Suppression of Cytokine Signaling by SOCS3: Characterization of the Mode of Inhibition and the Basis of Its Specificity

Total Page:16

File Type:pdf, Size:1020Kb

Suppression of Cytokine Signaling by SOCS3: Characterization of the Mode of Inhibition and the Basis of Its Specificity View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Elsevier - Publisher Connector Immunity Article Suppression of Cytokine Signaling by SOCS3: Characterization of the Mode of Inhibition and the Basis of Its Specificity Jeffrey J. Babon,1,2,5,* Nadia J. Kershaw,1,3,5 James M. Murphy,1,2,5 Leila N. Varghese,1,2 Artem Laktyushin,1 Samuel N. Young,1 Isabelle S. Lucet,4 Raymond S. Norton,1,2,6 and Nicos A. Nicola1,2,* 1Walter and Eliza Hall Institute of Medical Research, 1G Royal Pde, Parkville, 3052, VIC, Australia 2The University of Melbourne, Royal Parade, Parkville, 3050, VIC, Australia 3Ludwig Institute for Cancer Research, Royal Pde, Parkville, 3050, VIC, Australia 4Monash University, Wellington Rd, Clayton, 3800, VIC, Australia 5These authors contributed equally to this work 6Present address: Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia *Correspondence: [email protected] (J.J.B.), [email protected] (N.A.N.) DOI 10.1016/j.immuni.2011.12.015 SUMMARY Genetic deletion of each individual JAK leads to various immunological and hematopoietic defects; however, aberrant Janus kinases (JAKs) are key effectors in controlling activation of JAKs can be likewise pathological. Three myelopro- immune responses and maintaining hematopoiesis. liferative disorders (polycythemia vera, essential thrombocythe- SOCS3 (suppressor of cytokine signaling-3) is a mia, and primary myelofibrosis) are caused by a single point major regulator of JAK signaling and here we investi- mutation in JAK2 (JAK2V617F)(James et al., 2005; Levine et al., gate the molecular basis of its mechanism of action. 2005) that renders the kinase constitutively active and results We found that SOCS3 bound and directly inhibited in cytokine-independent activation of JAK-based signaling path- ways. An even more severe phenotype results from activation of the catalytic domains of JAK1, JAK2, and TYK2 JAK by oncogenic fusion, for example TEL-JAK2, which has but not JAK3 via an evolutionarily conserved motif been studied because of its role in childhood T and B cell acute unique to JAKs. Mutation of this motif led to the lymphoblastic leukemia (Lacronique et al., 2000). formation of an active kinase that could not be In order to prevent aberrant proliferation, JAK activity is regu- inhibited by SOCS3. Surprisingly, we found that lated in a number of ways. The primary negative regulators of the SOCS3 simultaneously bound JAK and the cytokine JAKs are a family of proteins known as the suppressors of cyto- receptor to which it is attached, revealing how spec- kine signaling (SOCS) (Endo et al., 1997; Hilton et al., 1998; Naka ificity is generated in SOCS action and explaining et al., 1997; Starr et al., 1997) whose expression is induced by why SOCS3 inhibits only a subset of cytokines. JAK-STAT activation and they then inhibit the signaling cascade, Importantly, SOCS3 inhibited JAKs via a noncompet- creating a negative feedback loop. itive mechanism, making it a template for the devel- All eight SOCS proteins (SOCS1-7 and CIS) contain a central SH2 domain and a C-terminal SOCS box domain (Hilton et al., opment of specific and effective inhibitors to treat 1998), which interacts with elongins B and C and Cullin5 to cata- JAK-based immune and proliferative diseases. lyze the ubiquitination of bound signaling proteins (Babon et al., 2009; Kamizono et al., 2001; Zhang et al., 1999). Elegant studies INTRODUCTION performed by Yoshimura and colleagues (Sasaki et al., 1999; Yasukawa et al., 1999) showed that the two most potent Both hematopoiesis and the immune response are regulated suppressors of signaling, SOCS1 and SOCS3, contain a short by the action of cytokines through activation of the Janus motif, upstream of their SH2 domain, known as the KIR (kinase kinase-signal transducer and activator of transcription-sup- inhibitory region), which allows them to suppress signaling by pressor of cytokine signaling (JAK-STAT-SOCS) signal trans- direct inhibition of JAK catalytic activity. This is their dominant duction pathway (O’Shea and Murray, 2008). mode of action in vivo (Boyle et al., 2007; Zhang et al., 2001). There are four mammalian JAKs (JAK1-3 and TYK2), each Initial characterization of the KIR noted its amino acid sequence consisting of four domains (Figure S1 available online; Wilks similarity to the activation loop of JAKs (Sasaki et al., 1999; and Harpur, 1994). The N-terminal FERM domain binds constitu- Yasukawa et al., 1999). Like most tyrosine kinases, JAKs contain tively to the appropriate membrane-bound receptor whereas the an activation loop that blocks the catalytic cleft. Autophosphor- C-terminal kinase (catalytic) domain phosphorylates substrate ylation of this loop causes its translocation away from the cata- proteins. Between these are a noncanonical SH2 domain and lytic site and allows substrate access, thus activating the kinase. a pseudokinase domain, the most distinctive feature of the Consequently, it was proposed that the SH2 domain of SOCS1 JAK family. This domain has recently been shown to be catalyt- and SOCS3 binds the activation loop tyrosine phosphate and ically active (Ungureanu et al., 2011) and it regulates the activity the KIR acts as a pseudosubstrate to block the active site (Sasaki of the JH1 domain (Saharinen et al., 2000). et al., 1999; Yasukawa et al., 1999). Immunity 36, 239–250, February 24, 2012 ª2012 Elsevier Inc. 239 Immunity Mechanism of Inhibition of Janus Kinase by SOCS3 Despite the ability of SOCS proteins to bind to and inhibit to be a good substrate for JAK2JH1 phosphorylation. To verify JAKs, deletion of individual SOCS genes in mice has revealed that phosphorylation of SOCS3 was not by itself the cause of an exquisite specificity for particular cytokine-receptor combina- decreased gp130cyt phosphorylation, the entire reaction was tions rather than specific JAKs. For example, SOCS1 inhibits spotted onto nitrocellulose membranes, allowing total phos- interferon g signaling without affecting IL-6 signaling whereas phorylation of all components to be quantified. SOCS3 had the converse is true for SOCS3 (Alexander et al., 1999; Croker a clearly titratable inhibitory effect on JAK-catalyzed phosphor- et al., 2003), yet both cytokine receptor systems utilize the ylation with an IC50 of 1 mM(Figure 1C). same JAKs (JAK1 and JAK2) (Murray, 2007). Moreover, the A limiting feature of these assays was that the concentration of binding affinities of the SOCS3 SH2 domain for phosphorylated SOCS3 required to inhibit JAK2JH1 was similar to the concentra- JAK peptides is several logs lower than that for certain cytokine tion of substrate. To ensure that it was not a SOCS3-substrate receptor phosphopeptides, and this binding is important in intact interaction that was responsible for inhibiting the phosphoryla- cells (Nicholson et al., 2000). tion reaction, we adopted a more robust enzyme-inhibition assay In this study we dissect both the mechanism and specificity of format where [Substrate] > > [Inhibitor] > > [Enzyme]. These JAK inhibition by SOCS3 by using biochemical, structural, and assays used high concentrations of a peptide substrate, resi- kinetic methods and resolve these apparent discrepancies. dues 693–708 of STAT5b (Saharinen et al., 2003; Zhao et al., We show that SOCS3 directly inhibits JAK1, JAK2, and TYK2 2010). SOCS3 inhibited phosphorylation of this peptide but not JAK3 because of the presence of a conserved three- substrate with the same IC50 of 1 mM(Figure 1D). These results residue motif on the former three JAK family members. By indicate that SOCS3 functions by blocking the ability of JAK2 to utilizing two distinct binding surfaces, SOCS3 is able to bind to phosphorylate protein substrates and is therefore a direct inhib- JAK and the cytokine receptor to which it is attached simulta- itor of its catalytic activity. neously, explaining why SOCS3 is specific for cytokines that signal through particular receptors. Intriguingly, inhibition occurs A SOCS1-SOCS3 Chimera Is a More Potent Inhibitor via a mechanism in which SOCS3 does not compete with either than SOCS3 ATP or substrate. This makes SOCS3 a noncompetitive tyrosine Replacing the KIR of SOCS3 with the corresponding region from kinase inhibitor and a new template for the future development SOCS1 resulted in a chimeric construct (termed SOCS1-3) that of a class of small-molecule JAK inhibitors with distinct advan- inhibited JAK2 kinase activity with higher affinity than did wild- tages over the current ATP analogs used to treat JAK-based type SOCS3 (IC50 values of 0.15 ± .02 and 1.2 ± 0.1 mM, respec- diseases. tively; see Figure 1D), despite there being a difference of only four residues. A truncated construct of SOCS3, SOCS322-185, RESULTS that lacks the SOCS box and hence elonginB and C binding, was equally effective at inhibiting JAK2. These results show SOCS3 Inhibits Substrate Phosphorylation by the Kinase that the kinase inhibitory activity of SOCS3 relies solely upon Domain of JAK2 its KIR and extended SH2 domain and not its SOCS box domain. To examine SOCS3 inhibition of JAK kinase activity, we devel- In addition, the KIR from SOCS1, but not SOCS4 or SOCS5, can oped an in vitro kinase assay consisting of three purified, act as a functional replacement, making it likely that SOCS1 acts recombinant components: enzyme (JAK2 catalytic domain, in a similar manner to SOCS3. JAK2JH1), substrate (gp130 cytoplasmic domain, gp130cyt), and inhibitor (SOCS3). Various chimeras of SOCS3 were also Both the KIR and SH2 Domain of SOCS3 Are Required for produced that had the key residues in the KIR, L22-S29, Kinase Inhibition but Phosphotyrosine Binding Is Not replaced by the corresponding residues of SOCS1, SOCS4, Previous work had highlighted the importance of F25A within the or SOCS5 (these are designated SOCS1-3, SOCS4-3, and KIR and R71 within the SH2 domain for SOCS3 function (Nichol- SOCS5-3; see Figure S1A).
Recommended publications
  • Quantitative Modelling Explains Distinct STAT1 and STAT3
    bioRxiv preprint doi: https://doi.org/10.1101/425868; this version posted September 24, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Title Quantitative modelling explains distinct STAT1 and STAT3 activation dynamics in response to both IFNγ and IL-10 stimuli and predicts emergence of reciprocal signalling at the level of single cells. 1,2, 3 1 1 1 1 1 2 Sarma U , Maitreye M , Bhadange S , Nair A , Srivastava A , Saha B , Mukherjee D . 1: National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India. 2 : Corresponding author. [email protected] , [email protected] 3: Present address. Labs, Persistent Systems Limited, Pingala – Aryabhata, Erandwane, Pune, 411004 India. bioRxiv preprint doi: https://doi.org/10.1101/425868; this version posted September 24, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Abstract Cells use IFNγ-STAT1 and IL-10-STAT3 pathways primarily to elicit pro and anti-inflammatory responses, respectively. However, activation of STAT1 by IL-10 and STAT3 by IFNγ is also observed. The regulatory mechanisms controlling the amplitude and dynamics of both the STATs in response to these functionally opposing stimuli remains less understood. Here, our experiments at cell population level show distinct early signalling dynamics of both STAT1 and STAT3(S/1/3) in responses to IFNγ and IL-10 stimulation.
    [Show full text]
  • SOCS3 Antibody A
    Revision 1 C 0 2 - t SOCS3 Antibody a e r o t S Orders: 877-616-CELL (2355) [email protected] Support: 877-678-TECH (8324) 3 2 Web: [email protected] 9 www.cellsignal.com 2 # 3 Trask Lane Danvers Massachusetts 01923 USA For Research Use Only. Not For Use In Diagnostic Procedures. Applications: Reactivity: Sensitivity: MW (kDa): Source: UniProt ID: Entrez-Gene Id: WB H M R Endogenous 26 Rabbit O14543 9021 Product Usage Information 5. Bjørbaek, C. et al. (1998) Mol Cell 1, 619-25. 6. Adams, T.E. et al. (1998) J Biol Chem 273, 1285-7. Application Dilution 7. Soriano, S.F. et al. (2002) J Exp Med 196, 311-21. 8. Emanuelli, B. et al. (2000) J Biol Chem 275, 15985-91. Western Blotting 1:1000 9. Stoiber, D. et al. (1999) J Immunol 163, 2640-7. 10. Stoiber, D. et al. (2001) J Immunol 166, 466-72. Storage 11. Roberts, A.W. et al. (2001) Proc Natl Acad Sci USA 98, 9324-9. 12. Seki, Y. et al. (2003) Nat Med 9, 1047-54. Supplied in 10 mM sodium HEPES (pH 7.5), 150 mM NaCl, 100 µg/ml BSA and 50% 13. Shouda, T. et al. (2001) J Clin Invest 108, 1781-8. glycerol. Store at –20°C. Do not aliquot the antibody. 14. Fang, M. et al. (2005) Cell Mol Immunol 2, 373-7. 15. Goren, I. et al. (2006) J Invest Dermatol 126, 477-85. Specificity / Sensitivity 16. Mori, H. et al. (2004) Nat Med 10, 739-43. 17.
    [Show full text]
  • Japanese Encephalitis Virus Upregulates the Expression of SOCS3 in Mouse Brain and Raw264.7 Cells
    Viruses2014, 6, 4280-4293; doi:10.3390/v6114280 OPEN ACCESS viruses ISSN 1999-4915 www.mdpi.com/journal/viruses Article Japanese Encephalitis Virus Upregulates the Expression of SOCS3 in Mouse Brain and Raw264.7 Cells Xiangmin Li 1,2, Qiaoyan Zhu 2, Qishu Cao 2, Huanchun Chen 1,2 and Ping Qian 1,2,* 1 State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, Hubei, China; E-Mails: [email protected] (X.L.); [email protected] (H.C.) 2 Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, China; E-Mails: [email protected] (Q.Z.); [email protected] (Q.C.) * Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel./Fax: +86-27-87282608. External Editor: Eric O. Freed Received: 29 August 2014; in revised form: 21 October 2014 / Accepted: 23 October 2014 / Published: 10 November 2014 Abstract: Japanese encephalitis virus (JEV) is one of the pathogens that can invade the central nervous system, causing acute infection and inflammation of brain. SOCS3 protein plays a vital role in immune processes and inflammation of the central nervous system. In this study, Raw264.7 cells and suckling mice were infected with JEV, and SOCS3 expression was analyzed by the gene expression profile, semiquantitative RT-PCR, qRT-PCR, immunohistochemistry (IHC) and Western blot. Results indicated that 520 genes were found to be differentially expressed (fold change ≥ 2.0, p < 0.05) in total. The differentially regulated genes were involved in biological processes, such as stimulus response, biological regulation and immune system processes.
    [Show full text]
  • Transcriptional Control of Tissue-Resident Memory T Cell Generation
    Transcriptional control of tissue-resident memory T cell generation Filip Cvetkovski Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2019 © 2019 Filip Cvetkovski All rights reserved ABSTRACT Transcriptional control of tissue-resident memory T cell generation Filip Cvetkovski Tissue-resident memory T cells (TRM) are a non-circulating subset of memory that are maintained at sites of pathogen entry and mediate optimal protection against reinfection. Lung TRM can be generated in response to respiratory infection or vaccination, however, the molecular pathways involved in CD4+TRM establishment have not been defined. Here, we performed transcriptional profiling of influenza-specific lung CD4+TRM following influenza infection to identify pathways implicated in CD4+TRM generation and homeostasis. Lung CD4+TRM displayed a unique transcriptional profile distinct from spleen memory, including up-regulation of a gene network induced by the transcription factor IRF4, a known regulator of effector T cell differentiation. In addition, the gene expression profile of lung CD4+TRM was enriched in gene sets previously described in tissue-resident regulatory T cells. Up-regulation of immunomodulatory molecules such as CTLA-4, PD-1, and ICOS, suggested a potential regulatory role for CD4+TRM in tissues. Using loss-of-function genetic experiments in mice, we demonstrate that IRF4 is required for the generation of lung-localized pathogen-specific effector CD4+T cells during acute influenza infection. Influenza-specific IRF4−/− T cells failed to fully express CD44, and maintained high levels of CD62L compared to wild type, suggesting a defect in complete differentiation into lung-tropic effector T cells.
    [Show full text]
  • Suppressor of Cytokine Signaling (SOCS) 5 Utilises Distinct Domains for Regulation of JAK1 and Interaction with the Adaptor Protein Shc-1
    Suppressor of Cytokine Signaling (SOCS) 5 Utilises Distinct Domains for Regulation of JAK1 and Interaction with the Adaptor Protein Shc-1 Edmond M. Linossi1,2, Indu R. Chandrashekaran3, Tatiana B. Kolesnik1,2, James M. Murphy1,2, Andrew I. Webb1,2, Tracy A. Willson1,2, Lukasz Kedzierski1,2, Alex N. Bullock4, Jeffrey J. Babon1,2, Raymond S. Norton3, Nicos A. Nicola1,2, Sandra E. Nicholson1,2* 1 Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia, 2 The University of Melbourne, Parkville, Victoria, Australia, 3 Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia, 4 Structural Genomics Consortium, University of Oxford, Oxford, United Kingdom Abstract Suppressor of Cytokine Signaling (SOCS)5 is thought to act as a tumour suppressor through negative regulation of JAK/ STAT and epidermal growth factor (EGF) signaling. However, the mechanism/s by which SOCS5 acts on these two distinct pathways is unclear. We show for the first time that SOCS5 can interact directly with JAK via a unique, conserved region in its N-terminus, which we have termed the JAK interaction region (JIR). Co-expression of SOCS5 was able to specifically reduce JAK1 and JAK2 (but not JAK3 or TYK2) autophosphorylation and this function required both the conserved JIR and additional sequences within the long SOCS5 N-terminal region. We further demonstrate that SOCS5 can directly inhibit JAK1 kinase activity, although its mechanism of action appears distinct from that of SOCS1 and SOCS3. In addition, we identify phosphoTyr317 in Shc-1 as a high-affinity substrate for the SOCS5-SH2 domain and suggest that SOCS5 may negatively regulate EGF and growth factor-driven Shc-1 signaling by binding to this site.
    [Show full text]
  • Original Article Identification of SOCS Family Members with Prognostic Values in Human Ovarian Cancer
    Am J Transl Res 2020;12(5):1824-1838 www.ajtr.org /ISSN:1943-8141/AJTR0102959 Original Article Identification of SOCS family members with prognostic values in human ovarian cancer Mengqi Yang1,2, Huanting Chen6, Lin Zhou3, Xiaobo Huang1,2,4, Fengxi Su1,2, Peng Wang1,5 1Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; 2Breast Tumor Center, Departments of 3Critical Care Medicine, 4Radiation Oncology, 5Emergency Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; 6Department of General Surgery, People’s Hospital of Shenzhen Baoan District, Affiliated Shenzhen Baoan Hospital of Southern Medical University, Shenzhen, China Received September 28, 2019; Accepted April 24, 2020; Epub May 15, 2020; Published May 30, 2020 Abstract: Background: Suppressor of cytokine signaling (SOCS) family proteins regulate cytokine responses through inhibition of multiple signaling pathways. The expression profiles and prognostic significance of SOCS members in ovarian cancer (OC) patients still remains unclear. Here, we aimed to provide a comprehensive understanding of the prognostic values of SOCS family members in OC and to discover promising therapeutic targets for OC. Methods: We firstly analyzed the KEGG pathway enrichment to reveal the possible pathways associated with SOCS family. Next, we used public databases including cBioPortal, Human Protein Atlas and Oncomine to investigate genetic altera- tions and mRNA expression of SOCS family in OC patients. More importantly, we explored the prognostic value of each individual SOCS members in OC patients using Kaplan Meier plotter database. Results: SOCS family was mark- edly enriched in JAK-STAT signaling pathway.
    [Show full text]
  • Supplementary Material DNA Methylation in Inflammatory Pathways Modifies the Association Between BMI and Adult-Onset Non- Atopic
    Supplementary Material DNA Methylation in Inflammatory Pathways Modifies the Association between BMI and Adult-Onset Non- Atopic Asthma Ayoung Jeong 1,2, Medea Imboden 1,2, Akram Ghantous 3, Alexei Novoloaca 3, Anne-Elie Carsin 4,5,6, Manolis Kogevinas 4,5,6, Christian Schindler 1,2, Gianfranco Lovison 7, Zdenko Herceg 3, Cyrille Cuenin 3, Roel Vermeulen 8, Deborah Jarvis 9, André F. S. Amaral 9, Florian Kronenberg 10, Paolo Vineis 11,12 and Nicole Probst-Hensch 1,2,* 1 Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland; [email protected] (A.J.); [email protected] (M.I.); [email protected] (C.S.) 2 Department of Public Health, University of Basel, 4001 Basel, Switzerland 3 International Agency for Research on Cancer, 69372 Lyon, France; [email protected] (A.G.); [email protected] (A.N.); [email protected] (Z.H.); [email protected] (C.C.) 4 ISGlobal, Barcelona Institute for Global Health, 08003 Barcelona, Spain; [email protected] (A.-E.C.); [email protected] (M.K.) 5 Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain 6 CIBER Epidemiología y Salud Pública (CIBERESP), 08005 Barcelona, Spain 7 Department of Economics, Business and Statistics, University of Palermo, 90128 Palermo, Italy; [email protected] 8 Environmental Epidemiology Division, Utrecht University, Institute for Risk Assessment Sciences, 3584CM Utrecht, Netherlands; [email protected] 9 Population Health and Occupational Disease, National Heart and Lung Institute, Imperial College, SW3 6LR London, UK; [email protected] (D.J.); [email protected] (A.F.S.A.) 10 Division of Genetic Epidemiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; [email protected] 11 MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, W2 1PG London, UK; [email protected] 12 Italian Institute for Genomic Medicine (IIGM), 10126 Turin, Italy * Correspondence: [email protected]; Tel.: +41-61-284-8378 Int.
    [Show full text]
  • The Genomics of Oral Poliovirus Vaccine
    THE GENOMICS OF ORAL POLIOVIRUS VACCINE RESPONSE IN BANGLADESHI INFANTS by Genevieve L. Wojcik, MHS A dissertation submitted to the Johns Hopkins University in conformity with the requirements for the degree of Doctor of Philosophy Baltimore, Maryland, USA October 2013 © Genevieve L. Wojcik All Rights Reserved Abstract The success of Oral Poliovirus Vaccine (OPV) in eradicating poliovirus has set an example for the immense potential of oral vaccines in preventing enteric infections. It is widely considered the standard for oral vaccines aiming to elicit a mucosal immune response. Despite being validated in diverse populations worldwide, there still remain some individuals that fail to mount an adequate response to vaccination with OPV. It has been hypothesized that this may be due to host genetics, as the heritability is estimated to be high (60%) and there have been ethnic differences in response. To address this question we conducted a genome-wide association study (GWAS) in 357 Bangladeshi children comparing individuals that fail to mount an immune response to high responders of OPV. Four different approaches were conducted to elucidate genetic risk loci: (1) a traditional GWAS analysis, (2) a correlation of the GWAS results with signatures of positive selection, (3) an application of gene-level methods to the GWAS results, and (4) an application of pathway-level methods to the GWAS results. Because there is no consensus as to the best gene- and pathway-level methods, a simulation experiment was conducted to systematically evaluate their relative performance. The traditional GWAS assessed the association of 6.6 million single nucleotide polymorphisms (SNPs) across the human genome, adjusted for stunting (height-for-age Z-score (HAZ) < -2).
    [Show full text]
  • SOCS4 Expressed by Recombinant HSV Protects Against Cytokine Storm in a Mouse Model
    ONCOLOGY REPORTS 41: 1509-1520, 2019 SOCS4 expressed by recombinant HSV protects against cytokine storm in a mouse model SHUQI REN1, XIAOQING CHEN2, RONGQUAN HUANG3, GRACE GUOYING ZHOU2,4 and ZHUQING YUAN1 1Department of Microbiology and Immunology, Guangzhou Medical University, Guangzhou, Guangdong 510182; 2Immvira Co., Ltd., Shenzhen International Institute for Biomedical Research, Shenzhen, Guangdong 518116; 3Department of Pathology; 4School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 510182, P.R. China Received June 20, 2018; Accepted November 30, 2018 DOI: 10.3892/or.2018.6935 Abstract. Oncolytic viruses are genetically engineered viruses be a useful regulator to inhibit cytokine overproduction, and designed for the treatment of solid tumors, and are often coupled that HSV-SOCS4 may provide a possible solution to control with the antitumor immunity of the host. The challenge of cytokine storm and its consequences following induction by using oncolytic herpes simplex virus (oHSV) as an efficacious oncolytic virus treatment. oncolytic agent is the potential host tissue damage caused by the production of a range of cytokines following intratumoral Introduction oHSV injection. An HSV-suppressor of cytokine signaling 4 (SOCS4) recombinant virus was created to investigate whether Oncolytic viruses (OVs) are genetically engineered viruses it inhibits cytokine storm. Recombinant HSV-SOCS4 and that selectively replicate in and kill cancer cells, and represent HSV-1(F) were used to infect mice, and levels of several a novel type of antitumor therapy (1-4). This approach has representative cytokines, including monocyte chemoattractant numerous advantages as a cancer therapeutic agent due to its protein-1, interleukin (IL)-1β, tumor necrosis factor-α, IL-6 mechanism-based selectivity, potential for mediating tumor and interferon γ, in serum and bronchoalveolar lavage fluid cell death and possibility of expressing additional therapeutic (BALF) of infected mice were determined, and immune cells transgenes at the tumor site (5,6).
    [Show full text]
  • (SOCS) Genes Are Downregulated in Breast Cancer Soudeh Ghafouri-Fard1, Vahid Kholghi Oskooei1, Iman Azari1 and Mohammad Taheri2,3*
    Ghafouri-Fard et al. World Journal of Surgical Oncology (2018) 16:226 https://doi.org/10.1186/s12957-018-1529-9 RESEARCH Open Access Suppressor of cytokine signaling (SOCS) genes are downregulated in breast cancer Soudeh Ghafouri-Fard1, Vahid Kholghi Oskooei1, Iman Azari1 and Mohammad Taheri2,3* Abstract Background: The suppressor of cytokine signaling (SOCS) family of proteins are inhibitors of the cytokine-activated Janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling pathway. We aimed at evaluation of expression of SOCS genes in breast cancer. Methods: We evaluated expression of SOCS1–3 and SOCS5 genes in breast cancer samples compared with the corresponding adjacent non-cancerous tissues (ANCTs). Results: All assessed SOCS genes were significantly downregulated in tumoral tissues compared with ANCTs. SOCS1 and SOCS2 genes were significantly overexpressed in higher grade samples, but SOCS3 had the opposite trend. Significant correlations were found between expression levels of SOCS genes. The SOCS1 and SOCS2 expression levels had the best specificity and sensitivity values respectively for breast cancer diagnosis. Conclusion: The current study provides further evidence for contribution of SOCS genes in breast cancer. Keywords: Suppressor of cytokine signaling, Breast cancer, Expression Introduction MCF-7 and HCC1937, two cell lines that are regarded The suppressor of cytokine signaling (SOCS) family of as prototypic breast cancer cell types. Moreover, they proteins have been recognized as potent inhibitors of have demonstrated responsiveness of SOCS1 and the cytokine-activated Janus kinase/signal transducers SOCS3 promoters to regulation by cytokine or growth and activators of transcription (JAK/STAT) signaling factor signals in spite of hypermethylation state of pathway through which they also suppress cytokine these promoters in these two cell lines [6].
    [Show full text]
  • Expression of SOCS1 and SOCS3 Genes Is Differentially Regulated in Breast Cancer Cells in Response to Proinflammatory Cytokine and Growth Factor Signals
    Oncogene (2007) 26, 1941–1948 & 2007 Nature Publishing Group All rights reserved 0950-9232/07 $30.00 www.nature.com/onc ORIGINAL ARTICLE Expression of SOCS1 and SOCS3 genes is differentially regulated in breast cancer cells in response to proinflammatory cytokine and growth factor signals MK Evans1, C-R Yu2, A Lohani1, RM Mahdi2, X Liu2, AR Trzeciak1 and CE Egwuagu2 1Laboratory of Cellular and Molecular Biology, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA and 2Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA DNA-hypermethylation of SOCS genes in breast, ova- Keywords: breast-cancer cells; SOCS; breast cancer; STAT; rian, squamous cell and hepatocellular carcinoma has led BRCA1; DNA hypermethylation; interferon to speculation that silencing of SOCS1 and SOCS3 genes might promote oncogenic transformation of epithelial tissues. To examine whether transcriptional silencing of SOCS genes is a common feature of human carcinoma, we have investigated regulation of SOCS genes expression by Introduction IFNc, IGF-1 and ionizing radiation, in a normal human mammary epithelial cell line (AG11134), two breast- Development of the mammary gland is regulated by cancer cell lines (MCF-7, HCC1937) and three prostate factors that coordinate proliferation, differentiation, cancer cell lines. Compared to normal breast cells, we maturation and apoptosis of mammary epithelial cells. observe a high level constitutive expression of SOCS2, Exquisite control of the initiation, strength and duration SOCS3, SOCS5, SOCS6, SOCS7, CIS and/or SOCS1 of signals from the diverse array of cytokines and genes in the human cancer cells. In MCF-7 and HCC1937 growth factors in mammalian breast is essential to the breast-cancer cells, transcription of SOCS1 is dramati- timely initiation of the menstrual cycle, lactation or cally up-regulated by IFNc and/or ionizing-radiation breast lobule involution (Medina, 2005).
    [Show full text]
  • Bioinformatics Analysis for the Identification of Differentially Expressed Genes and Related Signaling Pathways in H
    Bioinformatics analysis for the identification of differentially expressed genes and related signaling pathways in H. pylori-CagA transfected gastric cancer cells Dingyu Chen*, Chao Li, Yan Zhao, Jianjiang Zhou, Qinrong Wang and Yuan Xie* Key Laboratory of Endemic and Ethnic Diseases , Ministry of Education, Guizhou Medical University, Guiyang, China * These authors contributed equally to this work. ABSTRACT Aim. Helicobacter pylori cytotoxin-associated protein A (CagA) is an important vir- ulence factor known to induce gastric cancer development. However, the cause and the underlying molecular events of CagA induction remain unclear. Here, we applied integrated bioinformatics to identify the key genes involved in the process of CagA- induced gastric epithelial cell inflammation and can ceration to comprehend the potential molecular mechanisms involved. Materials and Methods. AGS cells were transected with pcDNA3.1 and pcDNA3.1::CagA for 24 h. The transfected cells were subjected to transcriptome sequencing to obtain the expressed genes. Differentially expressed genes (DEG) with adjusted P value < 0.05, | logFC |> 2 were screened, and the R package was applied for gene ontology (GO) enrichment and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. The differential gene protein–protein interaction (PPI) network was constructed using the STRING Cytoscape application, which conducted visual analysis to create the key function networks and identify the key genes. Next, the Submitted 20 August 2020 Kaplan–Meier plotter survival analysis tool was employed to analyze the survival of the Accepted 11 March 2021 key genes derived from the PPI network. Further analysis of the key gene expressions Published 15 April 2021 in gastric cancer and normal tissues were performed based on The Cancer Genome Corresponding author Atlas (TCGA) database and RT-qPCR verification.
    [Show full text]