Browse > Article
http://dx.doi.org/10.7314/APJCP.2014.15.19.8019

Promyelocytic Leukemia Gene Functions and Roles in Tumorigenesis  

Imani-Saber, Zeinab (Department of Medical Genetics, Shahid Beheshti University of Medical Sciences)
Ghafouri-Fard, Soudeh (Department of Medical Genetics, Shahid Beheshti University of Medical Sciences)
Publication Information
Asian Pacific Journal of Cancer Prevention / v.15, no.19, 2014 , pp. 8019-8026 More about this Journal
Abstract
The promyelocytic leukemia (PML) gene is a gene known to be a tumor suppressor, although recent data suggest that it has a dual function in tumorigenesis. It was initially discovered in acute promyelocytic leukemia (APL) in which a t(15; 17) chromosomal translocation fused it to the retinoic acid receptor alpha ($RAR{\alpha}$). It has been shown to be involved in various types of cancer. It has at least 6 nuclear isoforms and a cytoplasmic type with different characteristics. Its multiple functions in growth inhibition, apoptosis induction, replicative senescence, inhibition of oncogenic transformation, and suppression of migration and angiogenesis have made it a therapeutic target for cancer therapy. However, its dual role in the process of tumorigenesis has made this field challenging. In this review, we discuss PML structure, functions and expression in tumors.
Keywords
PML; cancers; structure; nuclear bodies; functions and; physiological roles;
Citations & Related Records
Times Cited By KSCI : 2  (Citation Analysis)
연도 인용수 순위
1 Cuchet-Lourenco D, Vanni E, Glass M, et al (2012). Herpes simplex virus 1 ubiquitin ligase ICP0 interacts with PML isoform I and induces its SUMO-independent degradation. J Virol, 86, 11209-22.   DOI
2 de Stanchina E, Querido E, Narita M, et al (2004). PML is a direct p53 target that modulates p53 effector functions. Molecular cell, 13, 523-35.   DOI
3 de The H CC, Lanotte M, Degos L, Dejean A (1990). The t(15;17) translocation of acute promyelocytic leukaemia fuses the retinoic acid receptor alpha gene to a novel transcribed locus. Natur, 347, 558-61.   DOI   ScienceOn
4 de Visser KE, Coussens LM (2006). The inflammatory tumor microenvironment and its impact on cancer development. Contrib Microbiol, 13, 118-37.
5 Dellaire G, Ching RW, Ahmed K, et al (2006b). Promyelocytic leukemia nuclear bodies behave as DNA damage sensors whose response to DNA double-strand breaks is regulated by NBS1 and the kinases ATM, Chk2, and ATR. J Cell Biol, 175, 55-66.   DOI
6 Fanelli M, Fantozzi A, De Luca P, et al (2004). The coiledcoil domain is the structural determinant for mammalian homologues of Drosophila Sina-mediated degradation of promyelocytic leukemia protein and other tripartite motif proteins by the proteasome. J Biol Chem, 279, 5374-9.   DOI
7 Dermime S, Bertazzoli C, Marchesi E, et al (1996). Lack of T-cell-mediated recognition of the fusion region of the pml/RAR-alpha hybrid protein by lymphocytes of acute promyelocytic leukemia patients. Clin Cancer Res, 2, 593-600.
8 Dyck JA, Maul GG, Miller WH, Jr., et al (1994). A novel macromolecular structure is a target of the promyelocyteretinoic acid receptor oncoprotein. Cell, 76, 333-43.   DOI
9 Bischof O, Kirsh O, Pearson M, et al (2002). Deconstructing PML-induced premature senescence. Embo J, 21, 3358-69.   DOI
10 Ben-Baruch A (2006). The multifaceted roles of chemokines in malignancy. Cancer Metastasis Rev, 25, 357-71.   DOI
11 Bernardi R, Guernah I, Jin D, et al (2006). PML inhibits HIF-1alpha translation and neoangiogenesis through repression of mTOR. Nature, 442, 779-85.   DOI
12 Bernardi R, Pandolfi PP (2007). Structure, dynamics and functions of promyelocytic leukaemia nuclear bodies. Nat Rev Mol Cell Biol, 8, 1006-16.   DOI
13 Best JL, Ganiatsas S, Agarwal S, et al (2002). SUMO-1 protease-1 regulates gene transcription through PML. Mol Cell, 10, 843-55.   DOI
14 Carbone R, Pearson M, Minucci S, et al (2002). PML NBs associate with the hMre11 complex and p53 at sites of irradiation induced DNA damage. Oncogene, 21, 1633-40.   DOI
15 Chen RH, Lee YR, Yuan WC (2012). The role of PML ubiquitination in human malignancies. J Biomed Sci, 19, 81.   DOI
16 Carracedo A, Weiss D, Leliaert AK, et al (2012). A metabolic prosurvival role for< i> PML in breast cancer. J clin Invest, 122, 3088-100.   DOI
17 Chang CC, Naik MT, Huang YS, et al (2011). Structural and functional roles of Daxx SIM phosphorylation in SUMO paralog-selective binding and apoptosis modulation. Mol Cell, 42, 62-74.   DOI
18 Chelbi-Alix M, Pelicano L, Quignon F, et al (1995). Induction of the PML protein by interferons in normal and APL cells. Leukemia, 9, 2027-33.
19 Cooke HJ, Smith BA (1986). Variability at the telomeres of the human X/Y pseudoautosomal region. Cold Spring Harb Symp Quant Biol, 51, 213-9.   DOI
20 Everett RD (2001). DNA viruses and viral proteins that interact with PML nuclear bodies. Oncogene, 20, 7266-73.   DOI
21 Gamell C, Jan Paul P, Haupt Y, et al (2014). PML tumour suppression and beyond: Therapeutic implications. FEBS Lett.
22 Geoffroy MC, Jaffray EG, Walker KJ, et al (2010). Arsenicinduced SUMO-dependent recruitment of RNF4 into PML nuclear bodies. Mol Biol Cell, 21, 4227-39.   DOI
23 Ghafouri-Fard S, Ghafouri-Fard S (2012a). Immunotherapy in nonmelanoma skin cancer. Immunotherapy, 4, 499-510.   DOI   ScienceOn
24 Ghafouri-Fard S, Ghafouri-Fard S (2012b). siRNA and cancer immunotherapy. Immunotherapy, 4, 907-17.   DOI
25 Ghafouri-Fard S, Modarressi MH, Yazarloo F (2012). Expression of testis-specific genes, TEX101 and ODF4, in chronic myeloid leukemia and evaluation of TEX101 immunogenicity. Ann. Saudi Med, 32, 256-61.
26 Ghafouri-Fard S, Shamsi R, Seifi-Alan M, et al (2014). Cancertestis genes as candidates for immunotherapy in breast cancer. Immunotherapy, 6, 165-79.   DOI
27 Hattersley N, Shen L, Jaffray EG, et al (2011). The SUMO protease SENP6 is a direct regulator of PML nuclear bodies. Mol Biol Cell, 22, 78-90.   DOI
28 Guan D, Factor D, Liu Y, et al (2013). The epigenetic regulator UHRF1 promotes ubiquitination-mediated degradation of the tumor-suppressor protein promyelocytic leukemia protein. Oncogene, 32, 3819-28.   DOI
29 Guo A, Salomoni P, Luo J, et al (2000). The function of PML in p53-dependent apoptosis. Nat Cell Biol, 2, 730-6.   DOI
30 Gurrieri C, Capodieci P, Bernardi R, et al (2004). Loss of the tumor suppressor PML in human cancers of multiple histologic origins. J Natl Cancer Inst, 96, 269-79.   DOI
31 Hubackova S, Krejcikova K, Bartek J, et al (2012). Interleukin 6 signaling regulates promyelocytic leukemia protein gene expression in human normal and cancer cells. J Biol Chem, 287, 26702-14.   DOI
32 Ishov AM, Sotnikov AG, Negorev D, et al (1999). PML is critical for ND10 formation and recruits the PML-interacting protein daxx to this nuclear structure when modified by SUMO-1. J Cell Biol, 147, 221-34.   DOI
33 Ito K, Bernardi R, Morotti A, et al (2008). PML targeting eradicates quiescent leukaemia-initiating cells. Nature, 453, 1072-8.   DOI
34 Jensen K, Shiels C, Freemont PS (2001). PML protein isoforms and the RBCC/TRIM motif. Oncogene, 20, 7223-33.   DOI
35 Jin G, Wang Y-J, Lin H-K (2013). Emerging cellular functions of cytoplasmic PML. Frontiers in oncology, 3, 147.
36 Kakizuka A, Miller WH, Jr., Umesono K, et al (1991). Chromosomal translocation t(15;17) in human acute promyelocytic leukemia fuses RAR alpha with a novel putative transcription factor, PML. Cell, 66, 663-74.   DOI   ScienceOn
37 Lallemand-Breitenbach V, de The H (2010). PML nuclear bodies. Cold Spring Harb Perspect Biol, 2, 000661.   DOI
38 Kamitani T, Kito K, Nguyen HP, et al (1998). Identification of three major sentrinization sites in PML. J Biol Chem, 273, 26675-82.   DOI
39 Kim HJ, Song DE, Lim SY, et al (2011). Loss of the promyelocytic leukemia protein in gastric cancer: implications for IP-10 expression and tumor-infiltrating lymphocytes. PLoS One, 6, 26264.   DOI
40 Koken MH, Puvion-Dutilleul F, Guillemin MC, et al (1994). The t(15;17) translocation alters a nuclear body in a retinoic acid-reversible fashion. Embo J, 13, 1073-83.
41 Lallemand-Breitenbach V, Jeanne M, Benhenda S, et al (2008). Arsenic degrades PML or PML-RARalpha through a SUMOtriggered RNF4/ubiquitin-mediated pathway. Nat Cell Biol, 10, 547-55.   DOI
42 Lee EY, Lee ZH, Song YW (2009). CXCL10 and autoimmune diseases. Autoimmun Rev, 8, 379-83.   DOI
43 Lee HE, Jee CD, Kim MA, et al (2007). Loss of promyelocytic leukemia protein in human gastric cancers. Cancer letters, 247, 103-9.   DOI
44 Lin H-K, Bergmann S, Pandolfi PP (2004). Cytoplasmic PML function in TGF-${\beta}$ signalling. Nature, 431, 205-11.   DOI
45 Maarifi G, Chelbi-Alix MK, Nisole S (2014). PML control of cytokine signaling. Cytokine Growth Factor Rev.
46 Maiuri MC, Tasdemir E, Criollo A, et al (2009). Control of autophagy by oncogenes and tumor suppressor genes. Cell Death Differ, 16, 87-93.   DOI   ScienceOn
47 Mallette FA, Goumard S, Gaumont-Leclerc MF, et al (2004). Human fibroblasts require the Rb family of tumor suppressors, but not p53, for PML-induced senescence. Oncogene, 23, 91-9.   DOI   ScienceOn
48 Nakamura TM, Morin GB, Chapman KB, et al (1997). Telomerase catalytic subunit homologs from fission yeast and human. Science, 277, 955-9.   DOI
49 Martin N, Benhamed M, Nacerddine K, et al (2012). Physical and functional interaction between PML and TBX2 in the establishment of cellular senescence. Embo J, 31, 95-109.   DOI
50 Mazza M, Pelicci PG (2013). Is PML a Tumor Suppressor? Front Oncol, 3, 174.
51 Osman Y, Takahashi M, Zheng Z, et al (1999). Dendritic cells stimulate the expansion of PML-RAR alpha specific cytotoxic T-lymphocytes: its applicability for antileukemia immunotherapy. J Exp Clin Cancer Res, 18, 485-92.
52 Meyerson M, Counter CM, Eaton EN, et al (1997). hEST2, the putative human telomerase catalytic subunit gene, is up-regulated in tumor cells and during immortalization. Cell, 90, 785-95.   DOI   ScienceOn
53 Nicewonger J, Suck G, Bloch D, et al (2004). Epstein-Barr virus (EBV) SM protein induces and recruits cellular Sp110b to stabilize mRNAs and enhance EBV lytic gene expression. J Virol, 78, 9412-22.   DOI   ScienceOn
54 Nisole S, Maroui MA, Mascle XH, et al (2013). Differential Roles of PML Isoforms. Front Oncol, 3, 125.
55 Padua RA, Larghero J, Robin M, et al (2003). PML-RARA-targeted DNA vaccine induces protective immunity in a mouse model of leukemia. Nature medicine, 9, 1413-7.   DOI
56 Potts PR, Yu H (2007). The SMC5/6 complex maintains telomere length in ALT cancer cells through SUMOylation of telomere-binding proteins. Nat Struct Mol Biol, 14, 581-90.   DOI
57 Quignon F, De Bels F, Koken M, et al (1998). PML induces a novel caspase-independent death process. Nat Genet, 20, 259-65.   DOI   ScienceOn
58 Salomoni P, Dvorkina M, Michod D (2012). Role of the promyelocytic leukaemia protein in cell death regulation. Cell Death Dis, 3, 247.   DOI
59 Rabellino A, Carter B, Konstantinidou G, et al (2012). The SUMO E3-ligase PIAS1 regulates the tumor suppressor PML and its oncogenic counterpart PML-RARA. Cancer Res, 72, 2275-84.   DOI
60 Reineke EL, Kao HY (2009). PML: An emerging tumor suppressor and a target with therapeutic potential. Cancer Ther, 7, 219-26.
61 Shen TH, Lin HK, Scaglioni PP, et al (2006). The mechanisms of PML-nuclear body formation. Mol Cell, 24, 331-9.   DOI
62 Reineke EL, Liu Y, Kao H-Y (2010). Promyelocytic leukemia protein controls cell migration in response to hydrogen peroxide and insulin-like growth factor-1. J Biol Chem, 285, 9485-92.   DOI
63 Satow R, Shitashige M, Jigami T, et al (2012). beta-catenin inhibits promyelocytic leukemia protein tumor suppressor function in colorectal cancer cells. Gastroenterology, 142, 572-81.   DOI
64 Scaglioni PP, Yung TM, Choi S, et al (2008). CK2 mediates phosphorylation and ubiquitin-mediated degradation of the PML tumor suppressor. Mol Cell Biochem, 316, 149-54.   DOI
65 Silzle T, Randolph GJ, Kreutz M, et al (2004). The fibroblast: sentinel cell and local immune modulator in tumor tissue. Int J Cancer, 108, 173-80.   DOI
66 Stadler M, Chelbi-Alix MK, Koken M, et al (1995). Transcriptional induction of the PML growth suppressor gene by interferons is mediated through an ISRE and a GAS element. Oncogene, 11, 2565-73.
67 Stehmeier P, Muller S (2009). Phospho-regulated SUMO interaction modules connect the SUMO system to CK2 signaling. Mol Cell, 33, 400-9.   DOI
68 Tatham MH, Geoffroy MC, Shen L, et al (2008). RNF4 is a poly-SUMO-specific E3 ubiquitin ligase required for arsenic-induced PML degradation. Nat Cell Biol, 10, 538-46.   DOI
69 Tabarestani S, Ghafouri-Fard S (2012). Cancer stem cells and response to therapy. Asian Pac J Cancer Prev, 13, 5951-8.
70 Tamura G (2006). Alterations of tumor suppressor and tumorrelated genes in the development and progression of gastric cancer. World J Gastroenterol, 12, 192-8.
71 Vernier M, Bourdeau V, Gaumont-Leclerc MF, et al (2011). Regulation of E2Fs and senescence by PML nuclear bodies. Genes Dev, 25, 41-50.   DOI
72 Tang MK, Liang YJ, Chan JY, et al (2013). Promyelocytic leukemia (PML) protein plays important roles in regulating cell adhesion, morphology, proliferation and migration. PLoS One, 8, 59477.   DOI
73 Trotman LC, Alimonti A, Scaglioni PP, et al (2006). Identification of a tumour suppressor network opposing nuclear Akt function. Nature, 441, 523-7.   DOI
74 Van Damme E, Laukens K, Dang TH, et al (2010). A manually curated network of the PML nuclear body interactome reveals an important role for PML-NBs in SUMOylation dynamics. Int J Biol Sci, 6, 51-67.
75 Wang S, Long J, Zheng CF (2012). The potential link between PML NBs and ICP0 in regulating lytic and latent infection of HSV-1. Protein Cell, 3, 372-82.   DOI
76 Wang Z-G, Ruggero D, Ronchetti S, et al (1998a). PML is essential for multiple apoptotic pathways. Nature genetics, 20, 266-72.   DOI
77 Wang ZG, Ruggero D, Ronchetti S, et al (1998b). PML is essential for multiple apoptotic pathways. Nat Genet, 20, 266-72.   DOI
78 Yang S, Kuo C, Bisi JE, et al (2002). PML-dependent apoptosis after DNA damage is regulated by the checkpoint kinase hCds1/Chk2. Nat Cell Biol, 4, 865-70.   DOI   ScienceOn
79 Wu G, Lee WH, Chen PL (2000). NBS1 and TRF1 colocalize at promyelocytic leukemia bodies during late S/G2 phases in immortalized telomerase-negative cells. Implication of NBS1 in alternative lengthening of telomeres. J Biol Chem, 275, 30618-22.   DOI
80 Wu S, Zhang X, Li ZM, et al (2013). Partial Least Squares Based Gene Expression Analysis in EBV- Positive and EBVNegative Posttransplant Lymphoproliferative Disorders. Asian Pac J Cancer Prev, 14, 6347-50.   DOI   ScienceOn
81 Wu WS, Xu ZX, Hittelman WN, et al (2003). Promyelocytic leukemia protein sensitizes tumor necrosis factor alphainduced apoptosis by inhibiting the NF-kappaB survival pathway. J Biol Chem, 278, 12294-304.   DOI
82 Yeager TR, Neumann AA, Englezou A, et al (1999). Telomerasenegative immortalized human cells contain a novel type of promyelocytic leukemia (PML) body. Cancer Res, 59, 4175-9.
83 Zhou W, Bao S (2014). PML-mediated signaling and its role in cancer stem cells. Oncogene, 33, 1475-84.   DOI