全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

The Role of Suppressors of Cytokine Signalling in Human Neoplasms

DOI: 10.1155/2014/630797

Full-Text   Cite this paper   Add to My Lib

Abstract:

Suppressors of cytokine signalling 1–7 (SOCS1–7) and cytokine-inducible SH2-containing protein (CIS) are a group of intracellular proteins that are well known as JAK-STAT and several other signalling pathways negative feedback regulators. More recently several members have been identified as tumour suppressors and dysregulation of their biological roles in controlling cytokine and growth factor signalling may contribute to the development of many solid organ and haematological malignancies. This review explores their biological functions and their possible tumour suppressing role in human neoplasms. 1. Cytokines and Their Signalling Pathways Cytokines are a large family of secreted soluble glycoproteins that regulate cellular growth and differentiation which are part of fundamental biological processes including embryonic development, immunity, wound healing, and haematopoiesis. Cytokines carry information about the biological status to target cells by interacting with receptors on the cell surface. Cellular responses to cytokine stimulation depend on the type of cytokine and the nature of the target cell and include proliferation, differentiation, effector function, and survival [1, 2]. Cytokines activate multiple intracellular signalling pathways in order to produce their physiological effects. One of the most studied pathways is that involving the receptor-associated janus kinases (JAKs) and the latent cytoplasmic transcription factors signal transducers and activators of transcription (STATs) [3, 4]. Genetic deletion experiments in mice have demonstrated that this pathway is critical for the actions of specific cytokines. For example, STAT1 is absolutely required for the actions of interferons, STAT4 is absolutely necessary for the actions of interleukin-12 (IL-12), STAT6 is required for the actions of interleukin-4 (IL-4), and JAK3 is required for the actions of cytokines that use the common γ receptor [5]. This cascade requires strict cellular control and loss of regulation can promote tumorigenesis and chronic inflammation. The threshold, magnitude, and specific responses elicited by cytokine stimulation are regulated by numerous mechanisms including tyrosine phosphatases, receptor internalisation, proteasomal degradation of signalling adaptor molecules, soluble receptor antagonists, and specific inhibitors, including the protein inhibitors of activated STATs (PIAS) and suppressor of cytokine signalling (SOCS) proteins. The expression of SOCS proteins can be induced by cytokine stimulation, and they not only serve to interfere with signalling

References

[1]  N. A. Nicola, “An introduction to the cytokines,” in Guidebook to Cytokines and Their Receptors, N. A. Nicola, Ed., pp. 1–7, Oxford University Press, Oxford, UK, 1994.
[2]  J. Lotem and L. Sachs, “Cytokine control of developmental programs in normal hematopoiesis and leukemia,” Oncogene, vol. 21, no. 21, pp. 3284–3294, 2002.
[3]  J. E. Darnell Jr., “STATs and gene regulation,” Science, vol. 277, no. 5332, pp. 1630–1635, 1997.
[4]  W. J. Leonard and J. J. O'Shea, “Jaks and STATs: biological implications,” Annual Review of Immunology, vol. 16, pp. 293–322, 1998.
[5]  A. C. Ward, I. Touw, and A. Yoshimura, “The Jak-Stat pathway in normal and perturbed hematopoiesis,” Blood, vol. 95, no. 1, pp. 19–29, 2000.
[6]  M. Aringer, A. Cheng, J. W. Nelson et al., “Janus kinases and their role in growth and disease,” Life Sciences, vol. 64, no. 24, pp. 2173–2186, 1999.
[7]  A. Weiss and J. Schlessinger, “Switching signals on or off by receptor dimerization,” Cell, vol. 94, no. 3, pp. 277–280, 1998.
[8]  G. P. Dunn, K. C. F. Sheehan, L. J. Old, and R. D. Schreiber, “IFN unresponsiveness in LNCaP cells due to the lack of JAK1 gene expression,” Cancer Research, vol. 65, no. 8, pp. 3447–3453, 2005.
[9]  S. Pestka, “The interferon receptors,” Seminars in Oncology, vol. 24, no. 3, supplement 9, pp. S9-18–S9-40, 1997.
[10]  O. Silvennoinen, J. N. Ihle, J. Schlessinger, and D. E. Levy, “Interferon-induced nuclear signalling by Jak protein tyrosine kinases,” Nature, vol. 366, no. 6455, pp. 583–585, 1993.
[11]  S. V. Kotenko and S. Pestka, “Jak-Stat signal transduction pathway through the eyes of cytokine class II receptor complexes,” Oncogene, vol. 19, no. 21, pp. 2557–2565, 2000.
[12]  R. Eulenfeld, A. Dittrich, C. Khouri et al., “Interleukin-6 signalling: more than Jaks and STATs,” European Journal of Cell Biology, vol. 91, no. 6-7, pp. 486–495, 2012.
[13]  T. Waldmann, Y. Tagaya, and R. Bamford, “Interleukin-2, interleukin-15, and their receptors,” International Reviews of Immunology, vol. 16, no. 3-4, pp. 205–226, 1998.
[14]  M. Martinez-Moczygemba, D. P. Huston, and J. T. Lei, “JAK kinases control IL-5 receptor ubiquitination, degradation, and internalization,” Journal of Leukocyte Biology, vol. 81, no. 4, pp. 1137–1148, 2007.
[15]  S. J. Frank, “Minireview: receptor dimerization in GH and erythropoietin action—it takes two to Tango, but how?” Endocrinology, vol. 143, no. 1, pp. 2–10, 2002.
[16]  J. N. Ihle, “STATs: signal transducers and activators of transcription,” Cell, vol. 84, no. 3, pp. 331–334, 1996.
[17]  K. Imada and W. J. Leonard, “The Jak-STAT pathway,” Molecular Immunology, vol. 37, no. 1-2, pp. 1–11, 2000.
[18]  K. Shuai, “Modulation of STAT signaling by STAT-interacting proteins,” Oncogene, vol. 19, no. 21, pp. 2638–2644, 2000.
[19]  U. Vinkemeier, I. Moarefi, J. E. Darnell Jr., and J. Kuriyan, “Structure of the amino-terminal protein interaction domain of STAT-4,” Science, vol. 279, no. 5353, pp. 1048–1052, 1998.
[20]  S. E. Nicholson and D. J. Hilton, “The SOCS proteins: a new family of negative regulators of signal transduction,” Journal of Leukocyte Biology, vol. 63, no. 6, pp. 665–668, 1998.
[21]  A. Yoshimura, “The CIS/JAB family: novel negative regulators of JAK signaling pathways,” Leukemia, vol. 12, no. 12, pp. 1851–1857, 1998.
[22]  D. J. Hilton, R. T. Richardson, W. S. Alexander et al., “Twenty proteins containing a C-terminal SOCS box form five structural classes,” Proceedings of the National Academy of Sciences of the United States of America, vol. 95, no. 1, pp. 114–119, 1998.
[23]  R. Starr, T. A. Willson, E. M. Viney et al., “A family of cytokine-inducible inhibitors of signalling,” Nature, vol. 387, no. 6636, pp. 917–921, 1997.
[24]  T. A. Endo, M. Masuhara, M. Yokouchi et al., “A new protein containing an SH2 domain that inhibits JAK kinases,” Nature, vol. 387, no. 6636, pp. 921–924, 1997.
[25]  X. P. Chen, J. A. Losman, and P. Rothman, “SOCS proteins, regulators of intracellular signaling,” Immunity, vol. 13, no. 3, pp. 287–290, 2000.
[26]  H. Yasukawa, A. Sasaki, and A. Yoshimura, “Negative regulation of cytokine signaling pathways,” Annual Review of Immunology, vol. 18, pp. 143–164, 2000.
[27]  K. Matuoka, H. Miki, K. Takahashi, and T. Takenawa, “A novel ligand for an SH3 domain of the adaptor protein Nck bears an SH2 domain and nuclear signaling motifs,” Biochemical and Biophysical Research Communications, vol. 239, no. 2, pp. 488–492, 1997.
[28]  H. Yasukawa, H. Misawa, H. Sakamoto et al., “The JAK-binding protein JAB inhibits Janus tyrosine kinase activity through binding in the activation loop,” The EMBO Journal, vol. 18, no. 5, pp. 1309–1320, 1999.
[29]  A. N. Bullock, M. C. Rodriguez, J. é. Debreczeni, Z. Songyang, and S. Knapp, “Structure of the SOCS4-ElonginB/C complex reveals a distinct SOCS box interface and the molecular basis for SOCS-dependent EGFR degradation,” Structure, vol. 15, no. 11, pp. 1493–1504, 2007.
[30]  J. J. Babon, E. J. McManus, S. Yao et al., “The structure of SOCS3 reveals the basis of the extended SH2 domain function and identifies an unstructured insertion that regulates stability,” Molecular Cell, vol. 22, no. 2, pp. 205–216, 2006.
[31]  A. Yoshimura, T. Ohkubo, T. Kiguchi et al., “A novel cytokine-inducible gene CIS encodes an SH2-containing protein that binds to tyrosinephosphorylated interleukin 3 and erythropoietin receptors,” The EMBO Journal, vol. 14, no. 12, pp. 2816–2826, 1995.
[32]  T. Naka, M. Narazaki, M. Hirata et al., “Structure and function of a new STAT-induced STAT inhibitor,” Nature, vol. 387, no. 6636, pp. 924–929, 1997.
[33]  M. Masuhara, H. Sakamoto, A. Matsumoto et al., “Cloning and characterization of novel CIS family genes,” Biochemical and Biophysical Research Communications, vol. 239, no. 2, pp. 439–446, 1997.
[34]  L. A. O'Sullivan, C. Liongue, R. S. Lewis, S. E. M. Stephenson, and A. C. Ward, “Cytokine receptor signaling through the Jak-Stat-Socs pathway in disease,” Molecular Immunology, vol. 44, no. 10, pp. 2497–2506, 2007.
[35]  J.-G. Zhang, A. Farley, S. E. Nicholson et al., “The conserved SOCS box motif in suppressors of cytokine signaling binds to elongins B and C and may couple bound proteins to proteasomal degradation,” Proceedings of the National Academy of Sciences of the United States of America, vol. 96, no. 5, pp. 2071–2076, 1999.
[36]  T. Kamura, K. Maenaka, S. Kotoshiba et al., “VHL-box and SOCS-box domains determine binding specificity for Cul2-Rbx1 and Cul5-Rbx2 modules of ubiquitin ligases,” Genes and Development, vol. 18, no. 24, pp. 3055–3065, 2004.
[37]  A. Matsumoto, Y. Seki, M. Kubo et al., “Suppression of STAT5 functions in liver, mammary glands, and T cells in cytokine-inducible SH2-containing protein 1 transgenic mice,” Molecular and Cellular Biology, vol. 19, no. 9, pp. 6396–6407, 1999.
[38]  M. J. Aman, T.-S. Migone, A. Sasaki et al., “CIS associates with the interleukin-2 receptor β chain and inhibits interleukin-2-dependent signaling,” The Journal of Biological Chemistry, vol. 274, no. 42, pp. 30266–30272, 1999.
[39]  A. Matsumoto, M. Masuhara, K. Mitsui et al., “CIS, a cytokine inducible SH2 protein, is a target of the JAK-STAT5 pathway and modulates STAT5 activation,” Blood, vol. 89, no. 9, pp. 3148–3154, 1997.
[40]  D. Lejeune, J.-B. Demoulin, and J.-C. Renauld, “Interleukin 9 induces expression of three cytokine signal inhibitors: cytokine-inducible SH2-containing protein, suppressor of cytokine signalling (SOCS)-2 and SOCS-3, but only SOCS-3 overexpression suppresses interleukin 9 signalling,” Biochemical Journal, vol. 353, part 1, pp. 109–116, 2001.
[41]  X. Shen, F. Hong, V.-A. Nguyen, and B. Gao, “IL-10 attenuates IFN-α-activated STAT1 in the liver: involvement of SOCS2 and SOCS3,” FEBS Letters, vol. 480, no. 2-3, pp. 132–136, 2000.
[42]  A. Pezet, H. Favre, P. A. Kelly, and M. Edery, “Inhibition and restoration of prolactin signal transduction by suppressors of cytokine signaling,” The Journal of Biological Chemistry, vol. 274, no. 35, pp. 24497–24502, 1999.
[43]  F. Dif, E. Saunier, B. Demeneix, P. A. Kelly, and M. Edery, “Cytokine-inducible SH2-containing protein suppresses PRL signaling by binding the PRL receptor,” Endocrinology, vol. 142, no. 12, pp. 5286–5293, 2001.
[44]  D. Helman, Y. Sandowski, Y. Cohen et al., “Cytokine-inducible SH2 protein (CIS3) and JAK2 binding protein (JAB) abolish prolactin receptor-mediated STAT5 signaling,” FEBS Letters, vol. 441, no. 2, pp. 287–291, 1998.
[45]  C. S. Zong, J. Chan, D. E. Levy, C. Horvath, H. B. Sadowski, and L.-H. Wang, “Mechanism of STAT3 activation by insulin-like growth factor I receptor,” The Journal of Biological Chemistry, vol. 275, no. 20, pp. 15099–15105, 2000.
[46]  P. A. Ram and D. J. Waxman, “Role of the cytokine-inducible SH2 protein CIS in desensitization of STAT5b signaling by continuous growth hormone,” The Journal of Biological Chemistry, vol. 275, no. 50, pp. 39487–39496, 2000.
[47]  P. A. Ram and D. J. Waxman, “SOCS/CIS protein inhibition of growth hormone-stimulated STAT5 signaling by multiple mechanisms,” The Journal of Biological Chemistry, vol. 274, no. 50, pp. 35553–35561, 1999.
[48]  S. Okabe, T. Tauchi, H. Morita, H. Ohashi, A. Yoshimura, and K. Ohyashiki, “Thrombopoietin induces an SH2-containing protein, CIS1, which binds to MP1: involvement of the ubiquitin proteosome pathway,” Experimental Hematology, vol. 27, no. 10, pp. 1542–1547, 1999.
[49]  M. G. Hunter, A. Jacob, L. C. O'Donnell et al., “Loss of SHIP and CIS recruitment to the granulocyte colony-stimulating factor receptor contribute to hyperproliferative responses in severe congenital neutropenia/acute myelogenous leukemia,” Journal of Immunology, vol. 173, no. 8, pp. 5036–5045, 2004.
[50]  C. Brender, M. Nielsen, G. R?pke et al., “Interferon-alpha induces transient suppressors of cytokine signalling expression in human T cells,” Experimental and Clinical Immunogenetics, vol. 18, no. 2, pp. 80–85, 2001.
[51]  V. Emilsson, J. R. S. Arch, R. P. de Groot, C. A. Lister, and M. A. Cawthorne, “Leptin treatment increases suppressors of cytokine signaling in central and peripheral tissues,” FEBS Letters, vol. 455, no. 1-2, pp. 170–174, 1999.
[52]  D. E. Isaksen, H. Baumann, P. A. Trobridge, A. G. Farr, S. D. Levin, and S. F. Ziegler, “Requirement for Stat5 in thymic stromal lymphopoietin-mediated signal transduction,” Journal of Immunology, vol. 163, no. 11, pp. 5971–5977, 1999.
[53]  C. Bj?rb?k, J. K. Elmquist, K. El-Haschimi et al., “Activation of SOCS-3 messenger ribonucleic acid in the hypothalamus by ciliary neurotrophic factor,” Endocrinology, vol. 140, no. 5, pp. 2035–2043, 1999.
[54]  J. A. Hansen, K. Lindberg, D. J. Hilton, J. H. Nielsen, and N. Billestrup, “Mechanism of inhibition of growth hormone receptor signaling by suppressor of cytokine signaling proteins,” Molecular Endocrinology, vol. 13, no. 11, pp. 1832–1843, 1999.
[55]  S. Li, S. Chen, X. Xu et al., “Cytokine-induced Src homology 2 protein (CIS) promotes T cell receptor-mediated proliferation and prolongs survival of activated T cells,” Journal of Experimental Medicine, vol. 191, no. 6, pp. 985–994, 2000.
[56]  B. Sporri, P. E. Kovanen, A. Sasaki, A. Yoshimura, and W. J. Leonard, “JAB/SOCS1/SSI-1 is an interleukin-2-induced inhibitor of IL-2 signaling,” Blood, vol. 97, no. 1, pp. 221–226, 2001.
[57]  J. A. Losman, X. P. Chen, D. Hilton, and P. Rothman, “Cutting edge: SOCS-1 is a potent inhibitor of IL-4 signal transduction,” Journal of Immunology, vol. 162, no. 7, pp. 3770–3774, 1999.
[58]  H. Dickensheets, N. Vazquez, F. Sheikh et al., “Suppressor of cytokine signaling-1 is an IL-4-inducible gene in macrophages and feedback inhibits IL-4 signaling,” Genes and Immunity, vol. 8, no. 1, pp. 21–27, 2007.
[59]  D. Hebenstreit, P. Luft, A. Schmiedlechner et al., “IL-4 and IL-13 induce SOCS-1 gene expression in A549 cells by three functional STAT6-binding motifs located upstream of the transcription initiation site,” Journal of Immunology, vol. 171, no. 11, pp. 5901–5907, 2003.
[60]  T. E. Adams, J. A. Hansen, R. Starr, N. A. Nicola, D. J. Hilton, and N. Billestrup, “Growth hormone preferentially induces the rapid, transient expression of SOCS-3, a novel inhibitor of cytokine receptor signaling,” The Journal of Biological Chemistry, vol. 273, no. 3, pp. 1285–1287, 1998.
[61]  M. M. Song and K. Shuai, “The suppressor of cytokine signaling (SOCS) 1 and SOCS3 but not SOCS2 proteins inhibit interferon-mediated antiviral and antiproliferative activities,” The Journal of Biological Chemistry, vol. 273, no. 52, pp. 35056–35062, 1998.
[62]  A. Crespo, M. B. Filla, S. W. Russell, and W. J. Murphy, “Indirect induction of suppressor of cytokine signalling-1 in macrophages stimulated with bacterial lipopolysaccharide: partial role of autocrine/paracrine interferon-α/β,” Biochemical Journal, vol. 349, no. 1, pp. 99–104, 2000.
[63]  Y. Morita, T. Naka, Y. Kawazoe et al., “Signals transducers and activators of transcription (STAT)-induced STAT inhibitor-1 (SSI-1)/suppressor of cytokine signaling-1 (SOCS-1) suppresses tumor necrosis factor α-induced cell death in fibroblasts,” Proceedings of the National Academy of Sciences of the United States of America, vol. 97, no. 10, pp. 5405–5410, 2000.
[64]  I. Kinjyo, T. Hanada, K. Inagaki-Ohara et al., “SOCS1/JAB is a negative regulator of LPS-induced macrophage activation,” Immunity, vol. 17, no. 5, pp. 583–591, 2002.
[65]  R. Nakagawa, T. Naka, H. Tsutsui et al., “SOCS-1 participates in negative regulation of LPS responses,” Immunity, vol. 17, no. 5, pp. 677–687, 2002.
[66]  C. L. Sadowski, T.-S. Choi, M. Le, T. T. Wheeler, L.-H. Wang, and H. B. Sadowski, “Insulin induction of SOCS-2 and SOCS-3 mRNA expression in C2C12 skeletal muscle cells is mediated by Stat5*,” The Journal of Biological Chemistry, vol. 276, no. 23, pp. 20703–20710, 2001.
[67]  E. S. Park, H. Kim, J. M. Suh et al., “Thyrotropin induces SOCS-1 (suppressor of cytokine signaling-1) and SOCS-3 in FRTL-5 thyroid cells,” Molecular Endocrinology, vol. 14, no. 3, pp. 440–448, 2000.
[68]  A. H. Dalpke, S. Opper, S. Zimmermann, and K. Heeg, “Suppressors of cytokine signaling (SOCS)-1 and SOCS-3 are induced by CpG-DNA and modulate cytokine responses in APCs1,” Journal of Immunology, vol. 166, no. 12, pp. 7082–7089, 2001.
[69]  P. de Sepulveda, K. Okkenhaug, J. la Rose, R. G. Hawley, P. Dubreuil, and R. Rottapel, “Socs1 binds to multiple signalling proteins and suppresses Steel factor-dependent proliferation,” The EMBO Journal, vol. 18, no. 4, pp. 904–915, 1999.
[70]  B. R. Dey, S. L. Spence, P. Nissley, and R. W. Furlanetto, “Interaction of human suppressor of cytokine signaling (SOCS)-2 with the insulin-like growth factor-I receptor,” The Journal of Biological Chemistry, vol. 273, no. 37, pp. 24095–24101, 1998.
[71]  S. Trop, P. de Sepulveda, J. C. Zú?iga-Pflücker, and R. Rottapel, “Overexpression of suppressor of cytokine signaling-1 impairs pre-T-cell receptor-induced proliferation but not differentiation of immature thymocytes,” Blood, vol. 97, no. 8, pp. 2269–2277, 2001.
[72]  J. L. Eyles, D. Metcalf, M. J. Grusby, D. J. Hilton, and R. Starr, “Negative regulation of interleukin-12 signaling by suppressor of cytokine signaling-1,” The Journal of Biological Chemistry, vol. 277, no. 46, pp. 43735–43740, 2002.
[73]  S. Ilangumaran, S. Ramanathan, J. la Rose, P. Poussier, and R. Rottapel, “Suppressor of cytokine signaling 1 regulates IL-15 receptor signaling in CD8+CD44high memory T lymphocytes,” Journal of Immunology, vol. 171, no. 5, pp. 2435–2445, 2003.
[74]  M. Motta, P. Accornero, and M. Baratta, “Leptin and prolactin modulate the expression of SOCS-1 in association with interleukin-6 and tumor necrosis factor-α in mammary cells: a role in differentiated secretory epithelium,” Regulatory Peptides, vol. 121, no. 1–3, pp. 163–170, 2004.
[75]  A. Mansell, R. Smith, S. L. Doyle et al., “Suppressor of cytokine signaling 1 negatively regulates Toll-like receptor signaling by mediating Mal degradation,” Nature Immunology, vol. 7, no. 2, pp. 148–155, 2006.
[76]  Y. Kawazoe, T. Naka, M. Fujimoto et al., “Signal transducer and activator of transcription (STAT)-induced STAT inhibitor 1 (SSI-1)/suppressor of cytokine signaling 1 (SOCS1) inhibits insulin signal transduction pathway through modulating insulin receptor substrate 1 (IRS-1) phosphorylation,” Journal of Experimental Medicine, vol. 193, no. 2, pp. 263–269, 2001.
[77]  Z. Dogusan, E. L. Hooghe-Peters, D. Berus, B. Velkeniers, and R. Hooghe, “Expression of SOCS genes in normal and leukemic human leukocytes stimulated by prolactin, growth hormone and cytokines,” Journal of Neuroimmunology, vol. 109, no. 1, pp. 34–39, 2000.
[78]  S. Minamoto, K. Ikegame, K. Ueno et al., “Cloning and functional analysis of new members of STAT induced STAT inhibitor (SSI) family: SSI-2 and SSI-3,” Biochemical and Biophysical Research Communications, vol. 237, no. 1, pp. 79–83, 1997.
[79]  G. M. Tannahill, J. Elliott, A. C. Barry, L. Hibbert, N. A. Cacalano, and J. A. Johnston, “SOCS2 can enhance interleukin-2 (IL-2) and IL-3 signaling by accelerating SOCS3 degradation,” Molecular and Cellular Biology, vol. 25, no. 20, pp. 9115–9126, 2005.
[80]  N. A. Nicola, S. E. Nicholson, D. Metcalf et al., “Negative regulation of cytokine signaling by the SOCS proteins,” Cold Spring Harbor Symposia on Quantitative Biology, vol. 64, pp. 397–404, 1999.
[81]  S. H. Lee, S. Yun, Z.-H. Piao et al., “Suppressor of cytokine signaling 2 regulates IL-15-primed human NK cell function via control of phosphorylated Pyk2,” Journal of Immunology, vol. 185, no. 2, pp. 917–928, 2010.
[82]  L. Wang, Z. Zhang, R. Zhang et al., “Erythropoietin up-regulates SOCS2 in neuronal progenitor cells derived from SVZ of adult rat,” NeuroReport, vol. 15, no. 8, pp. 1225–1229, 2004.
[83]  Y. Goldshmit, C. E. Walters, H. J. Scott, C. J. Greenhalgh, and A. M. Turnley, “SOCS2 induces neurite outgrowth by regulation of epidermal growth factor receptor activation,” The Journal of Biological Chemistry, vol. 279, no. 16, pp. 16349–16355, 2004.
[84]  Y. R. Boisclair, J. Wang, J. Shi, K. R. Hurst, and G. T. Ooi, “Role of the suppressor of cytokine signaling-3 in mediating the inhibitory effects of interleukin-1β on the growth hormone-dependent transcription of the acid-labile subunit gene in liver cells,” The Journal of Biological Chemistry, vol. 275, no. 6, pp. 3841–3847, 2000.
[85]  S. J. Cohney, D. Sanden, N. A. Cacalano et al., “SOCS-3 is tyrosine phosphorylated in response to interleukin-2 and suppresses STAT5 phosphorylation and lymphocyte proliferation,” Molecular and Cellular Biology, vol. 19, no. 7, pp. 4980–4988, 1999.
[86]  F. Magrangeas, O. Boisteau, S. Denis, Y. Jacques, and S. Minvielle, “Negative cross-talk between interleukin-3 and interleukin-11 is mediated by suppressor of cytokine signalling-3 (SOCS-3),” Biochemical Journal, vol. 353, part 2, pp. 223–230, 2001.
[87]  B. A. Croker, D. L. Krebs, J.-G. Zhang et al., “SOCS3 negatively regulates IL-6 signaling in vivo,” Nature Immunology, vol. 4, no. 6, pp. 540–545, 2003.
[88]  R. Lang, A.-L. Pauleau, E. Parganas et al., “SOCS3 regulates the plasticity of gp130 signaling,” Nature Immunology, vol. 4, no. 6, pp. 546–550, 2003.
[89]  H. Favre, A. Benhamou, J. Finidori, P. A. Kelly, and M. Edery, “Dual effects of suppressor of cytokine signaling (SOCS-2) on growth hormone signal transduction,” FEBS Letters, vol. 453, no. 1-2, pp. 63–66, 1999.
[90]  C. J. Auernhammer and S. Melmed, “Interleukin-11 stimulates proopiomelanocortin gene expression and adrenocorticotropin secretion in corticotroph cells: evidence for a redundant cytokine network in the hypothalamo-pituitary-adrenal axis,” Endocrinology, vol. 140, no. 4, pp. 1559–1566, 1999.
[91]  S. V. Kotenko, L. S. Izotova, O. V. Mirochnitchenko et al., “Identification, cloning, and characterization of a novel soluble receptor that binds IL-22 and neutralizes its activity,” Journal of Immunology, vol. 166, no. 12, pp. 7096–7103, 2001.
[92]  C. Paul, I. Seiliez, J. P. Thissen, and A. le Cam, “Regulation of expression of the rat SOCS-3 gene in hepatocytes by growth hormone, interleukin-6 and glucocorticoids: mRNA analysis and promoter characterization,” European Journal of Biochemistry, vol. 267, no. 19, pp. 5849–5857, 2000.
[93]  A. Sasaki, H. Yasukawa, T. Shouda, T. Kitamura, I. Dikic, and A. Yoshimura, “CIS3/SOCS-3 suppresses erythropoietin (EPO) signaling by binding the EPO receptor and JAK2,” The Journal of Biological Chemistry, vol. 275, no. 38, pp. 29338–29347, 2000.
[94]  J.-C. Marine, C. McKay, D. Wang et al., “SOCS3 is essential in the regulation of fetal liver erythropoiesis,” Cell, vol. 98, no. 5, pp. 617–627, 1999.
[95]  Q. Wang, Y. Miyakawa, N. Fox, and K. Kaushansky, “Interferon-α directly represses megakaryopoiesis by inhibiting thrombopoietin-induced signaling through induction of SOCS-1,” Blood, vol. 96, no. 6, pp. 2093–2099, 2000.
[96]  S. E. Nicholson, T. A. Willson, A. Farley et al., “Mutational analyses of the SOCS proteins suggest a dual domain requirement but distinct mechanisms for inhibition of LIF and IL-6 signal transduction,” The EMBO Journal, vol. 18, no. 2, pp. 375–385, 1999.
[97]  S. Faderl, D. Harris, Q. Van, H. M. Kantarjian, M. Talpaz, and Z. Estrov, “Granulocyte-macrophage colony-stimulating factor (GM-CSF) induces antiapoptotic and proapoptotic signals in acute myeloid leukemia,” Blood, vol. 102, no. 2, pp. 630–637, 2003.
[98]  F. Hong, V. A. Nguyen, and B. Gao, “Tumor necrosis factor alpha attenuates interferon alpha signaling in the liver: involvement of SOCS3 and SHP2 and implication in resistance to interferon therapy,” The FASEB Journal, vol. 15, no. 9, pp. 1595–1597, 2001.
[99]  N. A. Cacalano, D. Sanden, and J. A. Johnston, “Tyrosine-phosphorylated SOCS-3 inhibits STAT activation but binds to p120 RasGAP and activates Ras,” Nature Cell Biology, vol. 3, no. 5, pp. 460–465, 2001.
[100]  S. Tonko-Geymayer, O. Goupille, M. Tonko et al., “Regulation and function of the cytokine-inducible SH-2 domain proteins, CIS and SOCS3, in mammary epithelial cells,” Molecular Endocrinology, vol. 16, no. 7, pp. 1680–1695, 2002.
[101]  L. Terstegent, P. Gatsios, J. G. Bode, F. Schaper, P. C. Heinrich, and L. Graeve, “The inhibition of interleukin-6-dependent STAT activation by mitogen- activated protein kinases depends on tyrosine 759 in the cytoplasmic tail of glycoprotein 130,” The Journal of Biological Chemistry, vol. 275, no. 25, pp. 18810–18817, 2000.
[102]  C. Bj?rb?k, J. K. Elmquist, J. D. Frantz, S. E. Shoelson, and J. S. Flier, “Identification of SOCS-3 as a potential mediator of central leptin resistance,” Molecular Cell, vol. 1, no. 4, pp. 619–625, 1998.
[103]  C. Bj?rb?k, K. El-Haschimi, J. D. Frantz, and J. S. Flier, “The role of SOCS-3 in leptin signaling and leptin resistance,” The Journal of Biological Chemistry, vol. 274, no. 42, pp. 30059–30065, 1999.
[104]  B. Emanuelli, P. Peraldi, C. Filloux, D. Sawka-Verhelle, D. Hilton, and E. van Obberghen, “SOCS-3 is an insulin-induced negative regulator of insulin signaling,” The Journal of Biological Chemistry, vol. 275, no. 21, pp. 15985–15991, 2000.
[105]  F. Magrangeas, O. Boisteau, S. Denis, Y. Jacques, and S. Minvielle, “Negative regulation of onconstatin M signaling by suppressor of cytokine signaling (SOCS-3),” European Cytokine Network, vol. 12, no. 2, pp. 309–315, 2001.
[106]  B. Emanuelli, P. Peraldi, C. Filloux et al., “SOCS-3 inhibits insulin signaling and is up-regulated in response to tumor necrosis factor-α in the adipose tissue of obese mice,” The Journal of Biological Chemistry, vol. 276, no. 51, pp. 47944–47949, 2001.
[107]  I. Hamanaka, Y. Saito, H. Yasukawa et al., “Induction of JAB/SOCS-1/SSI-1 and CIS3/SOCS-3/SSI-3 is involved in gp130 resistance in cardiovascular system in rat treated with cardiotrophin-1 in vivo,” Circulation Research, vol. 88, no. 7, pp. 727–732, 2001.
[108]  A. E. Karlsen, S. G. R?nn, K. Lindberg et al., “Suppressor of cytokine signaling 3 (SOCS-3) protects β-cells against interleukin-1β- and interferon-γ-mediated toxicity,” Proceedings of the National Academy of Sciences of the United States of America, vol. 98, no. 21, pp. 12191–12196, 2001.
[109]  Z. Chen, A. Laurence, Y. Kanno et al., “Selective regulatory function of Socs3 in the formation of IL-17-secreting T cells,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 21, pp. 8137–8142, 2006.
[110]  C. Brender, G. M. Tannahill, B. J. Jenkins et al., “Suppressor of cytokine signaling 3 regulates CD8 T-cell proliferation by inhibition of interleukins 6 and 27,” Blood, vol. 110, no. 7, pp. 2528–2536, 2007.
[111]  B. A. Croker, D. Metcalf, L. Robb et al., “SOCS3 is a critical physiological negative regulator of G-CSF signaling and emergency granulopoiesis,” Immunity, vol. 20, no. 2, pp. 153–165, 2004.
[112]  C. Bj?rb?k, H. J. Lavery, S. H. Bates et al., “SOCS3 mediates feedback inhibition of the leptin receptor via Tyr985,” The Journal of Biological Chemistry, vol. 275, no. 51, pp. 40649–40657, 2000.
[113]  S. E. Nicholson, D. de Souza, L. J. Fabri et al., “Suppressor of cytokine signaling-3 preferentially binds to the SHP-2-binding site on the shared cytokine receptor subunit gp130,” Proceedings of the National Academy of Sciences of the United States of America, vol. 97, no. 12, pp. 6493–6498, 2000.
[114]  J. Schmitz, M. Weissenbach, S. Haan, P. C. Heinrich, and F. Schaper, “SOCS3 exerts its inhibitory function on interleukin-6 signal transduction through the SHP2 recruitment site of gp130,” The Journal of Biological Chemistry, vol. 275, no. 17, pp. 12848–12856, 2000.
[115]  J. J. Babon, N. J. Kershaw, J. M. Murphy et al., “Suppression of cytokine signaling by SOCS3: characterization of the mode of inhibition and the basis of its specificity,” Immunity, vol. 36, no. 2, pp. 239–250, 2012.
[116]  B. R. Dey, R. W. Furlanetto, and P. Nissley, “Suppressor of cytokine signaling (SOCS)-3 protein interacts with the insulin-like growth factor-I receptor,” Biochemical and Biophysical Research Communications, vol. 278, no. 1, pp. 38–43, 2000.
[117]  A. Matsumoto, Y.-I. Seki, R. Watanabe et al., “A role of suppressor of cytokine signaling 3 (SOCS3/CIS3/SSI3) in CD28-mediated interleukin 2 production,” Journal of Experimental Medicine, vol. 197, no. 4, pp. 425–436, 2003.
[118]  A. Banerjee, A. S. Banks, M. C. Nawijn, X. P. Chen, and P. B. Rothman, “Cutting edge: suppressor of cytokine signaling 3 inhibits activation of NFATp,” Journal of Immunology, vol. 168, no. 9, pp. 4277–4281, 2002.
[119]  S. E. Nicholson, D. Metcalf, N. S. Sprigg et al., “Suppressor of cytokine signaling (SOCS)-5 is a potential negative regulator of epidermal growth factor signaling,” Proceedings of the National Academy of Sciences of the United States of America, vol. 102, no. 7, pp. 2328–2333, 2005.
[120]  J. M. Sutherland, R. A. Keightley, B. Nixon et al., “Suppressor of cytokine signaling 4 (SOCS4): moderator of ovarian primordial follicle activation,” Journal of Cellular Physiology, vol. 227, no. 3, pp. 1188–1198, 2012.
[121]  E. Kario, M. D. Marmor, K. Adamsky et al., “Suppressors of cytokine signaling 4 and 5 regulate epidermal growth factor receptor signaling,” The Journal of Biological Chemistry, vol. 280, no. 8, pp. 7038–7048, 2005.
[122]  Y.-I. Seki, K. Hayashi, A. Matsumoto et al., “Expression of the suppressor of cytokine signaling-5 (SOCS5) negatively regulates IL-4-dependent STAT6 activation and Th2 differentiation,” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 20, pp. 13003–13008, 2002.
[123]  S. Gupta, K. Mishra, A. Surolia, and K. Banerjee, “Suppressor of cytokine signalling-6 promotes neurite outgrowth via JAK2/STAT5-mediated signalling pathway, involving negative feedback inhibition,” PLoS ONE, vol. 6, no. 11, Article ID e26674, 2011.
[124]  R. A. Mooney, J. Senn, S. Cameron et al., “Suppressors of cytokine signaling-1 and -6 associate with and inhibit the insulin receptor: a potential mechanism for cytokine-mediated insulin resistance,” The Journal of Biological Chemistry, vol. 276, no. 28, pp. 25889–25893, 2001.
[125]  J. U. Kazi, J. Sun, B. Phung, F. Zadjali, A. Flores-Morales, and L. Ronnstrand, “Suppressor of cytokine signaling 6 (SOCS6) negatively regulates Flt3 signal transduction through direct binding to phosphorylated tyrosines 591 and 919 of Flt3,” The Journal of Biological Chemistry, vol. 287, no. 43, pp. 36509–36517, 2012.
[126]  J. Bayle, S. Letard, R. Frank, P. Dubreuil, and P. de Sepulveda, “Suppressor of cytokine signaling 6 associates with KIT and regulates KIT receptor signaling,” The Journal of Biological Chemistry, vol. 279, no. 13, pp. 12249–12259, 2004.
[127]  Y. B. Choi, M. Son, M. Park, J. Shin, and G. Yun, “SOCS-6 negatively regulates T cell activation through targeting p56 lck to proteasomal degradation,” The Journal of Biological Chemistry, vol. 285, no. 10, pp. 7271–7280, 2010.
[128]  N. Martens, G. Uzan, M. Wery, R. Hooghe, E. L. Hooghe-Peters, and A. Gertler, “Suppressor of cytokine signaling 7 inhibits prolactin, growth hormone, and leptin signaling by interacting with STAT5 or STAT3 and attenuating their nuclear translocation,” The Journal of Biological Chemistry, vol. 280, no. 14, pp. 13817–13823, 2005.
[129]  D. L. Krebs, R. T. Uren, D. Metcalf et al., “SOCS-6 binds to insulin receptor substrate 4, and mice lacking the SOCS-6 gene exhibit mild growth retardation,” Molecular and Cellular Biology, vol. 22, no. 13, pp. 4567–4578, 2002.
[130]  A. S. Banks, J. Li, L. McKeag et al., “Deletion of SOCS7 leads to enhanced insulin action and enlarged islets of Langerhans,” The Journal of Clinical Investigation, vol. 115, no. 9, pp. 2462–2471, 2005.
[131]  B. E. Kremer, L. A. Adang, and I. G. Macara, “Septins regulate actin organization and cell-cycle arrest through nuclear accumulation of NCK mediated by SOCS7,” Cell, vol. 130, no. 5, pp. 837–850, 2007.
[132]  S. Tomic, N. Chughtai, and S. Ali, “SOCS-1, -2, -3: selective targets and functions downstream of the prolactin receptor,” Molecular and Cellular Endocrinology, vol. 158, no. 1-2, pp. 45–54, 1999.
[133]  H. Sakamoto, H. Yasukawa, M. Masuhara et al., “A Janus kinase inhibitor, JAB, is an interferon-γ-inducible gene and confers resistance to interferons,” Blood, vol. 92, no. 5, pp. 1668–1676, 1998.
[134]  B. Q. Vuong, T. L. Arenzana, B. M. Showalter et al., “SOCS-1 localizes to the microtubule organizing complex-associated 20S proteasome,” Molecular and Cellular Biology, vol. 24, no. 20, pp. 9092–9101, 2004.
[135]  D. Ungureanu, P. Saharinen, I. Junttila, D. J. Hilton, and O. Silvennoinen, “Regulation of Jak2 through the ubiquitin-proteasome pathway involves phosphorylation of Jak2 on Y1007 and interaction with SOCS-1,” Molecular and Cellular Biology, vol. 22, no. 10, pp. 3316–3326, 2002.
[136]  S. Kamizono, T. Hanada, H. Yasukawa et al., “The SOCS box of SOCS-1 accelerates ubiquitin-dependent proteolysis of TEL-JAK2,” The Journal of Biological Chemistry, vol. 276, no. 16, pp. 12530–12538, 2001.
[137]  J. Frantsve, J. Schwaller, D. W. Sternberg, J. Kutok, and D. G. Gilliland, “Socs-1 inhibits TEL-JAK2-mediated transformation of hematopoietic cells through inhibition of JAK2 kinase activity and induction of proteasome-mediated degradation,” Molecular and Cellular Biology, vol. 21, no. 10, pp. 3547–3557, 2001.
[138]  P. de Sepulveda, S. Ilangumaran, and R. Rottapel, “Suppressor of cytokine signaling-1 inhibits VAV function through protein degradation,” The Journal of Biological Chemistry, vol. 275, no. 19, pp. 14005–14008, 2000.
[139]  L. Rui, M. Yuan, D. Frantz, S. Shoelson, and M. F. White, “SOCS-1 and SOCS-3 block insulin signaling by ubiquitin-mediated degradation of IRS1 and IRS2,” The Journal of Biological Chemistry, vol. 277, no. 44, pp. 42394–42398, 2002.
[140]  J. Zhang, H. Li, J.-P. Yu, S. E. Wang, and X.-B. Ren, “Role of SOCS1 in tumor progression and therapeutic application,” International Journal of Cancer, vol. 130, no. 9, pp. 1971–1980, 2012.
[141]  M. Narazaki, M. Fujimoto, T. Matsumoto et al., “Three distinct domains of SSI-1/SOCS-1/JAB protein are required for its suppression of interleukin 6 signaling,” Proceedings of the National Academy of Sciences of the United States of America, vol. 95, no. 22, pp. 13130–13134, 1998.
[142]  K.-I. Ohya, S. Kajigaya, Y. Yamashita et al., “SOCS-1/JAB/SSI-1 can bind to and suppress Tec protein-tyrosine kinase,” The Journal of Biological Chemistry, vol. 272, no. 43, pp. 27178–27182, 1997.
[143]  J. E. Fenner, R. Starr, A. L. Cornish et al., “Suppressor of cytokine signaling 1 regulates the immune response to infection by a unique inhibition of type I interferon activity,” Nature Immunology, vol. 7, no. 1, pp. 33–39, 2006.
[144]  Y. Qing, A. P. Costa-Pereira, D. Watling, and G. R. Stark, “Role of tyrosine 441 of interferon-γ receptor subunit 1 in SOCS-1-mediated attenuation of STAT1 activation,” The Journal of Biological Chemistry, vol. 280, no. 3, pp. 1849–1853, 2005.
[145]  T. Wu, M. Xie, X. Wang, X. Jiang, J. Li, and H. Huang, “miR-155 modulates TNF-alpha-inhibited osteogenic differentiation by targeting SOCS1 expression,” Bone, vol. 51, no. 3, pp. 498–505, 2012.
[146]  L.-F. Lu, T.-H. Thai, D. P. Calado et al., “Foxp3-dependent microRNA155 confers competitive fitness to regulatory T cells by targeting SOCS1 protein,” Immunity, vol. 30, no. 1, pp. 80–91, 2009.
[147]  J. Harris, P. M. Stanford, K. Sutherland et al., “Socs2 and Elf5 mediate prolactin-induced mammary gland development,” Molecular Endocrinology, vol. 20, no. 5, pp. 1177–1187, 2006.
[148]  M. Vesterlund, F. Zadjali, T. Persson et al., “The SOCS2 ubiquitin ligase complex regulates growth hormone receptor levels,” PLoS ONE, vol. 6, no. 9, Article ID e25358, 2011.
[149]  D. Metcalf, C. J. Greenhalgh, E. Viney et al., “Gigantism in mice lacking suppressor of cytokine signalling-2,” Nature, vol. 405, no. 6790, pp. 1069–1073, 2000.
[150]  F. Verdier, S. Chrétien, O. Muller et al., “Proteasomes regulate erythropoietin receptor and signal transducer and activator of transcription 5 (STAT5) activation: possible involvement of the ubiquitinated Cis protein,” The Journal of Biological Chemistry, vol. 273, no. 43, pp. 28185–28190, 1998.
[151]  S. Gobert, S. Chretien, F. Gouilleux et al., “Identification of tyrosine residues within the intracellular domain of the erythropoietin receptor crucial for STAT5 activation,” The EMBO Journal, vol. 15, no. 10, pp. 2434–2441, 1996.
[152]  U. Klingmüller, S. Bergelson, J. G. Hsiao, and H. F. Lodish, “Multiple tyrosine residues in the cytosolic domain of the erythropoietin receptor promote activation of STAT5,” Proceedings of the National Academy of Sciences of the United States of America, vol. 93, no. 16, pp. 8324–8328, 1996.
[153]  T. Hanada, H. Yoshida, S. Kato et al., “Suppressor of cytokine signaling-1 is essential for suppressing dendritic cell activation and systemic autoimmunity,” Immunity, vol. 19, no. 3, pp. 437–450, 2003.
[154]  J. Piessevaux, L. de Ceuninck, D. Catteeuw, F. Peelman, and J. Tavernier, “Elongin B/C recruitment regulates substrate binding by CIS,” The Journal of Biological Chemistry, vol. 283, no. 31, pp. 21334–21346, 2008.
[155]  G. Hu, R. Zhou, J. Liu et al., “MicroRNA-98 and let-7 confer cholangiocyte expression of cytokine-inducible Src homology 2-containing protein in response to microbial challenge,” Journal of Immunology, vol. 183, no. 3, pp. 1617–1624, 2009.
[156]  Z. Wang, Y.-T. Zhou, T. Kakuma et al., “Leptin resistance of adipocytes in obesity: role of suppressors of cytokine signaling,” Biochemical and Biophysical Research Communications, vol. 277, no. 1, pp. 20–26, 2000.
[157]  R. Suzuki, H. Sakamoto, H. Yasukawa et al., “CIS3 and JAB have different regulatory roles in interleukin-6 mediated differentiation and STAT3 activation in M1 leukemia cells,” Oncogene, vol. 17, no. 17, pp. 2271–2278, 1998.
[158]  A. Sasaki, H. Yasukawa, A. Suzuki et al., “Cytokine-inducible SH2 protein-3 (CIS3/SOCS3) inhibits Janus tyrosine kinase by binding through the N-terminal kinase inhibitory region as well as SH2 domain,” Genes to Cells, vol. 4, no. 6, pp. 339–351, 1999.
[159]  N. J. Kershaw, J. M. Murphy, I. S. Lucet, N. A. Nicola, and J. J. Babon, “Regulation of Janus kinases by SOCS proteins,” Biochemical Society Transactions, vol. 41, no. 4, pp. 1042–1047, 2013.
[160]  D. de Souza, L. J. Fabri, A. Nash, D. J. Hilton, N. A. Nicola, and M. Baca, “SH2 domains from suppressor of cytokine signaling-3 and protein tyrosine phosphatase SHP-2 have similar binding specificities,” Biochemistry, vol. 41, no. 29, pp. 9229–9236, 2002.
[161]  T. Ohtani, K. Ishihara, T. Atsumi et al., “Dissection of signaling cascades through gp130 in vivo: reciprocal roles for STAT3- and SHP2-mediated signals in immune responses,” Immunity, vol. 12, no. 1, pp. 95–105, 2000.
[162]  K. Yamamoto, M. Yamaguchi, N. Miyasaka, and O. Miura, “SOCS-3 inhibits IL-12-induced STAT4 activation by binding through its SH2 domain to the STAT4 docking site in the IL-12 receptor β2 subunit,” Biochemical and Biophysical Research Communications, vol. 310, no. 4, pp. 1188–1193, 2003.
[163]  J. Piessevaux, D. Lavens, T. Montoye et al., “Functional cross-modulation between SOCS proteins can stimulate cytokine signaling,” The Journal of Biological Chemistry, vol. 281, no. 44, pp. 32953–32966, 2006.
[164]  M.-N. Hwang, C.-H. Min, H. S. Kim et al., “The nuclear localization of SOCS6 requires the N-terminal region and negatively regulates Stat3 protein levels,” Biochemical and Biophysical Research Communications, vol. 360, no. 2, pp. 333–338, 2007.
[165]  L. Li, L. M. Gr?nning, P. O. Anderson et al., “Insulin induces SOCS-6 expression and its binding to the p85 monomer of phosphoinositide 3-kinase, resulting in improvement in glucose metabolism,” The Journal of Biological Chemistry, vol. 279, no. 33, pp. 34107–34114, 2004.
[166]  G. Vlacich, M. C. Nawijn, G. C. Webb, and D. F. Steiner, “Pim3 negatively regulates glucose-stimulated insulin secretion,” Islets, vol. 2, no. 5, pp. 308–317, 2010.
[167]  R. Nishimura, W. Li, A. Kashishian et al., “Two signaling molecules share a phosphotyrosine-containing binding site in the platelet-derived growth factor receptor,” Molecular and Cellular Biology, vol. 13, no. 11, pp. 6889–6896, 1993.
[168]  M. Lettau, J. Pieper, and O. Janssen, “Nck adapter proteins: functional versatility in T cells,” Cell Communication and Signaling, vol. 7, no. 1, article 1, 2009.
[169]  C.-H. Lee, W. Li, R. Nishimura et al., “Nck associates with the SH2 domain-docking protein IRS-1 in insulin- stimulated cells,” Proceedings of the National Academy of Sciences of the United States of America, vol. 90, no. 24, pp. 11713–11717, 1993.
[170]  M. Amoui, B. P. Craddock, and W. T. Miller, “Differential phosphorylation of IRS-1 by insulin and insulin-like growth factor I receptors in Chinese hamster ovary cells,” Journal of Endocrinology, vol. 171, no. 1, pp. 153–162, 2001.
[171]  N. Martens, M. Wery, P. Wang et al., “The suppressor of cytokine signaling (SOCS)-7 interacts with the actin cytoskeleton through vinexin,” Experimental Cell Research, vol. 298, no. 1, pp. 239–248, 2004.
[172]  Q. Hu, D. Milfay, and L. T. Williams, “Binding of NCK to SOS and activation of Ras-dependent gene expression,” Molecular and Cellular Biology, vol. 15, no. 3, pp. 1169–1174, 1995.
[173]  V. Pazienza, M. Vinciguerra, A. Andriulli, and A. Mangia, “Hepatitis C virus core protein genotype 3a increases SOCS-7 expression through PPAR-γ in Huh-7 cells,” Journal of General Virology, vol. 91, no. 7, pp. 1678–1686, 2010.
[174]  J. A. Del Campo and M. Romero-Gómez, “Steatosis and insulin resistance in hepatitis C: a way out for the virus?” World Journal of Gastroenterology, vol. 15, no. 40, pp. 5014–5019, 2009.
[175]  J. Wauman, A.-S. de Smet, D. Catteeuw, D. Belsham, and J. Tavernier, “Insulin receptor substrate 4 couples the leptin receptor to multiple signaling pathways,” Molecular Endocrinology, vol. 22, no. 4, pp. 965–977, 2008.
[176]  E. Y. Skolnik, A. Batzer, N. Li et al., “The function of GRB2 in linking the insulin receptor to Ras signaling pathways,” Science, vol. 260, no. 5116, pp. 1953–1955, 1993.
[177]  D. Ge, A. C. Gao, Q. Zhang, S. Liu, Y. Xue, and Z. You, “LNCaP prostate cancer cells with autocrine interleukin-6 expression are resistant to IL-6-induced neuroendocrine differentiation due to increased expression of suppressors of cytokine signaling,” The Prostate, vol. 72, no. 12, pp. 1306–1316, 2012.
[178]  J. Knisz, A. Banks, L. McKeag, D. D. Metcalfe, P. B. Rothman, and J. M. Brown, “Loss of SOCS7 in mice results in severe cutaneous disease and increased mast cell activation,” Clinical Immunology, vol. 132, no. 2, pp. 277–284, 2009.
[179]  J. K. Howard and J. S. Flier, “Attenuation of leptin and insulin signaling by SOCS proteins,” Trends in Endocrinology and Metabolism, vol. 17, no. 9, pp. 365–371, 2006.
[180]  T. Nosaka, T. Kawashima, K. Misawa, K. Ikuta, A. L.-F. Mui, and T. Kitamura, “STAT5 as a molecular regulator of proliferation, differentiation and apoptosis in hematopoietic cells,” The EMBO Journal, vol. 18, no. 17, pp. 4754–4765, 1999.
[181]  G. Verdeil, D. Puthier, C. Nguyen, A.-M. Schmitt-Verhulst, and N. Auphan-Anezin, “STAT5-mediated signals sustain a TCR-initiated gene expression program toward differentiation of CD8 T cell effectors,” Journal of Immunology, vol. 176, no. 8, pp. 4834–4842, 2006.
[182]  R. Moriggl, D. J. Topham, S. Teglund et al., “Stat5 is required for IL-2-induced cell cycle progression of peripheral T cells,” Immunity, vol. 10, no. 2, pp. 249–259, 1999.
[183]  D. Metcalf, S. Mifsud, L. di Rago, N. A. Nicola, D. J. Hilton, and W. S. Alexander, “Polycystic kidneys and chronic inflammatory lesions are the delayed consequences of loss of the suppressor of cytokine signaling-1 (SOCS-1),” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 2, pp. 943–948, 2002.
[184]  A. L. Cornish, G. M. Davey, D. Metcalf et al., “Suppressor of cytokine signaling-1 has IFN-γ-independent actions in T cell homeostasis,” Journal of Immunology, vol. 170, no. 2, pp. 878–886, 2003.
[185]  C. Lee, T. B. Kolesnik, I. Caminschi et al., “Suppressor of cytokine signalling 1 (SOCS1) is a physiological regulator of the asthma response,” Clinical and Experimental Allergy, vol. 39, no. 6, pp. 897–907, 2009.
[186]  J. Horino, M. Fujimoto, F. Terabe et al., “Suppressor of cytokine signaling-1 ameliorates dextran sulfate sodium-induced colitis in mice,” International Immunology, vol. 20, no. 6, pp. 753–762, 2008.
[187]  A. L. Cornish, M. M. Chong, G. M. Davey et al., “Suppressor of cytokine signaling-1 regulates signaling in response to interleukin-2 and other γc-dependent cytokines in peripheral T cells,” The Journal of Biological Chemistry, vol. 278, no. 25, pp. 22755–22761, 2003.
[188]  H. Takatori, H. Nakajima, S.-I. Kagami et al., “Stat5a inhibits IL-12-induced Th1 cell differentiation through the induction of suppressor of cytokine signaling expression,” Journal of Immunology, vol. 174, no. 7, pp. 4105–4112, 2005.
[189]  H. Watanabe, M. Kubo, K. Numata et al., “Overexpression of suppressor of cytokine signaling-5 in T cells augments innate immunity during septic peritonitis,” Journal of Immunology, vol. 177, no. 12, pp. 8650–8657, 2006.
[190]  D. L. Krebs, D. Metcalf, T. D. Merson et al., “Development of hydrocephalus in mice lacking SOCS7,” Proceedings of the National Academy of Sciences of the United States of America, vol. 101, no. 43, pp. 15446–15451, 2004.
[191]  C. C. Khor, F. O. Vannberg, S. J. Chapman et al., “CISH and susceptibility to infectious diseases,” The New England Journal of Medicine, vol. 362, no. 22, pp. 2092–2101, 2010.
[192]  R. Starr, D. Metcalf, A. G. Elefanty et al., “Liver degeneration and lymphoid deficiencies in mice lacking suppressor of cytokine signaling-1,” Proceedings of the National Academy of Sciences of the United States of America, vol. 95, no. 24, pp. 14395–14399, 1998.
[193]  T. Naka, T. Matsumoto, M. Narazaki et al., “Accelerated apoptosis of lymphocytes by augmented induction of Bax in SSI-1 (STAT-induced STAT inhibitor-1) deficient mice,” Proceedings of the National Academy of Sciences of the United States of America, vol. 95, no. 26, pp. 15577–15582, 1998.
[194]  W. S. Alexander, R. Starr, J. E. Fenner et al., “SOCS1 is a critical inhibitor of interferon γ signaling and prevents the potentially fatal neonatal actions of this cytokine,” Cell, vol. 98, no. 5, pp. 597–608, 1999.
[195]  M. Fujimoto, T. Naka, R. Nakagawa et al., “Defective thymocyte development and perturbed homeostasis of T cells in STAT-induced STAT inhibitor-1/suppressors of cytokine signaling-1 transgenic mice,” Journal of Immunology, vol. 165, no. 4, pp. 1799–1806, 2000.
[196]  M. I. Ransome, Y. Goldshmit, P. F. Bartlett, M. J. Waters, and A. M. Turnley, “Comparative analysis of CNS populations in knockout mice with altered growth hormone responsiveness,” European Journal of Neuroscience, vol. 19, no. 8, pp. 2069–2079, 2004.
[197]  A. M. Turnley, C. H. Faux, R. L. Rietze, J. R. Coonan, and P. F. Bartlett, “Suppressor of cytokine signaling 2 regulates neuronal differentiation by inhibiting growth hormone signaling,” Nature Neuroscience, vol. 5, no. 11, pp. 1155–1162, 2002.
[198]  C. J. Greenhalgh, E. Rico-Bautista, M. Lorentzon et al., “SOCS2 negatively regulates growth hormone action in vitro and in vivo,” The Journal of Clinical Investigation, vol. 115, no. 2, pp. 397–406, 2005.
[199]  A. W. Roberts, L. Robb, S. Rakar et al., “Placental defects and embryonic lethality in mice lacking suppressor of cytokine signaling 3,” Proceedings of the National Academy of Sciences of the United States of America, vol. 98, no. 16, pp. 9324–9329, 2001.
[200]  Y. Takahashi, N. Carpino, J. C. Cross, M. Torres, E. Parganas, and J. N. Ihle, “SOCS3: an essential regulator of LIF receptor signaling in trophoblast giant cell differentiation,” The EMBO Journal, vol. 22, no. 3, pp. 372–384, 2003.
[201]  L. Robb, K. Boyle, S. Rakar et al., “Genetic reduction of embryonic leukemia-inhibitory factor production rescues placentation in SOCS3-null embryos but does not prevent inflammatory disease,” Proceedings of the National Academy of Sciences of the United States of America, vol. 102, no. 45, pp. 16333–16338, 2005.
[202]  Y.-I. Seki, H. Inoue, N. Nagata et al., “SOCS-3 regulates onset and maintenance of TH2-mediated allergic responses,” Nature Medicine, vol. 9, no. 8, pp. 1047–1054, 2003.
[203]  K. Lindberg, S. G. R?nn, D. Tornehave et al., “Regulation of pancreatic β-cell mass and proliferation by SOCS-3,” Journal of Molecular Endocrinology, vol. 35, no. 2, pp. 231–243, 2005.
[204]  C. Brender, R. Columbus, D. Metcalf et al., “SOCS5 is expressed in primary B and T lymphoid cells but is dispensable for lymphocyte production and function,” Molecular and Cellular Biology, vol. 24, no. 13, pp. 6094–6103, 2004.
[205]  A. Matsukawa, M. H. Kaplan, C. M. Hogaboam, N. W. Lukacs, and S. L. Kunkel, “Pivotal role of signal transducer and activator of transcription (Stat)4 and Stat6 in the innate immune response during sepsis,” Journal of Experimental Medicine, vol. 193, no. 6, pp. 679–688, 2001.
[206]  B. He, L. You, K. Uematsu et al., “SOCS-3 is frequently silenced by hypermethylation and suppresses cell growth in human lung cancer,” Proceedings of the National Academy of Sciences of the United States of America, vol. 100, no. 2, pp. 14133–14138, 2003.
[207]  T. Komazaki, H. Nagai, M. Emi et al., “Hypermethylation-associated inactivation of the SOCS-1 gene, a JAK/STAT inhibitor, in human pancreatic cancers,” Japanese Journal of Clinical Oncology, vol. 34, no. 4, pp. 191–194, 2004.
[208]  H. Yoshikawa, K. Matsubara, G.-S. Qian et al., “SOCS-1, a negative regulator of the JAK/STAT pathway, is silenced by methylation in human hepatocellular carcinoma and shows growth-suppression activity,” Nature Genetics, vol. 28, no. 1, pp. 29–35, 2001.
[209]  H. Nagai, T. Naka, Y. Terada et al., “Hypermethylation associated with inactivation of the SOCS-1 gene, a JAK/STAT inhibitor, in human hepatoblastomas,” Journal of Human Genetics, vol. 48, no. 2, pp. 65–69, 2003.
[210]  H. Wikman, E. Kettunen, J. K. Sepp?nen et al., “Identification of differentially expressed genes in pulmonary adenocarcinoma by using cDNA array,” Oncogene, vol. 21, no. 37, pp. 5804–5813, 2002.
[211]  F. Farabegoli, C. Ceccarelli, D. Santini, and M. Taffurelli, “Suppressor of cytokine signalling 2 (SOCS-2) expression in breast carcinoma,” Journal of Clinical Pathology, vol. 58, no. 10, pp. 1046–1050, 2005.
[212]  D. Watanabe, S. Ezoe, M. Fujimoto et al., “Suppressor of cytokine signalling-1 gene silencing in acute myeloid leukaemia and human haematopoietic cell lines,” British Journal of Haematology, vol. 126, no. 5, pp. 726–735, 2004.
[213]  O. Galm, H. Yoshikawa, M. Esteller, R. Osieka, and J. G. Herman, “SOCS-1, a negative regulator of cytokine signaling, is frequently silenced by methylation in multiple myeloma,” Blood, vol. 101, no. 7, pp. 2784–2788, 2003.
[214]  J. Roman-Gomez, A. Jimenez-Velasco, J. A. Castillejo et al., “The suppressor of cytokine signaling-1 is constitutively expressed in chronic myeloid leukemia and correlates with poor cytogenetic response to interferon-α,” Haematologica, vol. 89, no. 1, pp. 42–48, 2004.
[215]  M. Raccurt, S. P. Tam, P. Lau et al., “Suppressor of cytokine signalling gene expression is elevated in breast carcinoma,” British Journal of Cancer, vol. 89, no. 3, pp. 524–532, 2003.
[216]  M. K. Evans, C.-R. Yu, A. Lohani et al., “Expression of SOCS1 and SOCS3 genes is differentially regulated in breast cancer cells in response to proinflammatory cytokine and growth factor signals,” Oncogene, vol. 26, no. 13, pp. 1941–1948, 2007.
[217]  M. C. Haffner, B. Petridou, J. P. Peyrat et al., “Favorable prognostic value of SOCS2 and IGF-I in breast cancer,” BMC Cancer, vol. 7, article 136, 2007.
[218]  W. Komyod, M. B?hm, D. Metze, P. C. Heinrich, and I. Behrmann, “Constitutive suppressor of cytokine signaling 3 expression confers a growth advantage to a human melanoma cell line,” Molecular Cancer Research, vol. 5, no. 3, pp. 271–281, 2007.
[219]  C. Zheng, L. Li, M. Haak et al., “Gene expression profiling of CD34+ cells identifies a molecular signature of chronic myeloid leukemia blast crisis,” Leukemia, vol. 20, no. 6, pp. 1028–1034, 2006.
[220]  B. Schultheis, M. Carapeti-Marootian, A. Hochhaus, A. Wei?er, J. M. Goldman, and J. V. Melo, “Overexpression of SOCS-2 in advanced stages of chronic myeloid leukemia: possible inadequacy of a negative feedback mechanism,” Blood, vol. 99, no. 5, pp. 1766–1775, 2002.
[221]  J.-J. Schuringa, A. T. J. Wierenga, W. Kruijer, and E. Vellenga, “Constitutive Stat3, Tyr705, and Ser727 phosphorylation in acute myeloid leukemia cells caused by the autocrine secretion of interleukin-6,” Blood, vol. 95, no. 12, pp. 3765–3770, 2000.
[222]  C. Brender, M. Nielsen, K. Kaltoft et al., “STAT3-mediated constitutive expression of SOCS-3 in cutaneous T-cell lymphoma,” Blood, vol. 97, no. 4, pp. 1056–1062, 2001.
[223]  I. Sakai, K. Takeuchi, H. Yamauchi, H. Narumi, and S. Fujita, “Constitutive expression of SOCS3 confers resistance to IFN-α in chronic myelogenous leukemia cells,” Blood, vol. 100, no. 8, pp. 2926–2931, 2002.
[224]  J. H. Cho-Vega, G. Z. Rassidakis, H. M. Amin et al., “Suppressor of cytokine signaling 3 expression in anaplastic large cell lymphoma,” Leukemia, vol. 18, no. 11, pp. 1872–1878, 2004.
[225]  H. Neuwirt, M. Puhr, I. T. Cavarretta, M. Mitterberger, A. Hobisch, and Z. Culig, “Suppressor of cytokine signalling-3 is up-regulated by androgen in prostate cancer cell lines and inhibits androgen-mediated proliferation and secretion,” Endocrine-Related Cancer, vol. 14, no. 4, pp. 1007–1019, 2007.
[226]  I. Bellezza, H. Neuwirt, C. Nemes et al., “Suppressor of cytokine signaling-3 antagonizes cAMP effects on proliferation and apoptosis and is expressed in human prostate cancer,” American Journal of Pathology, vol. 169, no. 6, pp. 2199–2208, 2006.
[227]  M. Puhr, F. R. Santer, H. Neuwirt et al., “Down-regulation of suppressor of cytokine signaling-3 causes prostate cancer cell death through activation of the extrinsic and intrinsic apoptosis pathways,” Cancer Research, vol. 69, no. 18, pp. 7375–7384, 2009.
[228]  M. A. Weniger, I. Melzner, C. K. Menz et al., “Mutations of the tumor suppressor gene SOCS-1 in classical Hodgkin lymphoma are frequent and associated with nuclear phospho-STAT5 accumulation,” Oncogene, vol. 25, no. 18, pp. 2679–2684, 2006.
[229]  A. Mottok, C. Renné, K. Willenbrock, M.-L. Hansmann, and A. Br?uninger, “Somatic hypermutation of SOCS1 in lymphocyte-predominant Hodgkin lymphoma is accompanied by high JAK2 expression and activation of STAT6,” Blood, vol. 110, no. 9, pp. 3387–3390, 2007.
[230]  I. Melzner, A. J. Bucur, S. Brüderlein et al., “Biallelic mutation SOCS-1 impairs JAK2 degradation and sustains phospho-JAK2 action in the MedB-1 mediastinal lymphoma line,” Blood, vol. 105, no. 6, pp. 2535–2542, 2005.
[231]  C.-Y. Chen, W. Tsay, J.-L. Tang et al., “SOCSI methylation in patients with newly diagnosed acute myeloid leukemia,” Genes Chromosomes and Cancer, vol. 37, no. 3, pp. 300–305, 2003.
[232]  T.-C. Liu, S.-F. Lin, J.-G. Chang, M.-Y. Yang, S.-Y. Hung, and C.-S. Chang, “Epigenetic alteration of the SOCS1 gene in chronic myeloid leukaemia,” British Journal of Haematology, vol. 123, no. 4, pp. 654–661, 2003.
[233]  E. Jost, O. N. do, E. Dahl et al., “Epigenetic alterations complement mutation of JAK2 tyrosine kinase in patients with BCR/ABL-negative myeloproliferative disorders,” Leukemia, vol. 21, no. 3, pp. 505–510, 2007.
[234]  O. Bock, K. Hussein, K. Brakensiek et al., “The suppressor of cytokine signalling-1 (SOCS-1) gene is overexpressed in Philadelphia chromosome negative chronic myeloproliferative disorders,” Leukemia Research, vol. 31, no. 6, pp. 799–803, 2007.
[235]  O. Hatirnaz, U. Ure, C. Ar et al., “The SOCS-1 gene methylation in chronic myeloid leukemia patients,” American Journal of Hematology, vol. 82, no. 8, pp. 729–730, 2007.
[236]  R. C. Sobti, N. Singh, S. Hussain et al., “Aberrant promoter methylation and loss of suppressor of cytokine signalling-1 gene expression in the development of uterine cervical carcinogenesis,” Cellular Oncology, vol. 34, no. 6, pp. 533–543, 2011.
[237]  S. Hussain, N. Singh, I. Salam et al., “Methylation-mediated gene silencing of suppressor of cytokine signaling-1 (SOCS-1) gene in esophageal squamous cell carcinoma patients of Kashmir valley,” Journal of Receptors and Signal Transduction, vol. 31, no. 2, pp. 147–156, 2011.
[238]  T. Tischoff, U. R. Hengge, M. Vieth et al., “Methylation of SOCS-3 and SOCS-1 in the carcinogenesis of Barrett's adenocarcinoma,” Gut, vol. 56, no. 8, pp. 1047–1053, 2007.
[239]  H. Nagai, Y. S. Kim, N. Konishi et al., “Combined hypermethylation and chromosome loss associated with inactivation of SSI-1/SOCS-1/JAB gene in human hepatocellular carcinomas,” Cancer Letters, vol. 186, no. 1, pp. 59–65, 2002.
[240]  O. Okochi, K. Hibi, M. Sakai et al., “Methylation-mediated silencing of SOCS-1 gene in hepatocellular carcinoma derived from cirrhosis,” Clinical Cancer Research, vol. 9, no. 14, pp. 5295–5298, 2003.
[241]  T. Yoshida, H. Ogata, M. Kamio et al., “SOCS1 is a suppressor of liver fibrosis and hepatitis-induced carcinogenesis,” Journal of Experimental Medicine, vol. 199, no. 12, pp. 1701–1707, 2004.
[242]  H. Zhou, R. Miki, M. Eeva et al., “Reciprocal regulation of SOCS1 and SOCS3 enhances resistance to ionizing radiation in glioblastoma multiforme,” Clinical Cancer Research, vol. 13, no. 8, pp. 2344–2353, 2007.
[243]  K. D. Sutherland, G. J. Lindeman, D. Y. H. Choong et al., “Differential hypermethylation of SOCS genes in ovarian and breast carcinomas,” Oncogene, vol. 23, no. 46, pp. 7726–7733, 2004.
[244]  Y. Oshimo, K. Kuraoka, H. Nakayama et al., “Epigenetic inactivation of SOCS-1 by CpG island hypermethylation in human gastric carcinoma,” International Journal of Cancer, vol. 112, no. 6, pp. 1003–1009, 2004.
[245]  H. Nagai, Y. S. Kim, K.-T. Lee et al., “Inactivation of SSI-1, a JAK/STAT inhibitor, in human hepatocellular carcinomas, as revealed by two-dimensional electrophoresis,” Journal of Hepatology, vol. 34, no. 3, pp. 416–421, 2001.
[246]  B. J. Camp, S. T. Dyhrman, V. A. Memoli, L. A. Mott, and R. J. Barth Jr., “In situ cytokine production by breast cancer tumor-infiltrating lymphocytes,” Annals of Surgical Oncology, vol. 3, no. 2, pp. 176–184, 1996.
[247]  T. Hanada, T. Kobayashi, T. Chinen et al., “IFNγ-dependent, spontaneous development of colorectal carcinomas in SOCS1-deficient mice,” Journal of Experimental Medicine, vol. 203, no. 6, pp. 1391–1397, 2006.
[248]  R. Rottapel, S. Ilangumaran, C. Neale et al., “The tumor suppressor activity of SOCS-1,” Oncogene, vol. 21, no. 28, pp. 4351–4362, 2002.
[249]  V. Calabrese, F. A. Mallette, X. Deschênes-Simard et al., “SOCS1 links cytokine signaling to p53 and senescence,” Molecular Cell, vol. 36, no. 5, pp. 754–767, 2009.
[250]  F. A. Mallette, V. Calabrese, S. Ilangumaran, and G. Ferbeyre, “SOCS1, a novel interaction partner of p53 controlling oncogene-induced senescence,” Aging, vol. 2, no. 7, pp. 445–452, 2010.
[251]  Y. Gui, M. Yeganeh, S. Ramanathan et al., “SOCS1 controls liver regeneration by regulating HGF signaling in hepatocytes,” Journal of Hepatology, vol. 55, no. 6, pp. 1300–1308, 2011.
[252]  F. Bogazzi, F. Ultimieri, F. Raggi et al., “Changes in the expression of suppressor of cytokine signalling (SOCS) 2 in the colonic mucosa of acromegalic patients are associated with hyperplastic polyps,” Clinical Endocrinology, vol. 70, no. 6, pp. 898–906, 2009.
[253]  A. Yoshimura, “Signal transduction of inflammatory cytokines and tumor development,” Cancer Science, vol. 97, no. 6, pp. 439–447, 2006.
[254]  H. Ogata, T. Kobayashi, T. Chinen et al., “Deletion of the SOCS3 gene in liver parenchymal cells promotes hepatitis-induced hepatocarcinogenesis,” Gastroenterology, vol. 131, no. 1, pp. 179–193, 2006.
[255]  A. Weber, U. R. Hengge, W. Bardenheuer et al., “SOCS-3 is frequently methylated in head and neck squamous cell carcinoma and its precursor lesions and causes growth inhibition,” Oncogene, vol. 24, no. 44, pp. 6699–6708, 2005.
[256]  M. Puhr, F. R. Santer, H. Neuwirt, G. Marcias, A. Hobisch, and Z. Culig, “SOCS-3 antagonises the proliferative and migratory effects of fibroblast growth factor-2 in prostate cancer by inhibition of p44/p42 MAPK signalling,” Endocrine-Related Cancer, vol. 17, no. 2, pp. 525–538, 2010.
[257]  K. Iwahori, S. Serada, M. Fujimoto et al., “Overexpression of SOCS3 exhibits preclinical antitumor activity against malignant pleural mesothelioma,” International Journal of Cancer, vol. 129, no. 4, pp. 1005–1017, 2011.
[258]  W. Sasi, W. G. Jiang, A. Sharma, and K. Mokbel, “Higher expression levels of SOCS 1,3,4,7 are associated with earlier tumour stage and better clinical outcome in human breast cancer,” BMC Cancer, vol. 10, article 178, 2010.
[259]  D. F. Calvisi, S. Ladu, A. Gorden et al., “Mechanistic and prognostic significance of aberrant methylation in the molecular pathogenesis of human hepatocellular carcinoma,” The Journal of Clinical Investigation, vol. 117, no. 9, pp. 2713–2722, 2007.
[260]  D. Kobayashi, S. Nomoto, Y. Kodera et al., “Suppressor of cytokine signaling 4 detected as a novel gastric cancer suppressor gene using double combination array analysis,” World Journal of Surgery, vol. 36, no. 2, pp. 362–372, 2012.
[261]  C. J. Scheitz, T. S. Lee, D. J. McDermitt, and T. Tumbar, “Defining a tissue stem cell-driven Runx1/Stat3 signalling axis in epithelial cancer,” The EMBO Journal, vol. 31, no. 21, pp. 4124–4139, 2012.
[262]  M. G. Francipane, V. Eterno, V. Spina et al., “Suppressor of cytokine signaling 3 sensitizes anaplastic thyroid cancer to standard chemotherapy,” Cancer Research, vol. 69, no. 15, pp. 6141–6148, 2009.
[263]  R.-H. Lai, Y.-W. Hsiao, M.-J. Wang et al., “SOCS6, down-regulated in gastric cancer, inhibits cell proliferation and colony formation,” Cancer Letters, vol. 288, no. 1, pp. 75–85, 2010.
[264]  K. B. Sriram, J. E. Larsen, S. M. Savarimuthu Francis et al., “Array-comparative genomic hybridization reveals loss of SOCS6 is associated with poor prognosis in primary lung squamous cell carcinoma,” PLoS ONE, vol. 7, no. 2, Article ID e30398, 2012.
[265]  S. Yoon, Y.-S. Yi, S. S. Kim, J.-H. Kim, W. S. Park, and S. W. Nam, “SOCS5 and SOCS6 have similar expression patterns in normal and cancer tissues,” Tumor Biology, vol. 33, no. 1, pp. 215–221, 2012.
[266]  S. R. Hubbard, “Juxtamembrane autoinhibition in receptor tyrosine kinases,” Nature Reviews Molecular Cell Biology, vol. 5, no. 6, pp. 464–470, 2004.
[267]  N. J. Dibb, S. M. Dilworth, and C. D. Mol, “Switching on kinases: oncogenic activation of BRAF and the PDGFR family,” Nature Reviews Cancer, vol. 4, no. 9, pp. 718–727, 2004.
[268]  F. Zadjali, A. C. W. Pike, M. Vesterlund et al., “Structural basis for c-KIT inhibition by the suppressor of cytokine signaling 6 (SOCS6) ubiquitin ligase,” The Journal of Biological Chemistry, vol. 286, no. 1, pp. 480–490, 2011.
[269]  X. H. Liu, S. B. Xu, J. Yuan et al., “Defective interleukin-4/Stat6 activity correlates with increased constitutive expression of negative regulators SOCS-3, SOCS-7, and CISH in colon cancer cells,” Journal of Interferon and Cytokine Research, vol. 29, no. 12, pp. 809–816, 2009.
[270]  I. Melzner, M. A. Weniger, A. J. Bucur et al., “Biallelic deletion within 16p13.13 including SOCS-1 in Karpas1106P mediastinal B-cell lymphonia line is associated with delayed degradation of JAK2 protein,” International Journal of Cancer, vol. 118, no. 8, pp. 1941–1944, 2006.
[271]  A. Yoshimura, T. Naka, and M. Kubo, “SOCS proteins, cytokine signalling and immune regulation,” Nature Reviews Immunology, vol. 7, no. 6, pp. 454–465, 2007.
[272]  T. Kishimoto and H. Kikutani, “Knocking the SOCS off a tumor suppressor,” Nature Genetics, vol. 28, no. 1, pp. 4–5, 2001.
[273]  T. Torisu, M. Nakaya, S. Watanabe et al., “Suppressor of cytokine signaling 1 protects mice against concanavalin A-induced hepatitis by inhibiting apoptosis,” Hepatology, vol. 47, no. 5, pp. 1644–1654, 2008.
[274]  J. S. Lee, N. S. Paek, O. S. Kwon, and K. B. Hahm, “Anti-inflammatory actions of probiotics through activating suppressor of cytokine signaling (SOCS) expression and signaling in Helicobacter pylori infection: a novel mechanism,” Journal of Gastroenterology and Hepatology, vol. 25, no. 1, pp. 194–202, 2010.
[275]  A. Yoshimura, “Regulation of cytokine signaling by the SOCS and Spred family proteins,” Keio Journal of Medicine, vol. 58, no. 2, pp. 73–83, 2009.
[276]  Y. Niwa, H. Kanda, Y. Shikauchi et al., “Methylation silencing of SOCS-3 promotes cell growth and migration by enhancing JAK/STAT and FAK signalings in human hepatocellular carcinoma,” Oncogene, vol. 24, no. 42, pp. 6406–6417, 2005.
[277]  T. Tokita, C. Maesawa, T. Kimura et al., “Methylation status of the SOCS3 gene in human malignant melanomas,” International Journal of Oncology, vol. 30, no. 3, pp. 689–694, 2007.
[278]  H. Isomoto, “Epigenetic alterations in cholangiocarcinoma-sustained IL-6/STAT3 signaling in cholangio- carcinoma due to SOCS3 epigenetic silencing,” Digestion, vol. 79, supplement 1, pp. 2–8, 2009.
[279]  H. Isomoto, J. L. Mott, S. Kobayashi et al., “Sustained IL-6/STAT-3 signaling in cholangiocarcinoma cells due to SOCS-3 epigenetic silencing,” Gastroenterology, vol. 132, no. 1, pp. 384–396, 2007.
[280]  H. Ogata, T. Chinen, T. Yoshida et al., “Loss of SOCS3 in the liver promotes fibrosis by enhancing STAT3-mediated TGF-β1 production,” Oncogene, vol. 25, no. 17, pp. 2520–2530, 2006.
[281]  J. Bollrath, T. J. Phesse, V. A. von Burstin et al., “gp130-mediated Stat3 activation in enterocytes regulates cell survival and cell-cycle progression during colitis-associated tumorigenesis,” Cancer Cell, vol. 15, no. 2, pp. 91–102, 2009.
[282]  S. Grivennikov, E. Karin, J. Terzic et al., “IL-6 and STAT3 are required for survival of intestinal epithelial cells and development of colitis associated cancer,” Cancer Cell, vol. 15, no. 2, pp. 103–113, 2009.
[283]  R. J. Rigby, J. G. Simmons, C. J. Greenhalgh, W. S. Alexander, and P. K. Lund, “Suppressor of cytokine signaling 3 (SOCS3) limits damage-induced crypt hyper-proliferation and inflammation-associated tumorigenesis in the colon,” Oncogene, vol. 26, no. 33, pp. 4833–4841, 2007.
[284]  Y. Li, J. Deuring, M. P. Peppelenbosch, E. J. Kuipers, C. de Haar, and C. J. van der Woude, “IL-6-induced DNMT1 activity mediates SOCS3 promoter hypermethylation in ulcerative colitis-related colorectal cancer,” Carcinogenesis, vol. 33, no. 10, pp. 1889–1896, 2012.
[285]  A. Ventura, D. G. Kirsch, M. E. McLaughlin et al., “Restoration of p53 function leads to tumour regression in vivo,” Nature, vol. 445, no. 7128, pp. 661–665, 2007.
[286]  D. Hanahan and R. A. Weinberg, “The hallmarks of cancer,” Cell, vol. 100, no. 1, pp. 57–70, 2000.
[287]  R. Sotillo, E. Hernando, E. Díaz-Rodríguez et al., “Mad2 overexpression promotes aneuploidy and tumorigenesis in mice,” Cancer Cell, vol. 11, no. 1, pp. 9–23, 2007.
[288]  O. Galm, S. Wilop, J. Reichelt et al., “DNA methylation changes in multiple myeloma,” Leukemia, vol. 18, no. 10, pp. 1687–1692, 2004.
[289]  L. Zheng and W.-H. Lee, “Retinoblastoma tumor suppressor and genome stability,” Advances in Cancer Research, vol. 85, pp. 13–50, 2002.
[290]  S. Mazoyer, “Genomic rearrangements in the BRCA1 and BRCA2 genes,” Human Mutation, vol. 25, no. 5, pp. 415–422, 2005.
[291]  T. F. Barth, I. Melzner, S. Wegener et al., “Biallelic mutation of SOCS-1 impairs JAK2 degradation and sustains phospho-JAK2 action in MedB-1 mediastinal lymphoma line,” Verhandlungen der Deutschen Gesellschaft für Pathologie, vol. 89, pp. 234–244, 2005.
[292]  D. Capello, A. Gloghini, G. Baldanzi et al., “Alterations of negative regulators of cytokine signalling in immunodeficiency-related non-Hodgkin lymphoma,” Hematological Oncology, vol. 31, no. 1, pp. 22–28, 2013.
[293]  M. Esteller, “Epigenetics provides a new generation of oncogenes and tumour-suppressor genes,” British Journal of Cancer, vol. 94, supplement 2, pp. 179–183, 2006.
[294]  B. Peng, D. R. Hodge, S. B. Thomas et al., “Epigenetic silencing of the human nucleotide excision repair gene, hHR23B, in interleukin-6-responsive multiple myeloma KAS-6/1 cells,” The Journal of Biological Chemistry, vol. 280, no. 6, pp. 4182–4187, 2005.
[295]  H. Wehbe, R. Henson, F. Meng, J. Mize-Berge, and T. Patel, “Interleukin-6 contributes to growth in cholangiocarcinoma cells by aberrant promoter methylation and gene expression,” Cancer Research, vol. 66, no. 21, pp. 10517–10524, 2006.
[296]  Y. Souma, T. Nishida, S. Serada et al., “Antiproliferative effect of SOCS-1 through the suppression of STAT3 and p38 MAPK activation in gastric cancer cells,” International Journal of Cancer, vol. 131, no. 6, pp. 1287–1296, 2012.
[297]  B. Yang, M. Guo, J. G. Herman, and D. P. Clark, “Aberrant promoter methylation profiles of tumor suppressor genes in hepatocellular carcinoma,” American Journal of Pathology, vol. 163, no. 3, pp. 1101–1107, 2003.
[298]  K. Brakensiek, F. L?nger, B. Schlegelberger, H. Kreipe, and U. Lehmann, “Hypermethylation of the suppressor of cytokine signalling-1 (SOCS-1) in myelodysplastic syndrome,” British Journal of Haematology, vol. 130, no. 2, pp. 209–217, 2005.
[299]  S. Wilop, T. B. van Gemmeren, M. H. F. M. Lentjes et al., “Methylation-associated dysregulation of the suppressor of cytokine signaling-3 gene in multiple myeloma,” Epigenetics, vol. 6, no. 8, pp. 1047–1052, 2011.
[300]  N. Fukushima, N. Sato, F. Sahin, G. H. Su, R. H. Hruban, and M. Goggins, “Aberrant methylation of suppressor of cytokine signalling-1 (SOCS-1) gene in pancreatic ductal neoplasms,” British Journal of Cancer, vol. 89, no. 2, pp. 338–343, 2003.
[301]  B. Li, W. Liu, L. Wang et al., “CpG island methylator phenotype associated with tumor recurrence in tumor-node-metastasis stage I hepatocellular carcinoma,” Annals of Surgical Oncology, vol. 17, no. 7, pp. 1917–1926, 2010.
[302]  D. Capello, C. Deambrogi, D. Rossi et al., “Epigenetic inactivation of suppressors of cytokine signalling in Philadelphia-negative chronic myeloproliferative disorders,” British Journal of Haematology, vol. 141, no. 4, pp. 504–511, 2008.
[303]  Y. Li, C. de Haar, M. Chen et al., “Disease-related expression of the IL6/STAT3/SOCS3 signalling pathway in ulcerative colitis and ulcerative colitis-related carcinogenesis,” Gut, vol. 59, no. 2, pp. 227–235, 2010.
[304]  C. Lindemann, O. Hackmann, S. Delic, N. Schmidt, G. Reifenberger, and M. J. Riemenschneider, “SOCS3 promoter methylation is mutually exclusive to EGFR amplification in gliomas and promotes glioma cell invasion through STAT3 and FAK activation,” Acta Neuropathologica, vol. 122, no. 2, pp. 241–251, 2011.
[305]  F. Pierconti, M. Martini, F. Pinto et al., “Epigenetic silencing of SOCS3 identifies a subset of prostate cancer with an aggressive behavior,” The Prostate, vol. 71, no. 3, pp. 318–325, 2011.
[306]  Q. Yuan, P. D. Li, B. H. Li et al., “Differential IL-4/Stat6 activities correlate with differential expression of regulatory genes SOCS-1, SHP-1, and PP2A in colon cancer cells,” Journal of Cancer Research and Clinical Oncology, vol. 135, no. 1, pp. 131–140, 2009.
[307]  S. B. Xu, X. H. Liu, B. H. Li et al., “DNA methylation regulates constitutive expression of Stat6 regulatory genes SOCS-1 and SHP-1 in colon cancer cells,” Journal of Cancer Research and Clinical Oncology, vol. 135, no. 12, pp. 1791–1798, 2009.
[308]  I. Storojeva, J.-L. Boulay, K. Heinimann et al., “Prognostic and predictive relevance of microsatellite instability in colorectal cancer,” Oncology Reports, vol. 14, no. 1, pp. 241–249, 2005.
[309]  I. Storojeva, J.-L. Boulay, P. Ballabeni et al., “Prognostic and predictive relevance of DNAM-1, SOCS6 and CADH-7 genes on chromosome 18q in colorectal cancer,” Oncology, vol. 68, no. 2-3, pp. 246–255, 2005.
[310]  X. Qiu, G. Guo, K. Chen et al., “A requirement for SOCS-1 and SOCS-3 phosphorylation in Bcr-Abl-induced tumorigenesis,” Neoplasia, vol. 14, no. 6, pp. 547–558, 2012.
[311]  A. Limnander, N. N. Danial, and P. B. Rothman, “v-Abl signaling disrupts SOCS-1 function in transformed pre-B cells,” Molecular Cell, vol. 15, no. 3, pp. 329–341, 2004.
[312]  J.-L. Chen, A. Limnander, and P. B. Rothman, “Pim-1 and Pim-2 kinases are required for efficient pre-B-cell transformation by v-Abl oncogene,” Blood, vol. 111, no. 3, pp. 1677–1685, 2008.
[313]  R. Amaravadi and C. B. Thompson, “The survival kinases Akt and Pim as potential pharmacological targets,” The Journal of Clinical Investigation, vol. 115, no. 10, pp. 2618–2624, 2005.
[314]  M. Adam, V. Pogacic, M. Bendit et al., “Targeting PIM kinases impairs survival of hematopoietic cells transformed by kinase inhibitor-sensitive and kinase inhibitor-resistant forms of Fms-like tyrosine kinase 3 and BCR/ABL,” Cancer Research, vol. 66, no. 7, pp. 3828–3835, 2006.
[315]  X. P. Chen, J. A. Losman, S. Cowan et al., “Pim serine/threonine kinases regulate the stability of Socs-1 protein,” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 4, pp. 2175–2180, 2002.
[316]  E. White, “The pims and outs of survival signaling: role for the Pim-2 protein kinase in the suppression of apoptosis by cytokines,” Genes and Development, vol. 17, no. 15, pp. 1813–1816, 2003.
[317]  H. Mikkers, M. Nawijn, J. Allen et al., “Mice deficient for all PIM kinases display reduced body size and impaired responses to hematopoietic growth factors,” Molecular and Cellular Biology, vol. 24, no. 13, pp. 6104–6115, 2004.
[318]  S. Haan, P. Ferguson, U. Sommer et al., “Tyrosine phosphorylation disrupts elongin interaction and accelerates SOCS3 degradation,” The Journal of Biological Chemistry, vol. 278, no. 34, pp. 31972–31979, 2003.
[319]  M. Shi, J. C. Cooper, and C.-L. Yu, “A constitutively active Lck kinase promotes cell proliferation and resistance to apoptosis through signal transducer and activator of transcription 5b activation,” Molecular Cancer Research, vol. 4, no. 1, pp. 39–45, 2006.
[320]  J. C. Cooper, M. Shi, F.-Y. Chueh, S. Venkitachalam, and C.-L. Yu, “Enforced SOCS1 and SOCS3 expression attenuates Lck-mediated cellular transformation,” International Journal of Oncology, vol. 36, no. 5, pp. 1201–1208, 2010.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413