinal nematode infection (GNI) is the most important disease affecting the small ruminant industry in U.S. The environmental conditions in the southern United States are ideal for the survival of the most pathogenic gastrointestinal nematode, Haemonchus contortus. Host genetic variation for resistance to H. contortus allows selective breeding for increased resistance of animals. This selection process increases the prevalence of particular alleles in sheep and goats and creates unique genetic patterns in the genome of these species. The aim of this study was to identify loci with divergent allelic frequencies in a candidate gene panel of 100 genes using two different approaches (frequentist and Bayesian) to estimate Fst outliers in three different breeds of sheep and goats exposed to H. contortus. Results Our results for sheep populations showed SNPs under selection in C3AR1, CSF3, SOCS2, NOS2, STAT5B, TGFB2 and IL2RA genes using frequentist and Bayesian approaches. For goats, SNPs in CD1D, ITGA9, IL12A, IL13RA1, CD86 and TGFB2 genes were under selection. Common signatures of selection in both species were observed in NOS2, TGFB2 and TLR4 genes. Directional selection was present in all SNPs evaluated in the present study. Conclusions A total of 13 SNPs within 7 genes of our candidate gene panel related to H. contortus exposure were identified under selection in sheep populations. For goats, 11 SNPs within 7 genes were identified under selection. Results from this study support the hypothesis that resistance to H. contortus is likely to be controlled by many loci. Shared signatures of selection related to mechanisms of immune protection against H. contortus infection in sheep and goats could be useful targets in breeding programs aimed to produce resistant animals with low FEC.
Collapse
Affiliation(s)
| | - Yoko Tsukahara
- American Institute for Goat Research, Langston University, Langston, OK, USA
| | - Rodrigo R Amadeu
- Horticultural Sciences Department, University of Florida, Gainesville, FL, USA
| | - Arthur L Goetsch
- American Institute for Goat Research, Langston University, Langston, OK, USA
| | - Terry A Gipson
- American Institute for Goat Research, Langston University, Langston, OK, USA
| | - Tilahun Sahlu
- American Institute for Goat Research, Langston University, Langston, OK, USA
| | - Richard Puchala
- American Institute for Goat Research, Langston University, Langston, OK, USA
| | - Zaisen Wang
- American Institute for Goat Research, Langston University, Langston, OK, USA
| | - Steve P Hart
- American Institute for Goat Research, Langston University, Langston, OK, USA
| | - Raluca G Mateescu
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA
| |
Collapse
|
150
|
Rojas-Sanchez G, Cotzomi-Ortega I, Pazos-Salazar NG, Reyes-Leyva J, Maycotte P. Autophagy and Its Relationship to Epithelial to Mesenchymal Transition: When Autophagy Inhibition for Cancer Therapy Turns Counterproductive. BIOLOGY 2019; 8:biology8040071. [PMID: 31554173 PMCID: PMC6956138 DOI: 10.3390/biology8040071] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/14/2019] [Accepted: 09/20/2019] [Indexed: 02/06/2023]
Abstract
The manipulation of autophagy for cancer therapy has gained recent interest in clinical settings. Although inhibition of autophagy is currently being used in clinical trials for the treatment of several malignancies, autophagy has been shown to have diverse implications for normal cell homeostasis, cancer cell survival, and signaling to cells in the tumor microenvironment. Among these implications and of relevance for cancer therapy, the autophagic process is known to be involved in the regulation of protein secretion, in tumor cell immunogenicity, and in the regulation of epithelial-to-mesenchymal transition (EMT), a critical step in the process of cancer cell invasion. In this work, we have reviewed recent evidence linking autophagy to the regulation of EMT in cancer and normal epithelial cells, and have discussed important implications for the manipulation of autophagy during cancer therapy.
Collapse
Affiliation(s)
- Guadalupe Rojas-Sanchez
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, Puebla 72570, Mexico.
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Km 4.5 Carretera Atlixco-Metepec HGZ5, Puebla 74360, Mexico.
| | - Israel Cotzomi-Ortega
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, Puebla 72570, Mexico.
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Km 4.5 Carretera Atlixco-Metepec HGZ5, Puebla 74360, Mexico.
| | - Nidia G Pazos-Salazar
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, Puebla 72570, Mexico.
| | - Julio Reyes-Leyva
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Km 4.5 Carretera Atlixco-Metepec HGZ5, Puebla 74360, Mexico.
| | - Paola Maycotte
- Consejo Nacional de Ciencia y Tecnología (CONACYT)-CIBIOR, IMSS, Puebla 74360, Mexico.
| |
Collapse
|