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...

Androgen Receptor-Target Genes in African American Prostate Cancer Disparities

DOI: 10.1155/2013/763569

Full-Text   Cite this paper   Add to My Lib

Abstract:

The incidence and mortality rates of prostate cancer (PCa) are higher in African American (AA) compared to Caucasian American (CA) men. To elucidate the molecular mechanisms underlying PCa disparities, we employed an integrative approach combining gene expression profiling and pathway and promoter analyses to investigate differential transcriptomes and deregulated signaling pathways in AA versus CA cancers. A comparison of AA and CA PCa specimens identified 1,188 differentially expressed genes. Interestingly, these transcriptional differences were overrepresented in signaling pathways that converged on the androgen receptor (AR), suggesting that the AR may be a unifying oncogenic theme in AA PCa. Gene promoter analysis revealed that 382 out of 1,188 genes contained cis-acting AR-binding sequences. Chromatin immunoprecipitation confirmed STAT1, RHOA, ITGB5, MAPKAPK2, CSNK2A,1 and PIK3CB genes as novel AR targets in PCa disparities. Moreover, functional screens revealed that androgen-stimulated AR binding and upregulation of RHOA, ITGB5, and PIK3CB genes were associated with increased invasive activity of AA PCa cells, as siRNA-mediated knockdown of each gene caused a loss of androgen-stimulated invasion. In summation, our findings demonstrate that transcriptional changes have preferentially occurred in multiple signaling pathways converging (“transcriptional convergence”) on AR signaling, thereby contributing to AR-target gene activation and PCa aggressiveness in AAs. 1. Introduction Prostate cancer (PCa) is the most commonly diagnosed noncutaneous cancer and, after lung and bronchus cancers, the second leading cause of cancer deaths among American men [1, 2]. In the United States, it is estimated that 241,740 men will be newly diagnosed with prostate cancer, and 28,170 will succumb to this disease in 2012 (http://www.cancer.gov/cancertopics/types/prostate). The human androgen receptor (AR) plays a critical role in the growth and differentiation of the normal prostate gland as well as in the development of PCa [3, 4]. AR expression has been observed in nearly all primary PCa cases [5–7]. Previous studies have also shown that the cellular AR levels are correlated to primary and metastatic lesions and associated with disease progression to castration-resistant PCa (CRPCa) [8–10]. In the US, the African American (AA) population exhibits higher incidence and mortality rates of PCa compared to the Caucasian American (CA) population [11]. Accumulating evidence has suggested that biological factors may in part play a critical role in PCa health disparities that

References

[1]  A. Jemal, R. Siegel, E. Ward, Y. Hao, J. Xu, and M. J. Thun, “Cancer statistics, 2009,” CA: Cancer Journal for Clinicians, vol. 59, no. 4, pp. 225–249, 2009.
[2]  A. Jemal, R. Siegel, E. Ward et al., “Cancer statistics, 2008,” CA: Cancer Journal for Clinicians, vol. 58, no. 2, pp. 71–96, 2008.
[3]  M. M. Shen and C. Abate-Shen, “Molecular genetics of prostate cancer: new prospects for old challenges,” Genes and Development, vol. 24, no. 18, pp. 1967–2000, 2010.
[4]  E. P. Gelmann, “Molecular biology of the androgen receptor,” Journal of Clinical Oncology, vol. 20, no. 13, pp. 3001–3015, 2002.
[5]  J. A. R. de Winter, P. J. A. Janssen, H. M. E. B. Sleddens et al., “Androgen receptor status in localized and locally progressive hormone refractory human prostate cancer,” American Journal of Pathology, vol. 144, no. 4, pp. 735–746, 1994.
[6]  G. W. Chodak, D. M. Kranc, L. A. Puy, H. Takeda, K. Johnson, and C. Chang, “Nuclear localization of androgen receptor in heterogeneous samples of normal, hyperplastic and neoplastic human prostate,” Journal of Urology, vol. 147, no. 3, pp. 798–803, 1992.
[7]  M. V. Sadi, P. C. Walsh, and E. R. Barrack, “Immunohistochemical study of androgen receptors in metastatic prostate cancer: comparison of receptor content and response to hormonal therapy,” Cancer, vol. 67, no. 12, pp. 3057–3064, 1991.
[8]  S. M. Henshall, D. I. Quinn, C. S. Lee et al., “Altered expression of androgen receptor in the malignant epithelium and adjacent stroma is associated with early relapse in prostate cancer,” Cancer Research, vol. 61, no. 2, pp. 423–427, 2001.
[9]  C. Ricciardelli, C. S. Choong, G. Buchanan et al., “Androgen receptor levels in prostate cancer epithelial and peritumoral stromal cells identify non-organ confined disease,” Prostate, vol. 63, no. 1, pp. 19–28, 2005.
[10]  C. D. Chen, D. S. Welsbie, C. Tran, et al., “Molecular determinants of resistance to antiandrogen therapy,” Nature Medicine, vol. 10, no. 1, pp. 33–39, 2004.
[11]  S. Reddy, M. Shapiro, R. Morton, and O. W. Brawley, “Prostate cancer in black and white Americans,” Cancer and Metastasis Reviews, vol. 22, no. 1, pp. 83–86, 2003.
[12]  R. Ross, L. Bernstein, and H. Judd, “Serum testosterone levels in healthy young black and white men,” Journal of the National Cancer Institute, vol. 76, no. 1, pp. 45–48, 1986.
[13]  L. Ellis and H. Nyborg, “Racial/ethnic variations in male testosterone levels: a probable contributor to group differences in health,” Steroids, vol. 57, no. 2, pp. 72–75, 1992.
[14]  K. E. Gaston, D. Kim, S. Singh, O. H. Ford, and J. L. Mohler, “Racial differences in androgen receptor protein expression in men with clinically localized prostate cancer,” Journal of Urology, vol. 170, no. 3, pp. 990–993, 2003.
[15]  J. K. V. Reichardt, N. Makridakis, B. E. Henderson, M. C. Yu, M. C. Pike, and R. K. Ross, “Genetic variability of the human SRD5A2 gene: implications for prostate cancer risk,” Cancer Research, vol. 55, no. 18, pp. 3973–3975, 1995.
[16]  N. M. Makridakis, R. K. Ross, M. C. Pike et al., “Association of mis-sense substitution in SRD5A2 gene with prostate cancer in African-American and Hispanic men in Los Angeles, USA,” Lancet, vol. 354, no. 9183, pp. 975–978, 1999.
[17]  A. Edwards, H. A. Hammond, L. Jin, C. T. Caskey, and R. Chakraborty, “Genetic variation at five trimeric and tetrameric tandem repeat loci in four human population groups,” Genomics, vol. 12, no. 2, pp. 241–253, 1992.
[18]  N. L. Chamberlain, E. D. Driver, and R. L. Miesfeld, “The length and location of CAG trinucleotide repeats in the androgen receptor N-terminal domain affect transactivation function,” Nucleic Acids Research, vol. 22, no. 15, pp. 3181–3186, 1994.
[19]  O. Sartor, Q. Zheng, and J. A. Eastham, “Androgen receptor gene CAG repeat length varies in a race-specific fashion in men without prostate cancer,” Urology, vol. 53, no. 2, pp. 378–380, 1999.
[20]  R. A. Irvine, M. C. Yu, R. K. Ross, and G. A. Coetzee, “The CAG and GGC microsatellites of the androgen receptor gene are in linkage disequilibrium in men with prostate cancer,” Cancer Research, vol. 55, no. 9, pp. 1937–1940, 1995.
[21]  E. Giovannucci, M. J. Stampfer, K. Krithivas et al., “The CAG repeat within the androgen receptor gene and its relationship to prostate cancer,” Proceedings of the National Academy of Sciences of the United States of America, vol. 94, no. 7, pp. 3320–3333, 1997.
[22]  S. A. Ingles, R. K. Ross, M. C. Yu et al., “Association of prostate cancer risk with genetic polymorphisms in vitamin D receptor and androgen receptor,” Journal of the National Cancer Institute, vol. 89, no. 2, pp. 166–170, 1997.
[23]  E. A. Platz, E. Giovannucci, D. M. Dahl et al., “The androgen receptor gene GGN microsatellite and prostate cancer risk,” Cancer Epidemiology Biomarkers and Prevention, vol. 7, no. 5, pp. 379–384, 1998.
[24]  Y. P. Yu, D. Landsittel, L. Jing et al., “Gene expression alterations in prostate cancer predicting tumor aggression and preceding development of malignancy,” Journal of Clinical Oncology, vol. 22, no. 14, pp. 2790–2799, 2004.
[25]  J. Luo, D. J. Duggan, Y. Chen et al., “Human prostate cancer and benign prostatic hyperplasia: molecular dissection by gene expression profiling,” Cancer Research, vol. 61, no. 12, pp. 4683–4688, 2001.
[26]  J. B. Welsh, L. M. Sapinoso, A. I. Su et al., “Analysis of gene expression identifies candidate markers and pharmacological targets in prostate cancer,” Cancer Research, vol. 61, no. 16, pp. 5974–5978, 2001.
[27]  J. Lapointe, C. Li, J. P. Higgins, et al., “Gene expression profiling identifies clinically relevant subtypes of prostate cancer,” Proceedings of the National Academy of Sciences of the United States of America, vol. 101, no. 3, pp. 811–816, 2004.
[28]  A. J. Stephenson, A. Smith, M. W. Kattan et al., “Integration of gene expression profiling and clinical variables to predict prostate carcinoma recurrence after radical prostatectomy,” Cancer, vol. 104, no. 2, pp. 290–298, 2005.
[29]  D. Singh, P. G. Febbo, K. Ross et al., “Gene expression correlates of clinical prostate cancer behavior,” Cancer Cell, vol. 1, no. 2, pp. 203–209, 2002.
[30]  S. Nanni, V. Benvenuti, A. Grasselli et al., “Endothelial NOS, estrogen receptor β, and HIFs cooperate in the activation of a prognostic transcriptional pattern in aggressive human prostate cancer,” Journal of Clinical Investigation, vol. 119, no. 5, pp. 1093–1108, 2009.
[31]  E. K. Markert, H. Mizuno, A. Vazquez, and A. J. Levine, “Molecular classification of prostate cancer using curated expression signatures,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 52, pp. 21276–21281, 2011.
[32]  T. A. Wallace, R. L. Prueitt, M. Yi et al., “Tumor immunobiological differences in prostate cancer between African-American and European-American men,” Cancer Research, vol. 68, no. 3, pp. 927–936, 2008.
[33]  R. R. Reams, D. Agrawal, M. B. Davis et al., “Microarray comparison of prostate tumor gene expression in African-American and Caucasian American males: a pilot project study,” Infectious Agents and Cancer, vol. 4, supplement 1, article S3, 2009.
[34]  S. Ngan, E. A. Stronach, A. Photiou, J. Waxman, S. Ali, and L. Buluwela, “Microarray coupled to quantitative RT-PCR analysis of androgen-regulated genes in human LNCaP prostate cancer cells,” Oncogene, vol. 28, no. 19, pp. 2051–2063, 2009.
[35]  B. D. Wang, C. L. B. Kline, D. M. Pastor et al., “Prostate apoptosis response protein 4 sensitizes human colon cancer cells to chemotherapeutic 5-FU through mediation of an NFκB and microRNA network,” Molecular Cancer, vol. 9, article 98, 2010.
[36]  H. Teramoto, M. D. Castellone, R. L. Malek et al., “Autocrine activation of an osteopontin-CD44-Rac pathway enhances invasion and transformation by H-RasV12,” Oncogene, vol. 24, no. 3, pp. 489–501, 2005.
[37]  M. D. Abramoff, P. J. Magalhaes, and S. J. Ram, “Image Processing with ImageJ,” Biophotonics International, vol. 11, pp. 36–42, 2004.
[38]  C. D. House, C. J. Vaske, A. M. Schwartz et al., “Voltage-gated Na+ channel SCN5A is a key regulator of a gene transcriptional network that controls colon cancer invasion,” Cancer Research, vol. 70, no. 17, pp. 6957–6967, 2010.
[39]  K. J. Livak and T. D. Schmittgen, “Analysis of relative gene expression data using real-time quantitative PCR and the 2-ΔΔCT method,” Methods, vol. 25, no. 4, pp. 402–408, 2001.
[40]  N. Fujimoto, S. Yeh, H. Y. Kang et al., “Cloning and characterization of androgen receptor coactivator, ARA55, in human prostate,” The Journal of Biological Chemistry, vol. 274, no. 12, pp. 8316–8321, 1999.
[41]  Y. Miyoshi, H. Ishiguro, H. Uemura et al., “Expression of AR associated protein 55 (ARA55) and androgen receptor in prostate cancer,” Prostate, vol. 56, no. 4, pp. 280–286, 2003.
[42]  D. Wu, T. L. Foreman, C.W. Gregory, et al., “Protein kinase cepsilon has the potential to advance the recurrence of human prostate cancer,” Cancer Research, vol. 62, no. 8, pp. 2423–2429, 2002.
[43]  M. Garcia-Marcos, P. Ghosh, and M. G. Farquhar, “Molecular basis of a novel oncogenic mutation in GNAO1,” Oncogene, vol. 30, no. 23, pp. 2691–2696, 2011.
[44]  E. R. Sharlow, K. V. Giridhar, C. R. LaValle et al., “Potent and selective disruption of protein kinase D functionality by a benzoxoloazepinolone,” The Journal of Biological Chemistry, vol. 283, no. 48, pp. 33516–33526, 2008.
[45]  M. Alur, M. M. Nguyen, S. E. Eggener et al., “Suppressive roles of calreticulin in prostate cancer growth and metastasis,” American Journal of Pathology, vol. 175, no. 2, pp. 882–890, 2009.
[46]  S. Korenchuk, J. E. Lehr, L. McLean et al., “VCaP, a cell-based model system of human prostate cancer,” In Vivo, vol. 15, no. 2, pp. 163–168, 2001.
[47]  S. Koochekpour, G. A. Maresh, A. Katner et al., “Establishment and characterization of a primary androgen-responsive African-American prostate cancer cell line, E006AA,” Prostate, vol. 60, no. 2, pp. 141–152, 2004.
[48]  N. M. Navone, M. Olive, M. Ozen et al., “Establishment of two human prostate cancer cell lines derived from a single bone metastasis,” Clinical Cancer Research, vol. 3, no. 12 I, pp. 2493–2500, 1997.
[49]  K. R. Chng, C. W. Chang, S. K. Tan, et al., “A transcriptional repressor co-regulatory network governing androgen response in prostate cancers,” The EMBO Journal, vol. 31, no. 12, pp. 2810–2823, 2012.
[50]  R. A. S. Hemat, Principles of Orthomolecularism, Urotext, 2004.
[51]  H. Touge, H. Chikumi, T. Igishi et al., “Diverse activation states of RhoA in human lung cancer cells: contribution of G protein coupled receptors,” International Journal of Oncology, vol. 30, no. 3, pp. 709–715, 2007.
[52]  S. Wee, D. Wiederschain, S. M. Maira et al., “PTEN-deficient cancers depend on PIK3CB,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 35, pp. 13057–13062, 2008.
[53]  R. J. Crowder, C. Phommaly, Y. Tao et al., “PIK3CA and PIK3CB inhibition produce synthetic lethality when combined with estrogen deprivation in estrogen receptor-positive breast cancer,” Cancer Research, vol. 69, no. 9, pp. 3955–3962, 2009.
[54]  M. T. Abraham, M. A. Kuriakose, P. G. Sacks et al., “Motility-related proteins as markers for head and neck squamous cell cancer,” Laryngoscope, vol. 111, no. 7, pp. 1285–1289, 2001.
[55]  T. Kamai, K. Arai, S. Sumi et al., “The rho/rho-kinase pathway is involved in the progression of testicular germ cell tumour,” British Journal of Urology International, vol. 89, no. 4, pp. 449–453, 2002.
[56]  J. C. Hodge, J. Bub, S. Kaul, A. Kajdacsy-Balla, and P. F. Lindholm, “Requirement of RhoA activity for increased nuclear factor κB activity and PC-3 human prostate cancer cell invasion,” Cancer Research, vol. 63, no. 6, pp. 1359–1364, 2003.
[57]  Y. S. Hwang, J. C. Hodge, N. Sivapurapu, and P. F. Lindholm, “Lysophosphatidic acid stimulates PC-3 prostate cancer cell matrigel invasion through activation of RhoA and NF-κB activity,” Molecular Carcinogenesis, vol. 45, no. 7, pp. 518–529, 2006.
[58]  C. Zhou, M. T. Ling, T. Kin-Wah Lee, K. Man, X. Wang, and Y. C. Wong, “FTY720, a fungus metabolite, inhibits invasion ability of androgen-independent prostate cancer cells through inactivation of RhoA-GTPase,” Cancer Letters, vol. 233, no. 1, pp. 36–47, 2006.
[59]  R. Zheng, A. Iwase, R. Shen et al., “Neuropeptide-stimulated cell migration in prostate cancer cells is mediated by RhoA kinase signaling and inhibited by neutral endopeptidase,” Oncogene, vol. 25, no. 44, pp. 5942–5952, 2006.
[60]  D. L. Greenberg, G. J. Mize, and T. K. Takayama, “Protease-activated receptor mediated RhoA signaling and cytoskeletal reorganization in LNCaP cells,” Biochemistry, vol. 42, no. 3, pp. 702–709, 2003.
[61]  K. F. Decker, D. Zheng, Y. He, T. Bowman, J. R. Edwards, and L. Jia, “Persistent androgen receptor-mediated transcription in castration-resistant prostate cancer under androgen-deprived conditions,” Nucleic Acids Research, vol. 40, no. 21, pp. 10765–10779, 2012.
[62]  D. A. Hosack, G. Dennis, B. T. Sherman, H. C. Lane, and R. A. Lempicki, “Identifying biological themes within lists of genes with EASE,” Genome Biology, vol. 4, no. 10, article R70, 2003.
[63]  R. M. Kypta and J. Waxman, “Wnt/beta-catenin signalling in prostate cancer,” Nature Reviews Urology, vol. 9, pp. 418–428, 2012.
[64]  D. Gioeli, J. W. Mandell, G. R. Petroni, H. F. Frierson, and M. J. Weber, “Activation of mitogen-activated protein kinase associated with prostate cancer progression,” Cancer Research, vol. 59, no. 2, pp. 279–284, 1999.
[65]  D. Gioeli, “Signal transduction in prostate cancer progression,” Clinical Science, vol. 108, no. 4, pp. 293–308, 2005.
[66]  D. J. Mulholland, S. Dedhar, H. Wu, and C. C. Nelson, “PTEN and GSK3β: key regulators of progression to androgen-independent prostate cancer,” Oncogene, vol. 25, no. 3, pp. 329–337, 2006.
[67]  M. M. Shen and C. Abate-Shen, “Pten inactivation and the emergence of androgen-independent prostate cancer,” Cancer Research, vol. 67, no. 14, pp. 6535–6538, 2007.
[68]  Y. Wen, M. C. T. Hu, K. Makino et al., “HER-2/neu promotes androgen-independent survival and growth of prostate cancer cells through the Akt pathway,” Cancer Research, vol. 60, no. 24, pp. 6841–6845, 2000.
[69]  C. I. Truica, S. Byers, and E. P. Gelmann, “β-catenin affects androgen receptor transcriptional activity and ligand specificity,” Cancer Research, vol. 60, no. 17, pp. 4709–4713, 2000.
[70]  D. J. Mulholland, H. Cheng, K. Reid, P. S. Rennie, and C. C. Nelson, “The androgen receptor can promote β-catenin nuclear translocation independently of adenomatous polyposis coli,” The Journal of Biological Chemistry, vol. 277, no. 20, pp. 17933–17943, 2002.
[71]  D. J. Mulholland, J. T. Read, P. S. Rennie, M. E. Cox, and C. C. Nelson, “Functional localization and competition between the androgen receptor and T-cell factor for nuclear β-catenin: a means for inhibition of the Tcf signaling axis,” Oncogene, vol. 22, no. 36, pp. 5602–5613, 2003.
[72]  G. Chen, N. Shukeir, A. Potti et al., “Up-regulation of Wnt-1 and β-catenin production in patients with advanced metastatic prostate carcinoma: potential pathogenetic and prognostic implications,” Cancer, vol. 101, no. 6, pp. 1345–1356, 2004.
[73]  Z. Culig, A. Hobisch, M. V. Cronauer et al., “Androgen receptor activation in prostatic tumor cell lines by insulin-like growth factor-I, keratinocyte growth factor, and epidermal growth factor,” Cancer Research, vol. 54, no. 20, pp. 5474–5478, 1994.
[74]  G. Wang, S. J. M. Jones, M. A. Marra, and M. D. Sadar, “Identification of genes targeted by the androgen and PKA signaling pathways in prostate cancer cells,” Oncogene, vol. 25, no. 55, pp. 7311–7323, 2006.
[75]  C. E. Massie, A. Lynch, A. Ramos-Montoya et al., “The androgen receptor fuels prostate cancer by regulating central metabolism and biosynthesis,” The EMBO Journal, vol. 30, no. 13, pp. 2719–2733, 2011.
[76]  B. Lin, J. Wang, X. Hong et al., “Integrated expression profiling and ChIP-seq analyses of the growth inhibition response program of the androgen receptor,” PLoS ONE, vol. 4, no. 8, Article ID e6589, 2009.
[77]  C. E. Massie, B. Adryan, N. L. Barbosa-Morais et al., “New androgen receptor genomic targets show an interaction with the ETS1 transcription factor,” EMBO Reports, vol. 8, no. 9, pp. 871–878, 2007.
[78]  L. J. Schmidt, K. Duncan, N. Yadav, et al., “RhoA as a mediator of clinically relevant androgen action in prostate cancer cells,” Molecular Endocrinology, vol. 26, no. 5, pp. 716–735.
[79]  R. B. Nagle, J. D. Knox, C. Wolf, G. T. Bowden, and A. E. Cress, “Adhesion molecules, extracellular matrix, and proteases in prostate carcinoma,” Journal of Cellular Biochemistry, vol. 56, no. 19, pp. 232–237, 1994.
[80]  J. D. Knox, A. E. Cress, V. Clark et al., “Differential expression of extracellular matrix molecules and the α6-integrins in the normal and neoplastic prostate,” American Journal of Pathology, vol. 145, no. 1, pp. 167–174, 1994.
[81]  S. J. Murant, J. Handley, M. Stower, N. Reid, O. Cussenot, and N. J. Maitland, “Co-ordinated changes in expression of cell adhesion molecules in prostate cancer,” European Journal of Cancer A, vol. 33, no. 2, pp. 263–271, 1997.
[82]  D. Q. Zheng, A. S. Woodard, M. Fornaro, G. Tallini, and L. R. Languino, “Prostatic carcinoma cell migration via α(v)β3 integrin is modulated by a focal adhesion kinase pathway,” Cancer Research, vol. 59, no. 7, pp. 1655–1664, 1999.
[83]  I. P. Gorlov, J. Byun, O. Y. Gorlova, A. M. Aparicio, E. Efstathiou, and C. J. Logothetis, “Candidate pathways and genes for prostate cancer: a meta-analysis of gene expression data,” BMC Medical Genomics, vol. 2, article 48, 2009.
[84]  Q. Zhu, H. Youn, J. Tang et al., “Phosphoinositide 3-OH kinase p85α and p110β are essential for androgen receptor transactivation and tumor progression in prostate cancers,” Oncogene, vol. 27, no. 33, pp. 4569–4579, 2008.
[85]  S. Jia, Z. Liu, S. Zhang et al., “Essential roles of PI(3)K-p110β in cell growth, metabolism and tumorigenesis,” Nature, vol. 454, no. 7205, pp. 776–779, 2008.
[86]  K. M. Hill, S. Kalifa, J. R. Das et al., “The role of PI 3-kinase p110β in AKT signally, cell survival, and proliferation in human prostate cancer cells,” Prostate, vol. 70, no. 7, pp. 755–764, 2010.
[87]  N. El-Hashemite, H. Zhang, V. Walker, K. M. Hoffmeister, and D. J. Kwiatkowski, “Perturbed IFN-γ-Jak-signal transducers and activators of transcription signaling in tuberous sclerosis mouse models: synergistic effects of rapamycin-IFN-γ treatment,” Cancer Research, vol. 64, no. 10, pp. 3436–3443, 2004.
[88]  R. Buettner, L. B. Mora, and R. Jove, “Activated STAT signaling in human tumors provides novel molecular targets for therapeutic intervention,” Clinical Cancer Research, vol. 8, no. 4, pp. 945–954, 2002.
[89]  J. J. O'Shea, “Jaks, STATs, cytokine signal transduction, and immunoregulation: are we there yet?” Immunity, vol. 7, no. 1, pp. 1–11, 1997.
[90]  O. Cochet, C. Frelin, J. F. Peyron, and V. Imbert, “Constitutive activation of STAT proteins in the HDLM-2 and L540 Hodgkin lymphoma-derived cell lines supports cell survival,” Cellular Signalling, vol. 18, no. 4, pp. 449–455, 2006.
[91]  S. G. Patterson, S. Wei, X. Chen et al., “Novel role of Stat1 in the development of docetaxel resistance in prostate tumor cells,” Oncogene, vol. 25, no. 45, pp. 6113–6122, 2006.
[92]  L. D. Wood, D. W. Parsons, S. Jones et al., “The genomic landscapes of human breast and colorectal cancers,” Science, vol. 318, no. 5853, pp. 1108–1113, 2007.
[93]  S. Jones, X. Zhang, D. W. Parsons et al., “Core signaling pathways in human pancreatic cancers revealed by global genomic analyses,” Science, vol. 321, no. 5897, pp. 1801–1806, 2008.
[94]  L. Ding, G. Getz, D. A. Wheeler et al., “Somatic mutations affect key pathways in lung adenocarcinoma,” Nature, vol. 455, no. 7216, pp. 1069–1075, 2008.
[95]  R. McLendon, A. Friedman, D. Bigner et al., “Comprehensive genomic characterization defines human glioblastoma genes and core pathways,” Nature, vol. 455, no. 7216, pp. 1061–1068, 2008.
[96]  P. S. Hammerman, D. N. Hayes, M. D. Wilkerson, et al., “Comprehensive genomic characterization of squamous cell lung cancers,” Nature, vol. 489, no. 7417, pp. 519–525, 2012.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413