全部 标题 作者
关键词 摘要

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

查看量下载量

相关文章

更多...

The Role of BCL2 Family of Apoptosis Regulator Proteins in Acute and Chronic Leukemias

DOI: 10.1155/2012/524308

Full-Text   Cite this paper   Add to My Lib

Abstract:

The disturbance of apoptosis molecular signaling pathways is involved in carcinogenesis. BCL2 family of proteins is the hallmark of apoptosis regulation. In the last decade, new members of BCL2 gene family were discovered and cloned and were found to be differentially expressed in many types of cancer. BCL2 protein family, through its role in regulation of apoptotic pathways, is possibly related to cancer pathophysiology and resistance to conventional chemotherapy. It is well known that leukemias are haematopoietic malignancies characterized by biological diversity, varied cytogenetics, different immunophenotype profiles, and diverse outcome. Current research focuses on the prognostic impact and specific role of these proteins in the pathogenesis of leukemias. The understanding of the molecular pathways that participate in the biology of leukemias may lead to the design of new therapies which may improve patients' survival. In the present paper, we describe current knowledge on the role of BCL2 apoptosis regulator proteins in acute and chronic leukemias. 1. Introduction Apoptosis, or programmed cell death, is a cell-suicide program, distinct from necrosis, which is activated in physiological processes such as tissue development and differentiation as well as in pathophysiological conditions. The term is used to describe the situation in which a cell actively pursues a course toward death upon receiving certain stimuli. The morphological changes of apoptosis found in most cell types include nuclear pyknosis, DNA fragmentation and chromatin condensation, cytoskeleton destruction, membrane blebbing, and eventually the formation of membrane apoptotic bodies, which are phagocytosed by macrophages and other cells without promoting inflammatory response [1]. The mechanism of apoptosis is evolutionarily conserved and is executed by a family of proteins, called caspases. Caspases are cysteine proteases that are cleaved after an Asp residue in their substrates. They are synthesized as latent zymogenes and activated by proteolytic cleavage; their activation is mainly regulated by the BCL2 family proteins [2–4]. BCL2 gene (otherwise B-cell lymphoma 2 gene, bcl-2) was first discovered in follicular B-cell lymphoma as a gene which is linked to the immunoglobulin heavy chain locus at the breakpoints of t(14;18) translocation [5]; the result of this translocation is the enhanced BCL2 protein transcription. In normal cells this gene is located on chromosome segment 18q21.3. BCL2 protein was found to inhibit cell death. This discovery was a revolution in the way of

References

[1]  J. Yuan, M. Lipinski, and A. Degterev, “Diversity in the mechanisms of neuronal cell death,” Neuron, vol. 40, no. 2, pp. 401–413, 2003.
[2]  K. C. Zimmermann and D. R. Green, “How cells die: apoptosis pathways,” Journal of Allergy and Clinical Immunology, vol. 108, supplement 4, pp. S99–S103, 2001.
[3]  S. W. G. Tait and D. R. Green, “Mitochondria and cell death: outer membrane permeabilization and beyond,” Nature Reviews Molecular Cell Biology, vol. 11, no. 9, pp. 621–632, 2010.
[4]  D. R. Green and J. C. Reed, “Mitochondria and apoptosis,” Science, vol. 281, no. 5381, pp. 1309–1312, 1998.
[5]  Y. Tsujimoto, J. Gorham, and J. Cossman, “The t(14;18) chromosome translocations involved in B-cell neoplasms result from mistakes in VDJ joining,” Science, vol. 229, no. 4720, pp. 1390–1393, 1985.
[6]  S. Cory and J. M. Adams, “The BCL2 family: regulators of the cellular life-or-death switch,” Nature Reviews Cancer, vol. 2, no. 9, pp. 647–656, 2002.
[7]  B. M. Pickering, S. De Mel, M. Lee, et al., “Pharmacological inhibitors of NF-kappaB accelerate apoptosis in chronic lymphocytic leukaemia cells,” Oncogene, vol. 26, no. 8, pp. 1166–1177, 2007.
[8]  H. Walczak and P. H. Krammer, “The CD95 (APO-1/Fas) and the TRAIL (APO-2L) apoptosis systems,” Experimental Cell Research, vol. 256, no. 1, pp. 58–66, 2000.
[9]  H. Thomadaki and A. Scorilas, “BCL2 family of apoptosis-related genes: functions and clinical implications in cancer,” Critical Reviews in Clinical Laboratory Sciences, vol. 43, no. 1, pp. 1–67, 2006.
[10]  A. Scorilas, L. Kyriakopoulou, G. M. Yousef, L. K. Ashworth, A. Kwamie, and E. P. Diamandis, “Molecular cloning, physical mapping, and expression analysis of a novel gene, BCL2L12, encoding a proline-rich protein with a highly conserved BH2 domain of the Bcl-2 family,” Genomics, vol. 72, no. 2, pp. 217–221, 2001.
[11]  M. Talieri, E. P. Diamandis, N. Katsaros, D. Gourgiotis, and A. Scorilas, “Expression of BCL2L12, a new member of apoptosis-related genes, in breast tumors,” Thrombosis and Haemostasis, vol. 89, no. 6, pp. 1081–1088, 2003.
[12]  K. Mathioudaki, A. Scorilas, A. Papadokostopoulou, et al., “Expression analysis of BCL2L12, a new member of apoptosis-related genes, in colon cancer,” Biological Chemistry, vol. 385, no. 9, pp. 779–783, 2004.
[13]  H. Thomadaki, M. Talieri, and A. Scorilas, “Prognostic value of the apoptosis related genes BCL2 and BCL2L12 in breast cancer,” Cancer Letters, vol. 247, no. 1-2, pp. 48–55, 2007.
[14]  H. Thomadaki, M. Talieri, and A. Scorilas, “Treatment of MCF-7 cells with taxol and etoposide induces distinct alterations in the expression of apoptosis-related genes BCL2, BCL2L12, BAX, CASPASE-9 and FAS,” Biological Chemistry, vol. 387, no. 8, pp. 1081–1086, 2006.
[15]  A. Sklavounou, E. Chrysomali, A. Scorilas, and A. Karameris, “TNF-alpha expression and apoptosis-regulating proteins in oral lichen planus: a comparative immunohistochemical evaluation,” Journal of Oral Pathology and Medicine, vol. 29, no. 8, pp. 370–375, 2000.
[16]  I. Kalomenidis, D. Orphanidou, G. Papamichalis, et al., “Combined expression of p53, Bcl-2, and p21WAF-1 proteins in lung cancer and premalignant lesions: association with clinical characteristics,” Lung, vol. 179, no. 5, pp. 265–278, 2001.
[17]  J. C. Reed, “Proapoptotic multidomain Bcl-2/Bax-family proteins: mechanisms, physiological roles, and therapeutic opportunities,” Cell Death and Differentiation, vol. 13, no. 8, pp. 1378–1386, 2006.
[18]  J. M. Adams and S. Cory, “The Bcl-2 protein family: arbiters of cell survival,” Science, vol. 281, no. 5381, pp. 1322–1326, 1998.
[19]  R. J. Youle and A. Strasser, “The BCL-2 protein family: opposing activities that mediate cell death,” Nature Reviews Molecular Cell Biology, vol. 9, no. 1, pp. 47–59, 2008.
[20]  J. C. Reed, “Apoptosis mechanisms: implications for cancer drug discovery,” Oncology, vol. 18, supplement 10, no. 13, pp. 11–20, 2004.
[21]  S. J. Korsmeyer, “Bcl-2: an antidote to programmed cell death,” Cancer Surveys, vol. 15, pp. 105–118, 1992.
[22]  L. Khemtemourian, M. A. Sani, K. Bathany, G. Grobner, and E. J. Dufourc, “Synthesis and secondary structure in membranes of the Bcl-2 anti-apoptotic domain BH4,” Journal of Peptide Science, vol. 12, no. 1, pp. 58–64, 2006.
[23]  M. Simonen, H. Keller, and J. Heim, “The BH3 domain of Bax is sufficient for interaction of Bax with itself and with other family members and it is required for induction of apoptosis,” European Journal of Biochemistry, vol. 249, no. 1, pp. 85–91, 1997.
[24]  D. S. Bellows, B. N. Chau, L. Percy, Y. Lazebnik, W. H. Burns, and J. M. Hardwick, “Antiapoptotic herpesvirus Bcl-2 homologs escape caspase-mediated conversion to proapoptotic proteins,” Journal of Virology, vol. 74, no. 11, pp. 5024–5031, 2000.
[25]  H. F. Yang-Yen, “Mcl-1: a highly regulated cell death and survival controller,” Journal of Biomedical Science, vol. 13, no. 2, pp. 201–204, 2006.
[26]  D. P. Stewart, B. Koss, M. Bathina, R. M. Perciavalle, K. Bisanz, and J. T. Opferman, “Ubiquitin-independent degradation of antiapoptotic MCL-1,” Molecular and Cellular Biology, vol. 30, no. 12, pp. 3099–3110, 2010.
[27]  C. Akgul, “Mcl-1 is a potential therapeutic target in multiple types of cancer,” Cellular and Molecular Life Sciences, vol. 66, no. 8, pp. 1326–1336, 2009.
[28]  B. Leibowitz and J. Yu, “Mitochondrial signaling in cell death via the Bcl-2 family,” Cancer Biology and Therapy, vol. 9, no. 6, pp. 417–422, 2010.
[29]  A. M. Petros, E. T. Olejniczak, and S. W. Fesik, “Structural biology of the Bcl-2 family of proteins,” Biochimica et Biophysica Acta, vol. 1644, no. 2-3, pp. 83–94, 2004.
[30]  L. D. Walensky, “BCL-2 in the crosshairs: tipping the balance of life and death,” Cell Death and Differentiation, vol. 13, no. 8, pp. 1339–1350, 2006.
[31]  M. Klee and F. X. Pimentel-Muinos, “Bcl-X(L) specifically activates Bak to induce swelling and restructuring of the endoplasmic reticulum,” Journal of Cell Biology, vol. 168, no. 5, pp. 723–734, 2005.
[32]  J. E. Chipuk, T. Kuwana, L. Bouchier-Hayes, et al., “Direct activation of Bax by p53 mediates mitochondrial membrane permeabilization and apoptosis,” Science, vol. 303, no. 5660, pp. 1010–1014, 2004.
[33]  D. E. Wood, A. Thomas, L. A. Devi, et al., “Bax cleavage is mediated by calpain during drug-induced apoptosis,” Oncogene, vol. 17, no. 9, pp. 1069–1078, 1998.
[34]  J. Zha, H. Harada, E. Yang, J. Jockel, and S. J. Korsmeyer, “Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14-3-3 not BCL-X(L),” Cell, vol. 87, no. 4, pp. 619–628, 1996.
[35]  Y. Zhu, G. Y. Yang, B. Ahlemeyer, et al., “Transforming growth factor-beta 1 increases bad phosphorylation and protects neurons against damage,” Journal of Neuroscience, vol. 22, no. 10, pp. 3898–3909, 2002.
[36]  S. Zinkel, A. Gross, and E. Yang, “BCL2 family in DNA damage and cell cycle control,” Cell Death and Differentiation, vol. 13, no. 8, pp. 1351–1359, 2006.
[37]  S. S. Zinkel, C. C. Ong, D. O. Ferguson, et al., “Proapoptotic BID is required for myeloid homeostasis and tumor suppression,” Genes and Development, vol. 17, no. 2, pp. 229–239, 2003.
[38]  K. Wang, X. M. Yin, D. T. Chao, C. L. Milliman, and S. J. Korsmeyer, “BID: a novel BH3 domain-only death agonist,” Genes and Development, vol. 10, no. 22, pp. 2859–2869, 1996.
[39]  X. Luo, I. Budihardjo, H. Zou, C. Slaughter, and X. Wang, “Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors,” Cell, vol. 94, no. 4, pp. 481–490, 1998.
[40]  A. B. Werner, S. W. G. Tait, E. De Vries, E. Eldering, and J. Borst, “Requirement for aspartate-cleaved bid in apoptosis signaling by DNA-damaging anti-cancer regimens,” Journal of Biological Chemistry, vol. 279, no. 27, pp. 28771–28780, 2004.
[41]  T. Kuwana, L. Bouchier-Hayes, J. E. Chipuk, et al., “BH3 domains of BH3-only proteins differentially regulate Bax-mediated mitochondrial membrane permeabilization both directly and indirectly,” Molecular Cell, vol. 17, no. 4, pp. 525–535, 2005.
[42]  I. Wojcik, M. Szybka, E. Golanska, et al., “Abnormalities of the P53, MDM2, BCL2 and BAX genes in acute leukemias,” Neoplasma, vol. 52, no. 4, pp. 318–324, 2005.
[43]  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.
[44]  D. Campana, E. Coustan-Smith, A. Manabe, et al., “Prolonged survival of B-lineage acute lymphoblastic leukemia cells is accompanied by overexpression of bcl-2 protein,” Blood, vol. 81, no. 4, pp. 1025–1031, 1993.
[45]  S. Aref, O. Salama, Y. Al-Tonbary, and A. Mansour, “Assessment of bcl-2 expression ass modulator of Fas mediated apoptosis in acute leukemia,” Hematology, vol. 9, no. 2, pp. 113–121, 2004.
[46]  G. R. Sahu and B. R. Das, “Prognostic significance of p53 and Bcl-2 in acute lymphoblastic leukemia,” Oncology Reports, vol. 9, no. 6, pp. 1391–1398, 2002.
[47]  L. Campos, O. Sabido, C. Sebban, et al., “Expression of BCL-2 proto-oncogene in adult acute lymphoblastic leukemia,” Leukemia, vol. 10, no. 3, pp. 434–438, 1996.
[48]  L. A. Hogarth and A. G. Hall, “Increased BAX expression is associated with an increased risk of relapse in childhood acute lymphocytic leukemia,” Blood, vol. 93, no. 8, pp. 2671–2678, 1999.
[49]  E. Coustan-Smith, A. Kitanaka, C. H. Pui, et al., “Clinical relevance of BCL-2 overexpression in childhood acute lymphoblastic leukemia,” Blood, vol. 87, no. 3, pp. 1140–1146, 1996.
[50]  M. I. Del Principe, G. Del Poeta, L. Maurillo, et al., “P-glycoprotein and BCL-2 levels predict outcome in adult acute lymphoblastic leukaemia,” British Journal of Haematology, vol. 121, no. 5, pp. 730–738, 2003.
[51]  H. Feng, D. L. Stachura, R. M. White, et al., “T-lymphoblastic lymphoma cells express high levels of BCL2, S1P1, and ICAM1, leading to a blockade of tumor cell intravasation,” Cancer Cell, vol. 18, no. 4, pp. 353–366, 2010.
[52]  A. Prokop, T. Wieder, I. Sturm, et al., “Relapse in childhood acute lymphoblastic leukemia is associated with a decrease of the Bax/Bcl-2 ratio and loss of spontaneous caspase-3 processing in vivo,” Leukemia, vol. 14, no. 9, pp. 1606–1613, 2000.
[53]  P. J. Swanson, S. L. Kuslak, W. Fang, et al., “Fatal acute lymphoblastic leukemia in mice transgenic for B cell-restricted bcl-xL and c-myc,” Journal of Immunology, vol. 172, no. 11, pp. 6684–6691, 2004.
[54]  R. Addeo, M. Caraglia, A. Baldi, et al., “Prognostic role of bcl-xL and p53 in childhood acute lymphoblastic leukemia (ALL),” Cancer Biology and Therapy, vol. 4, no. 1, pp. 32–38, 2005.
[55]  J. F. Mata, V. S. Silveira, E. C. Mateo, et al., “Low mRNA expression of the apoptosis-related genes CASP3, CASP8, and FAS is associated with low induction treatment response in childhood acute lymphoblastic leukemia (ALL),” Pediatric Blood and Cancer, vol. 55, no. 1, pp. 100–107, 2010.
[56]  E. Laane, E. Tani, E. Bjorklund, et al., “Flow cytometric immunophenotyping including Bcl-2 detection on fine needle aspirates in the diagnosis of reactive lymphadenopathy and non-Hodgkin's lymphoma,” Cytometry Part B, vol. 64, no. 1, pp. 34–42, 2005.
[57]  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.
[58]  A. Holleman, M. L. Den Boer, R. X. De Menezes, et al., “The expression of 70 apoptosis genes in relation to lineage, genetic subtype, cellular drug resistance, and outcome in childhood acute lymphoblastic leukemia,” Blood, vol. 107, no. 2, pp. 769–776, 2006.
[59]  M. Andreeff, S. Jiang, X. Zhang, et al., “Expression of Bcl-2-related genes in normal and AML progenitors: changes induced by chemotherapy and retinoic acid,” Leukemia, vol. 13, no. 11, pp. 1881–1892, 1999.
[60]  D. Milojkovic, S. Devereux, N. B. Westwood, G. J. Mufti, N. S. B. Thomas, and A. G. S. Buggins, “Antiapoptotic microenvironment of acute myeloid leukemia,” Journal of Immunology, vol. 173, no. 11, pp. 6745–6752, 2004.
[61]  A. G. S. Buggins, D. Milojkovic, M. J. Arno, et al., “Microenvironment produced by acute myeloid leukemia cells prevents T cell activation and proliferation by inhibition of NF-kappaB, c-Myc, and pRb pathways,” Journal of Immunology, vol. 167, no. 10, pp. 6021–6030, 2001.
[62]  A. Venditti, G. Del Poeta, L. Maurillo, et al., “Combined analysis of bcl-2 and MDR1 proteins in 256 cases of acute myeloid leukemia,” Haematologica, vol. 89, no. 8, pp. 934–939, 2004.
[63]  C. Wuchter, L. Karawajew, V. Ruppert, et al., “Clinical significance of CD95, Bcl-2 and Bax expression and CD95 function in adult de novo acute myeloid leukemia in context of P-glycoprotein function, maturation stage, and cytogenetics,” Leukemia, vol. 13, no. 12, pp. 1943–1953, 1999.
[64]  S. M. Kornblau, P. F. Thall, Z. Estrov, et al., “The prognostic impact of BCL2 protein expression in acute myelogenous leukemia varies with cytogenetics,” Clinical Cancer Research, vol. 5, no. 7, pp. 1758–1766, 1999.
[65]  L. Klampfer, J. Zhang, A. O. Zelenetz, H. Uchida, and S. D. Nimer, “The AML1/ETO fusion protein activates transcription of BCL-2,” Proceedings of the National Academy of Sciences of the United States of America, vol. 93, no. 24, pp. 14059–14064, 1996.
[66]  M. Shikami, H. Miwa, K. Nishii, et al., “Low BCL-2 expression in acute leukemia with t(8;21) chromosomal abnormality,” Leukemia, vol. 13, no. 3, pp. 358–368, 1999.
[67]  T. Karakas, U. Maurer, E. Weidmann, C. C. Miething, D. Hoelzer, and L. Bergmann, “High expression of bcl-2 mRNA as a determinant of poor prognosis in acute myeloid leukemia,” Annals of Oncology, vol. 9, no. 2, pp. 159–165, 1998.
[68]  T. Kohler, C. Schill, M. W. Deininger, et al., “High bad and bax mRNA expression correlate with negative outcome in acute myeloid leukemia (AML),” Leukemia, vol. 16, no. 1, pp. 22–29, 2002.
[69]  Y. L. Ong, M. F. McMullin, K. E. M. Bailie, T. R. J. Lappin, F. G. C. Jones, and A. E. Irvine, “High bax expression is a good prognostic indicator in acute myeloid leukaemia,” British Journal of Haematology, vol. 111, no. 1, pp. 182–189, 2000.
[70]  H. Yamaguchi, K. Inokuchi, and K. Dan, “The study for loss of bcl-xs expression as a prognostic factor in acute myeloid leukemia,” Leukemia Research, vol. 26, no. 12, pp. 1119–1123, 2002.
[71]  S. M. Kornblau, H. T. Vu, P. Ruvolo, et al., “BAX and PKCalpha modulate the prognostic impact of BCL2 expression in acute myelogenous leukemia,” Clinical Cancer Research, vol. 6, no. 4, pp. 1401–1409, 2000.
[72]  C. Rochlitz, A. Lohri, M. Bacchi, et al., “Axl expression is associated with adverse prognosis and with expression of Bcl-2 and CD34 in de novo acute myeloid leukemia (AML): results from a multicenter trial of the Swiss Group for Clinical Cancer Research (SAKK),” Leukemia, vol. 13, no. 9, pp. 1352–1358, 1999.
[73]  D. Bradbury, S. Rogers, R. Kozlowski, G. Bowen, I. A. G. Reilly, and N. H. Russell, “Interleukin-1 is one factor which regulates autocrine production of GM-CSF by the blast cells of acute myeloblastic leukaemia,” British Journal of Haematology, vol. 76, no. 4, pp. 488–493, 1990.
[74]  S. Spiegel and S. Milstien, “Sphingosine-1-phosphate: an enigmatic signalling lipid,” Nature Reviews Molecular Cell Biology, vol. 4, no. 5, pp. 397–407, 2003.
[75]  J. Turzanski, M. Grundy, N. H. Russell, and M. Pallis, “Interleukin-1beta maintains an apoptosis-resistant phenotype in the blast cells of acute myeloid leukaemia via multiple pathways,” Leukemia, vol. 18, no. 10, pp. 1662–1670, 2004.
[76]  H. Asao and X. Y. Fu, “Interferon-gamma has dual potentials in inhibiting or promoting cell proliferation,” Journal of Biological Chemistry, vol. 275, no. 2, pp. 867–874, 2000.
[77]  S. Faderl, D. Harris, Q. Van, H. M. Kantarjian, M. Talpaz, and Z. Estrov, “Granulocyte-macrophage colony-stimulating factor (GM-CSF) induces antiapoptotic and proapoptotic signals in acute myeloid leukemia,” Blood, vol. 102, no. 2, pp. 630–637, 2003.
[78]  M. Milella, Z. Estrov, S. M. Kornblau, et al., “Synergistic induction of apoptosis by simultaneous disruption of the Bcl-2 and MEK/MAPK pathways in acute myelogenous leukemia,” Blood, vol. 99, no. 9, pp. 3461–3464, 2002.
[79]  I. N. Karnolsky, “Cytogenetic abnormalities in chronic lymphocytic leukemia,” Folia Medica, vol. 42, no. 3, pp. 5–10, 2000.
[80]  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.
[81]  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.
[82]  H. Nuckel, U. H. Frey, M. Bau, et al., “Association of a novel regulatory polymorphism in the BCL2 gene promoter with disease progression and survival in chronic lymphocytic leukemia,” Blood, vol. 109, no. 1, pp. 290–297, 2007.
[83]  A. Majid, O. Tsoulakis, R. Walewska, et al., “BCL2 expression in chronic lymphocytic leukemia: lack of association with the BCL2 promoter single nucleotide polymorphism,” Blood, vol. 111, no. 2, pp. 874–877, 2008.
[84]  C. Pepper, T. Hoy, and P. Bentley, “Elevated Bcl-2/Bax are a consistent feature of apoptosis resistance in B-cell chronic lymphocytic leukaemia and are correlated with in vivo chemoresistance,” Leukemia and Lymphoma, vol. 28, no. 3-4, pp. 355–361, 1998.
[85]  S. Skogsberg, G. Tobin, A. Krober, et al., “The G(-248)A polymorphism in the promoter region of the Bax gene does not correlate with prognostic markers or overall survival in chronic lymphocytic leukemia,” Leukemia, vol. 20, no. 1, pp. 77–81, 2006.
[86]  O. Moshynska, K. Sankaran, P. Pahwa, and A. Saxena, “Prognostic significance of a short sequence insertion in the MCL-1 promoter in chronic lymphocytic leukemia,” Journal of the National Cancer Institute, vol. 96, no. 9, pp. 673–682, 2004.
[87]  G. Packham and F. K. Stevenson, “Bodyguards and assassins: Bcl-2 family proteins and apoptosis control in chronic lymphocytic leukaemia,” Immunology, vol. 114, no. 4, pp. 441–449, 2005.
[88]  S. Kitada, J. Andersen, S. Akar, et al., “Expression of apoptosis-regulating proteins in chronic lymphocytic leukemia: correlations with in vitro and in vivo chemoresponses,” Blood, vol. 91, no. 9, pp. 3379–3389, 1998.
[89]  L. E. Robertson, W. Plunkett, K. McConnell, M. J. Keating, and T. J. McDonnell, “Bcl-2 expression in chronic lymphocytic leukemia and its correlation with the induction of apoptosis and clinical outcome,” Leukemia, vol. 10, no. 3, pp. 456–459, 1996.
[90]  S. Molica, A. Dattilo, C. Giulino, D. Levato, and L. Levato, “Increased bcl-2/bax ratio in B-cell chronic lymphocytic leukemia is associated with a progressive pattern of disease,” Haematologica, vol. 83, no. 12, pp. 1122–1124, 1998.
[91]  M. Aguilar-Santelises, M. E. Rottenberg, N. Lewin, H. Mellstedt, and M. Jondal, “bcl-2, Bax and p53 expression in B-CLL in relation to in vitro survival and clinical progression,” International Journal of Cancer, vol. 69, no. 2, pp. 114–119, 1996.
[92]  S. Molica, A. Mannella, G. Crispino, A. Dattilo, and D. Levato, “Comparative flow cytometric evaluation of bcl-2 oncoprotein in CD5+ and CD5- B-cell lymphoid chronic leukemias,” Haematologica, vol. 82, no. 5, pp. 555–559, 1997.
[93]  S. Faderl, M. J. Keating, K. A. Do, et al., “Expression profile of 11 proteins and their prognostic significance in patients with chronic lymphocytic leukemia (CLL),” Leukemia, vol. 16, no. 6, pp. 1045–1052, 2002.
[94]  J. R. Faria, M. Yamamoto, R. M. D. Faria, J. Kerbauy, and J. S. R. Oliveira, “Fludarabine induces apoptosis in chronic lymphocytic leukemia—the role of P53, Bcl-2, Bax, Mcl-1 and Bag-1 proteins,” Brazilian Journal of Medical and Biological Research, vol. 39, no. 3, pp. 327–333, 2006.
[95]  A. A. Morales, A. Olsson, F. Celsing, A. Osterborg, M. Jondal, and L. M. Osorio, “High expression of bfl-1 contributes to the apoptosis resistant phenotype in B-cell chronic lymphocytic leukemia,” International Journal of Cancer, vol. 113, no. 5, pp. 730–737, 2005.
[96]  A. Olsson, M. Norberg, A. Okvist, et al., “Upregulation of bfl-1 is a potential mechanism of chemoresistance in B-cell chronic lymphocytic leukaemia,” British Journal of Cancer, vol. 97, no. 6, pp. 769–777, 2007.
[97]  A. Saxena, S. Viswanathan, O. Moshynska, P. Tandon, K. Sankaran, and D. P. Sheridan, “Mcl-1 and Bcl-2/Bax ratio are associated with treatment response but not with Rai stage in B-cell chronic lymphocytic leukemia,” American Journal of Hematology, vol. 75, no. 1, pp. 22–33, 2004.
[98]  M. Klobusicka, J. Kusenda, and O. Babusikova, “Immunocytochemical detection of bcl-2 and p53 proteins in B-chronic lymphocytic leukemia patients,” Neoplasma, vol. 49, no. 6, pp. 387–393, 2002.
[99]  A. Lazaridou, C. Miraxtsi, J. Korantzis, N. Eleftheriadis, and J. I. Christakis, “Simultaneous detection of BCL-2 protein, trisomy 12, retinoblastoma and p53 monoallelic gene deletions in B-cell chronic lymphocytic leukemia by fluorescence in situ hybridization (FISH): relation to disease status,” Leukemia and Lymphoma, vol. 36, no. 5-6, pp. 503–512, 2000.
[100]  S. Duensing, A. Nasarek, J. Grosse, A. Ganser, and J. Atzpodien, “Elevated plasma levels of interleukin-1 receptor antagonist are associated with decreased cellular BCL-2 oncoprotein expression in B-chronic lymphocytic leukemia,” Cancer Biotherapy and Radiopharmaceuticals, vol. 13, no. 5, pp. 383–385, 1998.
[101]  R. R. Furman, Z. Asgary, J. O. Mascarenhas, H. C. Liou, and E. J. Schattner, “Modulation of NF-kappa B activity and apoptosis in chronic lymphocytic leukemia B cells,” Journal of Immunology, vol. 164, no. 4, pp. 2200–2206, 2000.
[102]  S. Cuni, P. Perez-Aciego, G. Perez-Chacon, et al., “A sustained activation of PI3K/NF-kappaB pathway is critical for the survival of chronic lymphocytic leukemia B cells,” Leukemia, vol. 18, no. 8, pp. 1391–1400, 2004.
[103]  J. Zhuang, S. F. Hawkins, M. A. Glenn, et al., “Akt is activated in chronic lymphocytic leukemia cells and delivers a pro-survival signal: the therapeutic potential of Akt inhibition,” Haematologica, vol. 95, no. 1, pp. 110–118, 2010.
[104]  S. Jaiswal, D. Traver, T. Miyamoto, K. Akashi, E. Lagasse, and I. L. Weissman, “Expression of BCR/ABL and BCL-2 in myeloid progenitors leads to myeloid leukemias,” Proceedings of the National Academy of Sciences of the United States of America, vol. 100, no. 17, pp. 10002–10007, 2003.
[105]  H. Handa, U. P. Hegde, V. M. Kotelnikov, et al., “Bcl-2 and c-myc expression, cell cycle kinetics and apoptosis during the progression of chronic myelogenous leukemia from diagnosis to blastic phase,” Leukemia Research, vol. 21, no. 6, pp. 479–489, 1997.
[106]  S. O. Freytag, “Enforced expression of the c-myc oncogene inhibits cell differentiation by precluding entry into a distinct predifferentiation state in G0/G1,” Molecular and Cellular Biology, vol. 8, no. 4, pp. 1614–1624, 1988.
[107]  K. Shuai, J. Halpern, J. T. Hoeve, X. Rao, and C. L. Sawyers, “Constitutive activation of STAT5 by the BCR-ABL oncogene in chronic myelogenous leukemia,” Oncogene, vol. 13, no. 2, pp. 247–254, 1996.
[108]  J. E. Darnell Jr., “STATs and gene regulation,” Science, vol. 277, no. 5332, pp. 1630–1635, 1997.
[109]  M. S. Neshat, A. B. Raitano, H. G. Wang, J. C. Reed, and C. L. Sawyers, “The survival function of the Bcr-Abl oncogene is mediated by Bad-dependent and -independent pathways: roles for phosphatidylinositol 3-kinase and Raf,” Molecular and Cellular Biology, vol. 20, no. 4, pp. 1179–1186, 2000.
[110]  K. Keeshan, T. G. Cotter, and S. L. McKenna, “High Bcr-Abl expression prevents the translocation of Bax and Bad to the mitochondrion,” Leukemia, vol. 16, no. 9, pp. 1725–1734, 2002.
[111]  T. Shinjyo, R. Kuribara, T. Inukai, et al., “Downregulation of Bim, a proapoptotic 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.
[112]  R. Kuribara, H. Honda, H. Matsui, et al., “Roles of Bim in apoptosis of normal and Bcr-Abl-expressing hematopoietic progenitors,” Molecular and Cellular Biology, vol. 24, no. 14, pp. 6172–6183, 2004.
[113]  K. J. Aichberger, M. Mayerhofer, M. T. Krauth, et al., “Identification of mcl-1 as a BCR/ABL-dependent target in chronic myeloid leukemia (CML): evidence for cooperative antileukemic effects of imatinib and mcl-1 antisense oligonucleotides,” Blood, vol. 105, no. 8, pp. 3303–3311, 2005.
[114]  S. M. O'Brien, C. C. Cunningham, A. K. Golenkov, A. G. Turkina, S. C. Novick, and K. R. Rai, “Phase I to II multicenter study of oblimersen sodium, a Bcl-2 antisense oligonucleotide, in patients with advanced chronic lymphocytic leukemia,” Journal of Clinical Oncology, vol. 23, no. 30, pp. 7697–7702, 2005.
[115]  N. W. C. van de Donk, M. M. J. Kamphuis, M. van Dijk, H. P. E. Borst, A. C. Bloem, and H. M. Lokhorst, “Chemosensitization of myeloma plasma cells by an antisense-mediated downregulation of Bcl-2 protein,” Leukemia, vol. 17, no. 1, pp. 211–219, 2003.
[116]  M. R. Smith, Y. Abubakr, R. Mohammad, T. Xie, M. Hamdan, and A. al-Katib, “Antisense oligodeoxyribonucleotide down-regulation of bcl-2 gene expression inhibits growth of the low-grade non-Hodgkin's lymphoma cell line WSU-FSCCL,” Cancer Gene Therapy, vol. 2, no. 3, pp. 207–212, 1995.
[117]  M. J. Keating, S. O'Brien, M. Albitar, et al., “Early results of a chemoimmunotherapy regimen of fludarabine, cyclophosphamide, and rituximab as initial therapy for chronic lymphocytic leukemia,” Journal of Clinical Oncology, vol. 23, no. 18, pp. 4079–4088, 2005.
[118]  A. Chanan-Khan, “Bcl-2 antisense therapy in B-cell malignancies,” Blood Reviews, vol. 19, no. 4, pp. 213–221, 2005.
[119]  G. Marcucci, W. Stock, G. Dai, et al., “Phase I study of oblimersen sodium, an antisense to Bcl-2, in untreated older patients with acute myeloid leukemia: pharmacokinetics, pharmacodynamics, and clinical activity,” Journal of Clinical Oncology, vol. 23, no. 15, pp. 3404–3411, 2005.
[120]  G. Marcucci, J. C. Byrd, G. Dai, et al., “Phase 1 and pharmacodynamic studies of G3139, a Bcl-2 antisense oligonucleotide, in combination with chemotherapy in refractory or relapsed acute leukemia,” Blood, vol. 101, no. 2, pp. 425–432, 2003.
[121]  F. J. Giles, “New drugs in acute myeloid leukemia,” Current Oncology Reports, vol. 4, no. 5, pp. 369–374, 2002.
[122]  K. V. Floros, M. Talieri, and A. Scorilas, “Topotecan and methotrexate alter expression of the apoptosis-related genes BCL2, FAS and BCL2L12 in leukemic HL-60 cells,” Biological Chemistry, vol. 387, no. 12, pp. 1629–1633, 2006.
[123]  K. V. Floros, H. Thomadaki, D. Florou, M. Talieri, and A. Scorilas, “Alterations in mRNA expression of apoptosis-related genes BCL2, BAX, FAS, caspase-3, and the novel member BCL2L12 after treatment of human leukemic cell line HL60 with the antineoplastic agent etoposide,” Annals of the New York Academy of Sciences, vol. 1090, pp. 89–97, 2006.
[124]  K. V. Floros, H. Thomadaki, N. Katsaros, M. Taileri, and A. Scorilas, “mRNA expression analysis of a variety of apoptosis-related genes, including the novel gene of the BCL2-family, BCL2L12, in HL-60 leukemia cells after treatment with carboplatin and doxorubicin,” Biological Chemistry, vol. 385, no. 11, pp. 1099–1103, 2004.
[125]  K. V. Floros, H. Thomadaki, G. Lallas, N. Katsaros, M. Talieri, and A. Scorilas, “Cisplatin-induced apoptosis in HL-60 human promyelocytic leukemia cells: differential expression of BCL2 and novel apoptosis-related gene BCL2L12,” Annals of the New York Academy of Sciences, vol. 1010, pp. 153–158, 2003.
[126]  H. Thomadaki, A. Karaliota, C. Litos, and A. Scorilas, “Enhanced antileukemic activity of the novel complex 2,5-dihydroxybenzoate molybdenum(VI) against 2,5-dihydroxybenzoate, polyoxometalate of Mo(VI), and tetraphenylphosphonium in the human HL-60 and K562 leukemic cell lines,” Journal of Medicinal Chemistry, vol. 50, no. 6, pp. 1316–1321, 2007.
[127]  H. Thomadaki, A. Karaliota, C. Litos, and A. Scorilas, “Enhanced concentration-dependent cytotoxic effect of the dinuclear copper(II) complex of L-carnitine [Cu2(L-carnitine) 2Cl2(H2O)2]Cl2, compared to L-carnitine or copper chloride dihydrate, in human leukemic cell lines,” Journal of Medicinal Chemistry, vol. 51, no. 13, pp. 3713–3719, 2008.
[128]  H. Thomadaki, K. V. Floros, and A. Scorilas, “Molecular response of HL-60 cells to mitotic inhibitors vincristine and taxol visualized with apoptosis-related gene expressions, including the new member BCL2L12,” Annals of the New York Academy of Sciences, vol. 1171, pp. 276–283, 2009.
[129]  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.
[130]  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.
[131]  M. H. Kang, Z. Wan, Y. H. Kang, R. Sposto, and C. P. Reynolds, “Mechanism of synergy of N-(4-hydroxyphenyl)retinamide and ABT-737 in acute lymphoblastic leukemia cell lines: Mcl-1 inactivation,” Journal of the National Cancer Institute, vol. 100, no. 8, pp. 580–595, 2008.
[132]  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.
[133]  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.
[134]  M. Vogler, M. Butterworth, A. Majid, et al., “Concurrent up-regulation of BCL-XL and BCL2A1 induces approximately 1000-fold resistance to ABT-737 in chronic lymphocytic leukemia,” Blood, vol. 113, no. 18, pp. 4403–4413, 2009.
[135]  M. Vogler, D. Dinsdale, X. M. Sun, et al., “A novel paradigm for rapid ABT-737-induced apoptosis involving outer mitochondrial membrane rupture in primary leukemia and lymphoma cells,” Cell Death and Differentiation, vol. 15, no. 5, pp. 820–830, 2008.
[136]  M. Vogler, S. D. Furdas, M. Jung, T. Kuwana, M. J. S. Dyer, and G. M. Cohen, “Diminished sensitivity of chronic lymphocytic leukemia cells to ABT-737 and ABT-263 due to albumin binding in blood,” Clinical Cancer Research, vol. 16, no. 16, pp. 4217–4225, 2010.
[137]  V. Del Gaizo 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.
[138]  K. D. Mason, S. L. Khaw, K. C. Rayeroux, et al., “The BH3 mimetic compound, ABT-737, synergizes with a range of cytotoxic chemotherapy agents in chronic lymphocytic leukemia,” Leukemia, vol. 23, no. 11, pp. 2034–2041, 2009.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413