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...

MicroRNAs and Glucocorticoid-Induced Apoptosis in Lymphoid Malignancies

DOI: 10.1155/2013/348212

Full-Text   Cite this paper   Add to My Lib

Abstract:

The initial response of lymphoid malignancies to glucocorticoids (GCs) is a critical parameter predicting successful treatment. Although being known as a strong inducer of apoptosis in lymphoid cells for almost a century, the signaling pathways regulating the susceptibility of the cells to GCs are only partly revealed. There is still a need to develop clinical tests that can predict the outcome of GC therapy. In this paper, I discuss important parameters modulating the pro-apoptotic effects of GCs, with a specific emphasis on the microRNA world comprised of small players with big impacts. The journey through the multifaceted complexity of GC-induced apoptosis brings forth explanations for the differential treatment response and raises potential strategies for overcoming drug resistance. 1. Introduction 1.1. Glucocorticoids in the Treatment of Lymphoid Malignancies Glucocorticoids (GCs) are among the most effective drugs used in the treatment of hematopoietic malignancies of the lymphoid lineage in virtue of their ability to induce apoptosis of these cancerous cells [1–3]. The main hematopoietic cancer types that respond well to GC therapy include T acute lymphoblastic leukemia (T-ALL), chronic B lymphocytic leukemia (CLL), multiple myeloma (MM), Hodgkin’s lymphoma (HL), and non-Hodgkin’s lymphoma (NHL). GCs appear, however, to have little value in the treatment of acute or chronic myeloid leukemia (AML/CML). A major drawback of GC therapy is the gradual development of resistance to GC during treatment that limits the clinical utility of this drug. Poor response to a 7-day monotherapy with the GC prednisone is one of the strongest predictors of adverse outcomes in the treatment of pediatric ALL [2, 4]. A great challenge today is to develop strategies that can overcome the drug resistant phenotype. For this purpose it is important to understand the underlying mechanisms of GC resistance and the signaling pathways regulating apoptosis induced by GCs. Besides inducing apoptosis of lymphoid cells, GCs are used in palliative care. GC treatment produces rapid symptomatic improvements, including relief of fever, sweats, lethargy, weakness, and other nonspecific effects of cancer. GCs decrease the severity of chemotherapy-induced emesis. GCs are also used in the clinics for other medical conditions such as autoimmune diseases, asthma, ulcerative colitis, chronic obstructive pulmonary disease, kidney diseases, and rheumatologic disorders due to their strong anti-inflammatory and immunosuppressive properties. GC therapy is hampered by a variety of metabolic and

References

[1]  C. H. Pui, A. J. Gajjar, J. R. Kane, I. A. Qaddoumi, and A. S. Pappo, “Challenging issues in pediatric oncology,” Nature Reviews Clinical Oncology, vol. 8, pp. 540–549, 2011.
[2]  H. Inaba and C. H. Pui, “Glucocorticoid use in acute lymphoblastic leukaemia,” The Lancet Oncology, vol. 11, no. 11, pp. 1096–1106, 2010.
[3]  J. O. Armitage, “The aggressive peripheral T-cell lymphomas: 2012 update on diagnosis, risk stratification, and management,” American Journal of Hematology, vol. 87, pp. 511–519, 2012.
[4]  N. Jiang, G. S. Koh, J. Y. Suang Lim et al., “BIM is a prognostic biomarker for early prednisolone response in pediatric acute lymphoblastic leukemia,” Experimental Hematology, vol. 39, no. 3, pp. 321.e3–329.e3, 2011.
[5]  D. den Uyl, I. E. M. Bultink, and W. F. Lems, “Advances in glucocorticoid-induced osteoporosis,” Current Rheumatology Reports, vol. 13, no. 3, pp. 233–240, 2011.
[6]  W. G. Dixon and N. Bansback, “Understanding the side effects of glucocorticoid therapy: shining a light on a drug everyone thinks they know,” Annals of the Rheumatic Diseases, vol. 71, no. 11, pp. 1761–1764, 2012.
[7]  J. L. Beaudry and M. C. Riddell, “Effects of glucocorticoids and exercise on pancreatic β-cell function and diabetes development,” Diabetes/Metabolism Research and Reviews, vol. 28, no. 7, pp. 560–573, 2012.
[8]  A. J?rns, C. Sennholz, O. Naujok, and S. Lenzen, “Beta cell mass regulation in the rat pancreas through glucocorticoids and thyroid hormones,” Pancreas, vol. 39, no. 8, pp. 1167–1172, 2010.
[9]  R. S. Weinstein, “Glucocorticoid-induced osteoporosis and osteonecrosis,” Endocrinology and Metabolism Clinics of North America, vol. 41, no. 3, pp. 595–611, 2012.
[10]  M. C. Lansang and L. K. Hustak, “Glucocorticoid-induced diabetes and adrenal suppression: how to detect and manage them,” Cleveland Clinic Journal of Medicine, vol. 78, no. 11, pp. 748–756, 2011.
[11]  E. Reich, A. Tamary, R. V. Sionov, and D. Melloul, “Involvement of thioredoxin-interacting protein (TXNIP) in glucocorticoid-mediated beta cell death,” Diabetologia, vol. 55, pp. 1048–1057, 2012.
[12]  L. X. Wang, Y. P. Wang, Z. Chen et al., “Exendin-4 protects murine pancreatic β-cells from dexamethasone-induced apoptosis through PKA and PI-3K signaling,” Diabetes Research and Clinical Practice, vol. 90, no. 3, pp. 297–304, 2010.
[13]  F. Ranta, D. Avram, S. Berchtold et al., “Dexamethasone induces cell death in insulin-secreting cells, an effect reversed by exendin-4,” Diabetes, vol. 55, no. 5, pp. 1380–1390, 2006.
[14]  P. Moutsatsou, E. Kassi, and A. G. Papavassiliou, “Glucocorticoid receptor signaling in bone cells,” Trends in Molecular Medicine, vol. 18, pp. 348–359, 2012.
[15]  S. I. Yun, H. Y. Yoon, S. Y. Jeong, and Y. S. Chung, “Glucocorticoid induces apoptosis of osteoblast cells through the activation of glycogen synthase kinase 3β,” Journal of Bone and Mineral Metabolism, vol. 27, no. 2, pp. 140–148, 2009.
[16]  H. Li, W. Qian, X. Weng, et al., “Glucocorticoid receptor and sequential P53 activation by dexamethasone mediates apoptosis and cell cycle arrest of osteoblastic MC3T3-E1 cells,” PLoS ONE, vol. 7, Article ID e37030, 2012.
[17]  E. Y. H. Wong and J. Herbert, “Raised circulating corticosterone inhibits neuronal differentiation of progenitor cells in the adult hippocampus,” Neuroscience, vol. 137, no. 1, pp. 83–92, 2006.
[18]  J. L. Mayer, L. Klumpers, S. Maslam, E. R. de Kloet, M. Jo?ls, and P. J. Lucassen, “Brief treatment with the glucocorticoid receptor antagonist mifepristone normalises the corticosterone-induced reduction of adult hippocampal neurogenesis,” Journal of Neuroendocrinology, vol. 18, no. 8, pp. 629–631, 2006.
[19]  M. Hallek, K. Fischer, G. Fingerle-Rowson et al., “Addition of rituximab to fludarabine and cyclophosphamide in patients with chronic lymphocytic leukaemia: a randomised, open-label, phase 3 trial,” The Lancet, vol. 376, no. 9747, pp. 1164–1174, 2010.
[20]  T. Robak, A. Dmoszynska, P. Solal-Céligny et al., “Rituximab plus fludarabine and cyclophosphamide prolongs progression-free survival compared with fludarabine and cyclophosphamide alone in previously treated chronic lymphocytic leukemia,” Journal of Clinical Oncology, vol. 28, no. 10, pp. 1756–1765, 2010.
[21]  A. R. Pettitt, R. Jackson, S. Carruthers, et al., “Alemtuzumab in combination with methylprednisolone is a highly effective induction regimen for patients with chronic lymphocytic leukemia and deletion of TP53: final results of the national cancer research institute CLL206 trial,” Journal of Clinical Oncology, vol. 30, pp. 1647–1655, 2012.
[22]  N. Skoetz, K. Bauer, T. Elter et al., “Alemtuzumab for patients with chronic lymphocytic leukaemia,” Cochrane Database of Systematic Reviews, vol. 2, Article ID CD008078, 2012.
[23]  A. Cortelezzi, M. Sciume, and G. Reda, “Lenalidomide in the treatment of chronic lymphocytic leukemia,” Advances in Hematology, vol. 2012, Article ID 393864, 2012.
[24]  M. Gentile, A. G. Recchia, E. Vigna et al., “Lenalidomide in the treatment of chronic lymphocytic leukemia,” Expert Opinion on Investigational Drugs, vol. 20, no. 2, pp. 273–286, 2011.
[25]  A. Younes, N. L. Bartlett, J. P. Leonard et al., “Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas,” The New England Journal of Medicine, vol. 363, no. 19, pp. 1812–1821, 2010.
[26]  I. N. Micallef, M. J. Maurer, G. A. Wiseman, et al., “Epratuzumab with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone chemotherapy in patients with previously untreated diffuse large B-cell lymphoma,” Blood, vol. 118, pp. 4053–4061, 2011.
[27]  M. Cives, A. Milano, F. Dammacco, and F. Silvestris, “Lenalidomide in multiple myeloma: current experimental and clinical data,” European Journal of Haematology, vol. 88, pp. 279–291, 2012.
[28]  L. J. Scott and K. A. Lyseng-Williamson, “Spotlight on lenalidomide in relapsed or refractory multiple myeloma,” BioDrugs, vol. 25, pp. 333–337, 2011.
[29]  P. S. Bachmann, R. G. Piazza, M. E. Janes et al., “Epigenetic silencing of BIM in glucocorticoid poor-responsive pediatric acute lymphoblastic leukemia, and its reversal by histone deacetylase inhibition,” Blood, vol. 116, no. 16, pp. 3013–3022, 2010.
[30]  R. V. Sionov, R. Spokoini, S. Kfir-Erenfeld, O. Cohen, and E. Yefenof, “Chapter 6 mechanisms regulating the susceptibility of hematopoietic malignancies to glucocorticoid-induced apoptosis,” Advances in Cancer Research, vol. 101, pp. 127–248, 2008.
[31]  S. Kfir-Erenfeld, R. V. Sionov, R. Spokoini, O. Cohen, and E. Yefenof, “Protein kinase networks regulating glucocorticoid-induced apoptosis of hematopoietic cancer cells: fundamental aspects and practical considerations,” Leukemia and Lymphoma, vol. 51, no. 11, pp. 1968–2005, 2010.
[32]  J. C. Reed, “Bcl-2-family proteins and hematologic malignancies: history and future prospects,” Blood, vol. 111, no. 7, pp. 3322–3330, 2008.
[33]  T. Oltersdorf, S. W. Elmore, A. R. Shoemaker et al., “An inhibitor of Bcl-2 family proteins induces regression of solid tumours,” Nature, vol. 435, no. 7042, pp. 677–681, 2005.
[34]  V. Del Gaizo Moore, K. D. Schlis, S. E. Sallan, S. A. Armstrong, and A. Letai, “BCL-2 dependence and ABT-737 sensitivity in acute lymphoblastic leukemia,” Blood, vol. 111, no. 4, pp. 2300–2309, 2008.
[35]  J. Deng, N. Carlson, K. Takeyama, P. Dal Cin, M. Shipp, and A. Letai, “BH3 profiling identifies three distinct classes of apoptotic blocks to predict response to ABT-737 and conventional chemotherapeutic agents,” Cancer Cell, vol. 12, no. 2, pp. 171–185, 2007.
[36]  P. N. Kelly, S. Grabow, A. R. Delbridge, J. M. Adams, and A. Strasser, “Prophylactic treatment with the BH3 mimetic ABT-737 impedes Myc-driven lymphomagenesis in mice,” Cell Death and Differentiation, vol. 20, pp. 57–63, 2013.
[37]  L. C. Spender and G. J. Inman, “Phosphoinositide 3-kinase/AKT/mTORC1/2 signaling determines sensitivity of Burkitt's lymphoma cells to BH3 mimetics,” Molecular Cancer Research, vol. 10, pp. 347–359, 2012.
[38]  V. D. G. Moore, J. R. Brown, M. Certo, T. M. Love, C. D. Novina, and A. Letai, “Chronic lymphocytic leukemia requires BCL2 to sequester prodeath BIM, explaining sensitivity to BCL2 antagonist ABT-737,” Journal of Clinical Investigation, vol. 117, no. 1, pp. 112–121, 2007.
[39]  M. H. Kang, H. K. Kang, B. Szymanska et al., “Activity of vincristine, L-ASP, and dexamethasone against acute lymphoblastic leukemia is enhanced by the BH3-mimetic ABT-737 in vitro and in vivo,” Blood, vol. 110, no. 6, pp. 2057–2066, 2007.
[40]  B. Szymanska, U. Wilczynska-Kalak, M. H. Kang, et al., “Pharmacokinetic modeling of an induction regimen for in vivo combined testing of novel drugs against pediatric acute lymphoblastic leukemia xenografts,” PLoS ONE, vol. 7, Article ID e33894, 2012.
[41]  M. P. Kline, S. V. Rajkumar, M. M. Timm et al., “ABT-737, an inhibitor of Bcl-2 family proteins, is a potent inducer of apoptosis in multiple myeloma cells,” Leukemia, vol. 21, no. 7, pp. 1549–1560, 2007.
[42]  D. Chauhan, M. Velankar, M. Brahmandam et al., “A novel Bcl-2/Bcl-XL/Bcl-w inhibitor ABT-737 as therapy in multiple myeloma,” Oncogene, vol. 26, no. 16, pp. 2374–2380, 2007.
[43]  L. Gandhi, D. R. Camidge, M. R. de Oliveira et al., “Phase I study of navitoclax (ABT-263), a novel bcl-2 family inhibitor, in patients with small-cell lung cancer and other solid tumors,” Journal of Clinical Oncology, vol. 29, no. 7, pp. 909–916, 2011.
[44]  A. W. Roberts, J. F. Seymour, J. Brown, et al., “Substantial susceptibility of chronic lymphocytic leukemia to BCL2 inhibition: results of a phase I study of navitoclax in patients with relapsed or refractory disease,” Journal of Clinical Oncology, vol. 30, pp. 488–496, 2012.
[45]  C. Tse, A. R. Shoemaker, J. Adickes et al., “ABT-263: a potent and orally bioavailable Bcl-2 family inhibitor,” Cancer Research, vol. 68, no. 9, pp. 3421–3428, 2008.
[46]  W. H. Wilson, O. A. O'Connor, M. S. Czuczman et al., “Navitoclax, a targeted high-affinity inhibitor of BCL-2, in lymphoid malignancies: a phase 1 dose-escalation study of safety, pharmacokinetics, pharmacodynamics, and antitumour activity,” The Lancet Oncology, vol. 11, no. 12, pp. 1149–1159, 2010.
[47]  S. Ackler, Y. Xiao, M. J. Mitten et al., “ABT-263 and rapamycin act cooperatively to kill lymphoma cells in vitro and in vivo,” Molecular Cancer Therapeutics, vol. 7, no. 10, pp. 3265–3274, 2008.
[48]  D. Yecies, N. E. Carlson, J. Deng, and A. Letai, “Acquired resistance to ABT-737 in lymphoma cells that up-regulate MCL-1 and BFL-1,” Blood, vol. 115, no. 16, pp. 3304–3313, 2010.
[49]  J. L. Coloff, A. N. Macintyre, A. G. Nichols et al., “Akt-dependent glucose metabolism promotes Mcl-1 synthesis to maintain cell survival and resistance to Bcl-2 inhibition,” Cancer Research, vol. 71, no. 15, pp. 5204–5213, 2011.
[50]  O. Meynet, M. Beneteau, M. A. Jacquin et al., “Glycolysis inhibition targets Mcl-1 to restore sensitivity of lymphoma cells to ABT-737-induced apoptosis,” Leukemia, vol. 26, pp. 1145–1147, 2012.
[51]  S. M. O'Brien, D. F. Claxton, M. Crump et al., “Phase I study of obatoclax mesylate (GX15-070), a small molecule pan Bcl-2 family antagonist, in patients with advanced chronic lymphocytic leukemia,” Blood, vol. 113, no. 2, pp. 299–305, 2009.
[52]  A. D. Schimmer, S. O'Brien, H. Kantarjian et al., “A phase i study of the pan bcl-2FamilyInhibitor obatoclax mesylate in patients with advanced hematologic malignancies,” Clinical Cancer Research, vol. 14, no. 24, pp. 8295–8301, 2008.
[53]  J. Joudeh and D. Claxton, “Obatoclax mesylate: pharmacology and potential for therapy of hematological neoplasms,” Expert Opinion on Investigational Drugs, vol. 21, pp. 363–373, 2012.
[54]  M. Nguyen, R. C. Marcellus, A. Roulston et al., “Small molecule obatoclax (GX15-070) antagonizes MCL-1 and overcomes MCL-1-mediated resistance to apoptosis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 49, pp. 19512–19517, 2007.
[55]  P. Pérez-Galán, G. Roué, N. Villamor, E. Campo, and D. Colomer, “The BH3-mimetic GX15-070 synergizes with bortezomib in mantle cell lymphoma by enhancing Noxa-mediated activation of Bak,” Blood, vol. 109, no. 10, pp. 4441–4449, 2007.
[56]  S. Chen, Y. Dai, X. Y. Pei, et al., “CDK inhibitors upregulate BH3-only proteins to sensitize human myeloma cells to BH3 mimetic therapies,” Cancer Research, vol. 72, pp. 4225–4237, 2012.
[57]  M. Rahmani, M. M. Aust, E. Attkisson et al., “Inhibition of Bcl-2 anti-apoptotic members by obatoclax potently enhances sorafenib-induced apoptosis in human myeloid leukemia cells through a Bim-dependent process,” Blood, vol. 119, pp. 6089–6098, 2012.
[58]  N. Heidari, M. A. Hicks, and H. Harada, “GX15-070 (obatoclax) overcomes glucocorticoid resistance in acute lymphoblastic leukemia through induction of apoptosis and autophagy,” Cell Death and Disease, vol. 1, no. 9, article e76, 2010.
[59]  L. Bonapace, B. C. Bornhauser, M. Schmitz et al., “Induction of autophagy-dependent necroptosis is required for childhood acute lymphoblastic leukemia cells to overcome glucocorticoid resistance,” Journal of Clinical Investigation, vol. 120, no. 4, pp. 1310–1323, 2010.
[60]  R. Kang, H. J. Zeh, M. T. Lotze, and D. Tang, “The Beclin 1 network regulates autophagy and apoptosis,” Cell Death and Differentiation, vol. 18, no. 4, pp. 571–580, 2011.
[61]  W. Declercq, T. Vanden Berghe, and P. Vandenabeele, “RIP kinases at the crossroads of cell death and survival,” Cell, vol. 138, no. 2, pp. 229–232, 2009.
[62]  M. Fakler, S. Loeder, M. Vogler et al., “Small molecule XIAP inhibitors cooperate with TRAIL to induce apoptosis in childhood acute leukemia cells and overcome Bcl-2-mediated resistance,” Blood, vol. 113, no. 8, pp. 1710–1722, 2009.
[63]  L. P. Frenzel, M. Patz, C. P. Pallasch et al., “Novel X-linked inhibitor of apoptosis inhibiting compound as sensitizer for TRAIL-mediated apoptosis in chronic lymphocytic leukaemia with poor prognosis,” British Journal of Haematology, vol. 152, no. 2, pp. 191–200, 2011.
[64]  S. Loeder, A. Drensek, I. Jeremias, K. M. Debatin, and S. Fulda, “Small molecule XIAP inhibitors sensitize childhood acute leukemia cells for CD95-induced apoptosis,” International Journal of Cancer, vol. 126, no. 9, pp. 2216–2228, 2010.
[65]  P. Hundsdoerfer, I. Dietrich, K. Schmelz, C. Eckert, and G. Henze, “XIAP expression is post-transcriptionally upregulated in childhood ALL and is associated with glucocorticoid response in T-cell ALL,” Pediatric Blood and Cancer, vol. 55, no. 2, pp. 260–266, 2010.
[66]  M. L. Deftos, Y. W. He, E. W. Ojala, and M. J. Bevan, “Correlating notch signaling with thymocyte maturation,” Immunity, vol. 9, no. 6, pp. 777–786, 1998.
[67]  R. Spokoini, S. Kfir-Erenfeld, E. Yefenof, and R. V. Sionov, “Glycogen synthase kinase-3 plays a central role in mediating glucocorticoid-induced apoptosis,” Molecular Endocrinology, vol. 24, no. 6, pp. 1136–1150, 2010.
[68]  H. Sade, S. Krishna, and A. Sarin, “The anti-apoptotic effect of Notch-1 requires p56lck-dependent, Akt/PKB-mediated signaling in T cells,” The Journal of Biological Chemistry, vol. 279, no. 4, pp. 2937–2944, 2004.
[69]  F. Radtke, A. Wilson, G. Stark et al., “Deficient T cell fate specification in mice with an induced inactivation of Notch1,” Immunity, vol. 10, no. 5, pp. 547–558, 1999.
[70]  F. Radtke, N. Fasnacht, and H. R. MacDonald, “Notch signaling in the immune system,” Immunity, vol. 32, no. 1, pp. 14–27, 2010.
[71]  L. Yin, O. C. Velazquez, and Z. J. Liu, “Notch signaling: emerging molecular targets for cancer therapy,” Biochemical Pharmacology, vol. 80, no. 5, pp. 690–701, 2010.
[72]  E. Ersvaer, K. J. Hatfield, H. Reikvam, and O. Bruserud, “Future perspectives: therapeutic targeting of notch signalling may become a strategy in patients receiving stem cell transplantation for hematologic malignancies,” Bone Marrow Research, vol. 2011, Article ID 570796, 15 pages, 2011.
[73]  W. S. Pear, J. C. Aster, M. L. Scott et al., “Exclusive development of T cell neoplasms in mice transplanted with bone marrow expressing activated Notch alleles,” Journal of Experimental Medicine, vol. 183, no. 5, pp. 2283–2291, 1996.
[74]  D. Bellavia, A. F. Campese, E. Alesse et al., “Constitutive activation of NF-κB and T-cell leukemia/lymphoma in Notch3 transgenic mice,” EMBO Journal, vol. 19, no. 13, pp. 3337–3348, 2000.
[75]  W. S. Pear and J. C. Aster, “T cell acute lymphoblastic leukemia/lymphoma: a human cancer commonly associated with aberrant NOTCH1 signaling,” Current Opinion in Hematology, vol. 11, no. 6, pp. 426–433, 2004.
[76]  A. A. Ferrando, “The role of NOTCH1 signaling in T-ALL,” Hematology/the Education Program of the American Society of Hematology, pp. 353–361, 2009.
[77]  G. Tzoneva and A. A. Ferrando, “Recent advances on NOTCH signaling in T-ALL,” Current Topics in Microbiology and Immunology, vol. 360, pp. 163–182, 2012.
[78]  J. Pancewicz and C. Nicot, “Current views on the role of Notch signaling and the pathogenesis of human leukemia,” BMC Cancer, vol. 11, article 502, 2011.
[79]  M. Paganin and A. Ferrando, “Molecular pathogenesis and targeted therapies for NOTCH1-induced T-cell acute lymphoblastic leukemia,” Blood Reviews, vol. 25, no. 2, pp. 83–90, 2011.
[80]  L. W. Ellisen, J. Bird, D. C. West et al., “TAN-1, the human homolog of the Drosophila Notch gene, is broken by chromosomal translocations in T lymphoblastic neoplasms,” Cell, vol. 66, no. 4, pp. 649–661, 1991.
[81]  A. P. Weng, A. A. Ferrando, W. Lee et al., “Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia,” Science, vol. 306, no. 5694, pp. 269–271, 2004.
[82]  Z. Wang, H. Inuzuka, J. Zhong et al., “Tumor suppressor functions of FBW7 in cancer development and progression,” FEBS Letters, vol. 586, pp. 1409–1418, 2012.
[83]  T. Palomero and A. Ferrando, “Therapeutic targeting of NOTCH1 signaling in T-cell acute lymphoblastic leukemia,” Clinical Lymphoma & Myeloma, vol. 9, supplement 3, pp. S205–210, 2009.
[84]  K. Cullion, K. M. Draheim, N. Hermance et al., “Targeting the Notch1 and mTOR pathways in a mouse T-ALL model,” Blood, vol. 113, no. 24, pp. 6172–6181, 2009.
[85]  V. Manfè, E. Biskup, A. Rosbjerg et al., “miR-122 regulates p53/Akt signalling and the chemotherapy-induced apoptosis in cutaneous T-cell lymphoma,” PLoS ONE, vol. 7, Article ID e29541, 2012.
[86]  M. R. Kamstrup, E. Ralfkiaer, G. L. Skovgaard, and R. Gniadecki, “Potential involvement of Notch1 signalling in the pathogenesis of primary cutaneous CD30-positive lymphoproliferative disorders,” British Journal of Dermatology, vol. 158, no. 4, pp. 747–753, 2008.
[87]  H. Kogoshi, T. Sato, T. Koyama, N. Nara, and S. Tohda, “Gamma-secretase inhibitors suppress the growth of leukemia and lymphoma cells,” Oncology Reports, vol. 18, no. 1, pp. 77–80, 2007.
[88]  P. J. Real, V. Tosello, T. Palomero et al., “γ-secretase inhibitors reverse glucocorticoid resistance in T cell acute lymphoblastic leukemia,” Nature Medicine, vol. 15, no. 1, pp. 50–58, 2009.
[89]  T. Palomero, M. Dominguez, and A. A. Ferrando, “The role of the PTEN/AKT pathway in NOTCH1-induced leukemia,” Cell Cycle, vol. 7, no. 8, pp. 965–970, 2008.
[90]  T. Palomero, M. L. Sulis, M. Cortina et al., “Mutational loss of PTEN induces resistance to NOTCH1 inhibition in T-cell leukemia,” Nature Medicine, vol. 13, no. 10, pp. 1203–1210, 2007.
[91]  P. Wei, M. Walls, M. Qiu et al., “Evaluation of selective γ-secretase inhibitor PF-03084014 for its antitumor efficacy and gastrointestinal safety to guide optimal clinical trial design,” Molecular Cancer Therapeutics, vol. 9, no. 6, pp. 1618–1628, 2010.
[92]  J. B. Samon, M. Castillo-Martin, M. Hadler, et al., “Preclinical analysis of the gamma-secretase inhibitor PF-03084014 in combination with glucocorticoids in T-cell acute lymphoblastic leukemia,” Molecular Cancer Therapeutics, vol. 11, pp. 1565–1575, 2012.
[93]  R. E. Moellering, M. Cornejo, T. N. Davis et al., “Direct inhibition of the NOTCH transcription factor complex,” Nature, vol. 462, pp. 182–188, 2009.
[94]  A. Gutierrez and A. T. Look, “NOTCH and PI3K-AKT pathways intertwined,” Cancer Cell, vol. 12, no. 5, pp. 411–413, 2007.
[95]  S. K. Mungamuri, X. Yang, A. D. Thor, and K. Somasundaram, “Survival signaling by Notch1: mammalian target of rapamycin (mTOR)-dependent inhibition of p53,” Cancer Research, vol. 66, no. 9, pp. 4715–4724, 2006.
[96]  S. M. Chan, A. P. Weng, R. Tibshirani, J. C. Aster, and P. J. Utz, “Notch signals positively regulate activity of the mTOR pathway in T-cell acute lymphoblastic leukemia,” Blood, vol. 110, no. 1, pp. 278–286, 2007.
[97]  R. V. Sionov, “The kinome and glucocorticoid-induced apoptosis,” Chinese Journal of Cancer, vol. 27, no. 11, pp. 1121–1129, 2008.
[98]  A. S. Garza, A. L. Miller, B. H. Johnson, and E. B. Thompson, “Converting cell lines representing hematological malignancies from glucocorticoid-resistant to glucocorticoid-sensitive: signaling pathway interactions,” Leukemia Research, vol. 33, no. 5, pp. 717–727, 2009.
[99]  E. B. Thompson, “Stepping stones in the path of glucocorticoid-driven apoptosis of lymphoid cells,” Acta Biochimica et Biophysica Sinica, vol. 40, no. 7, pp. 595–600, 2008.
[100]  D. Ruggero, L. Montanaro, L. Ma et al., “The translation factor eIF-4E promotes tumor formation and cooperates with c-Myc in lymphomagenesis,” Nature Medicine, vol. 10, no. 5, pp. 484–486, 2004.
[101]  H. G. Wendel, E. De Stanchina, J. S. Fridman et al., “Survival signalling by Akt and eIF4E in oncogenesis and cancer therapy,” Nature, vol. 428, no. 6980, pp. 332–337, 2004.
[102]  F. Vega, L. J. Medeiros, V. Leventaki et al., “Activation of mammalian target of rapamycin signaling pathway contributes to tumor cell survival in anaplastic lymphoma kinase-positive anaplastic large cell lymphoma,” Cancer Research, vol. 66, no. 13, pp. 6589–6597, 2006.
[103]  E. Peponi, E. Drakos, G. Reyes, V. Leventaki, G. Z. Rassidakis, and L. J. Medeiros, “Activation of mammalian target of rapamycin signaling promotes cell cycle progression and protects cells from apoptosis in mantle cell lymphoma,” American Journal of Pathology, vol. 169, no. 6, pp. 2171–2180, 2006.
[104]  A. Gutierrez, T. Sanda, R. Grebliunaite et al., “High frequency of PTEN, PI3K, and AKT abnormalities in T-cell acute lymphoblastic leukemia,” Blood, vol. 114, no. 3, pp. 647–650, 2009.
[105]  A. Silva, J. A. Yunes, B. A. Cardoso et al., “PTEN posttranslational inactivation and hyperactivation of the PI3K/Akt pathway sustain primary T cell leukemia viability,” Journal of Clinical Investigation, vol. 118, no. 11, pp. 3762–3774, 2008.
[106]  D. Guo, Q. Teng, and C. Ji, “NOTCH and phosphatidylinositide 3-kinase/phosphatase and tensin homolog deleted on chromosome ten/AKT/mammalian target of rapamycin (mTOR) signaling in T-cell development and T-cell acute lymphoblastic leukemia,” Leukemia and Lymphoma, vol. 52, no. 7, pp. 1200–1210, 2011.
[107]  W. L. Zhao, “Targeted therapy in T-cell malignancies: dysregulation of the cellular signaling pathways,” Leukemia, vol. 24, no. 1, pp. 13–21, 2010.
[108]  A. L. Miller, A. S. Garza, B. H. Johnson, and E. B. Thompson, “Pathway interactions between MAPKs, mTOR, PKA, and the glucocorticoid receptor in lymphoid cells,” Cancer Cell International, vol. 7, article 3, 2007.
[109]  A. H. Beesley, M. J. Firth, J. Ford et al., “Glucocorticoid resistance in T-lineage acute lymphoblastic leukaemia is associated with a proliferative metabolism,” British Journal of Cancer, vol. 100, no. 12, pp. 1926–1936, 2009.
[110]  A. Batista, J. T. Barata, E. Raderschall et al., “Targeting of active mTOR inhibits primary leukemia T cells and synergizes with cytotoxic drugs and signaling inhibitors,” Experimental Hematology, vol. 39, no. 4, pp. 457.e3–472.e3, 2011.
[111]  C. Zhang, Y. K. Ryu, T. Z. Chen, C. P. Hall, D. R. Webster, and M. H. Kang, “Synergistic activity of rapamycin and dexamethasone in vitro and in vivo in acute lymphoblastic leukemia via cell-cycle arrest and apoptosis,” Leukemia Research, vol. 36, pp. 342–349, 2012.
[112]  G. Fuka, H. P. Kantner, R. Grausenburger, et al., “Silencing of ETV6/RUNX1 abrogates PI3K/AKT/mTOR signaling and impairs reconstitution of leukemia in xenografts,” Leukemia, vol. 26, pp. 927–933, 2012.
[113]  L. Gu, C. Zhou, H. Liu et al., “Rapamycin sensitizes T-ALL cells to dexamethasone-induced apoptosis,” Journal of Experimental and Clinical Cancer Research, vol. 29, no. 1, article 150, 2010.
[114]  L. Gu, J. Gao, Q. Li et al., “Rapamycin reverses NPM-ALK-induced glucocorticoid resistance in lymphoid tumor cells by inhibiting mTOR signaling pathway, enhancing G1 cell cycle arrest and apoptosis,” Leukemia, vol. 22, no. 11, pp. 2091–2096, 2008.
[115]  N. López-Royuela, P. Balsas, P. Galán-Malo, A. Anel, I. Marzo, and J. Naval, “Bim is the key mediator of glucocorticoid-induced apoptosis and of its potentiation by rapamycin in human myeloma cells,” Biochimica et Biophysica Acta, vol. 1803, no. 2, pp. 311–322, 2010.
[116]  H. Yan, P. Frost, Y. Shi et al., “Mechanism by which mammalian target of rapamycin inhibitors sensitize multiple myeloma cells to dexamethasone-induced apoptosis,” Cancer Research, vol. 66, no. 4, pp. 2305–2313, 2006.
[117]  T. Str?mberg, A. Dimberg, A. Hammarberg et al., “Rapamycin sensitizes multiple myeloma cells to apoptosis induced by dexamethasone,” Blood, vol. 103, no. 8, pp. 3138–3147, 2004.
[118]  C. Schult, M. Dahlhaus, A. Glass et al., “The dual kinase inhibitor NVP-BEZ235 in combination with cytotoxic drugs exerts anti-proliferative activity towards acute lymphoblastic leukemia cells,” Anticancer Research, vol. 32, pp. 463–474, 2012.
[119]  A. Kim, S. Park, J. E. Lee et al., “The dual PI3K and mTOR inhibitor NVP-BEZ235 exhibits anti-proliferative activity and overcomes bortezomib resistance in mantle cell lymphoma cells,” Leukemia Research, vol. 36, pp. 912–920, 2012.
[120]  S. Kfir, R. V. Sionov, E. Zafrir, Y. Zilberman, and E. Yefenof, “Staurosporine sensitizes T lymphoma cells to glucocorticoid-induced apoptosis: role of Nur77 and Bcl-2,” Cell Cycle, vol. 6, no. 24, pp. 3086–3096, 2007.
[121]  P. Robak and T. Robak, “A targeted therapy for protein and lipid kinases in chronic lymphocytic leukemia,” Current Medicinal Chemistry, vol. 19, no. 31, pp. 5294–5318, 2012.
[122]  S. Trudel, Z. H. Li, E. Wei et al., “CHIR-258, a novel, multitargeted tyrosine kinase inhibitor for the potential treatment of t(4;14) multiple myeloma,” Blood, vol. 105, no. 7, pp. 2941–2948, 2005.
[123]  M. Ohanian, J. Cortes, H. Kantarjian, and E. Jabbour, “Tyrosine kinase inhibitors in acute and chronic leukemias,” Expert Opinion on Pharmacotherapy, vol. 13, pp. 927–938, 2012.
[124]  G. P. Carlo, M. Cristina, and E. M. Pogliani, “Crizotinib in anaplastic large-cell lymphoma,” The New England Journal of Medicine, vol. 364, no. 8, pp. 775–776, 2011.
[125]  J. A. Burger, “Inhibiting B-cell receptor signaling pathways in chronic lymphocytic leukemia,” Current Hematologic Malignancy Reports, vol. 7, pp. 26–33, 2012.
[126]  R. E. Davis, V. N. Ngo, G. Lenz et al., “Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma,” Nature, vol. 463, no. 7277, pp. 88–92, 2010.
[127]  L. A. Honigberg, A. M. Smith, M. Sirisawad et al., “The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy,” Proceedings of the National Academy of Sciences of the United States of America, vol. 107, no. 29, pp. 13075–13080, 2010.
[128]  E. S. Winer, R. R. Ingham, and J. J. Castillo, “PCI-32765: a novel Bruton's tyrosine kinase inhibitor for the treatment of lymphoid malignancies,” Expert Opinion on Investigational Drugs, vol. 21, pp. 355–361, 2012.
[129]  J. A. Woyach, A. J. Johnson, and J. C. Byrd, “The B-cell receptor signaling pathway as a therapeutic target in CLL,” Blood, vol. 120, pp. 1175–1184, 2012.
[130]  S. Ponader, S. S. Chen, J. J. Buggy et al., “The Bruton tyrosine kinase inhibitor PCI-32765 thwarts chronic lymphocytic leukemia cell survival and tissue homing in vitro and in vivo,” Blood, vol. 119, pp. 1182–1189, 2012.
[131]  S. E. M. Herman, A. L. Gordon, E. Hertlein et al., “Bruton tyrosine kinase represents a promising therapeutic target for treatment of chronic lymphocytic leukemia and is effectively targeted by PCI-32765,” Blood, vol. 117, no. 23, pp. 6287–6296, 2011.
[132]  Y. T. Tai, B. Y. Chang, S. Y. Kong, et al., “Bruton tyrosine kinase inhibition is a novel therapeutic strategy targeting tumor in the bone marrow microenvironment in multiple myeloma,” Blood, vol. 120, pp. 1877–1887, 2012.
[133]  J. W. Friedberg, J. Sharman, J. Sweetenham et al., “Inhibition of Syk with fostamatinib disodium has significant clinical activity in non-Hodgkin lymphoma and chronic lymphocytic leukemia,” Blood, vol. 115, no. 13, pp. 2578–2585, 2010.
[134]  R. M. Young, I. R. Hardy, R. L. Clarke et al., “Mouse models of non-hodgkin lymphoma reveal Syk as an important therapeutic target,” Blood, vol. 113, no. 11, pp. 2508–2516, 2009.
[135]  W. Wang, M. Corrigan-Cummins, J. Hudson, et al., “MicroRNA profiling of follicular lymphoma identifies microRNAs related to cell proliferation and tumor response,” Haematologica, vol. 97, pp. 586–594, 2012.
[136]  B. J. Lannutti, S. A. Meadows, S. E. M. Herman et al., “CAL-101, a p110δ selective phosphatidylinositol-3-kinase inhibitor for the treatment of B-cell malignancies, inhibits PI3K signaling and cellular viability,” Blood, vol. 117, no. 2, pp. 591–594, 2011.
[137]  H. Ikeda, T. Hideshima, M. Fulciniti et al., “PI3K/p110δ is a novel therapeutic target in multiple myeloma,” Blood, vol. 116, no. 9, pp. 1460–1468, 2010.
[138]  J. J. Castillo, M. Furman, and E. S. Winer, “CAL-101: a phosphatidylinositol-3-kinase p110-delta inhibitor for the treatment of lymphoid malignancies,” Expert Opinion on Investigational Drugs, vol. 21, pp. 15–22, 2012.
[139]  S. T. Jou, N. Carpino, Y. Takahashi et al., “Essential, nonredundant role for the phosphoinositide 3-kinase p110δ in signaling by the B-cell receptor complex,” Molecular and Cellular Biology, vol. 22, no. 24, pp. 8580–8591, 2002.
[140]  I. Ringshausen, F. Schneller, C. Bogner et al., “Constitutively activated phosphatidylinositol-3 kinase (PI-3K) is involved in the defect of apoptosis in B-CLL: association with protein kinase Cδ,” Blood, vol. 100, no. 10, pp. 3741–3748, 2002.
[141]  S. D. Pauls, S. T. Lafarge, I. Landego, T. Zhang, and A. J. Marshall, “The phosphoinositide 3-kinase signaling pathway in normal and malignant B cells: activation mechanisms, regulation and impact on cellular functions,” Frontiers in Immunology, vol. 3, article 224, 2012.
[142]  G. V. Baracho, A. V. Miletic, S. A. Omori, M. H. Cato, and R. C. Rickert, “Emergence of the PI3-kinase pathway as a central modulator of normal and aberrant B cell differentiation,” Current Opinion in Immunology, vol. 23, no. 2, pp. 178–183, 2011.
[143]  B. Accordi, V. Espina, M. Giordan et al., “Functional protein network activation mapping reveals new potential molecular drug targets for poor prognosis pediatric BCP-ALL,” PLoS ONE, vol. 5, no. 10, Article ID e13552, 2010.
[144]  M. W. Harr, P. F. Caimi, K. S. McColl et al., “Inhibition of Lck enhances glucocorticoid sensitivity and apoptosis in lymphoid cell lines and in chronic lymphocytic leukemia,” Cell Death and Differentiation, vol. 17, no. 9, pp. 1381–1391, 2010.
[145]  G. G?rgün, E. Calabrese, T. Hideshima et al., “Anovel Aurora-A kinase inhibitor MLN8237 induces cytotoxicity and cell-cycle arrest in multiple myeloma,” Blood, vol. 115, no. 25, pp. 5202–5213, 2010.
[146]  J. N. Kuznetsov, G. J. Leclerc, G. M. Leclerc, and J. C. Barredo, “AMPK and Akt determine apoptotic cell death following perturbations of one-carbon metabolism by regulating ER stress in acute lymphoblastic leukemia,” Molecular Cancer Therapeutics, vol. 10, no. 3, pp. 437–447, 2011.
[147]  A. F. Santidrián, D. M. González-Gironès, D. Iglesias-Serret et al., “AICAR induces apoptosis independently of AMPK and p53 through up-regulation of the BH3-only proteins BIM and NOXAin chronic lymphocytic leukemia cells,” Blood, vol. 116, no. 16, pp. 3023–3032, 2010.
[148]  C. Campàs, J. M. López, A. F. Santidrián et al., “Acadesine activates AMPK and induces apoptosis in B-cell chronic lymphocytic leukemia cells but not in T lymphocytes,” Blood, vol. 101, no. 9, pp. 3674–3680, 2003.
[149]  C. Stefanelli, I. Stanic', F. Bonavita et al., “Inhibition of glucocorticoid-induced apoptosis with 5-aminoimidazole-4-carboxamide ribonucleoside, a cell-permeable activator of AMP-Activated protein kinase,” Biochemical and Biophysical Research Communications, vol. 243, no. 3, pp. 821–826, 1998.
[150]  G. Schlossmacher, A. Stevens, and A. White, “Glucocorticoid receptor-mediated apoptosis: mechanisms of resistance in cancer cells,” Journal of Endocrinology, vol. 211, pp. 17–25, 2011.
[151]  C. W. Distelhorst, “Recent insights into the mechanism of glucocorticosteroid-induced apoptosis,” Cell Death and Differentiation, vol. 9, no. 1, pp. 6–19, 2002.
[152]  L. K. Smith and J. A. Cidlowski, “Glucocorticoid-induced apoptosis of healthy and malignant lymphocytes,” Progress in Brain Research, vol. 182, pp. 1–30, 2010.
[153]  S. Schmidt, J. Rainer, C. Ploner, E. Presul, S. Riml, and R. Kofler, “Glucocorticoid-induced apoptosis and glucocorticoid resistance: molecular mechanisms and clinical relevance,” Cell Death and Differentiation, vol. 11, no. 1, pp. S45–S55, 2004.
[154]  R. Kumar and E. B. Thompson, “Transactivation functions of the N-terminal domains of nuclear hormone receptors: protein folding and coactivator interactions,” Molecular Endocrinology, vol. 17, no. 1, pp. 1–10, 2003.
[155]  J. D. Turner, S. R. Alt, L. Cao et al., “Transcriptional control of the glucocorticoid receptor: CpG islands, epigenetics and more,” Biochemical Pharmacology, vol. 80, no. 12, pp. 1860–1868, 2010.
[156]  M. J. M. Schaaf and J. A. Cidlowski, “Molecular mechanisms of glucocorticoid action and resistance,” Journal of Steroid Biochemistry and Molecular Biology, vol. 83, no. 1–5, pp. 37–48, 2002.
[157]  P. de Lange, C. M. Segeren, J. W. Koper et al., “Expression in hematological malignancies of a glucocorticoid receptor splice variant that augments glucocorticoid receptor-mediated effects in transfected cells,” Cancer Research, vol. 61, no. 10, pp. 3937–3941, 2001.
[158]  J. Zhou and J. A. Cidlowski, “The human glucocorticoid receptor: one gene, multiple proteins and diverse responses,” Steroids, vol. 70, no. 5–7, pp. 407–417, 2005.
[159]  M. Lauten, I. Fernandez-Munoz, K. Gerdes et al., “Kinetics of the in vivo expression of glucocorticoid receptor splice variants during prednisone treatment in childhood acute lymphoblastic leukaemia,” Pediatric Blood and Cancer, vol. 52, no. 4, pp. 459–463, 2009.
[160]  N. Z. Lu and J. A. Cidlowski, “Glucocorticoid receptor isoforms generate transcription specificity,” Trends in Cell Biology, vol. 16, no. 6, pp. 301–307, 2006.
[161]  D. Duma, C. M. Jewell, and J. A. Cidlowski, “Multiple glucocorticoid receptor isoforms and mechanisms of post-translational modification,” Journal of Steroid Biochemistry and Molecular Biology, vol. 102, no. 1–5, pp. 11–21, 2006.
[162]  S. Vandevyver, L. Dejager, and C. Libert, “On the trail of the glucocorticoid receptor: into the nucleus and back,” Traffic, vol. 13, pp. 364–374, 2012.
[163]  C. P. Fitzsimons, S. Ahmed, C. F. W. Wittevrongel et al., “The microtubule-associated protein doublecortin-like regulates the transport of the glucocorticoid receptor in neuronal progenitor cells,” Molecular Endocrinology, vol. 22, no. 2, pp. 248–262, 2008.
[164]  H. N. Daghestani, G. Zhu, P. A. Johnston, S. N. Shinde, J. L. Brodsky, and B. W. Day, “Characterization of inhibitors of glucocorticoid receptor nuclear translocation: a model of cytoplasmic dynein-mediated cargo transport,” ASSAY and Drug Development Technologies, vol. 10, pp. 46–60, 2012.
[165]  N. C. Nicolaides, Z. Galata, T. Kino, G. P. Chrousos, and E. Charmandari, “The human glucocorticoid receptor: molecular basis of biologic function,” Steroids, vol. 75, no. 1, pp. 1–12, 2010.
[166]  L. Cao, S. C. Leija, R. Kumsta et al., “Transcriptional control of the human glucocorticoid receptor: identification and analysis of alternative promoter regions,” Human Genetics, vol. 129, no. 5, pp. 533–543, 2011.
[167]  K. B. Pedersen and W. V. Vedeckis, “Quantification and glucocorticoid regulation of glucocorticoid receptor transcripts in two human leukemic cell lines,” Biochemistry, vol. 42, no. 37, pp. 10978–10990, 2003.
[168]  C. D. Geng, J. R. Schwartz, and W. V. Vedeckis, “A conserved molecular mechanism is responsible for the auto-up-regulation of glucocorticoid receptor gene promoters,” Molecular Endocrinology, vol. 22, no. 12, pp. 2624–2642, 2008.
[169]  C. D. Geng and W. V. Vedeckis, “A new, lineage specific, autoup-regulation mechanism for human glucocorticoid receptor gene expression in 697 pre-b-acute lymphoblastic leukemia cells,” Molecular Endocrinology, vol. 25, no. 1, pp. 44–57, 2011.
[170]  W. J. E. Tissing, J. P. P. Meijerink, B. Brinkhof et al., “Glucocorticoid-induced glucocorticoid-receptor expression and promoter usage is not linked to glucocorticoid resistance in childhood ALL,” Blood, vol. 108, no. 3, pp. 1045–1049, 2006.
[171]  S. Greenstein, K. Ghias, N. L. Krett, and S. T. Rosen, “Mechanisms of glucocorticoid-mediated apoptosis in hematological malignancies,” Clinical Cancer Research, vol. 8, no. 6, pp. 1681–1694, 2002.
[172]  P. A. Moalli and S. T. Rosen, “Glucocorticoid receptors and resistance to glucocorticoids in hematologic malignancies,” Leukemia and Lymphoma, vol. 15, no. 5-6, pp. 363–374, 1994.
[173]  B. Sánchez-Vega and V. Gandhi, “Glucocorticoid resistance in a multiple myeloma cell line is regulated by a transcription elongation block in the glucocorticoid receptor gene (NR3C1),” British Journal of Haematology, vol. 144, no. 6, pp. 856–864, 2009.
[174]  S. Sharma and A. Liechtenstein, “Dexamethasone-induced apoptotic mechanisms in myeloma cells investigated by analysis of mutant glucocorticoid receptors,” Blood, vol. 112, no. 4, pp. 1338–1345, 2008.
[175]  S. Geley, B. L. Hartmann, M. Hala, E. M. C. Strasser-Wozak, K. Kapelari, and R. Kofler, “Resistance to glucocorticoid-induced apoptosis in human T-cell acute lymphoblastic leukemia CEM-C1 cells is due to insufficient glucocorticoid receptor expression,” Cancer Research, vol. 56, no. 21, pp. 5033–5038, 1996.
[176]  J. Ramdas, W. Liu, and J. M. Harmon, “Glucocorticoid-induced cell death requires autoinduction of glucocorticoid receptor expression in human leukemic T cells,” Cancer Research, vol. 59, no. 6, pp. 1378–1385, 1999.
[177]  J. R. Schwartz, P. J. Sarvaiya, and W. V. Vedeckis, “Glucocorticoid receptor knock down reveals a similar apoptotic threshold but differing gene regulation patterns in T-cell and pre-B-cell acute lymphoblastic leukemia,” Molecular and Cellular Endocrinology, vol. 320, no. 1-2, pp. 76–86, 2010.
[178]  G. Gruber, M. Carlet, E. Türtscher et al., “Levels of glucocorticoid receptor and its ligand determine sensitivity and kinetics of glucocorticoid-induced leukemia apoptosis,” Leukemia, vol. 23, no. 4, pp. 820–823, 2009.
[179]  Y. Zilberman, E. Zafrir, H. Ovadia, E. Yefenof, R. Guy, and R. V. Sionov, “The glucocorticoid receptor mediates the thymic epithelial cell-induced apoptosis of CD4+8+ thymic lymphoma cells,” Cellular Immunology, vol. 227, no. 1, pp. 12–23, 2004.
[180]  P. A. Moalli, S. Pillay, D. Weiner, R. Leikin, and S. T. Rosen, “A mechanism of resistance to glucocorticoids in multiple myeloma: transient expression of a truncated glucocorticoid receptor mRNA,” Blood, vol. 79, no. 1, pp. 213–222, 1992.
[181]  M. Gomi, K. Moriwaki, S. Katagiri, Y. Kurata, and E. B. Thompson, “Glucocorticoid effects on myeloma cells in culture: correlation of growth inhibition with induction of glucocorticoid receptor messenger RNA,” Cancer Research, vol. 50, no. 6, pp. 1873–1878, 1990.
[182]  P. S. Bachmann, R. Gorman, K. L. MacKenzie, L. Lutze-Mann, and R. B. Lock, “Dexamethasone resistance in B-cell precursor childhood acute lymphoblastic leukemia occurs downstream of ligand-induced nuclear translocation of the glucocorticoid receptor,” Blood, vol. 105, no. 6, pp. 2519–2526, 2005.
[183]  P. S. Bachmann, R. Gorman, R. A. Papa et al., “Divergent mechanisms of glucocorticoid resistance in experimental models of pediatric acute lymphoblastic leukemia,” Cancer Research, vol. 67, no. 9, pp. 4482–4490, 2007.
[184]  E. G. Haarman, G. J. L. Kaspers, R. Pieters, M. M. A. Rottier, and A. J. P. Veerman, “Glucocorticoid receptor alpha, beta and gamma expression vs in vitro glucocorticod resistance in childhood leukemia,” Leukemia, vol. 18, no. 3, pp. 530–537, 2004.
[185]  A. H. Beesley, R. E. Weller, S. Senanayake, M. Welch, and U. R. Kees, “Receptor mutation is not a common mechanism of naturally occurring glucocorticoid resistance in leukaemia cell lines,” Leukemia Research, vol. 33, no. 2, pp. 321–325, 2009.
[186]  M. Labuda, A. Gahier, V. Gagné, A. Moghrabi, D. Sinnett, and M. Krajinovic, “Polymorphisms in glucocorticoid receptor gene and the outcome of childhood acute lymphoblastic leukemia (ALL),” Leukemia Research, vol. 34, no. 4, pp. 492–497, 2010.
[187]  E. Cuzzoni, S. De Iudicibus, F. Bartoli, A. Ventura, and G. Decorti, “Association between BclI polymorphism in the NR3C1 gene and in vitro individual variations in lymphocyte responses to methylprednisolone,” British Journal of Clinical Pharmacology, vol. 73, pp. 651–655, 2012.
[188]  K. L. Burnstein, C. M. Jewell, and J. A. Cidlowski, “Human glucocorticoid receptor cDNA contains sequences sufficient for receptor down-regulation,” The Journal of Biological Chemistry, vol. 265, no. 13, pp. 7284–7291, 1990.
[189]  K. L. Burnstein, C. M. Jewell, M. Sar, and J. A. Cidlowski, “Intragenic sequences of the human glucocorticoid receptor complementary DNA mediate hormone-inducible receptor messenger RNA down-regulation through multiple mechanisms,” Molecular Endocrinology, vol. 8, no. 12, pp. 1764–1773, 1994.
[190]  L. Davies, N. Karthikeyan, J. T. Lynch et al., “Cross talk of signaling pathways in the regulation of the glucocorticoid receptor function,” Molecular Endocrinology, vol. 22, no. 6, pp. 1331–1344, 2008.
[191]  H. Faus and B. Haendler, “Post-translational modifications ofsteroid receptors,” Biomedicine and Pharmacotherapy, vol. 60, no. 9, pp. 520–528, 2006.
[192]  A. D. Wallace and J. A. Cidlowski, “Proteasome-mediated glucocorticoid receptor degradation restricts transcriptional signaling by glucocorticoids,” The Journal of Biological Chemistry, vol. 276, no. 46, pp. 42714–42721, 2001.
[193]  M. Itoh, M. Adachi, H. Yasui, M. Takekawa, H. Tanaka, and K. Imai, “Nuclear export of glucocorticoid receptor is enhanced by c-Jun N-terminal kinase-mediated phosphorylation,” Molecular Endocrinology, vol. 16, no. 10, pp. 2382–2392, 2002.
[194]  W. Chen, T. Dang, R. D. Blind et al., “Glucocorticoid receptor phosphorylation differentially affects target gene expression,” Molecular Endocrinology, vol. 22, no. 8, pp. 1754–1766, 2008.
[195]  A. L. Miller, M. S. Webb, A. J. Copik et al., “p38 mitogen-activated protein kinase (MAPK) is a key mediator in glucocorticoid-induced apoptosis of lymphoid cells: correlation between p38 MAPK activation and site-specific phosphorylation of the human glucocorticoid receptor at serine 211,” Molecular Endocrinology, vol. 19, no. 6, pp. 1569–1583, 2005.
[196]  S. Hsu, M. Qi, and D. B. DeFranco, “Cell cycle regulation of glucocorticoid receptor function,” EMBO Journal, vol. 11, no. 9, pp. 3457–3468, 1992.
[197]  J. E. Bodwell, J. C. Webster, C. M. Jewell, J. A. Cidlowski, J. M. Hu, and A. Munck, “Glucocorticoid receptor phosphorylation: overview, function and cell cycle-dependence,” Journal of Steroid Biochemistry and Molecular Biology, vol. 65, no. 1–6, pp. 91–99, 1998.
[198]  M. D. Li, H. B. Ruan, J. P. Singh, et al., “O-GlcNAc transferase is involved in glucocorticoid receptor-mediated transrepression,” The Journal of Biological Chemistry, vol. 287, pp. 12904–12912, 2012.
[199]  M. S. Webb, A. L. Miller, B. H. Johnson et al., “Gene networks in glucocorticoid-evoked apoptosis of leukemic cells,” Journal of Steroid Biochemistry and Molecular Biology, vol. 85, no. 2–5, pp. 183–193, 2003.
[200]  R. D. Medh, M. S. Webb, A. L. Miller et al., “Gene expression profile of human lymphoid CEM cells sensitive and resistant to glucocorticoid-evoked apoptosis,” Genomics, vol. 81, no. 6, pp. 543–555, 2003.
[201]  S. Schmidt, J. Rainer, S. Riml et al., “Identification of glucocorticoid-response genes in children with acute lymphoblastic leukemia,” Blood, vol. 107, no. 5, pp. 2061–2069, 2006.
[202]  E. B. Thompson and B. H. Johnson, “Regulation of a distinctive set of genes in glucocorticoid-evoked apoptosis in CEM human lymphoid cells,” Recent Progress in Hormone Research, vol. 58, pp. 175–197, 2003.
[203]  J. Rainer, J. Lelong, D. Bindreither et al., “Research resource: transcriptional response to glucocorticoids in childhood acute lymphoblastic leukemia,” Molecular Endocrinology, vol. 26, pp. 178–193, 2012.
[204]  D. W. Chen, V. Saha, J. Z. Liu, J. M. Schwartz, and M. Krstic-Demonacos, “Erg and AP-1 as determinants of glucocorticoid response in acute lymphoblastic leukemia,” Oncogene. In press.
[205]  P. Bouillet, D. Metcalf, and D. C. S. Huang, “Pro-apoptotic Bcl-2 relative Bim required for certain apoptotic responses, leukocyte homeostasis, and to preclude autoimmunity,” Science, vol. 286, no. 5445, pp. 1735–1738, 1999.
[206]  J. K. Molitoris, K. S. McColl, and C. W. Distelhorst, “Glucocorticoid-mediated repression of the oncogenic microRNA cluster miR-17~92 contributes to the induction of Bim and initiation of apoptosis,” Molecular Endocrinology, vol. 25, pp. 409–420, 2011.
[207]  M. T. Abrams, N. M. Robertson, K. Yoon, and E. Wickstrom, “Inhibition of glucocorticoid-induced apoptosis by targeting the major splice variants of BIM mRNA with small interfering RNA and short hairpin RNA,” The Journal of Biological Chemistry, vol. 279, no. 53, pp. 55809–55817, 2004.
[208]  Z. Wang, M. H. Malone, H. He, K. S. McColl, and C. W. Distelhorst, “Microarray analysis uncovers the induction of the pro-apoptotic BH3-only protein Bim in multiple models of glucocorticoid-induced apoptosis,” The Journal of Biological Chemistry, vol. 278, no. 26, pp. 23861–23867, 2003.
[209]  R. V. Sionov, S. Kfir, E. Zafrir, O. Cohen, Y. Zilberman, and E. Yefenof, “Glucocorticoid-induced apoptosis revisited: a novel role for glucocorticoid receptor translocation to the mitochondria,” Cell Cycle, vol. 5, no. 10, pp. 1017–1026, 2006.
[210]  L. A. O'Reilly, L. Cullen, J. Visvader et al., “The pro-apoptotic BH3-only protein Bim is expressed in hematopoietic, epithelial, neuronal, and germ cells,” American Journal of Pathology, vol. 157, no. 2, pp. 449–461, 2000.
[211]  Y. N. Zhao, X. Guo, Z. G. Ma, L. Gu, J. Ge, and Q. Li, “Pro-apoptotic protein BIM in apoptosis of glucocorticoid-sensitive and -resistant acute lymphoblastic leukemia CEM cells,” Medical Oncology, vol. 28, no. 4, pp. 1609–1617, 2011.
[212]  D. Bhojwani, H. Kang, R. X. Menezes et al., “Gene expression signatures predictive of early response and outcome in high-risk childhood acute lymphoblastic leukemia: a Children's Oncology Group Study on Behalf of the Dutch Childhood Oncology Group and the German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia,” Journal of Clinical Oncology, vol. 26, no. 27, pp. 4376–4384, 2008.
[213]  H. Tagawa, S. Karnan, R. Suzuki et al., “Genome-wide array-based CGH for mantle cell lymphoma: identification of homozygous deletions of the pro-apoptotic gene BIM,” Oncogene, vol. 24, no. 8, pp. 1348–1358, 2005.
[214]  C. Mestre-Escorihuela, F. Rubio-Moscardo, J. A. Richter et al., “Homozygous deletions localize novel tumor suppressor genes in B-cell lymphomas,” Blood, vol. 109, no. 1, pp. 271–280, 2007.
[215]  L. Happo, A. Strasser, and S. Cory, “BH3-only proteins in apoptosis at a glance,” Journal of Cell Science, vol. 125, pp. 1081–1087, 2012.
[216]  J. E. Chipuk, T. Moldoveanu, F. Llambi, M. J. Parsons, and D. R. Green, “The BCL-2 family reunion,” Molecular Cell, vol. 37, no. 3, pp. 299–310, 2010.
[217]  S. N. Willis, J. I. Fletcher, T. Kaufmann et al., “Apoptosis initiated when BH3 ligands engage multiple Bcl-2 homologs, not Bax or Bak,” Science, vol. 315, no. 5813, pp. 856–859, 2007.
[218]  A. S. Gillings, K. Balmanno, C. M. Wiggins, M. Johnson, and S. J. Cook, “Apoptosis and autophagy: BIM as a mediator of tumour cell death in response to oncogene-targeted therapeutics,” FEBS Journal, vol. 276, no. 21, pp. 6050–6062, 2009.
[219]  T. Melarangi, J. Zhuang, K. Lin et al., “Glucocorticoid resistance in chronic lymphocytic leukaemia is associated with a failure of upregulated Bim/Bcl-2 complexes to activate Bax and Bak,” Cell Death and Disease, vol. 3, Article ID e372, 2012.
[220]  M. Erlacher, E. M. Michalak, P. N. Kelly et al., “BH3-only proteins Puma and Bim are rate-limiting for γ-radiation- and glucocorticoid-induced apoptosis of lymphoid cells in vivo,” Blood, vol. 106, no. 13, pp. 4131–4138, 2005.
[221]  V. Labi, M. Erlacher, S. Kiessling et al., “Loss of the BH3-only protein Bmf impairs B cell homeostasis and accelerates γ irradiation—induced thymic lymphoma development,” Journal of Experimental Medicine, vol. 205, no. 3, pp. 641–655, 2008.
[222]  J. C. Rathmell, T. Lindsten, W. X. Zong, R. M. Cinalli, and C. B. Thompson, “Deficiency in Bak and Bax perturbs thymic selection and lymphoid homeostasis,” Nature Immunology, vol. 3, no. 10, pp. 932–939, 2002.
[223]  Z. Wang, Y. P. Rong, M. H. Malone, M. C. Davis, F. Zhong, and C. W. Distelhorst, “Thioredoxin-interacting protein (txnip) is a glucocorticoid-regulated primary response gene involved in mediating glucocorticoid-induced apoptosis,” Oncogene, vol. 25, no. 13, pp. 1903–1913, 2006.
[224]  C. Ploner, J. Rainer, S. Lobenwein, S. Geley, and R. Kofler, “Repression of the BH3-only molecule PMAIP1/Noxa impairs glucocorticoid sensitivity of acute lymphoblastic leukemia cells,” Apoptosis, vol. 14, no. 6, pp. 821–828, 2009.
[225]  M. Mansha, M. Wasim, A. Kofler, and C. Ploner, “Expression and glucocorticoid-regulation of, “Bam”, a novel BH3-only transcript in acute lymphoblastic leukemia,” Molecular Biology Reports, vol. 39, pp. 6007–6013, 2012.
[226]  J. Lu, B. Quearry, and H. Harada, “p38-MAP kinase activation followed by BIM induction is essential for glucocorticoid-induced apoptosis in lymphoblastic leukemia cells,” FEBS Letters, vol. 580, no. 14, pp. 3539–3544, 2006.
[227]  P. F. Dijkers, R. H. Medema, J. W. J. Lammers, L. Koenderman, and P. J. Coffer, “Expression of the pro-apoptotic Bcl-2 family member Bim is regulated by the forkhead transcription factor FKHR-L1,” Current Biology, vol. 10, no. 19, pp. 1201–1204, 2000.
[228]  J. Gilley, P. J. Coffer, and J. Ham, “FOXO transcription factors directly activate bim gene expression and promote apoptosis in sympathetic neurons,” Journal of Cell Biology, vol. 162, no. 4, pp. 613–622, 2003.
[229]  A. Essafi, S. Fernández de Mattos, Y. A. M. Hassen et al., “Direct transcriptional regulation of Bim by FoxO3a mediates STI571-induced apoptosis in Bcr-Abl-expressing cells,” Oncogene, vol. 24, no. 14, pp. 2317–2329, 2005.
[230]  G. M. Wildey and P. H. Howe, “Runx1 is a co-activator with FOXO3 to mediate transforming growth factor β(TGFβ)-induced Bim transcription in hepatic cells,” The Journal of Biological Chemistry, vol. 284, no. 30, pp. 20227–20239, 2009.
[231]  N. Lützner, H. Kalbacher, A. Krones-Herzig, and F. Rosl, “FOXO3 is a glucocorticoid receptor target and regulates lkb1 and its own expression based on cellular amp levels via a positive autoregulatory loop,” PLoS ONE, vol. 7, Article ID e42166, 2012.
[232]  G. Tzivion, M. Dobson, and G. Ramakrishnan, “FoxO transcription factors; Regulation by AKT and 14-3-3 proteins,” Biochimica et Biophysica Acta, vol. 1813, no. 11, pp. 1938–1945, 2011.
[233]  X. Zhang, N. Tang, T. J. Hadden, and A. K. Rishi, “Akt, FoxO and regulation of apoptosis,” Biochimica et Biophysica Acta, vol. 1813, no. 11, pp. 1938–1945, 2011.
[234]  K. K. Ho, V. A. McGuire, C. Y. Koo, et al., “Phosphorylation of FOXO3a on Ser-7 by p38 promotes its nuclear localization in response to doxorubicin,” The Journal of Biological Chemistry, vol. 287, pp. 1545–1555, 2012.
[235]  B. Cai and Z. Xia, “p38 MAP kinase mediates arsenite-induced apoptosis through FOXO3a activation and induction of Bim transcription,” Apoptosis, vol. 13, no. 6, pp. 803–810, 2008.
[236]  J. Y. Yang, C. S. Zong, W. Xia et al., “ERK promotes tumorigenesis by inhibiting FOXO3a via MDM2-mediated degradation,” Nature Cell Biology, vol. 10, no. 2, pp. 138–148, 2008.
[237]  R. Ley, K. Balmanno, K. Hadfield, C. Weston, and S. J. Cook, “Activation of the ERK1/2 signaling pathway promotes phosphorylation and proteasome-dependent degradation of the BH3-only protein, Bim,” The Journal of Biological Chemistry, vol. 278, no. 21, pp. 18811–18816, 2003.
[238]  E. Dehan, F. Bassermann, D. Guardavaccaro et al., “βTrCP- and Rsk1/2-mediated degradation of BimEL inhibits apoptosis,” Molecular Cell, vol. 33, no. 1, pp. 109–116, 2009.
[239]  K. E. Ewings, K. Hadfield-Moorhouse, C. M. Wiggins et al., “ERK1/2-dependent phosphorylation of BimEL promotes its rapid dissociation from Mcl-1 and Bcl-xL,” EMBO Journal, vol. 26, no. 12, pp. 2856–2867, 2007.
[240]  K. Lei and R. J. Davis, “JNK phosphorylation of Bim-related members of the Bcl2 family induces Bax-dependent apoptosis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 100, no. 5, pp. 2432–2437, 2003.
[241]  G. V. Putcha, S. Le, S. Frank et al., “JNK-mediated BIM phosphorylation potentiates BAX-dependent apoptosis,” Neuron, vol. 38, no. 6, pp. 899–914, 2003.
[242]  Y. Engelbrecht, H. De Wet, K. Horsch, C. R. Langeveldt, F. S. Hough, and P. A. Hulley, “Glucocorticoids induce rapid up-regulation of mitogen-activated protein kinase phosphatase-1 and dephosphorylation of extracellular signal-regulated kinase and impair proliferation in human and mouse osteoblast cell lines,” Endocrinology, vol. 144, no. 2, pp. 412–422, 2003.
[243]  A. A. Rambal, Z. L. G. Panaguiton, L. Kramer, S. Grant, and H. Harada, “MEK inhibitors potentiate dexamethasone lethality in acute lymphoblastic leukemia cells through the pro-apoptotic molecule BIM,” Leukemia, vol. 23, no. 10, pp. 1744–1754, 2009.
[244]  F. Zhou and E. B. Thompson, “Role of c-jun induction in the glucocorticoid-evoked apoptotic pathway in human leukemic lymphoblasts,” Molecular Endocrinology, vol. 10, no. 3, pp. 306–316, 1996.
[245]  N. Heidari, A. V. Miller, M. A. Hicks, C. B. Marking, and H. Harada, “Glucocorticoid-mediated BIM induction and apoptosis are regulated by Runx2 and c-Jun in leukemia cells,” Cell Death and Disease, vol. 3, Article ID e349, 2012.
[246]  A. Ventura, A. G. Young, M. M. Winslow et al., “Targeted deletion reveals essential and overlapping functions of the miR-17~92 family of miRNA clusters,” Cell, vol. 132, no. 5, pp. 875–886, 2008.
[247]  C. Xiao, L. Srinivasan, D. P. Calado et al., “Lymphoproliferative disease and autoimmunity in mice with increased miR-17~92 expression in lymphocytes,” Nature Immunology, vol. 9, no. 4, pp. 405–414, 2008.
[248]  P. Mu, Y. C. Han, D. Betel et al., “Genetic dissection of the miR-17~92 cluster of microRNAs in Myc-induced B-cell lymphomas,” Genes & Development, vol. 23, pp. 2806–2811, 2009.
[249]  V. Olive, I. Jiang, and L. He, “miR-17~92, a cluster of miRNAs in the midst of the cancer network,” International Journal of Biochemistry and Cell Biology, vol. 42, no. 8, pp. 1348–1354, 2010.
[250]  G. van Haaften and R. Agami, “Tumorigenicity of the miR-17~92 cluster distilled,” Genes and Development, vol. 24, no. 1, pp. 1–4, 2010.
[251]  S. Mi, Z. Li, P. Chen et al., “Aberrant overexpression and function of the miR-17~92 cluster in MLL-rearranged acute leukemia,” Proceedings of the National Academy of Sciences of the United States of America, vol. 107, no. 8, pp. 3710–3715, 2010.
[252]  S. K. Lee and G. A. Calin, “Non-coding RNAs and cancer: new paradigms in oncology,” Discovery Medicine, vol. 11, no. 58, pp. 245–254, 2011.
[253]  K. J. Mavrakis, J. Van Der Meulen, A. L. Wolfe et al., “A cooperative microRNA-tumor suppressor gene network in acute T-cell lymphoblastic leukemia,” Nature Genetics, vol. 43, no. 8, p. 815, 2011.
[254]  W. K. K. Wu, C. W. Lee, C. H. Cho et al., “MicroRNA dysregulation in gastric cancer: a new player enters the game,” Oncogene, vol. 29, no. 43, pp. 5761–5771, 2010.
[255]  K. A. O'Donnell, E. A. Wentzel, K. I. Zeller, C. V. Dang, and J. T. Mendell, “c-Myc-regulated microRNAs modulate E2F1 expression,” Nature, vol. 435, no. 7043, pp. 839–843, 2005.
[256]  Y. Li, W. Tan, T. W. L. Neo et al., “Role of the miR-106b-25 microRNA cluster in hepatocellular carcinoma,” Cancer Science, vol. 100, no. 7, pp. 1234–1242, 2009.
[257]  C. Haftmann, A. B. Stittrich, E. Sgouroudis, et al., “Lymphocyte signaling: regulation of FoxO transcription factors by microRNAs,” Annals of the New York Academy of Sciences, vol. 1247, pp. 46–55, 2012.
[258]  I. K. Guttilla and B. A. White, “Coordinate regulation of FOXO1 by miR-27a, miR-96, and miR-182 in breast cancer cells,” The Journal of Biological Chemistry, vol. 284, no. 35, pp. 23204–23216, 2009.
[259]  M. F. Segura, D. Hanniford, S. Menendez et al., “Aberrant miR-182 expression promotes melanoma metastasis by repressing FOXO3 and microphthalmia-associated transcription factor,” Proceedings of the National Academy of Sciences of the United States of America, vol. 106, no. 6, pp. 1814–1819, 2009.
[260]  A. B. Stittrich, C. Haftmann, E. Sgouroudis et al., “The microRNA miR-182 is induced by IL-2 and promotes clonal expansion of activated helper T lymphocytes,” Nature Immunology, vol. 11, no. 11, pp. 1057–1062, 2010.
[261]  A. Yang, J. Ma, M. Wu, et al., “Aberrant microRNA-182 expression is associated with glucocorticoid resistance in lymphoblastic malignancies,” Leukemia & Lymphoma, vol. 53, no. 12, pp. 2465–2473, 2012.
[262]  L. Elia, R. Contu, M. Quintavalle et al., “Reciprocal regulation of microrna-1 and insulin-like growth factor-1 signal transduction cascade in cardiac and skeletal muscle in physiological and pathological conditions,” Circulation, vol. 120, no. 23, pp. 2377–2385, 2009.
[263]  M. Yamamoto, E. Kondo, M. Takeuchi et al., “miR-155, a modulator of FOXO3a protein expression, is underexpressed and cannot be upregulated by stimulation of HOZOT, a line of multifunctional Treg,” PLoS ONE, vol. 6, no. 2, Article ID e16841, 2011.
[264]  W. Kong, L. He, M. Coppola et al., “MicroRNA-155 regulates cell survival, growth, and chemosensitivity by targeting FOXO3a in breast cancer,” The Journal of Biological Chemistry, vol. 285, no. 23, pp. 17869–17879, 2010.
[265]  R. M. O'Connell, D. S. Rao, A. A. Chaudhuri, and D. Baltimore, “Physiological and pathological roles for microRNAs in the immune system,” Nature Reviews Immunology, vol. 10, no. 2, pp. 111–122, 2010.
[266]  T. H. Thai, D. P. Calado, S. Casola et al., “Regulation of the germinal center response by MicroRNA-155,” Science, vol. 316, no. 5824, pp. 604–608, 2007.
[267]  R. Dai, Y. Zhang, D. Khan et al., “Identification of a common lupus disease-associated microRNA expression pattern in three different murine models of lupus,” PLoS ONE, vol. 5, no. 12, p. e14302, 2010.
[268]  K. Wang and P. F. Li, “Foxo3a regulates apoptosis by negatively targeting miR-21,” The Journal of Biological Chemistry, vol. 285, no. 22, pp. 16958–16966, 2010.
[269]  F. Meng, R. Henson, H. Wehbe-Janek, K. Ghoshal, S. T. Jacob, and T. Patel, “MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer,” Gastroenterology, vol. 133, no. 2, pp. 647–658, 2007.
[270]  D. Iliopoulos, S. A. Jaeger, H. A. Hirsch, M. L. Bulyk, and K. Struhl, “STAT3 activation of miR-21 and miR-181b-1 via PTEN and CYLD are part of the epigenetic switch linking inflammation to cancer,” Molecular Cell, vol. 39, no. 4, pp. 493–506, 2010.
[271]  J. Ma, Y. Xie, Y. Shi, W. Qin, B. Zhao, and Y. Jin, “Glucocorticoid-induced apoptosis requires FOXO3A activity,” Biochemical and Biophysical Research Communications, vol. 377, no. 3, pp. 894–898, 2008.
[272]  R. V. Sionov, O. Cohen, S. Kfir, Y. Zilberman, and E. Yefenof, “Role of mitochondrial glucocorticoid receptor in glucocorticoid-induced apoptosis,” Journal of Experimental Medicine, vol. 203, no. 1, pp. 189–201, 2006.
[273]  G. Talabér, F. Boldizsár, D. Bartis et al., “Mitochondrial translocation of the glucocorticoid receptor in double-positive thymocytes correlates with their sensitivity to glucocorticoid-induced apoptosis,” International Immunology, vol. 21, no. 11, pp. 1269–1276, 2009.
[274]  J. Du, Y. Wang, R. Hunter et al., “Dynamic regulation of mitochondrial function by glucocorticoids,” Proceedings of the National Academy of Sciences of the United States of America, vol. 106, no. 9, pp. 3543–3548, 2009.
[275]  M. E. Tome, K. Lee, M. C. Jaramillo, and M. M. Briehl, “Mitochondria are the primary source of the H2O2 signal for glucocorticoid-induced apoptosis of lymphoma cells,” Experimental and Therapeutic Medicine, vol. 4, pp. 237–242, 2012.
[276]  M. E. Tome, M. C. Jaramillo, and M. M. Briehl, “Hydrogen peroxide signaling is required for glucocorticoid-induced apoptosis in lymphoma cells,” Free Radical Biology and Medicine, vol. 51, pp. 2048–2059, 2011.
[277]  A. M. G. Psarra, S. Hermann, G. Panayotou, and G. Spyrou, “Interaction of mitochondrial thioredoxin with glucocorticoid receptor and NF-κB modulates glucocorticoid receptor and NF-κB signalling in HEK-293 cells,” Biochemical Journal, vol. 422, no. 3, pp. 521–531, 2009.
[278]  A. M. G. Psarra and C. E. Sekeris, “Glucocorticoids induce mitochondrial gene transcription in HepG2 cells. Role of the mitochondrial glucocorticoid receptor,” Biochimica et Biophysica Acta, vol. 1813, no. 10, pp. 1814–1821, 2011.
[279]  K. Eberhart, J. Rainer, D. Bindreither et al., “Glucocorticoid-induced alterations in mitochondrial membrane properties and respiration in childhood acute lymphoblastic leukemia,” Biochimica et Biophysica Acta, vol. 1807, no. 6, pp. 719–725, 2011.
[280]  C. Fujita, F. Ichikawa, T. Teratani et al., “Direct effects of corticosterone on ATP production by mitochondria from immortalized hypothalamic GT1-7 neurons,” Journal of Steroid Biochemistry and Molecular Biology, vol. 117, no. 1–3, pp. 50–55, 2009.
[281]  I. M. E. Beck, W. V. Berghe, L. Vermeulen, K. R. Yamamoto, G. Haegeman, and K. De Bosscher, “Crosstalk in inflammation: the interplay of glucocorticoid receptor-based mechanisms and kinases and phosphatases,” Endocrine Reviews, vol. 30, no. 7, pp. 830–882, 2009.
[282]  U. Nuutinen, A. Ropponen, S. Suoranta et al., “Dexamethasone-induced apoptosis and up-regulation of Bim is dependent on glycogen synthase kinase-3,” Leukemia Research, vol. 33, no. 12, pp. 1714–1717, 2009.
[283]  U. Nuutinen, V. Postila, M. M?tt? et al., “Inhibition of PI3-kinase-Akt pathway enhances dexamethasone-induced apoptosis in a human follicular lymphoma cell line,” Experimental Cell Research, vol. 312, no. 3, pp. 322–330, 2006.
[284]  C. Rubio-Pati?o, C. M. Palmeri, A. Perez-Perarnau, et al., “Glycogen synthase kinase-3beta is involved in ligand-dependent activation of transcription and cellular localization of the glucocorticoid receptor,” Molecular Endocrinology, vol. 26, pp. 1508–1520, 2012.
[285]  F. S. Wang, J. Y. Ko, L. H. Weng, D. W. Yeh, H. J. Ke, and S. L. Wu, “Inhibition of glycogen synthase kinase-3β attenuates glucocorticoid-induced bone loss,” Life Sciences, vol. 85, no. 19-20, pp. 685–692, 2009.
[286]  B. C. Bornhauser, L. Bonapace, D. Lindholm et al., “Low-dose arsenic trioxide sensitizes glucocorticoid-resistant acute lymphoblastic leukemia cells to dexamethasone via an Akt-dependent pathway,” Blood, vol. 110, no. 6, pp. 2084–2091, 2007.
[287]  G. Wei, D. Twomey, J. Lamb et al., “Gene expression-based chemical genomics identifies rapamycin as a modulator of MCL1 and glucocorticoid resistance,” Cancer Cell, vol. 10, no. 4, pp. 331–342, 2006.
[288]  N. Morishita, H. Tsukahara, K. Chayama et al., “Activation of Akt is associated with poor prognosis and chemotherapeutic resistance in pediatric B-precursor acute lymphoblastic leukemia,” Pediatric Blood & Cancer, vol. 59, pp. 83–89, 2012.
[289]  N. Hay and N. Sonenberg, “Upstream and downstream of mTOR,” Genes and Development, vol. 18, no. 16, pp. 1926–1945, 2004.
[290]  K. Lee, K. T. Nam, S. H. Cho, et al., “Vital roles of mTOR complex 2 in Notch-driven thymocyte differentiation and leukemia,” Journal of Experimental Medicine, vol. 209, pp. 713–728, 2012.
[291]  A. M. Martelli, C. Evangelisti, F. Chiarini, and J. A. McCubrey, “The phosphatidylinositol 3-kinase/Akt/mTOR signaling network as a therapeutic target in acute myelogenous leukemia patients,” Oncotarget, vol. 1, no. 2, pp. 89–103, 2010.
[292]  A. Carnero, “The PKB/AKT pathway in cancer,” Current Pharmaceutical Design, vol. 16, no. 1, pp. 34–44, 2010.
[293]  S. Hafsi, F. M. Pezzino, S. Candido et al., “Gene alterations in the PI3K/PTEN/AKT pathway as a mechanism of drug-resistance,” International Journal of Oncology, vol. 40, pp. 639–644, 2012.
[294]  D. A. Guertin and D. M. Sabatini, “Defining the role of mTOR in cancer,” Cancer Cell, vol. 12, no. 1, pp. 9–22, 2007.
[295]  D. D. Sarbassov, D. A. Guertin, S. M. Ali, and D. M. Sabatini, “Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex,” Science, vol. 307, no. 5712, pp. 1098–1101, 2005.
[296]  Z. Wang, M. H. Malone, M. J. Thomenius, F. Zhong, F. Xu, and C. W. Distelhorst, “Dexamethasone-induced gene 2 (dig2) is a novel pro-survival stress gene induced rapidly by diverse apoptotic signals,” The Journal of Biological Chemistry, vol. 278, no. 29, pp. 27053–27058, 2003.
[297]  T. Shinjyo, R. Kuribara, T. Inukai et al., “Downregulation of Bim, a pro-apoptotic relative of Bcl-2, is a pivotal step in cytokine-initiated survival signaling in murine hematopoietic progenitors,” Molecular and Cellular Biology, vol. 21, no. 3, pp. 854–864, 2001.
[298]  P. Argyriou, P. Economopoulou, and S. Papageorgiou, “The role of mTOR inhibitors for the treatment of B-Cell lymphomas,” Advances in Hematology, vol. 2012, Article ID 435342, 13 pages, 2012.
[299]  L. Salmena, A. Carracedo, and P. P. Pandolfi, “Tenets of PTEN tumor suppression,” Cell, vol. 133, no. 3, pp. 403–414, 2008.
[300]  P. Y. Jotta, M. A. Ganazza, A. Silva et al., “Negative prognostic impact of PTEN mutation in pediatric T-cell acute lymphoblastic leukemia,” Leukemia, vol. 24, no. 1, pp. 239–242, 2010.
[301]  J. E. Fata, S. Debnath, E. C. Jenkins, Jr, and M. V. Fournier, “Nongenomic mechanisms of PTEN regulation,” International Journal of Cell Biology, vol. 2012, Article ID 379685, 10 pages, 2012.
[302]  J. M. García, J. Silva, C. Pe?a et al., “Promoter methylation of the PTEN gene is a common molecular change in breast cancer,” Genes Chromosomes and Cancer, vol. 41, no. 2, pp. 117–124, 2004.
[303]  Y. H. Kang, S. L. Hye, and H. K. Woo, “Promoter methylation and silencing of PTEN in gastric carcinoma,” Laboratory Investigation, vol. 82, no. 3, pp. 285–291, 2002.
[304]  P. M. Brauer and A. L. Tyner, “RAKing in AKT: a tumor suppressor function for the intracellular tyrosine kinase FRK,” Cell Cycle, vol. 8, no. 17, pp. 2728–2732, 2009.
[305]  J. Torres and R. Pulido, “The tumor suppressor PTEN is phosphorylated by the protein kinase CK2 at its C terminus. Implications for PTEN stability to proteasome-mediated degradation,” The Journal of Biological Chemistry, vol. 276, no. 2, pp. 993–998, 2001.
[306]  H. Maccario, N. M. Perera, L. Davidson, C. P. Downes, and N. R. Leslie, “PTEN is destabilized by phosphorylation on Thr366,” Biochemical Journal, vol. 405, no. 3, pp. 439–444, 2007.
[307]  X. Wang, L. C. Trotman, T. Koppie et al., “NEDD4-1 is a proto-oncogenic ubiquitin ligase for PTEN,” Cell, vol. 128, no. 1, pp. 129–139, 2007.
[308]  H. Guo, G. Qiao, H. Ying et al., “E3 ubiquitin ligase Cbl-b regulates Pten via Nedd4 in T cells independently of its ubiquitin ligase activity,” Cell Reports, vol. 1, pp. 472–482, 2012.
[309]  C. Van Themsche, V. Leblanc, S. Parent, and E. Asselin, “X-linked inhibitor of apoptosis protein (XIAP) regulates PTEN ubiquitination, content, and compartmentalization,” The Journal of Biological Chemistry, vol. 284, no. 31, pp. 20462–20466, 2009.
[310]  J. Redondo-Mu?oz, E. Escobar-Díaz, M. H. Del Cerro et al., “Induction of B-chronic lymphocytic leukemia cell apoptosis by arsenic trioxide involves suppression of the phosphoinositide 3-kinase/Akt survival pathway via c-jun-NH2 terminal kinase activation and PTEN upregulation,” Clinical Cancer Research, vol. 16, no. 17, pp. 4382–4391, 2010.
[311]  S. Maddika, S. Kavela, N. Rani et al., “WWP2 is an E3 ubiquitin ligase for PTEN,” Nature Cell Biology, vol. 13, no. 6, pp. 728–733, 2011.
[312]  S. F. Ahmed, S. Deb, I. Paul et al., “The chaperone-assisted E3 ligase C terminus of Hsc70-interacting protein (CHIP) targets PTEN for proteasomal degradation,” The Journal of Biological Chemistry, vol. 287, pp. 15996–16006, 2012.
[313]  Z. Ni, J. Tang, Z. Cai et al., “A new pathway of glucocorticoid action for asthma treatment through the regulation of PTEN expression,” Respiratory Research, vol. 12, p. 47, 2011.
[314]  L. Poliseno, L. Salmena, L. Riccardi, et al., “Identification of the miR-106b~25 microRNA cluster as a proto-oncogenic PTEN-targeting intron that cooperates with its host gene MCM7 in transformation,” Science Signaling, vol. 3, Article ID ra29, 2010.
[315]  J. T. Huse, C. Brennan, D. Hambardzumyan et al., “The PTEN-regulating microRNA miR-26a is amplified in high-grade glioma and facilitates gliomagenesis in vivo,” Genes and Development, vol. 23, no. 11, pp. 1327–1337, 2009.
[316]  C. Wang, Z. Bian, D. Wei, and J. G. Zhang, “miR-29b regulates migration of human breast cancer cells,” Molecular and Cellular Biochemistry, vol. 352, no. 1-2, pp. 197–207, 2011.
[317]  P. T. Jindra, J. Bagley, J. G. Godwin, and J. Iacomini, “Costimulation-dependent expression of microRNA-214 increases the ability of T cells to proliferate by targeting Pten,” Journal of Immunology, vol. 185, no. 2, pp. 990–997, 2010.
[318]  H. Yang, W. Kong, L. He et al., “MicroRNA expression profiling in human ovarian cancer: miR-214 induces cell survival and cisplatin resistance by targeting PTEN,” Cancer Research, vol. 68, no. 5, p. 1609, 2008.
[319]  M. Kato, S. Putta, M. Wang et al., “TGF-β activates Akt kinase through a microRNA-dependent amplifying circuit targeting PTEN,” Nature Cell Biology, vol. 11, no. 7, pp. 881–889, 2009.
[320]  M. Incoronato, M. Garofalo, L. Urso et al., “miR-212 increases tumor necrosis factor-related apoptosis-inducing ligand sensitivity in non-small cell lung cancer by targeting the anti-apoptotic protein PED,” Cancer Research, vol. 70, no. 9, pp. 3638–3646, 2010.
[321]  M. Garofalo, G. Di Leva, G. Romano et al., “miR-221&222 Regulate TRAIL Resistance and Enhance Tumorigenicity through PTEN and TIMP3 Downregulation,” Cancer Cell, vol. 16, no. 6, pp. 498–509, 2009.
[322]  J. K. Brunelle and A. Letai, “Control of mitochondrial apoptosis by the Bcl-2 family,” Journal of Cell Science, vol. 122, no. 4, pp. 437–441, 2009.
[323]  S. Krishna, I. C. C. Low, and S. Pervaiz, “Regulation of mitochondrial metabolism: yet another facet in the biology of the oncoprotein Bcl-2,” Biochemical Journal, vol. 435, no. 3, pp. 545–551, 2011.
[324]  H. Feng, H. Xiang, Y. W. Mao et al., “Human Bcl-2 activates ERK signaling pathway to regulate activating protein-1, lens epithelium-derived growth factor and downstream genes,” Oncogene, vol. 23, no. 44, pp. 7310–7321, 2004.
[325]  G. Vairo, T. J. Soos, T. M. Upton et al., “Bcl-2 retards cell cycle entry through p27(Kip1), pRB relative p130, and altered E2F regulation,” Molecular and Cellular Biology, vol. 20, no. 13, pp. 4745–4753, 2000.
[326]  N. Katayama, N. Mahmud, K. Nishi et al., “Bcl-2 in cell-cycle regulation of hematopoietic cells by transforming growth factor-β1,” Leukemia and Lymphoma, vol. 39, no. 5-6, pp. 601–605, 2000.
[327]  Y. Janumyan, Q. Cui, L. Yan, C. G. Sansam, M. Valentin, and E. Yang, “G0 function of BCL2 and BCL-xL requires BAX, BAK, and p27 phosphorylation by mirk, revealing a novel role of BAX and BAK in quiescence regulation,” The Journal of Biological Chemistry, vol. 283, no. 49, pp. 34108–34120, 2008.
[328]  C. Greider, A. Chattopadhyay, C. Parkhurst, and E. Yang, “BCL-xL and BCL2 delay Myc-induced cell cycle entry through elevation of p27 and inhibition of G1 cyclin-dependent kinases,” Oncogene, vol. 21, no. 51, pp. 7765–7775, 2002.
[329]  H. G. Wang, U. R. Rapp, and J. C. Reed, “Bcl-2 targets the protein kinase Raf-1 to mitochondria,” Cell, vol. 87, no. 4, pp. 629–638, 1996.
[330]  J. C. Reed, “Double identity for proteins of the Bcl-2 family,” Nature, vol. 387, no. 6635, pp. 773–776, 1997.
[331]  C. Batsi, S. Markopoulou, E. Kontargiris et al., “Bcl-2 blocks 2-methoxyestradiol induced leukemia cell apoptosis by a p27Kip1-dependent G1/S cell cycle arrest in conjunction with NF-κB activation,” Biochemical Pharmacology, vol. 78, no. 1, pp. 33–44, 2009.
[332]  R. V. Sionov, S. Kfir-Erenfeld, R. Spokoini, and E. Yefenof, “A role for bcl-2 in notch1-dependent transcription in thymic lymphoma cells,” Advances in Hematology, vol. 2012, Article ID 435241, 5 pages, 2012.
[333]  Z. Wang, A. S. Azmi, A. Ahmad et al., “TW-37, a small-molecule inhibitor of Bcl-2, inhibits cell growth and induces apoptosis in pancreatic cancer: involvement of notch-1 signaling pathway,” Cancer Research, vol. 69, no. 7, pp. 2757–2765, 2009.
[334]  N. Cheng, C. I. van de Wetering, and C. M. Knudson, “p27 deficiency cooperates with Bcl-2 but not Bax to promote T-cell lymphoma,” PLoS ONE, vol. 3, no. 4, Article ID e1911, 2008.
[335]  C. A. Tucker, A. I. Kapanen, G. Chikh et al., “Silencing Bcl-2 in models of mantle cell lymphoma is associated with decreases in cyclin D1, nuclear factor-κB, p53, bax, and p27 levels,” Molecular Cancer Therapeutics, vol. 7, no. 4, pp. 749–758, 2008.
[336]  Y. M. Janumyan, C. G. Sansam, A. Chattopadhyay et al., “Bcl-xL/Bcl-2 coordinately regulates apoptosis, cell cycle arrest and cell cycle entry,” EMBO Journal, vol. 22, no. 20, pp. 5459–5470, 2003.
[337]  B. L. Hartmann, S. Geley, M. L?ffler et al., “Bcl-2 interferes with the execution phase, but not upstream events, in glucocorticoid-induced leukemia apoptosis,” Oncogene, vol. 18, no. 3, pp. 713–719, 1999.
[338]  M. Rogalińska and Z. M. Kiliańska, “Targeting Bcl-2 in CLL,” Current Medicinal Chemistry, vol. 19, no. 30, pp. 5109–5115, 2012.
[339]  F. Tzifi, C. Economopoulou, D. Gourgiotis, A. Ardavanis, S. Papageorgiou, and A. Scorilas, “The role of BCL2 family of apoptosis regulator proteins in acute and chronic leukemias,” Advances in Hematology, vol. 2012, Article ID 524308, 15 pages, 2012.
[340]  C. Ploner, J. Rainer, H. Niederegger et al., “The BCL2 rheostat in glucocorticoid-induced apoptosis of acute lymphoblastic leukemia,” Leukemia, vol. 22, no. 2, pp. 370–377, 2008.
[341]  P. Menendez, A. Vargas, C. Bueno et al., “Quantitative analysis of bcl-2 expression in normal and leukemic human B-cell differentiation,” Leukemia, vol. 18, no. 3, pp. 491–498, 2004.
[342]  Y. Tsujimoto, L. R. Finger, and J. Yunis, “Cloning of the chromosome breakpoint of neoplastic B cells with the t(14;18) chromosome translocation,” Science, vol. 226, no. 4678, pp. 1097–1099, 1984.
[343]  Y. Tsujimoto, J. Cossman, E. Jaffe, and C. M. Croce, “Involvement of the bcl-2 gene in human follicular lymphoma,” Science, vol. 228, no. 4706, pp. 1440–1443, 1985.
[344]  M. Nambiar and S. C. Raghavan, “Mechanism of fragility at BCL2 gene minor breakpoint cluster region during t(14,18) chromosomal translocation,” Journal of Biological Chemistry, vol. 287, pp. 8688–8701, 2012.
[345]  G. A. Calin, C. D. Dumitru, M. Shimizu et al., “Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia,” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 24, pp. 15524–15529, 2002.
[346]  A. Cimmino, G. A. Calin, M. Fabbri et al., “miR-15 and miR-16 induce apoptosis by targeting BCL2,” Proceedings of the National Academy of Sciences of the United States of America, vol. 102, no. 39, pp. 13944–13949, 2005.
[347]  H. Hermeking, “p53 enters the microRNA world,” Cancer Cell, vol. 12, no. 5, pp. 414–418, 2007.
[348]  L. Li, L. Yuan, J. Luo, J. Gao, J. Guo, and X. Xie, “MiR-34a inhibits proliferation and migration of breast cancer through down-regulation of Bcl-2 and SIRT1,” Clinical and Experimental Medicine . In press.
[349]  S. Willimott and S. D. Wagner, “miR-125b and miR-155 contribute to BCL2 repression and proliferation in response to CD40 ligand (CD154) in human leukemic B-cells,” The Journal of Biological Chemistry, vol. 287, pp. 2608–2617, 2012.
[350]  J. Gong, J. P. Zhang, B. Li et al., “MicroRNA-125b promotes apoptosis by regulating the expression of Mcl-1, Bcl-w and IL-6R,” Oncogene. In press.
[351]  A. Zhao, Q. Zeng, X. Xie et al., “MicroRNA-125b induces cancer cell apoptosis through suppression of Bcl-2 expression,” Journal of Genetics and Genomics, vol. 39, pp. 29–35, 2012.
[352]  L. H. Liu, H. Li, J. P. Li et al., “miR-125b suppresses the proliferation and migration of osteosarcoma cells through down-regulation of STAT3,” Biochemical and Biophysical Research Communications, vol. 416, pp. 31–38, 2011.
[353]  M. Zhou, Z. Liu, Y. Zhao et al., “MicroRNA-125b confers the resistance of breast cancer cells to paclitaxel through suppression of pro-apoptotic Bcl-2 antagonist killer 1 (Bak1) expression,” The Journal of Biological Chemistry, vol. 285, no. 28, pp. 21496–21507, 2010.
[354]  H. Zhang, X. Q. Luo, D. D. Feng et al., “Upregulation of microRNA-125b contributes to leukemogenesis and increases drug resistance in pediatric acute promyelocytic leukemia,” Molecular Cancer, vol. 10, article 108, 2011.
[355]  E. Surdziel, M. Cabanski, I. Dallmann et al., “Enforced expression of miR-125b affects myelopoiesis by targeting multiple signaling pathways,” Blood, vol. 117, no. 16, pp. 4338–4348, 2011.
[356]  H. F. Xia, T. Z. He, C. M. Liu et al., “MiR-125b expression affects the proliferation and apoptosis of human glioma cells by targeting Bmf,” Cellular Physiology and Biochemistry, vol. 23, no. 4–6, pp. 347–358, 2009.
[357]  M. Bousquet, M. H. Harris, B. Zhou, and H. F. Lodish, “MicroRNA miR-125b causes leukemia,” Proceedings of the National Academy of Sciences of the United States of America, vol. 107, no. 50, pp. 21558–21563, 2010.
[358]  D. X. Zhu, W. Zhu, C. Fang et al., “miR-181a/b significantly enhances drug sensitivity in chronic lymphocytic leukemia cells via targeting multiple anti-apoptosis genes,” Carcinogenesis, vol. 33, pp. 1294–1301, 2012.
[359]  H. Li, L. Hui, and W. Xu, “miR-181a sensitizes a multidrug-resistant leukemia cell line K562/A02 to daunorubicin by targeting BCL-2,” Acta Biochimica et Biophysica Sinica, vol. 44, pp. 269–277, 2012.
[360]  Y. B. Ouyang, Y. Lu, S. Yue, and R. G. Giffard, “miR-181 targets multiple Bcl-2 family members and influences apoptosis and mitochondrial function in astrocytes,” Mitochondrion, vol. 12, pp. 213–219, 2012.
[361]  S. Shimizu, T. Takehara, H. Hikita et al., “The let-7 family of microRNAs inhibits Bcl-xL expression and potentiates sorafenib-induced apoptosis in human hepatocellular carcinoma,” Journal of Hepatology, vol. 52, no. 5, pp. 698–704, 2010.
[362]  H. Nakano, T. Miyazawa, K. Kinoshita, Y. Yamada, and T. Yoshida, “Functional screening identifies a microRNA, miR-491 that induces apoptosis by targeting Bcl-XLin colorectal cancer cells,” International Journal of Cancer, vol. 127, no. 5, pp. 1072–1080, 2010.
[363]  H. Guan, P. Zhang, C. Liu et al., “Characterization and functional analysis of the human microRNA let-7a2 promoter in lung cancer A549 cell lines,” Molecular Biology Reports, vol. 38, no. 8, pp. 5327–5334, 2011.
[364]  A. Holleman, M. H. Cheok, M. L. Den Boer et al., “Gene-expression patterns in drug-resistant acute lymphoblastic leukemia cells and response to treatment,” The New England Journal of Medicine, vol. 351, no. 6, pp. 533–542, 2004.
[365]  R. W. Stam, M. L. Den Boer, P. Schneider et al., “Association of high-level MCL-1 expression with in vitro and in vivo prednisone resistance in MLL-rearranged infant acute lymphoblastic leukemia,” Blood, vol. 115, no. 5, pp. 1018–1025, 2010.
[366]  L. W. Thomas, C. Lam, and S. W. Edwards, “Mcl-1; the molecular regulation of protein function,” FEBS Letters, vol. 584, no. 14, pp. 2981–2989, 2010.
[367]  M. Taniai, A. Grambihler, H. Higuchi et al., “Mcl-1 mediates tumor necrosis factor-related apoptosis-inducing ligand resistance in human cholangiocarcinoma cells,” Cancer Research, vol. 64, no. 10, pp. 3517–3524, 2004.
[368]  J. T. Opferman and D. R. Green, “DUB-le trouble for cell survival,” Cancer Cell, vol. 17, no. 2, pp. 117–119, 2010.
[369]  Q. Ding, X. He, J. M. Hsu et al., “Degradation of Mcl-1 by β-TrCP mediates glycogen synthase kinase 3-induced tumor suppression and chemosensitization,” Molecular and Cellular Biology, vol. 27, no. 11, pp. 4006–4017, 2007.
[370]  U. Maurer, C. Charvet, A. S. Wagman, E. Dejardin, and D. R. Green, “Glycogen synthase kinase-3 regulates mitochondrial outer membrane permeabilization and apoptosis by destabilization of MCL-1,” Molecular Cell, vol. 21, no. 6, pp. 749–760, 2006.
[371]  Q. Zhong, W. Gao, F. Du, and X. Wang, “Mule/ARF-BP1, a BH3-only E3 ubiquitin ligase, catalyzes the polyubiquitination of Mcl-1 and regulates apoptosis,” Cell, vol. 121, no. 7, pp. 1085–1095, 2005.
[372]  H. Inuzuka, H. Fukushima, S. Shaik, and W. Wei, “Novel insights into the molecular mechanisms governing Mdm2 ubiquitination and destruction,” Oncotarget, vol. 1, no. 7, pp. 685–690, 2010.
[373]  M. Schwickart, X. Huang, J. R. Lill et al., “Deubiquitinase USP9X stabilizes MCL1 and promotes tumour cell survival,” Nature, vol. 463, no. 7277, pp. 103–107, 2010.
[374]  P. Agrawal, Y. T. Chen, B. Schilling, B. W. Gibson, and R. E. Hughes, “Ubiquitin-specific peptidase 9, X-linked (USP9X) modulates activity of mammalian target of rapamycin (mTOR),” Journal of Biological Chemistry, vol. 287, pp. 21164–21175, 2012.
[375]  P. E. Czabotar, E. F. Lee, M. F. Van Delft et al., “Structural insights into the degradation of Mcl-1 induced by BH3 domains,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 15, pp. 6217–6222, 2007.
[376]  P. Gomez-Bougie, E. Menoret, P. Juin, C. Dousset, C. Pellat-Deceunynck, and M. Amiot, “Noxa controls Mule-dependent Mcl-1 ubiquitination through the regulation of the Mcl-1/USP9X interaction,” Biochemical and Biophysical Research Communications, vol. 413, pp. 460–464, 2011.
[377]  J. R. Mills, Y. Hippo, F. Robert et al., “mTORC1 promotes survival through translational control of Mcl-1,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 31, pp. 10853–10858, 2008.
[378]  K. Inoki, H. Ouyang, T. Zhu et al., “TSC2 integrates wnt and energy signals via a coordinated phosphorylation by AMPK and GSK3 to regulate cell growth,” Cell, vol. 126, no. 5, pp. 955–968, 2006.
[379]  S. N. Willis, L. Chen, G. Dewson et al., “Pro-apoptotic Bak is sequestered by Mcl-1 and Bcl-xL, but not Bcl-2, until displaced by BH3-only proteins,” Genes and Development, vol. 19, no. 11, pp. 1294–1305, 2005.
[380]  C. Desjobert, M. H. Renalier, J. Bergalet et al., “MiR-29a down-regulation in ALK-positive anaplastic large cell lymphomas contributes to apoptosis blockade through MCL-1 overexpression,” Blood, vol. 117, no. 24, pp. 6627–6637, 2011.
[381]  Y. K. Zhang, H. Wang, Y. Leng et al., “Overexpression of microRNA-29b induces apoptosis of multiple myeloma cells through down regulating Mcl-1,” Biochemical and Biophysical Research Communications, vol. 414, pp. 233–239, 2011.
[382]  J. L. Mott, S. Kobayashi, S. F. Bronk, and G. J. Gores, “mir-29 regulates Mcl-1 protein expression and apoptosis,” Oncogene, vol. 26, no. 42, pp. 6133–6140, 2007.
[383]  R. Garzon, C. E. A. Heaphy, V. Havelange et al., “MicroRNA 29b functions in acute myeloid leukemia,” Blood, vol. 114, no. 26, pp. 5331–5341, 2009.
[384]  H. J. Wang, H. J. Ruan, X. J. He et al., “MicroRNA-101 is down-regulated in gastric cancer and involved in cell migration and invasion,” European Journal of Cancer, vol. 46, no. 12, pp. 2295–2303, 2010.
[385]  R. Visone, L. Z. Rassenti, A. Veronese et al., “Karyotype-specific microRNA signature in chronic lymphocytic leukemia,” Blood, vol. 114, no. 18, pp. 3872–3879, 2009.
[386]  M. Crawford, K. Batte, L. Yu et al., “MicroRNA 133B targets pro-survival molecules MCL-1 and BCL2L2 in lung cancer,” Biochemical and Biophysical Research Communications, vol. 388, no. 3, pp. 483–489, 2009.
[387]  J. Chen, X. Zhang, C. Lentz et al., “miR-193b Regulates Mcl-1 in melanoma,” American Journal of Pathology, vol. 179, pp. 2162–2168, 2011.
[388]  Y. Saito, H. Suzuki, H. Tsugawa et al., “Chromatin remodeling at Alu repeats by epigenetic treatment activates silenced microRNA-512-5p with downregulation of Mcl-1 in human gastric cancer cells,” Oncogene, vol. 28, no. 30, pp. 2738–2744, 2009.
[389]  J. A. Spijkers-Hagelstein, P. Schneider, E. Hulleman et al., “Elevated S100A8/S100A9 expression causes glucocorticoid resistance in MLL-rearranged infant acute lymphoblastic leukemia,” Leukemia, vol. 26, pp. 1255–1265, 2012.
[390]  N. Minagawa, E. A. Kruglov, J. A. Dranoff, M. E. Robert, G. J. Gores, and M. H. Nathanson, “The anti-apoptotic protein Mcl-1 inhibits mitochondrial Ca2+ signals,” The Journal of Biological Chemistry, vol. 280, no. 39, pp. 33637–33644, 2005.
[391]  M. C. Bassik, L. Scorrano, S. A. Oakes, T. Pozzan, and S. J. Korsmeyer, “Phosphorylation of BCL-2 regulates ER Ca2+ homeostasis and apoptosis,” EMBO Journal, vol. 23, no. 5, pp. 1207–1216, 2004.
[392]  M. J. Thomenius and C. W. Distelhorst, “Bcl-2 on the endoplasmic reticulum: protecting the mitochondria from a distance,” Journal of Cell Science, vol. 116, no. 22, pp. 4493–4499, 2003.
[393]  C. Giorgi, F. Baldassari, A. Bononi, et al., “Mitochondrial Ca2+ and apoptosis,” Cell Calcium, vol. 52, pp. 36–43, 2012.
[394]  Y. P. Rong, A. S. Aromolaran, G. Bultynck et al., “Targeting Bcl-2-IP3 receptor interaction to reverse Bcl-2's inhibition of apoptotic calcium signals,” Molecular Cell, vol. 31, no. 2, pp. 255–265, 2008.
[395]  C. Li, X. Wang, H. Vais, C. B. Thompson, J. K. Foskett, and C. White, “Apoptosis regulation by Bcl-xL modulation of mammalian inositol 1,4,5-trisphosphate receptor channel isoform gating,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 30, pp. 12565–12570, 2007.
[396]  E. F. Eckenrode, J. Yang, G. V. Velmurugan, J. Kevin Foskett, and C. White, “Apoptosis protection by Mcl-1 and Bcl-2 modulation of inositol 1,4,5-trisphosphate receptor-dependent Ca2+ signaling,” The Journal of Biological Chemistry, vol. 285, no. 18, pp. 13678–13684, 2010.
[397]  C. Li, C. J. Fox, S. R. Master, V. P. Bindokas, L. A. Chodosh, and C. B. Thompson, “Bcl-XL affects Ca2+ homeostasis by altering expression of inositol 1,4,5-trisphosphate receptors,” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 15, pp. 9830–9835, 2002.
[398]  M. C. Jaramillo, J. B. Frye, J. D. Crapo, M. M. Briehl, and M. E. Tome, “Increased manganese superoxide dismutase expression or treatment with manganese porphyrin potentiates dexamethasone-induced apoptosis in lymphoma cells,” Cancer Research, vol. 69, no. 13, pp. 5450–5457, 2009.
[399]  M. E. Tome, A. F. Baker, G. Powis, C. M. Payne, and M. M. Briehl, “Catalase-overexpressing thymocytes are resistant to glucocorticoid-induced apoptosis and exhibit increased net tumor growth,” Cancer Research, vol. 61, no. 6, pp. 2766–2773, 2001.
[400]  M. E. Tome, N. W. Lutz, and M. M. Briehl, “Overexpression of catalase or Bcl-2 alters glucose and energy metabolism concomitant with dexamethasone resistance,” Biochimica et Biophysica Acta, vol. 1693, no. 1, pp. 57–72, 2004.
[401]  I. C. Low, J. Kang, and S. Pervaiz, “Bcl-2: a prime regulator of mitochondrial redox metabolism in cancer cells,” Antioxidants & Redox Signaling, vol. 15, pp. 2975–2987, 2011.
[402]  J. C. Aster, S. C. Blacklow, and W. S. Pear, “Notch signalling in T-cell lymphoblastic leukaemia/lymphoma and other haematological malignancies,” Journal of Pathology, vol. 223, no. 2, pp. 262–273, 2011.
[403]  A. Malyukova, T. Dohda, N. Von Der Lehr et al., “The tumor suppressor gene hCDC4 is frequently mutated in human T-cell acute lymphoblastic leukemia with functional consequences for Notch signaling,” Cancer Research, vol. 67, pp. 5611–5616, 2007.
[404]  C. ?berg, J. Li, A. Pauley, E. Wolf, M. Gurney, and U. Lendahl, “The Notch intracellular domain is ubiquitinated and negatively regulated by the mammalian Sel-10 homolog,” The Journal of Biological Chemistry, vol. 276, no. 38, pp. 35847–35853, 2001.
[405]  A. Ferrando, “NOTCH mutations as prognostic markers in T-ALL,” Leukemia, vol. 24, no. 12, pp. 2003–2004, 2010.
[406]  E. C. Lai, “Protein degradation: four E3s for the Notch pathway,” Current Biology, vol. 12, no. 2, pp. R74–R78, 2002.
[407]  S. J. Bray, “Notch signalling: a simple pathway becomes complex,” Nature Reviews Molecular Cell Biology, vol. 7, no. 9, pp. 678–689, 2006.
[408]  J. Nie, M. A. McGill, M. Dermer, S. E. Dho, C. D. Wolting, and C. J. McGlade, “LNX functions as a RING type E3 ubiquitin ligase that targets the cell fate determinant Numb for ubiquitin-dependent degradation,” EMBO Journal, vol. 21, no. 1-2, pp. 93–102, 2002.
[409]  M. Itoh, C. H. Kim, G. Palardy et al., “Mind bomb is a ubiquitin ligase that is essential for efficient activation of notch signaling by delta,” Developmental Cell, vol. 4, no. 1, pp. 67–82, 2003.
[410]  E. Pavlopoulos, C. Pitsouli, K. M. Klueg, M. A. T. Muskavitch, N. K. Moschonas, and C. Delidakis, “Neuralized encodes a peripheral membrane protein involved in delta signaling and endocytosis,” Developmental Cell, vol. 1, no. 6, pp. 807–816, 2001.
[411]  L. Qiu, C. Joazeiro, N. Fang et al., “Recognition and ubiquitination of Notch by Itch, a Hect-type E3 ubiquitin ligase,” The Journal of Biological Chemistry, vol. 275, no. 46, pp. 35734–35737, 2000.
[412]  A. H. Nwabo Kamdje and M. Krampera, “Notch signaling in acute lymphoblastic leukemia: any role for stromal microenvironment?” Blood, vol. 118, pp. 6506–6514, 2011.
[413]  A. H. N. Kamdje, F. Mosna, F. Bifari et al., “Notch-3 and Notch-4 signaling rescue from apoptosis human B-ALL cells in contact with human bone marrow-derived mesenchymal stromal cells,” Blood, vol. 118, no. 2, pp. 380–389, 2011.
[414]  Y. Nefedova, D. M. Sullivan, S. C. Bolick, W. S. Dalton, and D. I. Gabrilovich, “Inhibition of notch signaling induces apoptosis of myeloma cells and enhances sensitivity to chemotherapy,” Blood, vol. 111, no. 4, pp. 2220–2229, 2008.
[415]  M. T. Tetzlaff, W. Yu, M. Li et al., “Defective cardiovascular development and elevated cyclin E and Notch proteins in mice lacking the Fbw7 F-box protein,” Proceedings of the National Academy of Sciences of the United States of America, vol. 101, no. 10, pp. 3338–3345, 2004.
[416]  D. M. Koepp, L. K. Schaefer, X. Ye et al., “Phosphorylation-dependent ubiquitination of cyctin E by the SCFFbw7 ubiquitin ligase,” Science, vol. 294, no. 5540, pp. 173–177, 2001.
[417]  M. Baron, “Endocytic routes to Notch activation,” Seminars in Cell & Developmental Biology, vol. 23, pp. 437–442, 2012.
[418]  J. Jang, Y. I. Choi, J. Choi et al., “Notch1 confers thymocytes a resistance to GC-induced apoptosis through Deltex1 by blocking the recruitment of p300 to the SRG3 promoter,” Cell Death and Differentiation, vol. 13, no. 9, pp. 1495–1505, 2006.
[419]  C. Talora, A. F. Campese, D. Bellavia et al., “Notch signaling and diseases: n evolutionary journey from a simple beginning to complex outcomes,” Biochimica et Biophysica Acta, vol. 1782, no. 9, pp. 489–497, 2008.
[420]  T. Palomero and A. Ferrando, “Oncogenic NOTCH1 control of MYC and PI3K: challenges and opportunities for anti-NOTCH1 therapyinT-cell acute lymphoblastic leukemias and lymphomas,” Clinical Cancer Research, vol. 14, no. 17, pp. 5314–5317, 2008.
[421]  N. Chadwick, L. Zeef, V. Portillo et al., “Notch protection against apoptosis in T-ALL cells mediated by GIMAP5,” Blood Cells, Molecules, and Diseases, vol. 45, no. 3, pp. 201–209, 2010.
[422]  N. Chadwick, L. Zeef, V. Portillo et al., “Identification of novel Notch target genes in T cell leukaemia,” Molecular Cancer, vol. 8, article 35, 2009.
[423]  T. Nitta, M. Nasreen, T. Seike et al., “IAN family critically regulates survival and development of T lymphocytes,” PLoS Biology, vol. 4, no. 4, p. e103, 2006.
[424]  T. Zenz, A. Roessner, A. Thomas et al., “hlan5: the human ortholog to the rat lan4/lddm1/lyp is a new member of the Ian family that is overexpressed in B-cell lymphoid malignancies,” Genes and Immunity, vol. 5, no. 2, pp. 109–116, 2004.
[425]  M. Keita, C. Leblanc, D. Andrews, and S. Ramanathan, “GIMAP5 regulates mitochondrial integrity from a distinct subcellular compartment,” Biochemical and Biophysical Research Communications, vol. 361, no. 2, pp. 481–486, 2007.
[426]  S. Filén and R. Lahesmaa, “GIMAP proteins in T-lymphocytes,” Journal of Signal Transduction, vol. 2010, Article ID 268589, 10 pages, 2010.
[427]  M. S. Hyun, L. M. Minter, H. C. Ok et al., “Notch1 augments NF-κB activity by facilitating its nuclear retention,” EMBO Journal, vol. 25, no. 1, pp. 129–138, 2006.
[428]  T. Palomero, K. L. Wei, D. T. Odom et al., “NOTCH1 directly regulates c-MYC and activates a feed-forward-loop transcriptional network promoting leukemic cell growth,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 48, pp. 18261–18266, 2006.
[429]  V. M. Sharma, J. A. Calvo, K. M. Draheim et al., “Notch1 contributes to mouse T-cell leukemia by directly inducing the expression of c-myc,” Molecular and Cellular Biology, vol. 26, no. 21, pp. 8022–8031, 2006.
[430]  A. P. Weng, J. M. Millholland, Y. Yashiro-Ohtani et al., “c-Myc is an important direct target of Notch1 in T-cell acute lymphoblastic leukemia/lymphoma,” Genes and Development, vol. 20, no. 15, pp. 2096–2109, 2006.
[431]  S. Cialfi, R. Palermo, S. Manca et al., “Glucocorticoid sensitivity of T-cell lymphoblastic leukemia/lymphoma is associated with glucocorticoid receptor-mediated inhibition of Notch1 expression,” Leukemia. In press.
[432]  J. S. Mo, E. J. Ann, J. H. Yoon et al., “Serum- and glucocorticoid-inducible kinase 1 (SGK1) controls notch1 signaling by downregulation of protein stability through Fbw7 ubiquitin ligase,” Journal of Cell Science, vol. 124, no. 1, pp. 100–112, 2011.
[433]  C. Kox, M. Zimmermann, M. Stanulla et al., “The favorable effect of activating NOTCH1 receptor mutations on long-term outcome in T-ALL patients treated on the ALL-BFM 2000 protocol can be separated from FBXW7 loss of function,” Leukemia, vol. 24, no. 12, pp. 2005–2013, 2010.
[434]  L. Zuurbier, I. Homminga, V. Calvert et al., “NOTCH1 and/or FBXW7 mutations predict for initial good prednisone response but not for improved outcome in pediatric T-cell acute lymphoblastic leukemia patients treated on DCOG or COALL protocols,” Leukemia, vol. 24, no. 12, pp. 2014–2022, 2010.
[435]  V. Asnafi, A. Buzyn, S. Le Noir et al., “NOTCH1/FBXW7 mutation identifies a large subgroup with favorable outcome in adult T-cell acute lymphoblastic leukemia (T-ALL): a Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL) study,” Blood, vol. 113, no. 17, pp. 3918–3924, 2009.
[436]  E. Clappier, S. Collette, N. Grardel et al., “NOTCH1 and FBXW7 mutations have a favorable impact on early response to treatment, but not on outcome, in children with T-cell acute lymphoblastic leukemia (T-ALL) treated on EORTC trials 58881 and 58951,” Leukemia, vol. 24, no. 12, pp. 2023–2031, 2010.
[437]  Y. Tang, Y. Wang, L. Chen, N. Weintraub, and Y. Pan, “Cross talk between the notch signaling and noncoding RNA on the fate of stem cells,” Progress in Molecular Biology and Translational Science, vol. 111, pp. 175–193, 2012.
[438]  M. Lerner, J. Lundgren, S. Akhoondi et al., “MiRNA-27a controls FBW7/hCDC4-dependent cyclin E degradation and cell cycle progression,” Cell Cycle, vol. 10, no. 13, pp. 2172–2183, 2011.
[439]  Q. Wang, D. C. Li, Z. F. Li et al., “Upregulation of miR-27a contributes to the malignant transformation of human bronchial epithelial cells induced by SV40 small T antigen,” Oncogene, vol. 30, pp. 3875–3886, 2011.
[440]  X. Li, T. Sanda, A. Thomas Look, C. D. Novina, and H. von Boehmer, “Repression of tumor suppressor miR-451 is essential for NOTCH1-induced oncogenesis in T-ALL,” Journal of Experimental Medicine, vol. 208, no. 4, pp. 663–675, 2011.
[441]  M. Ghisi, A. Corradin, K. Basso et al., “Modulation of microRNA expression in human T-cell development: targeting of NOTCH3 by miR-150,” Blood, vol. 117, no. 26, pp. 7053–7062, 2011.
[442]  B. Kefas, L. Comeau, D. H. Floyd et al., “The neuronal microRNA miR-326 acts in a feedback loop with Notch and has therapeutic potential against brain tumors,” Journal of Neuroscience, vol. 29, no. 48, pp. 15161–15168, 2009.
[443]  B. P. Lewis, I. H. Shih, M. W. Jones-Rhoades, D. P. Bartel, and C. B. Burge, “Prediction of mammalian microRNA targets,” Cell, vol. 115, no. 7, pp. 787–798, 2003.
[444]  P. de Antonellis, C. Medaglia, E. Cusanelli, et al., “MiR-34a targeting of Notch ligand delta-like 1 impairs CD15+/CD133+ tumor-propagating cells and supports neural differentiation in medulloblastoma,” PLoS ONE, vol. 6, Article ID e24584, 2011.
[445]  V. Manfè, L. M. Holst, A. Rosbjerg, M. R. Kamstrup, B. Kaczkowski, and R. Gniadecki, “Changes in oncomiR expression in CTCL cell lines during apoptosis induced by Notch inhibition,” Leukemia Research, vol. 34, no. 9, pp. e235–e236, 2010.
[446]  S. U. Mertens-Talcott, S. Chintharlapalli, X. Li, and S. Safe, “The oncogenic microRNA-27a targets genes that regulate specificity protein transcription factors and the G2-M checkpoint in MDA-MB-231 breast cancer cells,” Cancer Research, vol. 67, no. 22, pp. 11001–11011, 2007.
[447]  C. V. Dang, “MYC on the path to cancer,” Cell, vol. 149, pp. 22–35, 2012.
[448]  R. D. Medh, A. Wang, F. Zhou, and E. B. Thompson, “Constitutive expression of ectopic c-Myc delays glucocorticoid-evoked apoptosis of human leukemic CEM-C7 cells,” Oncogene, vol. 20, no. 34, pp. 4629–4639, 2001.
[449]  D. W. Felsher and J. M. Bishop, “Reversible tumorigenesis by MYC in hematopoietic lineages,” Molecular Cell, vol. 4, no. 2, pp. 199–207, 1999.
[450]  M. Bonnet, M. Loosveld, B. Montpellier et al., “Posttranscriptional deregulation of MYC via PTEN constitutes a major alternative pathway of MYC activation in T-cell acute lymphoblastic leukemia,” Blood, vol. 117, no. 24, pp. 6650–6659, 2011.
[451]  J. Battey, C. Moulding, R. Taub, et al., “The human c-myc oncogene: structural consequences of translocation into the IgH locus in Burkitt-lymphoma,” Cell, vol. 34, no. 3, pp. 779–787, 1983.
[452]  D. Dominguez-Sola and R. Dalla-Favera, “Burkitt lymphoma: much more than MYC,” Cancer Cell, vol. 22, pp. 141–142, 2012.
[453]  E. M. Molyneux, R. Rochford, B. Griffin et al., “Burkitt's lymphoma,” The Lancet, vol. 379, pp. 1234–1244, 2012.
[454]  K. Yamamoto, H. Matsuoka, K. Yakushijin et al., “A novel five-way translocation, t(3,9,13,8,14)(q27,p13,q32,q24,q32), with concurrent MYC and BCL6 rearrangements in a primary bone marrow B-cell lymphoma,” Cancer Genetics, vol. 204, pp. 501–506, 2011.
[455]  N. Tomita, “BCL2 and MYC dual-hit lymphoma/leukemia,” Journal of Clinical and Experimental Hematopathology, vol. 51, pp. 7–12, 2011.
[456]  M. Snuderl, O. K. Kolman, Y. B. Chen et al., “B-cell lymphomas with concurrent IGH-BCL2 and MYC rearrangements are aggressive neoplasms with clinical and pathologic features distinct from Burkitt lymphoma and diffuse large B-cell lymphoma,” American Journal of Surgical Pathology, vol. 34, no. 3, pp. 327–340, 2010.
[457]  G. W. Slack and R. D. Gascoyne, “MYC and aggressive B-cell lymphomas,” Advances in Anatomic Pathology, vol. 18, no. 3, pp. 219–228, 2011.
[458]  S. Shiratori, T. Kondo, S. Fujisawa et al., “C-myc rearrangement in B-cell lymphoblastic lymphoma with the involvement of multiple extranodal lesions,” Leukemia and Lymphoma, vol. 52, no. 4, pp. 716–718, 2011.
[459]  M. L. Slovak, J. P. Ho, M. J. Pettenati et al., “Localization of amplified MYC gene sequences to double minute chromosomes in acute myelogenous leukemia,” Genes Chromosomes and Cancer, vol. 9, no. 1, pp. 62–67, 1994.
[460]  R. D. Medh, M. F. Saeed, B. H. Johnson, and E. B. Thompson, “Resistance of human leukemic CEM-C1 cells is overcome by synergism between glucocorticoid and protein kinase A pathways: correlation with c-Myc suppression,” Cancer Research, vol. 58, no. 16, pp. 3684–3693, 1998.
[461]  E. B. Thompson, R. Thulasi, M. F. Saeed, and B. H. Johnson, “Glucocorticoid antagonist RU 486 reverses agonist-induced apoptosis and c-myc repression in human leukemic CEM-C7 cells,” Annals of the New York Academy of Sciences, vol. 761, pp. 261–275, 1995.
[462]  M. J. Ausserlechner, P. Obexer, G. B?ck, S. Geley, and R. Kofler, “Cyclin D3 and c-MYC control glucocorticoid-induced cell cycle arrest but not apoptosis in lymphoblastic leukemia cells,” Cell Death and Differentiation, vol. 11, no. 2, pp. 165–174, 2004.
[463]  E. M. Blackwood and R. N. Eisenman, “Max: a helix-loop-helix zipper protein that forms a sequence-specific DNA-binding complex with Myc,” Science, vol. 251, no. 4998, pp. 1211–1217, 1991.
[464]  K. I. Zeller, X. Zhao, C. W. H. Lee et al., “Global mapping of c-Myc binding sites and target gene networks in human B cells,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 47, pp. 17834–17839, 2006.
[465]  S. Sander, L. Bullinger, and T. Wirth, “Repressing the repressor—a new mode of MYC action in lymphomagenesis,” Cell Cycle, vol. 8, no. 4, pp. 556–559, 2009.
[466]  D. Kleine-Kohlbrecher, S. Adhikary, and M. Eilers, “Mechanisms of transcriptional repression by Myc,” Current Topics in Microbiology and Immunology, vol. 302, pp. 51–62, 2006.
[467]  Y. Sylvestre, V. De Guire, E. Querido et al., “An E2F/miR-20a autoregulatory feedback loop,” The Journal of Biological Chemistry, vol. 282, no. 4, pp. 2135–2143, 2007.
[468]  Z. Yu, C. Wang, M. Wang et al., “A cyclin D1/microRNA 17/20 regulatory feedback loop in control of breast cancer cell proliferation,” Journal of Cell Biology, vol. 182, no. 3, pp. 509–517, 2008.
[469]  L. He, J. M. Thomson, M. T. Hemann et al., “A microRNA polycistron as a potential human oncogene,” Nature, vol. 435, no. 7043, pp. 828–833, 2005.
[470]  S. Polager and D. Ginsberg, “E2F—at the crossroads of life and death,” Trends in Cell Biology, vol. 18, no. 11, pp. 528–535, 2008.
[471]  M. Ofir, D. Hacohen, and D. Ginsberg, “miR-15 and miR-16 are direct transcriptional targets of E2F1 that limit E2F-induced proliferation by targeting cyclin E,” Molecular Cancer Research, vol. 9, no. 4, pp. 440–447, 2011.
[472]  D. Bashari, D. Hacohen, and D. Ginsberg, “JNK activation is regulated by E2F and promotes E2F1-induced apoptosis,” Cellular Signalling, vol. 23, no. 1, pp. 65–70, 2011.
[473]  T. C. Chang, D. Yu, Y. S. Lee et al., “Widespread microRNA repression by Myc contributes to tumorigenesis,” Nature Genetics, vol. 40, no. 1, pp. 43–50, 2008.
[474]  H. Zhu, H. Wu, X. Liu et al., “Regulation of autophagy by a beclin 1-targeted microRNA, miR-30a, in cancer cells,” Autophagy, vol. 5, no. 6, pp. 816–823, 2009.
[475]  Z. Zou, L. Wu, H. Ding et al., “MicroRNA-30a sensitizes tumor cells to cis-platinum via suppressing beclin 1-mediated autophagy,” The Journal of Biological Chemistry, vol. 287, pp. 4148–4156, 2012.
[476]  S. M. Johnson, H. Grosshans, J. Shingara et al., “RAS is regulated by the let-7 microRNA family,” Cell, vol. 120, no. 5, pp. 635–647, 2005.
[477]  S. L. Yong and A. Dutta, “The tumor suppressor microRNA let-7 represses the HMGA2 oncogene,” Genes and Development, vol. 21, no. 9, pp. 1025–1030, 2007.
[478]  C. Mayr, M. T. Hemann, and D. P. Bartel, “Disrupting the pairing between let-7 and Hmga2 enhances oncogenic transformation,” Science, vol. 315, no. 5818, pp. 1576–1579, 2007.
[479]  C. D. Johnson, A. Esquela-Kerscher, G. Stefani et al., “The let-7 microRNA represses cell proliferation pathways in human cells,” Cancer Research, vol. 67, no. 16, pp. 7713–7722, 2007.
[480]  P. Gao, I. Tchernyshyov, T. C. Chang et al., “C-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism,” Nature, vol. 458, no. 7239, pp. 762–765, 2009.
[481]  J. Lu, M. L. He, L. Wang et al., “MiR-26a inhibits cell growth and tumorigenesis of nasopharyngeal carcinoma through repression of EZH2,” Cancer Research, vol. 71, no. 1, pp. 225–233, 2011.
[482]  J. Ji, J. Shi, A. Budhu et al., “MicroRNA expression, survival, and response to interferon in liver cancer,” The New England Journal of Medicine, vol. 361, no. 15, pp. 1437–1447, 2009.
[483]  S. Sander, L. Bullinger, K. Klapproth et al., “MYC stimulates EZH2 expression by repression of its negative regulator miR-26a,” Blood, vol. 112, no. 10, pp. 4202–4212, 2008.
[484]  T. C. Chang, E. A. Wentzel, O. A. Kent et al., “Transactivation of miR-34a by p53 broadlyinfluences gene expression andpromotesapoptosis,” Molecular Cell, vol. 26, no. 5, pp. 745–752, 2007.
[485]  L. He, X. He, L. P. Lim et al., “A microRNA component of the p53 tumour suppressor network,” Nature, vol. 447, no. 7148, pp. 1130–1134, 2007.
[486]  N. Raver-Shapira, E. Marciano, E. Meiri et al., “Transcriptional activation of miR-34a contributes to p53-mediated apoptosis,” Molecular Cell, vol. 26, no. 5, pp. 731–743, 2007.
[487]  T. C. Chang, L. R. Zeitels, H. W. Hwang et al., “Lin-28B transactivation is necessary for Myc-mediated let-7 repression and proliferation,” Proceedings of the National Academy of Sciences of the United States of America, vol. 106, no. 9, pp. 3384–3389, 2009.
[488]  E. Piskounova, C. Polytarchou, J. E. Thornton et al., “Lin28A and Lin28B inhibit let-7 microRNA biogenesis by distinct mechanisms,” Cell, vol. 147, pp. 1066–1079, 2011.
[489]  S. R. Viswanathan, G. Q. Daley, and R. I. Gregory, “Selective blockade of microRNA processing by Lin28,” Science, vol. 320, no. 5872, pp. 97–100, 2008.
[490]  S. R. Viswanathan, J. T. Powers, W. Einhorn et al., “Lin28 promotes transformation and is associated with advanced human malignancies,” Nature Genetics, vol. 41, no. 7, pp. 843–848, 2009.
[491]  J. Yu, M. A. Vodyanik, K. Smuga-Otto et al., “Induced pluripotent stem cell lines derived from human somatic cells,” Science, vol. 318, no. 5858, pp. 1917–1920, 2007.
[492]  S. Dangi-Garimella, J. Yun, E. M. Eves et al., “Raf kinase inhibitory protein suppresses a metastasis signalling cascade involving LIN28 and let-7,” EMBO Journal, vol. 28, no. 4, pp. 347–358, 2009.
[493]  W. K. K. Wu, S. B. Coffelt, C. H. Cho et al., “The autophagic paradox in cancer therapy,” Oncogene, vol. 31, pp. 939–953, 2011.
[494]  B. Levine, S. Sinha, and G. Kroemer, “Bcl-2 family members: dual regulators of apoptosis and autophagy,” Autophagy, vol. 4, no. 5, pp. 600–606, 2008.
[495]  M. C. Maiuri, G. Le Toumelin, A. Criollo et al., “Functional and physical interaction between Bcl-XL and a BH3-like domain in Beclin-1,” EMBO Journal, vol. 26, no. 10, pp. 2527–2539, 2007.
[496]  S. Pattingre, A. Tassa, X. Qu et al., “Bcl-2 anti-apoptotic proteins inhibit Beclin 1-dependent autophagy,” Cell, vol. 122, no. 6, pp. 927–939, 2005.
[497]  E. Wirawan, L. Vande Walle, K. Kersse et al., “Caspase-mediated cleavage of Beclin-1 inactivates Beclin-1-induced autophagy and enhances apoptosis by promoting the release of pro-apoptotic factors from mitochondria,” Cell Death and Disease, vol. 1, no. 1, article e18, 2010.
[498]  J. Wang, “Beclin 1 bridges autophagy, apoptosis and differentiation,” Autophagy, vol. 4, no. 7, pp. 947–948, 2008.
[499]  X. Qu, J. Yu, G. Bhagat et al., “Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene,” Journal of Clinical Investigation, vol. 112, no. 12, pp. 1809–1820, 2003.
[500]  J. J. Huang, Y. J. Zhu, T. Y. Lin, W. Q. Jiang, H. Q. Huang, and Z. M. Li, “Beclin 1 expression predicts favorable clinical outcome in patients with diffuse large B-cell lymphoma treated with R-CHOP,” Human Pathology, vol. 42, no. 10, pp. 1459–1466, 2011.
[501]  E. Laane, K. P. Tamm, E. Buentke et al., “Cell death induced by dexamethasone in lymphoid leukemia is mediated through initiation of autophagy,” Cell Death and Differentiation, vol. 16, no. 7, p. 1071, 2009.
[502]  S. Swerdlow, K. McColl, Y. Rong, M. Lam, A. Gupta, and C. W. Distelhorst, “Apoptosis inhibition by Bcl-2 gives way to autophagy in glucocorticoid-treated lymphocytes,” Autophagy, vol. 4, no. 5, pp. 612–620, 2008.
[503]  J. K. Molitoris, K. S. McColl, S. Swerdlow et al., “Glucocorticoid elevation of dexamethasone-induced gene 2 (Dig2/RTP801/REDD1) protein mediates autophagy in lymphocytes,” The Journal of Biological Chemistry, vol. 286, pp. 30181–30189, 2011.
[504]  M. P. Deyoung, P. Horak, A. Sofer, D. Sgroi, and L. W. Ellisen, “Hypoxia regulates TSC1/2-mTOR signaling and tumor suppression through REDD1-mediated 14-3-3 shuttling,” Genes and Development, vol. 22, no. 2, pp. 239–251, 2008.
[505]  L. Galluzzi and G. Kroemer, “Necroptosis: a specialized pathway of programmed Necrosis,” Cell, vol. 135, no. 7, pp. 1161–1163, 2008.
[506]  H. Ye, X. Liu, M. Lv et al., “MicroRNA and transcription factor co-regulatory network analysis reveals miR-19 inhibits CYLD in T-cell acute lymphoblastic leukemia,” Nucleic Acids Research, vol. 40, pp. 5201–5214, 2012.
[507]  T. Otsuki, H. M. Clark, A. Wellmann, E. S. Jaffe, and M. Raffeld, “Involvement of CDKN2 (p16(INK4A)/MTS1) and p15(INK4B)/MTS2 in human leukemias and lymphomas,” Cancer Research, vol. 55, no. 7, pp. 1436–1440, 1995.
[508]  T. Okuda, S. A. Shurtleff, M. B. Valentine et al., “Frequent deletion of p16(INK4a)/MTS1 and p15(INK4b)/MTS2 in pediatric acute lymphoblastic leukemia,” Blood, vol. 85, no. 9, pp. 2321–2330, 1995.
[509]  U. R. Kees, P. R. Burton, C. Lü, and D. L. Baker, “Homozygous deletion of the p16/MTS1 gene in pediatric acute lymphoblastic leukemia is associated with unfavorable clinical outcome,” Blood, vol. 89, no. 11, pp. 4161–4166, 1997.
[510]  M. Fizzotti, G. Cimino, S. Pisegna et al., “Detection of homozygous deletions of the cyclin-dependent kinase 4 inhibitor (p16) gene in acute lymphoblastic leukemia and association with adverse prognostic features,” Blood, vol. 85, no. 10, pp. 2685–2690, 1995.
[511]  P. Obexer, J. Hagenbuchner, M. Rupp et al., “p16INK4A sensitizes human leukemia cells to FAS- and glucocorticoid-induced apoptosis via induction of BBC3/Puma and repression of MCL1 and BCL2,” The Journal of Biological Chemistry, vol. 284, no. 45, pp. 30933–30940, 2009.
[512]  F. Jardin, J. P. Jais, T. J. Molina et al., “Diffuse large B-cell lymphomas with CDKN2A deletion have a distinct gene expression signature and a poor prognosis under R-CHOP treatment: a GELA study,” Blood, vol. 116, no. 7, pp. 1092–1104, 2010.
[513]  K. J. Savage, N. A. Johnson, S. Ben-Neriah et al., “MYC gene rearrangements are associated with a poor prognosis in diffuse large B-cell lymphoma patients treated with R-CHOP chemotherapy,” Blood, vol. 114, no. 17, pp. 3533–3537, 2009.
[514]  U. Bissels, A. Bosio, and W. Wagner, “MicroRNAs are shaping the hematopoietic landscape,” Haematologica, vol. 97, pp. 160–167, 2012.
[515]  R. Garzon, S. K. Sandhu, and C. M. Croce, “Micro-RNA expression and function in lymphomas,” Advances in Hematology, vol. 2011, Article ID 347137, 2011.
[516]  S. Babashah and M. Soleimani, “The oncogenic and tumour suppressive roles of microRNAs in cancer and apoptosis,” European Journal of Cancer, vol. 47, no. 8, pp. 1127–1137, 2011.
[517]  A. L. Kasinski and F. J. Slack, “Epigenetics and genetics. MicroRNAs en route to the clinic: progress in validating and targeting microRNAs for cancer therapy,” Nature Reviews Cancer, vol. 11, pp. 849–864, 2011.
[518]  B. M. Ryan, A. I. Robles, and C. C. Harris, “Genetic variation in microRNA networks: the implications for cancer research,” Nature Reviews Cancer, vol. 10, no. 6, pp. 389–402, 2010.
[519]  G. A. Calin and C. M. Croce, “MicroRNA signatures in human cancers,” Nature Reviews Cancer, vol. 6, no. 11, pp. 857–866, 2006.
[520]  D. Schotte, J. C. K. Chau, G. Sylvester et al., “Identification of new microRNA genes and aberrant microRNA profiles in childhood acute lymphoblastic leukemia,” Leukemia, vol. 23, no. 2, pp. 313–322, 2009.
[521]  M. Lu, Q. Zhang, M. Deng et al., “An analysis of human microRNA and disease associations,” PLoS ONE, vol. 3, no. 10, Article ID e3420, 2008.
[522]  M. V. Iorio and C. M. Croce, “MicroRNAs in cancer: small molecules with a huge impact,” Journal of Clinical Oncology, vol. 27, no. 34, pp. 5848–5856, 2009.
[523]  J. Lu, G. Getz, E. A. Miska et al., “MicroRNA expression profiles classify human cancers,” Nature, vol. 435, no. 7043, pp. 834–838, 2005.
[524]  W. Filipowicz, S. N. Bhattacharyya, and N. Sonenberg, “Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight?” Nature Reviews Genetics, vol. 9, no. 2, pp. 102–114, 2008.
[525]  G. Di Leva, D. Briskin, and C. M. Croce, “MicroRNA in cancer: new hopes for antineoplastic chemotherapy,” Upsala Journal of Medical Sciences, vol. 117, pp. 202–216, 2012.
[526]  J. G. Ruby, C. H. Jan, and D. P. Bartel, “Intronic microRNA precursors that bypass Drosha processing,” Nature, vol. 448, no. 7149, pp. 83–86, 2007.
[527]  E. Berezikov, W. J. Chung, J. Willis, E. Cuppen, and E. C. Lai, “Mammalian mirtron genes,” Molecular Cell, vol. 28, no. 2, pp. 328–336, 2007.
[528]  L. Wu and J. G. Belasco, “Let me count the ways: mechanisms of gene regulation by miRNAs and siRNAs,” Molecular Cell, vol. 29, no. 1, pp. 1–7, 2008.
[529]  J. R. Buchan and R. Parker, “The two faces of miRNA,” Science, vol. 318, no. 5858, pp. 1877–1878, 2007.
[530]  S. H. Orkin and L. I. Zon, “Hematopoiesis: an evolving paradigm for stem cell biology,” Cell, vol. 132, no. 4, pp. 631–644, 2008.
[531]  H. F. Lodish, B. Zhou, G. Liu, and C. Z. Chen, “Micromanagement of the immune system by microRNAs,” Nature Reviews Immunology, vol. 8, no. 2, pp. 120–130, 2008.
[532]  M. A. Lindsay, “microRNAs and the immune response,” Trends in Immunology, vol. 29, no. 7, pp. 343–351, 2008.
[533]  R. Malumbres, K. A. Sarosiek, E. Cubedo et al., “Differentiation stage-specific expression of microRNAs in B lymphocytes and diffuse large B-cell lymphomas,” Blood, vol. 113, no. 16, pp. 3754–3764, 2009.
[534]  S. A. Muljo, K. Mark Ansel, C. Kanellopoulou, D. M. Livingston, A. Rao, and K. Rajewsky, “Aberrant T cell differentiation in the absence of Dicer,” Journal of Experimental Medicine, vol. 202, no. 2, pp. 261–269, 2005.
[535]  B. S. Cobb, A. Hertweck, J. Smith et al., “A role for Dicer in immune regulation,” Journal of Experimental Medicine, vol. 203, no. 11, pp. 2519–2527, 2006.
[536]  A. Liston, L. F. Lu, D. O'Carroll, A. Tarakhovsky, and A. Y. Rudensky, “Dicer-dependent microRNA pathway safeguards regulatory T cell function,” Journal of Experimental Medicine, vol. 205, no. 9, pp. 1993–2004, 2008.
[537]  S. B. Koralov, S. A. Muljo, G. R. Galler et al., “Dicer ablation affects antibody diversity and cell survival in the B lymphocyte lineage,” Cell, vol. 132, no. 5, pp. 860–874, 2008.
[538]  D. Cifuentes, H. Xue, D. W. Taylor et al., “A novel miRNA processing pathway independent of dicer requires argonaute2 catalytic activity,” Science, vol. 328, no. 5986, pp. 1694–1698, 2010.
[539]  J. R. Neilson, G. X. Y. Zheng, C. B. Burge, and P. A. Sharp, “Dynamic regulation of miRNA expression in ordered stages of cellular development,” Genes and Development, vol. 21, no. 5, pp. 578–589, 2007.
[540]  G. J. Wiegers, M. Knoflach, G. Bock, et al., “CD4+CD8+TCRlow thymocytes express low levels of glucocorticoid receptors while being sensitive to glucocorticoid-induced apoptosis,” European Journal of Immunology, vol. 31, pp. 2293–2301, 2001.
[541]  N. Rosenheimer-Goudsmid, Y. Haupt, E. Yefenof, Y. Zilberman, and R. Guy, “p53 and thymic ‘death by neglect’: thymic epithelial cell-induced apoptosis of CD4+8+ thymocytes is p53-independent,” Cell Death and Differentiation, vol. 7, no. 3, pp. 241–249, 2000.
[542]  J. D. Ashwell, F. W. M. Lu, and M. S. Vacchio, “Glucocorticoids in T cell development and function,” Annual Review of Immunology, vol. 18, pp. 309–345, 2000.
[543]  B. S. Cobb, T. B. Nesterova, E. Thompson et al., “T cell lineage choice and differentiation in the absence of the RNase III enzyme Dicer,” Journal of Experimental Medicine, vol. 201, no. 9, pp. 1367–1373, 2005.
[544]  A. Ceribelli, M. Satoh, and E. K. Chan, “MicroRNAs and autoimmunity,” Current Opinion in Immunology, vol. 24, no. 6, pp. 686–691, 2012.
[545]  N. Rusca and S. Monticelli, “MiR-146a in immunity and disease,” Molecular Biology International, vol. 2011, Article ID 437301, 7 pages, 2011.
[546]  Q. J. Li, J. Chau, P. J. R. Ebert et al., “miR-181a is an intrinsic modulator of T cell sensitivity and selection,” Cell, vol. 129, no. 1, pp. 147–161, 2007.
[547]  C. Xiao, D. P. Calado, G. Galler et al., “MiR-150 controls B cell differentiation by targeting the transcription factor c-Myb,” Cell, vol. 131, no. 1, pp. 146–159, 2007.
[548]  P. J. Ebert, S. Jiang, J. Xie, Q. J. Li, and M. M. Davis, “An endogenous positively selecting peptide enhances mature T cell responses and becomes an autoantigen in the absence of microRNA miR-181a,” Nature Immunology, vol. 10, no. 11, pp. 1162–1169, 2009.
[549]  R. Fragoso, T. Mao, S. Wang et al., “Modulating the strength and threshold of NOTCH oncogenic signals by mir-181a-1/b-1,” PLOS Genetics, vol. 8, Article ID e1002855, 2012.
[550]  C. Z. Chen, L. Li, H. F. Lodish, and D. P. Bartel, “MicroRNAs modulate hematopoietic lineage differentiation,” Science, vol. 303, no. 5654, pp. 83–86, 2004.
[551]  F. Cichocki, M. Felices, V. McCullar et al., “Cutting edge: microRNA-181 promotes human NK cell development by regulating Notch signaling,” Journal of Immunology, vol. 187, pp. 6171–6175, 2011.
[552]  X. Li, J. Zhang, L. Gao et al., “MiR-181 mediates cell differentiation by interrupting the Lin28 and let-7 feedback circuit,” Cell Death & Differentiation, vol. 19, pp. 378–386, 2012.
[553]  B. Zhou, S. Wang, C. Mayr, D. P. Bartel, and H. F. Lodish, “miR-150, a microRNA expressed in mature B and T cells, blocks early B cell development when expressed prematurely,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 17, pp. 7080–7085, 2007.
[554]  J. Lu, S. Guo, B. L. Ebert et al., “MicroRNA-mediated control of cell fate in megakaryocyte-erythrocyte progenitors,” Developmental Cell, vol. 14, no. 6, pp. 843–853, 2008.
[555]  N. A. Bezman, T. Chakraborty, T. Bender, and L. L. Lanier, “miR-150 regulates the development of NK and iNKT cells,” Journal of Experimental Medicine, vol. 208, pp. 2717–2731, 2011.
[556]  Q. Zheng, L. Zhou, and Q. S. Mi, “MicroRNA miR-150 is involved in Valpha14 invariant NKT cell development and function,” Journal of Immunology, vol. 188, pp. 2118–2126, 2012.
[557]  D. L. Zanette, F. Rivadavia, G. A. Molfetta et al., “miRNA expression profiles in chronic lymphocytic and acute lymphocytic leukemia,” Brazilian Journal of Medical and Biological Research, vol. 40, no. 11, pp. 1435–1440, 2007.
[558]  H. Zhao, D. Wang, W. Du, D. Gu, and R. Yang, “MicroRNA and leukemia: tiny molecule, great function,” Critical Reviews in Oncology/Hematology, vol. 74, no. 3, pp. 149–155, 2010.
[559]  Y. Wang, Z. Li, C. He et al., “MicroRNAs expression signatures are associated with lineage and survival in acute leukemias,” Blood Cells, Molecules, and Diseases, vol. 44, no. 3, pp. 191–197, 2010.
[560]  X. Agirre, A. Vilas-Zornoza, A. Jiménez-Velasco et al., “Epigenetic silencing of the tumor suppressor microRNA Hsa-miR-124a regulates CDK6 expression and confers a poor prognosis in acute lymphoblastic leukemia,” Cancer Research, vol. 69, no. 10, pp. 4443–4453, 2009.
[561]  S. Mi, J. Lu, M. Sun et al., “MicroRNA expression signatures accurately discriminate acute lymphoblastic leukemia from acute myeloid leukemia,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 50, pp. 19971–19976, 2007.
[562]  S. Babashah, M. Sadeghizadeh, M. R. Tavirani, S. Farivar, and M. Soleimani, “Aberrant microRNA expression and its implications in the pathogenesis of leukemias,” Cellular Oncology, vol. 35, no. 5, pp. 317–334, 2012.
[563]  G. A. Calin, M. Ferracin, A. Cimmino et al., “A microRNA signature associated with prognosis and progression in chronic lymphocytic leukemia,” The New England Journal of Medicine, vol. 353, no. 17, pp. 1793–1801, 2005.
[564]  L. Di Lisio, M. Sanchez-Beato, G. Gomez-Lopez, et al., “MicroRNA signatures in B-cell lymphomas,” Blood Cancer Journal, vol. 2, Article ID e57, 2012.
[565]  S. Li, H. F. Moffett, J. Lu et al., “Microrna expression profiling identifies activated B cell status in chronic lymphocytic leukemia cells,” PLoS ONE, vol. 6, no. 3, Article ID e16956, 2011.
[566]  V. Fulci, S. Chiaretti, M. Goldoni et al., “Quantitative technologies establish a novel microRNA profile of chronic lymphocytic leukemia,” Blood, vol. 109, no. 11, pp. 4944–4951, 2007.
[567]  M. Mraz, S. Pospisilova, K. Malinova, I. Slapak, and J. Mayer, “MicroRNAs in chronic lymphocytic leukemia pathogenesis and disease subtypes,” Leukemia and Lymphoma, vol. 50, no. 3, pp. 506–509, 2009.
[568]  S. Marton, M. R. Garcia, C. Robello et al., “Small RNAs analysis in CLL reveals a deregulation of miRNA expression and novel miRNA candidates of putative relevance in CLL pathogenesis,” Leukemia, vol. 22, no. 2, pp. 330–338, 2008.
[569]  Y. Pekarsky, U. Santanam, A. Cimmino et al., “Tcl1 expression in chronic lymphocytic leukemia is regulated by miR-29 and miR-181,” Cancer Research, vol. 66, no. 24, pp. 11590–11593, 2006.
[570]  T. Zenz, J. Mohr, E. Eldering et al., “miR-34a as part of the resistance network in chronic lymphocytic leukemia,” Blood, vol. 113, no. 16, pp. 3801–3808, 2009.
[571]  E. Tili, J. J. Michaille, Z. Luo et al., “The downregulation of miR-125b in chronic lymphocytic leukemias leads to metabolic adaptation of cells to a transformed state,” Blood, vol. 120, no. 13, pp. 2631–2638, 2012.
[572]  E. Barbarotto, T. D. Schmittgen, and G. A. Calin, “MicroRNAs and cancer: profile, profile, profile,” International Journal of Cancer, vol. 122, no. 5, pp. 969–977, 2008.
[573]  Y. Akao, Y. Nakagawa, Y. Kitade, T. Kinoshita, and T. Naoe, “Downregulation of microRNAs-143 and -145 in B-cell malignancies,” Cancer Science, vol. 98, no. 12, pp. 1914–1920, 2007.
[574]  F. Pichiorri, S. S. Suh, M. Ladetto et al., “MicroRNAs regulate critical genes associated with multiple myeloma pathogenesis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 35, pp. 12885–12890, 2008.
[575]  A. M. Roccaro, A. Sacco, B. Thompson et al., “MicroRNAs 15a and 16 regulate tumor proliferation in multiple myeloma,” Blood, vol. 113, no. 26, pp. 6669–6680, 2009.
[576]  F. Pichiorri, L. De Luca, and R. I. Aqeilan, “MicroRNAs: new players in multiple myeloma,” Frontiers in Genetics, vol. 2, article 22, 2011.
[577]  K. Unno, Y. Zhou, T. Zimmerman, L. C. Platanias, and A. Wickrema, “Identification of a novel microRNA cluster miR-193b-365 in multiple myeloma,” Leukemia and Lymphoma, vol. 50, no. 11, pp. 1865–1871, 2009.
[578]  N. C. Gutiérrez, M. E. Sarasquete, I. Misiewicz-Krzeminska et al., “Deregulation of microRNA expression in the different genetic subtypes of multiple myeloma and correlation with gene expression profiling,” Leukemia, vol. 24, no. 3, pp. 629–637, 2010.
[579]  M. Lionetti, M. Biasiolo, L. Agnelli et al., “Identification of microRNA expression patterns and definition of a microRNA/mRNA regulatory network in distinct molecular groups of multiple myeloma,” Blood, vol. 114, no. 25, pp. e20–e26, 2009.
[580]  C. H. Lawrie, S. Soneji, T. Marafioti et al., “MicroRNA expression distinguishes between germinal center B cell-like and activated B cell-like subtypes of diffuse large B cell lymphoma,” International Journal of Cancer, vol. 121, no. 5, pp. 1156–1161, 2007.
[581]  C. H. Lawrie, J. Chi, S. Taylor et al., “Expression of microRNAs in diffuse large B cell lymphoma is associated with immunophenotype, survival and transformation from follicular lymphoma,” Journal of Cellular and Molecular Medicine, vol. 13, no. 7, pp. 1248–1260, 2009.
[582]  A. Roehle, K. P. Hoefig, D. Repsilber et al., “MicroRNA signatures characterize diffuse large B-cell lymphomas and follicular lymphomas,” British Journal of Haematology, vol. 142, no. 5, pp. 732–744, 2008.
[583]  J. J. Zhao, J. Lin, T. Lwin et al., “MicroRNA expression profile and identification of miR-29 as a prognostic marker and pathogenetic factor by targeting CDK6 in mantle cell lymphoma,” Blood, vol. 115, no. 13, pp. 2630–2639, 2010.
[584]  J. Kluiver, S. Poppema, D. de Jong et al., “BIC and miR-155 are highly expressed in Hodgkin, primary mediastinal and diffuse large B cell lymphomas,” Journal of Pathology, vol. 207, no. 2, pp. 243–249, 2005.
[585]  C. Li, S. W. Kim, D. Rai et al., “Copy number abnormalities, MYC activity, and the genetic fingerprint of normal B cells mechanistically define the microRNA profile of diffuse large B-cell lymphoma,” Blood, vol. 113, no. 26, pp. 6681–6690, 2009.
[586]  M. F. Benner, E. Ballabio, M. S. van Kester et al., “Primary cutaneous anaplastic large cell lymphoma shows a distinct miRNA expression profile and reveals differences from tumor-stage mycosis fungoides,” Experimental Dermatology, vol. 21, pp. 632–634, 2012.
[587]  O. Merkel, F. Hamacher, D. Laimer et al., “Identification of differential and functionally active miRNAs in both anaplastic lymphoma kinase (ALK)+ and ALK- anaplastic large-cell lymphoma,” Proceedings of the National Academy of Sciences of the United States of America, vol. 107, no. 37, pp. 16228–16233, 2010.
[588]  H. Matsuyama, H. I. Suzuki, H. Nishimori et al., “miR-135b mediates NPM-ALK-driven oncogenicity and renders IL-17-producing immunophenotype to anaplastic large cell lymphoma,” Blood, vol. 118, pp. 6881–6892, 2011.
[589]  J. H. Gibcus, L. P. Tan, G. Harms et al., “Hodgkin lymphoma cell lines are characterized by a specific miRNA expression profile,” Neoplasia, vol. 11, no. 2, pp. 167–176, 2009.
[590]  A. Navarro, A. Gaya, A. Martinez et al., “MicroRNA expression profiling in classic Hodgkin lymphoma,” Blood, vol. 111, no. 5, pp. 2825–2832, 2008.
[591]  L. Di Lisio, G. Gómez-López, M. Sánchez-Beato et al., “Mantle cell lymphoma: transcriptional regulation by microRNAs,” Leukemia, vol. 24, no. 7, pp. 1335–1342, 2010.
[592]  R. L. Rossi, G. Rossetti, L. Wenandy et al., “Distinct microRNA signatures in human lymphocyte subsets and enforcement of the naive state in CD4+ T cells by the microRNA miR-125b,” Nature Immunology, vol. 12, pp. 796–803, 2011.
[593]  U. Klein, S. Casola, G. Cattoretti et al., “Transcription factor IRF4 controls plasma cell differentiation and class-switch recombination,” Nature Immunology, vol. 7, no. 7, pp. 773–782, 2006.
[594]  C. Angelin-Duclos, G. Cattoretti, K. I. Lin, and K. Calame, “Commitment of B lymphocytes to a plasma cell fate is associated with Blimp-1 expression in vivo,” Journal of Immunology, vol. 165, no. 10, pp. 5462–5471, 2000.
[595]  A. A. Chaudhuri, A. Y. So, A. Mehta et al., “Oncomir miR-125b regulates hematopoiesis by targeting the gene Lin28A,” Proceedings of the National Academy of Sciences United States of America, vol. 109, pp. 4233–4238, 2012.
[596]  J. H. Klusmann, Z. Li, K. B?hmer et al., “miR-125b-2 is a potential oncomiR on human chromosome 21 in megakaryoblastic leukemia,” Genes and Development, vol. 24, no. 5, pp. 478–490, 2010.
[597]  A. Rodriguez, E. Vigorito, S. Clare et al., “Requirement of bic/microRNA-155 for normal immune function,” Science, vol. 316, no. 5824, pp. 608–611, 2007.
[598]  R. M. O'Connell, D. S. Rao, A. A. Chaudhuri et al., “Sustained expression of microRNA-155 in hematopoietic stem cells causes a myeloproliferative disorder,” Journal of Experimental Medicine, vol. 205, no. 3, pp. 585–594, 2008.
[599]  E. Vigorito, K. L. Perks, C. Abreu-Goodger et al., “microRNA-155 regulates the generation of immunoglobulin class-switched plasma cells,” Immunity, vol. 27, no. 6, pp. 847–859, 2007.
[600]  T. Ruggiero, M. Trabucchi, F. De Santa et al., “LPS induces KH-type splicing regulatory protein-dependent processing of microRNA-155 precursors in macrophages,” FASEB Journal, vol. 23, no. 9, pp. 2898–2908, 2009.
[601]  S. Kohlhaas, O. A. Garden, C. Scudamore, M. Turner, K. Okkenhaug, and E. Vigorito, “Cutting edge: the Foxp3 target miR-155 contributes to the development of regulatory T cells1,” Journal of Immunology, vol. 182, no. 5, pp. 2578–2582, 2009.
[602]  S. Costinean, N. Zanesi, Y. Pekarsky et al., “Pre-B cell proliferation and lymphoblastic leukemia/high-grade lymphoma in Eμ-miR155 transgenic mice,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 18, pp. 7024–7029, 2006.
[603]  E. Tili, J. J. Michaille, D. Wernicke et al., “Mutator activity induced by microRNA-155 (miR-155) links inflammation and cancer,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 12, pp. 4908–4913, 2011.
[604]  T. Wu, A. Wieland, K. Araki et al., “Temporal expression of microRNA cluster miR-17~92 regulates effector and memory CD8+ T-cell differentiation,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, pp. 9965–9970, 2012.
[605]  V. Fulci, T. Colombo, S. Chiaretti et al., “Characterization of B- and T-lineage acute lymphoblastic leukemia by integrated analysis of microRNA and mRNA expression profiles,” Genes Chromosomes and Cancer, vol. 48, no. 12, pp. 1069–1082, 2009.
[606]  U. Ralfkiaer, P. H. Hagedorn, N. Bangsgaard, et al., “Diagnostic microRNA profiling in cutaneous T-cell lymphoma (CTCL),” Blood, vol. 118, pp. 5891–5900, 2011.
[607]  C. Cocco and I. Airoldi, “Cytokines and microRNA in pediatric B-acute lymphoblastic leukemia,” Cytokine and Growth Factor Reviews, vol. 22, no. 3, pp. 149–156, 2011.
[608]  J. Zhang, D. D. Jima, C. Jacobs et al., “Patterns of microRNA expression characterize stages of human B-cell differentiation,” Blood, vol. 113, no. 19, pp. 4586–4594, 2009.
[609]  D. D. Jima, J. Zhang, C. Jacobs et al., “Deep sequencing of the small RNA transcriptome of normal and malignant human B cells identifies hundreds of novel microRNAs,” Blood, vol. 116, no. 23, pp. e118–e127, 2010.
[610]  R. E. Culpin, S. J. Proctor, B. Angus, S. Crosier, J. J. Anderson, and T. Mainou-Fowler, “A 9 series microRNA signature differentiates between germinal centre and activated B-cell-like diffuse large B-cell lymphoma cell lines,” International Journal of Oncology, vol. 37, no. 2, pp. 367–376, 2010.
[611]  Y. Zhoua, L. Chena, B. Barlogiea et al., “High-risk myeloma is associated with global elevation of miRNAs and overexpression of EIF2C2/AGO2,” Proceedings of the National Academy of Sciences of the United States of America, vol. 107, no. 17, pp. 7904–7909, 2010.
[612]  S. L. Corthals, M. Jongen-Lavrencic, Y. de Knegt et al., “Micro-RNA-15a and micro-RNA-16 expression and chromosome 13 deletions in multiple myeloma,” Leukemia Research, vol. 34, no. 5, pp. 677–681, 2010.
[613]  J. Iqbal, Y. Shen, Y. Liu, et al., “Genome-wide miRNA profiling of mantle cell lymphoma reveals a distinct subgroup with poor prognosis,” Blood, vol. 119, pp. 4939–4948, 2012.
[614]  J. D. Schiffman, P. D. Lorimer, V. Rodic, et al., “Genome wide copy number analysis of paediatric Burkitt lymphoma using formalin-fixed tissues reveals a subset with gain of chromosome 13q and corresponding miRNA over expression,” British Journal of Haematology, vol. 155, pp. 477–486, 2011.
[615]  E. Leich, A. Zamo, H. Horn, et al., “MicroRNA profiles of t(14;18)-negative follicular lymphoma support a late germinal center B-cell phenotype,” Blood, vol. 118, pp. 5550–5558, 2011.
[616]  K. J. Mavrakis, A. L. Wolfe, E. Oricchio et al., “Genome-wide RNA-mediated interference screen identifies miR-19 targets in Notch-induced T-cell acute lymphoblastic leukaemia,” Nature Cell Biology, vol. 12, no. 4, pp. 372–379, 2010.
[617]  V. Olive, M. J. Bennett, J. C. Walker et al., “miR-19 is a key oncogenic component of miR-17~92,” Genes and Development, vol. 23, no. 24, pp. 2839–2849, 2009.
[618]  A. Ota, H. Tagawa, S. Karnan et al., “Identification and characterization of a novel gene, C13orf25, as a target for 13q31-q32 amplification in malignant lymphoma,” Cancer Research, vol. 64, no. 9, pp. 3087–3095, 2004.
[619]  Z. Mourelatos, J. Dostie, S. Paushkin et al., “miRNPs: a novel class of ribonucleoproteins containing numerous microRNAs,” Genes and Development, vol. 16, no. 6, pp. 720–728, 2002.
[620]  S. Nagel, L. Venturini, G. K. Przybylski et al., “Activation of miR-17~92 by NK-like homeodomain proteins suppresses apoptosis via reduction of E2F1 in T-cell acute lymphoblastic leukemia,” Leukemia and Lymphoma, vol. 50, no. 1, pp. 101–108, 2009.
[621]  S. T. Dorsam, C. M. Ferrell, G. P. Dorsam et al., “The transcriptome of the leukemogenic homeoprotein HOXA9 in human hematopoietic cells,” Blood, vol. 103, no. 5, pp. 1676–1684, 2004.
[622]  A. A. Ferrando, S. A. Armstrong, D. S. Neuberg et al., “Gene expression signatures in MLL-rearranged T-lineage and B-precursor acute leukemias: dominance of HOX dysregulation,” Blood, vol. 102, no. 1, pp. 262–268, 2003.
[623]  D. Schotte, E. A. M. Lange-Turenhout, D. J. P. M. Stumpel et al., “Expression of miR-196b is not exclusively MLL-driven but is especially linked to activation of HOXA genes in pediatric acute lymphoblastic leukemia,” Haematologica, vol. 95, no. 10, pp. 1675–1682, 2010.
[624]  G. W. Wong, G. C. Knowles, T. W. Mak, A. A. Ferrando, and J. C. Zuniga-Pflucker, “HES1 opposes a PTEN-dependent check on survival, differentiation, and proliferation of TCRbeta-selected mouse thymocytes,” Blood, vol. 120, pp. 1439–1448, 2012.
[625]  U. Koch and F. Radtke, “Notch in T-ALL: new players in a complex disease,” Trends in Immunology, vol. 32, no. 9, pp. 434–442, 2011.
[626]  X. Li, F. Gounari, A. Protopopov, K. Khazaie, and H. Von Boehmer, “Oncogenesis of T-ALL and nonmalignant consequences of overexpressing intracellular NOTCH1,” Journal of Experimental Medicine, vol. 205, no. 12, pp. 2851–2861, 2008.
[627]  V. M. Sharma, K. M. Draheim, and M. A. Kelliher, “The Notch1/c-Myc pathway in T cell leukemia,” Cell Cycle, vol. 6, no. 8, pp. 927–930, 2007.
[628]  Y. Tian, Y. Nan, L. Han et al., “MicroRNA miR-451 downregulates the PI3K/AKT pathway through CAB39 in human glioma,” International Journal of Oncology, vol. 40, pp. 1105–1112, 2012.
[629]  B. Salvatori, I. Iosue, N. Djodji Damas et al., “Critical role of c-Myc in acute myeloid leukemia involving direct regulation of miR-26a and histone methyltransferase EZH2,” Genes & Cancer, vol. 2, pp. 585–592, 2011.
[630]  S. Volinia, G. A. Calin, C. G. Liu et al., “A microRNA expression signature of human solid tumors defines cancer gene targets,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 7, pp. 2257–2261, 2006.
[631]  N. Dahiya, C. A. Sherman-Baust, T. L. Wang et al., “MicroRNA expression and identification of putative miRNA targets in ovarian cancer,” PLoS ONE, vol. 3, no. 6, Article ID e2436, 2008.
[632]  H. He, K. Jazdzewski, W. Li et al., “The role of microRNA genes in papillary thyroid carcinoma,” Proceedings of the National Academy of Sciences of the United States of America, vol. 102, no. 52, pp. 19075–19080, 2005.
[633]  X. Wang, S. Tang, S. Y. Le et al., “Aberrant expression of oncogenic and tumor-suppressive microRNAs in cervical cancer is required for cancer cell growth,” PLoS ONE, vol. 3, no. 7, Article ID e2557, 2008.
[634]  D. T. Starczynowski, F. Kuchenbauer, J. Wegrzyn et al., “MicroRNA-146a disrupts hematopoietic differentiation and survival,” Experimental Hematology, vol. 39, no. 2, pp. 167.e4–178.e4, 2011.
[635]  M. P. Boldin, K. D. Taganov, D. S. Rao et al., “miR-146a is a significant brake on autoimmunity, myeloproliferation, and cancer in mice,” Journal of Experimental Medicine, vol. 208, no. 6, pp. 1189–1201, 2011.
[636]  J. L. Zhao, D. S. Rao, M. P. Boldin, K. D. Taganov, R. M. O'Connell, and D. Baltimore, “NF-κB dysregulation in microRNA-146a-deficient mice drives the development of myeloid malignancies,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 22, pp. 9184–9189, 2011.
[637]  K. D. Taganov, M. P. Boldin, K. J. Chang, and D. Baltimore, “NF-κB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 33, pp. 12481–12486, 2006.
[638]  J. H. Paik, J. Y. Jang, Y. K. Jeon et al., “MicroRNA-146a downregulates NFκB activity via targeting TRAF6 and functions as a tumor suppressor having strong prognostic implications in NK/T cell lymphoma,” Clinical Cancer Research, vol. 17, no. 14, pp. 4761–4771, 2011.
[639]  P. Landgraf, M. Rusu, R. Sheridan et al., “A mammalian microRNA expression atlas based on small RNA library sequencing,” Cell, vol. 129, no. 7, pp. 1401–1414, 2007.
[640]  K. Nie, M. Gomez, P. Landgraf et al., “MicroRNA-mediated down-regulation of PRDM1/Blimp-1 in Hodgkin/Reed—sternberg cells: a potential pathogenetic lesion in Hodgkin lymphomas,” American Journal of Pathology, vol. 173, no. 1, pp. 242–252, 2008.
[641]  L. M. Guo, Y. Pu, Z. Han et al., “MicroRNA-9 inhibits ovarian cancer cell growth through regulation of NF-κB1,” FEBS Journal, vol. 276, no. 19, pp. 5537–5546, 2009.
[642]  E. Ruiz-Ballesteros, M. Mollejo, M. Mateo, P. Algara, P. Martínez, and M. A. Piris, “MicroRNA losses in the frequently deleted region of 7q in SMZL,” Leukemia, vol. 21, no. 12, pp. 2547–2549, 2007.
[643]  G. A. Calin, Y. Pekarsky, and C. M. Croce, “The role of microRNA and other non-coding RNA in the pathogenesis of chronic lymphocytic leukemia,” Best Practice and Research in Clinical Haematology, vol. 20, no. 3, pp. 425–437, 2007.
[644]  U. Klein, M. Lia, M. Crespo et al., “The DLEU2/miR-15a/16-1 cluster controls B cell proliferation and its deletion leads to chronic lymphocytic leukemia,” Cancer Cell, vol. 17, no. 1, pp. 28–40, 2010.
[645]  E. Salerno, B. J. Scaglione, F. D. Coffman et al., “Correcting miR-15a/16 genetic defect in New Zealand Black mouse model of CLL enhances drug sensitivity,” Molecular Cancer Therapeutics, vol. 8, no. 9, pp. 2684–2692, 2009.
[646]  J. Rainer, C. Ploner, S. Jesacher et al., “Glucocorticoid-regulated microRNAs and mirtrons in acute lymphoblastic leukemia,” Leukemia, vol. 23, no. 4, pp. 746–752, 2009.
[647]  M. Inomata, H. Tagawa, Y. M. Guo, Y. Kameoka, N. Takahashi, and K. Sawada, “MicroRNA-17-92 down-regulates expression of distinct targets in different B-cell lymphoma subtypes,” Blood, vol. 113, no. 2, pp. 396–402, 2009.
[648]  L. K. Smith, R. R. Shah, and J. A. Cidlowski, “Glucocorticoids modulate microRNA expression and processing during lymphocyte apoptosis,” The Journal of Biological Chemistry, vol. 285, no. 47, pp. 36698–36708, 2010.
[649]  E. Vreugdenhil, C. S. L. Verissimo, R. Mariman et al., “MicroRNA 18 and 124a down-regulate the glucocorticoid receptor: implications for glucocorticoid responsiveness in the brain,” Endocrinology, vol. 150, no. 5, pp. 2220–2228, 2009.
[650]  G. A. Calin and C. M. Croce, “Chronic lymphocytic leukemia: interplay between noncoding RNAs and protein-coding genes,” Blood, vol. 114, no. 23, pp. 4761–4770, 2009.
[651]  S. Zhu, M. L. Si, H. Wu, and Y. Y. Mo, “MicroRNA-21 targets the tumor suppressor gene tropomyosin 1 (TPM1),” The Journal of Biological Chemistry, vol. 282, no. 19, pp. 14328–14336, 2007.
[652]  M. Lionetti, L. Agnelli, L. Mosca et al., “Integrative high-resolution microarray analysis of human myeloma cell lines reveals deregulated miRNA expression associated with allelic imbalances and gene expression profiles,” Genes Chromosomes and Cancer, vol. 48, no. 6, pp. 521–531, 2009.
[653]  Y. Yamanaka, H. Tagawa, N. Takahashi et al., “Aberrant overexpression of microRNAs activate AKT signaling via down-regulation of tumor suppressors in natural killer-cell lymphoma/leukemia,” Blood, vol. 114, no. 15, pp. 3265–3275, 2009.
[654]  L. B. Frankel, N. R. Christoffersen, A. Jacobsen, M. Lindow, A. Krogh, and A. H. Lund, “Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells,” The Journal of Biological Chemistry, vol. 283, no. 2, pp. 1026–1033, 2008.
[655]  B. W. Han, D. D. Feng, Z. G. Li et al., “A set of miRNAs that involve in the pathways of drug resistance and leukemic stem-cell differentiation is associated with the risk of relapse and glucocorticoid response in childhood ALL,” Human Molecular Genetics, vol. 20, pp. 4903–4915, 2011.
[656]  Z. Wang, W. Wei, and F. H. Sarkar, “miR-23a, a critical regulator of, “migR”ation and metastasis in colorectal cancer,” Cancer Discovery, vol. 2, pp. 489–491, 2012.
[657]  M. Harada, K. Pokrovskaja-Tamm, S. Soderhall, M. Heyman, D. Grander, and M. Corcoran, “Involvement of miR17 pathway in glucocorticoid-induced cell death in pediatric acute lymphoblastic leukemia,” Leukemia & Lymphoma, vol. 53, pp. 2041–2050, 2012.
[658]  D. D. Feng, H. Zhang, P. Zhang et al., “Down-regulated miR-331-5p and miR-27a are associated with chemotherapy resistance and relapse in leukaemia,” Journal of Cellular and Molecular Medicine, vol. 15, pp. 2164–2175, 2011.
[659]  Y. Pekarsky and C. M. Croce, “Is miR-29 an oncogene or tumor suppressor in CLL?” Oncotarget, vol. 1, pp. 224–227, 2010.
[660]  S. Y. Park, J. H. Lee, M. Ha, J. W. Nam, and V. N. Kim, “miR-29 miRNAs activate p53 by targeting p85α and CDC42,” Nature Structural and Molecular Biology, vol. 16, no. 1, pp. 23–29, 2009.
[661]  Y. C. Han, C. Y. Park, G. Bhagat et al., “MicroRNA-29a induces aberrant self-renewal capacity in hematopoietic progenitors, biased myeloid development, and acute myeloid leukemia,” Journal of Experimental Medicine, vol. 207, no. 3, pp. 475–489, 2010.
[662]  J. L. Mott, S. Kurita, S. C. Cazanave, S. F. Bronk, N. W. Werneburg, and M. E. Fernandez-Zapico, “Transcriptional suppression of mir-29b-1/mir-29a promoter by c-Myc, hedgehog, and NF-kappaB,” Journal of Cellular Biochemistry, vol. 110, no. 5, pp. 1155–1164, 2010.
[663]  M. Fabbri, A. Bottoni, M. Shimizu et al., “Association of a microRNA/TP53 feedback circuitry with pathogenesis and outcome of b-cell chronic lymphocytic leukemia,” Journal of the American Medical Association, vol. 305, no. 1, pp. 59–67, 2011.
[664]  G. Zauli, R. Voltan, M. G. Di Iasio et al., “miR-34a induces the downregulation of both E2F1 and B-Myb oncogenes in leukemic cells,” Clinical Cancer Research, vol. 17, no. 9, pp. 2712–2724, 2011.
[665]  H. Hermeking, “The miR-34 family in cancer and apoptosis,” Cell Death and Differentiation, vol. 17, no. 2, pp. 193–199, 2010.
[666]  M. Yamakuchi, M. Ferlito, and C. J. Lowenstein, “miR-34a repression of SIRT1 regulates apoptosis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 36, pp. 13421–13426, 2008.
[667]  S. T. Hashimi, J. A. Fulcher, M. H. Chang, L. Gov, S. Wang, and B. Lee, “MicroRNA profiling identifies miR-34a and miR-21 and their target genes JAG1 and WNT1 in the coordinate regulation of dendritic cell differentiation,” Blood, vol. 114, no. 2, pp. 404–414, 2009.
[668]  I. Ivanovska, A. S. Ball, R. L. Diaz et al., “MicroRNAs in the miR-106b family regulate p21/CDKN1A and promote cell cycle progression,” Molecular and Cellular Biology, vol. 28, no. 7, pp. 2167–2174, 2008.
[669]  F. Petrocca, R. Visone, M. R. Onelli et al., “E2F1-regulated microRNAs impair TGFβ-dependent cell-cycle arrest and apoptosis in gastric cancer,” Cancer Cell, vol. 13, no. 3, pp. 272–286, 2008.
[670]  C. Wang, N. Yao, C. L. Lu, D. Li, and X. Ma, “Mouse microRNA-124 regulates the expression of Hes1 in P19 cells,” Frontiers in Bioscience, vol. 2, pp. 127–132, 2010.
[671]  M. Bousquet, D. Nguyen, C. Chen, L. Schields, and H. Lodish, “miR-125b transforms myeloid cell lines by repressing multiple mRNA targets,” Haematologica, vol. 97, no. 11, pp. 1713–1721, 2012.
[672]  A. Kotani, D. Ha, J. Hsieh et al., “miR-128b is a potent glucocorticoid sensitizer in MLL-AF4 acute lymphocytic leukemia cells and exerts cooperative effects with miR-221,” Blood, vol. 114, no. 19, pp. 4169–4178, 2009.
[673]  A. Kotani, D. Ha, D. Schotte, M. L. Den Boer, S. A. Armstrong, and H. F. Lodish, “A novel mutation in the miR-128b gene reduces miRNA processing and leads to glucocorticoid resistance of MLL-AF4 acute lymphocytic leukemia cells,” Cell Cycle, vol. 9, no. 6, pp. 1037–1042, 2010.
[674]  M. A. Tessel, A. L. Benham, N. L. Krett, S. T. Rosen, and P. H. Gunaratne, “Role for microRNAs in regulating glucocorticoid response and resistance in multiple myeloma,” Hormones and Cancer, vol. 2, no. 3, pp. 182–189, 2011.
[675]  K. W. Lai, K. X. Koh, M. Loh et al., “MicroRNA-130b regulates the tumour suppressor RUNX3 in gastric cancer,” European Journal of Cancer, vol. 46, no. 8, pp. 1456–1463, 2010.
[676]  V. Borgdorff, M. E. Lleonart, C. L. Bishop et al., “Multiple microRNAs rescue from Ras-induced senescence by inhibiting p21 Waf1/Cip1,” Oncogene, vol. 29, no. 15, pp. 2262–2271, 2010.
[677]  A. Navarro, T. Diaz, A. Martinez et al., “Regulation of JAK2 by miR-135a: prognostic impact in classic Hodgkin lymphoma,” Blood, vol. 114, no. 14, pp. 2945–2951, 2009.
[678]  M. Lv, X. Zhang, H. Jia et al., “An oncogenic role of miR-142-3p in human T-cell acute lymphoblastic leukemia (T-ALL) by targeting glucocorticoid receptor-alpha and cAMP/PKA pathways,” Leukemia, vol. 26, pp. 769–777, 2012.
[679]  B. Huang, J. Zhao, Z. Lei et al., “miR-142-3p restricts cAMP production in CD4+CD25- T cells and CD4+CD25+ TREG cells by targeting AC9 mRNA,” EMBO Reports, vol. 10, no. 2, pp. 180–185, 2009.
[680]  X. Huang, M. Yang, and J. Jin, “Triptolide enhances the sensitivity of multiple myeloma cells to dexamethasone via microRNAs,” Leukemia & Lymphoma, vol. 53, pp. 1188–1195, 2012.
[681]  J. L. Robertus, J. Kluiver, C. Weggemans et al., “MiRNA profiling in B non-Hodgkin lymphoma: a MYC-related miRNA profile characterizes Burkitt lymphoma,” British Journal of Haematology, vol. 149, no. 6, pp. 896–899, 2010.
[682]  M. Wang, L. P. Tan, M. K. Dijkstra et al., “miRNA analysis in B-cell chronic lymphocytic leukaemia: proliferation centres characterized by low miR-150 and high BIC/miR-155 expression,” Journal of Pathology, vol. 215, no. 1, pp. 13–20, 2008.
[683]  A. Watanabe, H. Tagawa, J. Yamashita et al., “The role of microRNA-150 as a tumor suppressor in malignant lymphoma,” Leukemia, vol. 25, pp. 1324–1334, 2011.
[684]  S. Belkaya, R. L. Silge, A. R. Hoover et al., “Dynamic modulation of thymic microRNAs in response to stress,” PLoS ONE, vol. 6, Article ID e27580, 2011.
[685]  M. Metzler, M. Wilda, K. Busch, S. Viehmann, and A. Borkhardt, “High expression of precursor MicroRNA-155/BIC RNA in children with Burkitt lymphoma,” Genes Chromosomes and Cancer, vol. 39, no. 2, pp. 167–169, 2004.
[686]  J. Kluiver, E. Haralambieva, D. De Jong et al., “Lack of BIC and microRNA miR-155 expression in primary cases of Burkitt lymphoma,” Genes Chromosomes and Cancer, vol. 45, no. 2, pp. 147–153, 2006.
[687]  K. Vargova, N. Curik, P. Burda et al., “MYB transcriptionally regulates the miR-155 host gene in chronic lymphocytic leukemia,” Blood, vol. 117, no. 14, pp. 3816–3825, 2011.
[688]  S. Costinean, S. K. Sandhu, I. M. Pedersen et al., “Src homology 2 domain-containing inositol-5-phosphatase and CCAAT enhancer-binding protein β are targeted by miR-155 in B cells of Eμ-MiR-155 transgenic mice,” Blood, vol. 114, no. 7, pp. 1374–1382, 2009.
[689]  G. Gatto, A. Rossi, D. Rossi, S. Kroening, S. Bonatti, and M. Mallardo, “Epstein-Barr virus latent membrane protein 1 trans-activates miR-155 transcription through the NF-κB pathway,” Nucleic Acids Research, vol. 36, no. 20, pp. 6608–6619, 2008.
[690]  Y. Zheng, S. Xiong, P. Jiang et al., “Glucocorticoids inhibit lipopolysaccharide-mediated inflammatory response by downregulating microRNA-155: a novel anti-inflammation mechanism,” Free Radical Biology and Medicine, vol. 52, pp. 1307–1317, 2012.
[691]  P. S. Eis, W. Tam, L. Sun et al., “Accumulation of miR-155 and BIC RNA in human B cell lymphomas,” Proceedings of the National Academy of Sciences of the United States of America, vol. 102, no. 10, pp. 3627–3632, 2005.
[692]  Z. Li, H. Huang, Y. Li, et al., “Up-regulation of a HOXA-PBX3 homeobox-gene signature following down-regulation of miR-181 is associated with adverse prognosis in patients with cytogenetically abnormal AML,” Blood, vol. 119, pp. 2314–2324, 2012.
[693]  A. J. Alencar, R. Malumbres, G. A. Kozloski et al., “MicroRNAs are independent predictors of outcome in diffuse large B-cell lymphoma patients treated with R-CHOP,” Clinical Cancer Research, vol. 17, no. 12, pp. 4125–4135, 2011.
[694]  X. Wang, E. Gocek, C. G. Liu, and G. P. Studzinski, “MicroRNAs181 regulate the expression of p27Kip1 in human myeloid leukemia cells induced to differentiate by 1,25-dihydroxyvitamin D3,” Cell Cycle, vol. 8, no. 5, pp. 736–741, 2009.
[695]  C. Le Sage, R. Nagel, D. A. Egan et al., “Regulation of the p27Kip1 tumor suppressor by miR-221 and miR-222 promotes cancer cell proliferation,” EMBO Journal, vol. 26, no. 15, pp. 3699–3708, 2007.
[696]  S. Galardi, N. Mercatelli, E. Giorda et al., “miR-221 and miR-222 expression affects the proliferation potential of human prostate carcinoma cell lines by targeting p27Kip1,” The Journal of Biological Chemistry, vol. 282, no. 32, pp. 23716–23724, 2007.
[697]  N. Felli, L. Fontana, E. Pelosi et al., “MicroRNAs 221 and 222 inhibit normal erythropoiesis and erythroleukemic cell growth via kit receptor down-modulation,” Proceedings of the National Academy of Sciences of the United States of America, vol. 102, no. 50, pp. 18081–18086, 2005.
[698]  R. Medina, S. K. Zaidi, C. G. Liu et al., “MicroRNAs 221 and 222 bypass quiescence and compromise cell survival,” Cancer Research, vol. 68, no. 8, pp. 2773–2780, 2008.
[699]  T. E. Miller, K. Ghoshal, B. Ramaswamy et al., “MicroRNA-221/222 confers tamoxifen resistance in breast cancer by targeting p27Kip1,” The Journal of Biological Chemistry, vol. 283, no. 44, pp. 29897–29903, 2008.
[700]  B. Stamatopoulos, N. Meuleman, B. Haibe-Kains et al., “microRNA-29c and microRNA-223 down-regulation has in vivo significance in chronic lymphocytic leukemia and improves disease risk stratification,” Blood, vol. 113, no. 21, pp. 5237–5245, 2009.
[701]  J. B. Johnnidis, M. H. Harris, R. T. Wheeler et al., “Regulation of progenitor cell proliferation and granulocyte function by microRNA-223,” Nature, vol. 451, no. 7182, pp. 1125–1129, 2008.
[702]  J. A. Pulikkan, V. Dengler, P. S. Peramangalam et al., “Cell-cycle regulator E2F1 and microRNA-223 comprise an autoregulatory negative feedback loop in acute myeloid leukemia,” Blood, vol. 115, no. 9, pp. 1768–1778, 2010.
[703]  F. Fazi, S. Racanicchi, G. Zardo et al., “Epigenetic silencing of the myelopoiesis regulator microRNA-223 by the AML1/ETO oncoprotein,” Cancer Cell, vol. 12, no. 5, pp. 457–466, 2007.
[704]  C. Y. Jia, H. H. Li, X. C. Zhu et al., “MiR-223 suppresses cell proliferation by targeting IGF-1R,” PLoS ONE, vol. 6, Article ID e27008, 2011.
[705]  Y. Xu, T. Sengupta, L. Kukreja, and A. C. Minella, “MicroRNA-223 regulates cyclin E activity by modulating expression of F-box and WD-40 domain protein 7,” The Journal of Biological Chemistry, vol. 285, no. 45, pp. 34439–34446, 2010.
[706]  J. Y. Yuan, F. Wang, J. Yu, G. H. Yang, X. L. Liu, and J. W. Zhang, “MicroRNA-223 reversibly regulates erythroid and megakaryocytic differentiation of K562 cells,” Journal of Cellular and Molecular Medicine, vol. 13, no. 11-12, pp. 4551–4559, 2009.
[707]  J. Laine, G. Künstle, T. Obata, M. Sha, and M. Noguchi, “The protooncogene TCL1 is an Akt kinase coactivator,” Molecular Cell, vol. 6, no. 2, pp. 395–407, 2000.
[708]  R. Munker and G. A. Calin, “MicroRNA profiling in cancer,” Clinical Science, vol. 121, no. 4, pp. 141–158, 2011.
[709]  M. T. Smonskey, A. W. Block, G. Deeb et al., “Monoallelic and biallelic deletions of 13q14. 3 in chronic lymphocytic leukemia: FISH vs miRNA RT-qPCR detection,” American Journal of Clinical Pathology, vol. 137, pp. 641–646, 2012.
[710]  D. Sampath, C. Liu, K. Vasan et al., “Histone deacetylases mediate the silencing of miR-15a, miR-16, and miR-29b in chronic lymphocytic leukemia,” Blood, vol. 119, pp. 1162–1172, 2012.
[711]  G. A. Calin, C. G. Liu, C. Sevignani et al., “MicroRNA profiling reveals distinct signatures in B cell chronic lymphocytic leukemias,” Proceedings of the National Academy of Sciences of the United States of America, vol. 101, no. 32, pp. 11755–11760, 2004.
[712]  N. Bandi, S. Zbinden, M. Gugger et al., “miR-15a and miR-16 are implicated in cell cycle regulation in a Rb-dependent manner and are frequently deleted or down-regulated in non-small cell lung cancer,” Cancer Research, vol. 69, no. 13, pp. 5553–5559, 2009.
[713]  E. S. Raveche, E. Salerno, B. J. Scaglione et al., “Abnormal microRNA-16 locus with synteny to human 13q14 linked to CLL in NZB mice,” Blood, vol. 109, no. 12, pp. 5079–5086, 2007.
[714]  S. Rossi, M. Shimizu, E. Barbarotto et al., “MicroRNA fingerprinting of CLL patients with chromosome 17p deletion identify a miR-21 score that stratifies early survival,” Blood, vol. 116, no. 6, pp. 945–952, 2010.
[715]  P. G. Longo, L. Laurenti, S. Gobessi, S. Sica, G. Leone, and D. G. Efremov, “The Akt/Mcl-1 pathway plays a prominent role in mediating anti-apoptotic signals downstream of the B-cell receptor in chronic lymphocytic leukemia B cells,” Blood, vol. 111, no. 2, pp. 846–855, 2008.
[716]  U. Santanam, N. Zanesi, A. Efanov et al., “Chronic lymphocytic leukemia modeled in mouse by targeted miR-29 expression,” Proceedings of the National Academy of Sciences of the United States of America, vol. 107, no. 27, pp. 12210–12215, 2010.
[717]  M. Fabbri, R. Garzon, A. Cimmino et al., “MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting DNA methyltransferases 3A and 3B,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 40, pp. 15805–15810, 2007.
[718]  M. Frenquelli, M. Muzio, C. Scielzo et al., “MicroRNA and proliferation control in chronic lymphocytic leukemia: functional relationship between miR-221/222 cluster and p27,” Blood, vol. 115, no. 19, pp. 3949–3959, 2010.
[719]  F. Fornari, L. Gramantieri, M. Ferracin et al., “MiR-221 controls CDKN1C/p57 and CDKN1B/p27 expression in human hepatocellular carcinoma,” Oncogene, vol. 27, no. 43, pp. 5651–5661, 2008.
[720]  G. Di Leva, P. Gasparini, C. Piovan et al., “MicroRNA cluster 221-222 and estrogen receptor α interactions in breast cancer,” Journal of the National Cancer Institute, vol. 102, no. 10, pp. 706–721, 2010.
[721]  N. Hamada, Y. Fujita, T. Kojima et al., “MicroRNA expression profiling of NGF-treated PC12 cells revealed a critical role for miR-221 in neuronal differentiation,” Neurochemistry International, vol. 60, pp. 743–750, 2012.
[722]  M. Garofalo, G. Romano, G. Di Leva, et al., “EGFR and MET receptor tyrosine kinase-altered microRNA expression induces tumorigenesis and gefitinib resistance in lung cancers,” Nature Medicine, vol. 18, pp. 74–82, 2012.
[723]  L. Gramantieri, F. Fornari, M. Ferracin et al., “MicroRNA-221 targets Bmf in hepatocellular carcinoma and correlates with tumor multifocality,” Clinical Cancer Research, vol. 15, no. 16, pp. 5073–5081, 2009.
[724]  A. Navarro, S. Beà, V. Fernández et al., “MicroRNA expression, chromosomal alterations, and immunoglobulin variable heavy chain hypermutations in mantle cell lymphomas,” Cancer Research, vol. 69, no. 17, pp. 7071–7078, 2009.
[725]  E. Rao, C. Jiang, M. Ji et al., “The miRNA-17 approximately 92 cluster mediates chemoresistance and enhances tumor growth in mantle cell lymphoma via PI3K/AKT pathway activation,” Leukemia, vol. 26, pp. 1064–1072, 2012.
[726]  Y. Li, X. Zhu, J. Gu et al., “Anti-miR-21 oligonucleotide sensitizes leukemic K562 cells to arsenic trioxide by inducing apoptosis,” Cancer Science, vol. 101, no. 4, pp. 948–954, 2010.
[727]  P. P. Medina, M. Nolde, and F. J. Slack, “OncomiR addiction in an in vivo model of microRNA-21-induced pre-B-cell lymphoma,” Nature, vol. 467, no. 7311, pp. 86–90, 2010.
[728]  M. E. Hatley, D. M. Patrick, M. R. Garcia et al., “Modulation of K-Ras-dependent lung tumorigenesis by MicroRNA-21,” Cancer Cell, vol. 18, no. 3, pp. 282–293, 2010.
[729]  X. Ma, M. Kumar, S. N. Choudhury et al., “Loss of the miR-21 allele elevates the expression of its target genes and reduces tumorigenesis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 25, pp. 10144–10149, 2011.
[730]  D. Sayed, S. Rane, J. Lypowy et al., “MicroRNA-21 targets Sprouty2 and promotes cellular outgrowths,” Molecular Biology of the Cell, vol. 19, no. 8, pp. 3272–3282, 2008.
[731]  I. A. Asangani, S. A. K. Rasheed, D. A. Nikolova et al., “MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer,” Oncogene, vol. 27, no. 15, pp. 2128–2136, 2008.
[732]  N. Valeri, P. Gasparini, C. Braconi et al., “MicroRNA-21 induces resistance to 5-fluorouracil by down-regulating human DNA MutS homolog 2 (hMSH2),” Proceedings of the National Academy of Sciences of the United States of America, vol. 107, no. 49, pp. 21098–21103, 2010.
[733]  T. Papagiannakopoulos, A. Shapiro, and K. S. Kosik, “MicroRNA-21 targets a network of key tumor-suppressive pathways in glioblastoma cells,” Cancer Research, vol. 68, no. 19, pp. 8164–8172, 2008.
[734]  H. S. Yang, A. P. Jansen, R. Nair et al., “A novel transformation suppressor, Pdcd4, inhibits AP-1 transactivation but not NF-κB or ODC transactivation,” Oncogene, vol. 20, no. 6, pp. 669–676, 2001.
[735]  R. G?ke, P. Barth, A. Schmidt, B. Samans, and B. Lankat-Buttgereit, “Programmed cell death protein 4 suppresses CDK1/cdc2 via induction of p21Waf1/Cip1,” American Journal of Physiology, vol. 287, no. 6, pp. C1541–C1546, 2004.
[736]  H. S. Yang, J. L. Knies, C. Stark, and N. H. Colburn, “Pdcd4 suppresses tumor phenotype in JB6 cells by inhibiting AP-1 transactivation,” Oncogene, vol. 22, no. 24, pp. 3712–3720, 2003.
[737]  Y. Chen, T. Kn?sel, G. Kristiansen et al., “Loss of PDCD4 expression in human lung cancer correlates with tumour progression and prognosis,” Journal of Pathology, vol. 200, no. 5, pp. 640–646, 2003.
[738]  F. Gao, P. Zhang, C. Zhou et al., “Frequent loss of PDCD4 expression in human glioma: possible role in the tumorigenesis of glioma,” Oncology Reports, vol. 17, no. 1, pp. 123–128, 2007.
[739]  H. Zhang, I. Ozaki, T. Mizuta et al., “Involvement of programmed cell death 4 in transforming growth factor-β1-induced apoptosis in human hepatocellular carcinoma,” Oncogene, vol. 25, no. 45, pp. 6101–6112, 2006.
[740]  E. Gottwein, N. Mukherjee, C. Sachse et al., “A viral microRNA functions as an orthologue of cellular miR-155,” Nature, vol. 450, no. 7172, pp. 1096–1099, 2007.
[741]  L. N. Dagan, X. Jiang, S. Bhatt, E. Cubedo, K. Rajewsky, and I. S. Lossos, “miR-155 regulates HGAL expression and increases lymphoma cell motility,” Blood, vol. 119, pp. 513–520, 2012.
[742]  D. Rai, S. W. Kim, M. R. McKeller, P. L. M. Dahia, and R. C. T. Aguiar, “Targeting of SMAD5 links microRNA-155 to the TGF-β pathway and lymphomagenesis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 107, no. 7, pp. 3111–3116, 2010.
[743]  X. Jiang, X. Lu, G. McNamara et al., “HGAL, a germinal center specific protein, decreases lymphoma cell motility by modulation of the RhoA signaling pathway,” Blood, vol. 116, no. 24, pp. 5217–5227, 2010.
[744]  X. Lu, K. Kazmierczak, X. Jiang et al., “Germinal center-specific protein human germinal center associated lymphoma directly interacts with both myosin and actin and increases the binding of myosin to actin,” FEBS Journal, vol. 278, no. 11, pp. 1922–1931, 2011.
[745]  J. Massagué, J. Seoane, and D. Wotton, “Smad transcription factors,” Genes and Development, vol. 19, no. 23, pp. 2783–2810, 2005.
[746]  R. Fonseca, E. Blood, M. Rue et al., “Clinical and biologic implications of recurrent genomic aberrations in myeloma,” Blood, vol. 101, no. 11, pp. 4569–4575, 2003.
[747]  W. M. Kuehl and P. L. Bergsagel, “Multiple myeloma: evolving genetic events and host interactions,” Nature Reviews Cancer, vol. 2, no. 3, pp. 175–187, 2002.
[748]  R. I. Aqeilan, G. A. Calin, and C. M. Croce, “MiR-15a and miR-16-1 in cancer: discovery, function and future perspectives,” Cell Death and Differentiation, vol. 17, no. 2, pp. 215–220, 2010.
[749]  B. M. Weiss, J. Abadie, P. Verma, R. S. Howard, and W. M. Kuehl, “A monoclonal gammopathy precedes multiple myeloma in most patients,” Blood, vol. 113, no. 22, pp. 5418–5422, 2009.
[750]  M. Chesi, D. F. Robbiani, M. Sebag et al., “AID-dependent activation of a MYC transgene induces multiple myeloma in a conditional mouse model of post-germinal center malignancies,” Cancer Cell, vol. 13, no. 2, pp. 167–180, 2008.
[751]  M. Chesi and P. L. Bergsagel, “Many multiple myelomas: making more of the molecular mayhem,” Hematology/the Education Program of the American Society of Hematology, vol. 2011, pp. 344–353, 2011.
[752]  M. E. Sarasquete, N. C. Gutiérrez, I. Misiewicz-Krzeminska et al., “Upregulation of dicer is more frequent in monoclonal gammopathies of undetermined significance than in multiple myeloma patients and is associated with longer survival in symptomatic myeloma patients,” Haematologica, vol. 96, no. 3, pp. 468–471, 2011.
[753]  J. Liu, M. A. Carmell, F. V. Rivas et al., “Argonaute2 is the catalytic engine of mammalian RNAi,” Science, vol. 305, no. 5689, pp. 1437–1441, 2004.
[754]  S. Diederichs and D. A. Haber, “Dual role for argonautes in microRNA processing and posttranscriptional regulation of microRNA expression,” Cell, vol. 131, no. 6, pp. 1097–1108, 2007.
[755]  B. Z. Katz, “Adhesion molecules—the lifelines of multiple myeloma cells,” Seminars in Cancer Biology, vol. 20, no. 3, pp. 186–195, 2010.
[756]  M. A. Frassanito, A. Cusmai, G. Iodice, and F. Dammacco, “Autocrine interleukin-6 production and highly malignant multiple myeloma: relation with resistance to drug-induced apoptosis,” Blood, vol. 97, no. 2, pp. 483–489, 2001.
[757]  T. Hideshima, P. Richardson, D. Chauhan et al., “The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma cells,” Cancer Research, vol. 61, no. 7, pp. 3071–3076, 2001.
[758]  A. C. Bharti, N. Donato, and B. B. Aggarwal, “Curcumin (diferuloylmethane) inhibits constitutive and IL-6-inducible STAT3 phosphorylation in human multiple myeloma cells,” Journal of Immunology, vol. 171, no. 7, pp. 3863–3871, 2003.
[759]  A. C. Bharti, N. Donato, S. Singh, and B. B. Aggarwal, “Curcumin (diferuloylmethane) down-regulates the constitutive activation of nuclear factor-κB and IκBα kinase in human multiple myeloma cells, leading to suppression of proliferation and induction of apoptosis,” Blood, vol. 101, no. 3, pp. 1053–1062, 2003.
[760]  R. Feinman, J. Koury, M. Thames, B. Barlogie, J. Epstein, and D. S. Siegel, “Role of NF-κB in the rescue of multiple myeloma cells from glucocorticoid-induced apoptosis by bcl-2,” Blood, vol. 93, no. 9, pp. 3044–3052, 1999.
[761]  D. L?ffler, K. Brocke-Heidrich, G. Pfeifer et al., “Interleukin-6-dependent survival of multiple myeloma cells involves the Stat3-mediated induction of microRNA-21 through a highly conserved enhancer,” Blood, vol. 110, no. 4, pp. 1330–1333, 2007.
[762]  X. Wang, C. Li, S. Ju, Y. Wang, H. Wang, and R. Zhong, “Myeloma cell adhesion to bone marrow stromal cells confers drug resistance by microRNA-21 up-regulation,” Leukemia & Lymphoma, vol. 52, pp. 1991–1998, 2011.
[763]  L. K. Linares, R. Kiernan, R. Triboulet et al., “Intrinsic ubiquitination activity of PCAF controls the stability of the oncoprotein Hdm2,” Nature Cell Biology, vol. 9, no. 3, pp. 331–338, 2007.
[764]  Y. H. Chen, D. Lavelle, J. DeSimone, S. Uddin, L. C. Platanias, and M. Hankewych, “Growth inhibition of a human myeloma cell line by all-trans retinoic acid is not mediated through downregulation of interleukin-6 receptors but through upregulation of p21(WAF1),” Blood, vol. 94, no. 1, pp. 251–259, 1999.
[765]  D. Lavelle, Y. H. Chen, M. Hankewych, and J. Desimone, “Histone deacetylase inhibitors increase p21WAF1 and induce apoptosis of human myeloma cell lines independent of decreased IL-6 receptor expression,” American Journal of Hematology, vol. 68, no. 3, pp. 170–178, 2001.
[766]  M. Lerner, M. Harada, J. Lovén et al., “DLEU2, frequently deleted in malignancy, functions as a critical host gene of the cell cycle inhibitory microRNAs miR-15a and miR-16-1,” Experimental Cell Research, vol. 315, no. 17, pp. 2941–2952, 2009.
[767]  L. Xia, D. Zhang, R. Du et al., “miR-15b and miR-16 modulate multidrug resistance by targeting BCL2 in human gastric cancer cells,” International Journal of Cancer, vol. 123, no. 2, pp. 372–379, 2008.
[768]  G. A. Calin, A. Cimmino, M. Fabbri et al., “MiR-15a and miR-16-1 cluster functions in human leukemia,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 13, pp. 5166–5171, 2008.
[769]  R. Ria, A. Vacca, F. Russo et al., “A VEGF-dependent autocrine loop mediates proliferation and capillarogenesis in bone marrow endothelial cells of patients with multiple myeloma,” Thrombosis and Haemostasis, vol. 92, no. 6, pp. 1438–1445, 2004.
[770]  A. Vacca, R. Ria, D. Ribatti et al., “A paracrine loop in the vascular endothelial growth factor pathway triggers tumor angiogenesis and growth in multiple myeloma,” Haematologica, vol. 88, no. 2, pp. 176–185, 2003.
[771]  J. D. Pearson, J. K. Lee, J. T. Bacani, R. Lai, and R. J. Ingham, “NPM-ALK: the prototypic member of a family of oncogenic fusion tyrosine kinases,” Journal of Signal Transduction, vol. 2012, Article ID 123253, 14 pages, 2012.
[772]  E. Campo, S. H. Swerdlow, N. L. Harris, S. Pileri, H. Stein, and E. S. Jaffe, “The 2008 WHO classification of lymphoid neoplasms and beyond: evolving concepts and practical applications,” Blood, vol. 117, no. 19, pp. 5019–5032, 2011.
[773]  A. Carbone, A. Gloghini, A. Aiello, A. Testi, and A. Cabras, “B-cell lymphomas with features intermediate between distinct pathologic entities. From pathogenesis to pathology,” Human Pathology, vol. 41, no. 5, pp. 621–631, 2010.
[774]  K. J. Savage, “Therapies for peripheral T-cell lymphomas,” Hematology American Society of Hematology Education Program, vol. 2011, pp. 515–524, 2011.
[775]  E. A. Raetz, S. L. Perkins, M. A. Carlson, K. P. Schooler, W. L. Carroll, and D. M. Virshup, “The nucleophosmin-anaplastic lymphoma kinase fusion protein induces c-Myc expression in pediatric anaplastic large cell lymphomas,” American Journal of Pathology, vol. 161, no. 3, pp. 875–883, 2002.
[776]  F. Z. Wang, F. Weber, C. Croce, C. G. Liu, X. Liao, and P. E. Pellett, “Human cytomegalovirus infection alters the expression of cellular MicroRNA species that affect its replication,” Journal of Virology, vol. 82, no. 18, pp. 9065–9074, 2008.
[777]  H. Su, J. R. Yang, T. Xu et al., “MicroRNA-101, down-regulated in hepatocellular carcinoma, promotes apoptosis and suppresses tumorigenicity,” Cancer Research, vol. 69, no. 3, pp. 1135–1142, 2009.
[778]  J. M. Friedman, G. Liang, C. C. Liu et al., “The putative tumor suppressor microRNA-101 modulates the cancer epigenome by repressing the polycomb group protein EZH2,” Cancer Research, vol. 69, no. 6, pp. 2623–2629, 2009.
[779]  S. Varambally, Q. Cao, R. S. Mani et al., “Genomic loss of microRNA-101 leads to overexpression of histone methyltransferase EZH2 in cancer,” Science, vol. 322, no. 5908, pp. 1695–1699, 2008.
[780]  S. Montes-Moreno, N. Martinez, B. Sanchez-Espiridión et al., “MiRNA expression in diffuse large B-cell lymphoma treated with chemoimmunotherapy,” Blood, vol. 118, no. 4, pp. 1034–1040, 2011.
[781]  G. Lenz, G. W. Wright, N. C. T. Emre et al., “Molecular subtypes of diffuse large B-cell lymphoma arise by distinct genetic pathways,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 36, pp. 13520–13525, 2008.
[782]  G. Lenz, G. Wright, S. S. Dave et al., “Stromal gene signatures in large-B-cell lymphomas,” The New England Journal of Medicine, vol. 359, no. 22, pp. 2313–2323, 2008.
[783]  J. P. Jais, C. Haioun, T. J. Molina et al., “The expression of 16 genes related to the cell of origin and immune response predicts survival in elderly patients with diffuse large B-cell lymphoma treated with CHOP and rituximab,” Leukemia, vol. 22, no. 10, pp. 1917–1924, 2008.
[784]  A. H. Banham, N. Beasley, E. Campo et al., “The FOXP1 winged helix transcription factor is a novel candidate tumor suppressor gene on chromosome 3p,” Cancer Research, vol. 61, no. 24, pp. 8820–8829, 2001.
[785]  A. L. Shaffer, A. Rosenwald, and L. M. Staudt, “Lymphoid malignancies: the dark side of B-cell differentiation,” Nature Reviews Immunology, vol. 2, no. 12, pp. 920–932, 2002.
[786]  I. Wlodarska, E. Veyt, P. De Paepe et al., “FOXP1, a gene highly expressed in a subset of diffuse large B-cell lymphoma, is recurrently targeted by genomic aberrations,” Leukemia, vol. 19, no. 8, pp. 1299–1305, 2005.
[787]  B. Streubel, U. Vinatzer, A. Lamprecht, M. Raderer, and A. Chott, “T(3;14)(p14.1;q32) involving IGH and FOXP1 is a novel recurrent chromosomal aberration in MALT lymphoma,” Leukemia, vol. 19, no. 4, pp. 652–658, 2005.
[788]  A. H. Banham, J. M. Connors, P. J. Brown et al., “Expression of the FOXP1 transcription factor is strongly associated with inferior survival in patients with diffuse large B-cell lymphoma,” Clinical Cancer Research, vol. 11, no. 3, pp. 1065–1072, 2005.
[789]  S. L. Barrans, J. A. L. Fenton, A. Banham, R. G. Owen, and A. S. Jack, “Strong expression of FOXP1 identifies a distinct subset of diffuse large B-cell lymphoma (DLBCL) patients with poor outcome,” Blood, vol. 104, no. 9, pp. 2933–2935, 2004.
[790]  J. Mandelbaum, G. Bhagat, H. Tang et al., “BLIMP1 is a tumor suppressor gene frequently disrupted in activated B cell-like diffuse large B cell lymphoma,” Cancer Cell, vol. 18, no. 6, pp. 568–579, 2010.
[791]  D. P. Calado, B. Zhang, L. Srinivasan et al., “Constitutive canonical NF-κB activation cooperates with disruption of BLIMP1 in the pathogenesis of activated B cell-like diffuse large cell lymphoma,” Cancer Cell, vol. 18, no. 6, pp. 580–589, 2010.
[792]  G. Martins and K. Calame, “Regulation and functions of Blimp-1 in T and B lymphocytes,” Annual Review of Immunology, vol. 26, pp. 133–169, 2008.
[793]  T. C. Kuo, A. L. Shaffer, J. Haddad, S. C. Yong, L. M. Staudt, and K. Calame, “Repression of BCL-6 is required for the formation of human memory B cells in vitro,” Journal of Experimental Medicine, vol. 204, no. 4, pp. 819–830, 2007.
[794]  C. Tunyaplin, A. L. Shaffer, C. D. Angelin-Duclos, X. Yu, L. M. Staudt, and K. L. Calame, “Direct repression of prdm1 by Bcl-6 inhibits plasmacytic differentiation,” Journal of Immunology, vol. 173, no. 2, pp. 1158–1165, 2004.
[795]  L. Pasqualucci, M. Compagno, J. Houldsworth et al., “Inactivation of the PRDM1/BLIMP1 gene in diffuse large B cell lymphoma,” Journal of Experimental Medicine, vol. 203, no. 2, pp. 311–317, 2006.
[796]  R. Küppers, “The biology of Hodgkin's lymphoma,” Nature Reviews Cancer, vol. 9, no. 1, pp. 15–27, 2009.
[797]  J. M. Gozgit, G. Bebernitz, P. Patil et al., “Effects of the JAK2 inhibitor, AZ960, on Pim/BAD/BCL-xL survival signaling in the human JAK2 V617F cell line SET-2,” The Journal of Biological Chemistry, vol. 283, no. 47, pp. 32334–32343, 2008.
[798]  C. M. Croce and G. A. Calin, “miRNAs, cancer, and stem cell division,” Cell, vol. 122, no. 1, pp. 6–7, 2005.
[799]  S. D. Linnstaedt, E. Gottwein, R. L. Skalsky, M. A. Luftig, and B. R. Cullen, “Virally induced cellular microRNA miR-155 plays a key role in B-cell immortalization by Epstein-Barr virus,” Journal of Virology, vol. 84, no. 22, pp. 11670–11678, 2010.
[800]  E. R. de Kloet, C. P. Fitzsimons, N. A. Datson, O. C. Meijer, and E. Vreugdenhil, “Glucocorticoid signaling and stress-related limbic susceptibility pathway: about receptors, transcription machinery and microRNA,” Brain Research, vol. 1293, pp. 129–141, 2009.
[801]  S. Uchida, A. Nishida, K. Hara et al., “Characterization of the vulnerability to repeated stress in Fischer 344 rats: possible involvement of microRNA-mediated down-regulation of the glucocorticoid receptor,” European Journal of Neuroscience, vol. 27, no. 9, pp. 2250–2261, 2008.
[802]  Y. Hayashita, H. Osada, Y. Tatematsu et al., “A polycistronic MicroRNA cluster, miR-17~92, is overexpressed in human lung cancers and enhances cell proliferation,” Cancer Research, vol. 65, no. 21, pp. 9628–9632, 2005.
[803]  H. Matsubara, T. Takeuchi, E. Nishikawa et al., “Apoptosis induction by antisense oligonucleotides against miR-17-5p and miR-20a in lung cancers overexpressing miR-17~92,” Oncogene, vol. 26, no. 41, pp. 6099–6105, 2007.
[804]  P. Sommer, P. Le Rouzic, H. Gillingham et al., “Glucocorticoid receptor overexpression exerts an anti-survival effect on human small cell lung cancer cells,” Oncogene, vol. 26, no. 50, pp. 7111–7121, 2007.
[805]  C. Ledderose, P. Mohnle, E. Limbeck et al., “Corticosteroid resistance in sepsis is influenced by microRNA-124-induced downregulation of glucocorticoid receptor-alpha,” Critical Care Medicine, vol. 40, no. 10, pp. 2745–2753, 2012.
[806]  B. Malzkorn, M. Wolter, F. Liesenberg et al., “Identification and functional characterization of microRNAs involved in the malignant progression of gliomas,” Brain Pathology, vol. 20, no. 3, pp. 539–550, 2010.
[807]  Z. Ji, F. C. Mei, A. L. Miller, E. B. Thompson, and X. Cheng, “Protein kinase A (PKA) isoform RIIβ mediates the synergistic killing effect of cAMP and glucocorticoid in acute lymphoblastic leukemia cells,” The Journal of Biological Chemistry, vol. 283, no. 32, pp. 21920–21925, 2008.
[808]  S. W. Kim, D. Rai, and R. C. Aguiar, “Gene set enrichment analysis unveils the mechanism for the phosphodiesterase 4B control of glucocorticoid response in B-cell lymphoma,” Clinical Cancer Research, vol. 17, pp. 6723–6732, 2011.
[809]  M. Yang, J. Huang, H. Z. Pan, and J. Jin, “Triptolide overcomes dexamethasone resistance and enhanced PS-341-induced apoptosis via PI3k/Akt/NF-κB pathways in human multiple myeloma cells,” International Journal of Molecular Medicine, vol. 22, no. 4, pp. 489–496, 2008.
[810]  W. Zhu, X. Shan, T. Wang, Y. Shu, and P. Liu, “MiR-181b modulates multidrug resistance by targeting BCL2 in human cancer cell lines,” International Journal of Cancer, vol. 127, no. 11, pp. 2520–2529, 2010.
[811]  D. L. Allen and A. S. Loh, “Posttranscriptional mechanisms involving microRNA-27a and b contribute to fast-specific and glucocorticoid-mediated myostatin expression in skeletal muscle,” American Journal of Physiology, vol. 300, no. 1, pp. C124–C137, 2011.
[812]  A. Rodriguez, S. Griffiths-Jones, J. L. Ashurst, and A. Bradley, “Identification of mammalian microRNA host genes and transcription units,” Genome Research, vol. 14, no. 10, pp. 1902–1910, 2004.
[813]  P. S. Linsley, J. Schelter, J. Burchard et al., “Transcripts targeted by the microRNA-16 family cooperatively regulate cell cycle progression,” Molecular and Cellular Biology, vol. 27, no. 6, pp. 2240–2252, 2007.
[814]  F. Fazi, A. Rosa, A. Fatica et al., “A minicircuitry comprised of microRNA-223 and transcription factors NFI-A and C/EBPα regulates human granulopoiesis,” Cell, vol. 123, no. 5, pp. 819–831, 2005.
[815]  W. Sun, W. Shen, S. Yang, F. Hu, H. Li, and T. H. Zhu, “MiR-223 and miR-142 attenuate hematopoietic cell proliferation, and miR-223 positively regulates miR-142 through LMO2 isoforms and CEBP-β,” Cell Research, vol. 20, no. 10, pp. 1158–1169, 2010.
[816]  J. Li, Y. Guo, X. Liang et al., “MicroRNA-223 functions as an oncogene in human gastric cancer by targeting FBXW7/hCdc4,” Journal of Cancer Research and Clinical Oncology, vol. 138, pp. 763–774, 2012.
[817]  G. D. Sempowski, L. P. Hale, J. S. Sundy et al., “Leukemia inhibitory factor, oncostatin M, IL-6, and stem cell factor mRNA expression in human thymus increases with age and is associated with thymic atrophy,” Journal of Immunology, vol. 164, no. 4, pp. 2180–2187, 2000.
[818]  G. D. Sempowski, M. E. Rhein, R. M. Scearce, and B. F. Haynes, “Leukemia inhibitory factor is a mediator of Escherichia coli lipopolysaccharide-induced acute thymic atrophy,” European Journal of Immunology, vol. 32, pp. 3066–3070, 2002.
[819]  D. P. Gearing, N. M. Gough, J. A. King et al., “Molecular cloning and expression of cDNA encoding a murine myeloid leukaemia inhibitory factor (LIF),” EMBO Journal, vol. 6, no. 13, pp. 3995–4002, 1987.
[820]  E. Tili, J. J. Michaille, A. Cimino et al., “Modulation of miR-155 and miR-125b levels following lipopolysaccharide/TNF- α stimulation and their possible roles in regulating the response to endotoxin shock,” Journal of Immunology, vol. 179, no. 8, pp. 5082–5089, 2007.
[821]  H. Kawashima, T. Numakawa, E. Kumamaru et al., “Glucocorticoid attenuates brain-derived neurotrophic factor-dependent upregulation of glutamate receptors via the suppression of microRNA-132 expression,” Neuroscience, vol. 165, no. 4, pp. 1301–1311, 2010.
[822]  N. Vo, M. E. Klein, O. Varlamova et al., “A cAMP-response element binding protein-induced microRNA regulates neuronal morphogenesis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 102, no. 45, pp. 16426–16431, 2005.
[823]  A. C. Rodrigues, X. Li, L. Radecki et al., “MicroRNA expression is differentially altered by xenobiotic drugs in different human cell lines,” Biopharmaceutics and Drug Disposition, vol. 32, no. 6, pp. 355–367, 2011.
[824]  A. K. Biswas and D. G. Johnson, “Transcriptional and nontranscriptional functions of E2F1 in response to DNA damage,” Cancer Research, vol. 72, pp. 13–17, 2012.
[825]  R. C. Y. Hui, A. R. Gomes, D. Constantinidou et al., “The forkhead transcription factor FOXO3a increases phosphoinositide-3 kinase/Akt activity in drug-resistant leukemic cells through induction of PIK3CA expression,” Molecular and Cellular Biology, vol. 28, no. 19, pp. 5886–5898, 2008.
[826]  K. Miyamoto, K. Y. Araki, K. Naka et al., “Foxo3a is essential for maintenance of the hematopoietic stem cell pool,” Cell Stem Cell, vol. 1, no. 1, pp. 101–112, 2007.
[827]  Z. Tothova, R. Kollipara, B. J. Huntly et al., “FoxOs are critical mediators of hematopoietic stem cell resistance tophysiologic oxidative stress,” Cell, vol. 128, no. 2, pp. 325–339, 2007.
[828]  K. Naka, T. Hoshii, T. Muraguchi et al., “TGF-β-FOXO signalling maintains leukaemia-initiating cells in chronic myeloid leukaemia,” Nature, vol. 463, no. 7281, pp. 676–680, 2010.
[829]  S. S. Myatt and E. W. F. Lam, “The emerging roles of forkhead box (Fox) proteins in cancer,” Nature Reviews Cancer, vol. 7, no. 11, pp. 847–859, 2007.
[830]  S. Chintharlapalli, S. Papineni, M. Abdelrahim et al., “Oncogenic microRNA-27a is a target for anticancer agent methyl 2-cyano-3,11-dioxo-18β-olean-1,12-dien-30-oate in colon cancer cells,” International Journal of Cancer, vol. 125, no. 8, pp. 1965–1974, 2009.
[831]  F. M. Raaphorst, “Self-renewal of hematopoietic and leukemic stem cells: a central role for the Polycomb-group gene Bmi-1,” Trends in Immunology, vol. 24, no. 10, pp. 522–524, 2003.
[832]  I. K. Park, D. Qian, M. Kiel et al., “Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells,” Nature, vol. 423, no. 6937, pp. 302–305, 2003.
[833]  J. Lessard and G. Sauvageau, “Bmi-1 determines the proliferative capacity of normal and leukaemic stem cells,” Nature, vol. 423, no. 6937, pp. 255–260, 2003.
[834]  C. H. Pui, J. M. Chessells, B. Camitta et al., “Clinical heterogeneity in childhood acute lymphoblastic leukemia with 11q23 rearrangements,” Leukemia, vol. 17, no. 4, pp. 700–706, 2003.
[835]  R. Pieters, M. Schrappe, P. De Lorenzo et al., “A treatment protocol for infants younger than 1 year with acute lymphoblastic leukaemia (Interfant-99): an observational study and a multicentre randomised trial,” The Lancet, vol. 370, no. 9583, pp. 240–250, 2007.
[836]  G. Meister, M. Landthaler, Y. Dorsett, and T. Tuschl, “Sequence-specific inhibition of microRNA-and siRNA-induced RNA silencing,” RNA, vol. 10, no. 3, pp. 544–550, 2004.
[837]  L. Dou, D. Zheng, J. Li et al., “Methylation-mediated repression of microRNA-143 enhances MLL-AF4 oncogene expression,” Oncogene, vol. 31, pp. 507–517, 2011.
[838]  I. A. Babar, C. J. Cheng, C. J. Booth et al., “Nanoparticle-based therapy in an in vivo microRNA-155 (miR-155)-dependent mouse model of lymphoma,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, pp. E1695–E1704, 2012.
[839]  C. Burnsides, J. Corry, J. Alexander et al., “Ex vivo stimulation of whole blood as a means to determine glucocorticoid sensitivity,” Journal of Inflammation Research, vol. 5, pp. 89–97, 2012.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413