scholarly journals Chromatin Redistribution of the DEK Oncoprotein Represses hTERT Transcription in Leukemias

Neoplasia ◽  
2014 ◽  
Vol 16 (1) ◽  
pp. 21-W14 ◽  
Author(s):  
Maroun Karam ◽  
Morgan Thenoz ◽  
Valérie Capraro ◽  
Jean-Philippe Robin ◽  
Christiane Pinatel ◽  
...  
Keyword(s):  
2007 ◽  
Vol 22 (6) ◽  
pp. 897-906 ◽  
Author(s):  
Sandra Isenmann ◽  
Dimitrios Cakouros ◽  
Andrew Zannettino ◽  
Songtao Shi ◽  
Stan Gronthos

2002 ◽  
Vol 13 (8) ◽  
pp. 2585-2597 ◽  
Author(s):  
Izumi Horikawa ◽  
P. LouAnn Cable ◽  
Sharlyn J. Mazur ◽  
Ettore Appella ◽  
Cynthia A. Afshari ◽  
...  

Regulation of the hTERT gene encoding the telomerase catalytic subunit plays an important role in human cell senescence, immortalization, and carcinogenesis. By examining the activity of various deleted or mutated hTERT promoter fragments, we show that an E-box element downstream of the transcription initiation site is critical to differential hTERT transcription between the telomerase/hTERT-positive renal cell carcinoma cell line (RCC23) and its telomerase/hTERT-negative counterpart containing a transferred, normal chromosome 3 (RCC23+3). This E-box element mediated repression of hTERT transcription in RCC23+3 but not in RCC23. A copy number–dependent enhancement of the repression suggested active repression, rather than loss of activation, in RCC23+3. Endogenous expression levels of c-Myc or Mad1, which could activate or repress hTERT transcription when overexpressed, did not account for the differential hTERT transcription. Gel mobility shift assays identified the upstream stimulatory factors (USFs) as a major E-box–binding protein complex in both RCC23 and RCC23+3 and, importantly, detected an RCC23+3-specific, E-box–binding factor that was distinct from the USF and Myc/Mad families. The E-box–mediated repression was also active in normal human fibroblasts and epithelial cells and inactive in some, but not all, telomerase/hTERT-positive cancer cells. These findings provide evidence for an endogenous, repressive mechanism that actively functions in telomerase/hTERT-negative normal cells and becomes defective during carcinogenic processes, e.g., by an inactivation of the telomerase repressor gene on chromosome 3.


2014 ◽  
Vol 32 (3) ◽  
pp. 1273-1280 ◽  
Author(s):  
YAFEI ZHANG ◽  
ANRAN ZHANG ◽  
CAIFEI SHEN ◽  
BICHENG ZHANG ◽  
ZHIGUO RAO ◽  
...  

2004 ◽  
Vol 45 (1-2) ◽  
pp. 23-32 ◽  
Author(s):  
Izumi Horikawa ◽  
Eriko Michishita ◽  
J. Carl Barrett
Keyword(s):  

Author(s):  
M. Maura ◽  
Y. Katakura ◽  
T. Miura ◽  
T. Fujiki ◽  
H. Shiraishi ◽  
...  

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2850-2850
Author(s):  
Franck Mortreux ◽  
Valérie Capraro ◽  
Agnès Lançon ◽  
Xavier Thomas ◽  
Eric Wattel

Abstract hTERT encodes telomerase reverse transcriptase which is the rate-limiting factor for telomerase activity (TA). Its expression is primarily regulated at the transcriptional level. High TA participates to the immortalization of malignant cells but the positive correlation between hTERT overexpression, increased TA and leukemogenesis does not seem to be a rule as hTERT underexpression has been evidenced in chronic myeloid leukemia, adult T-cell leukemia/lymphoma (ATLL), chronic lymphocytic leukemia, and in acute myeloid leukemia (AML). Certain oncogenes such as BCR-ABL or the HTLV-1-encoded oncoprotein Tax, which are respectively involved in the development of chronic myeloid leukemia and ATLL, negatively interfere with hTERT, resulting in its transcriptional repression. To investigate the molecular control of hTERT transcriptional repression during leukemogenesis, we carried out a proteomic screening of hTERT promoter occupancy in Tax+ and Tax− cells. Nuclear extracts (NEs) were prepared from HeLa cells transfected with a Tax or a control plasmid. NEs were incubated with the hTERT core promoter (hCP) and with an irrelevant control DNA fragment (CDF). For Tax+ and Tax− cells, protein interacting with the hCP or the CDF were purified and then analyzed by mass spectrometry. 270 proteins were identified from NEs incubated with hCP. Of those, 230 were present in both Tax+ and Tax− NEs whereas 22 and 18 were identified in NEs derived from Tax− and Tax+ cells. Values for CDF were 11, 1, and 0. The Tax+-hCP peptidome included DEK, a chromatin protein first identified as a fusion protein with CAN in AML with t(6;9). DEK is overexpressed in the majority of AML and displays multifunctional properties. Cotransfections and western blotting with proteins purified from NEs showed that DEK and Tax are present along the hCP in Tax+ cells but not in cells transfected with the control vector. DEK recruitment to the endogenous hTERT promoter was also evidenced by ChIP assays in Tax+- but not in Tax--cells. Cotransfection assays with a hTERT promoter-luciferase reporter plasmid and various concentrations of DEK and Tax expressors revealed that DEK inhibits hTERT transcription in a dose-dependent manner and that Tax and DEK synergize for repressing hTERT. Physical interaction between Tax and DEK was further demonstrated in vivo by co-immunoprecipitation assays. Cotransfections with modified reporter plasmids demonstrated that DEK represses hTERT transcription through the same E-box as Tax. We showed that a phosphorylation-dependent increased DNA binding activity of DEK stimulates its negative effect on hTERT transcription. SiRNA-mediated knockdown of DEK expression abolished the negative effect of Tax on hTERT, indicating that DEK is necessary for Tax-mediated hTERT repression. We quantified by qRTPCR the expression of 3 additional E-box containing genes known to be repressed by Tax, in cells stably expressing tax before and after siRNA-mediated knockdown of DEK: DEK knockdown resulted in a 3.5-, 2-, 1.5-, 14-fold increased expression of hTERT, lck, DNA polymerase beta and p18INK4C genes, respectively. Whether DEK could be involved in hTERT repression in other hematological malignancies than ATLL was finally demonstrated by identifying DEK as a member of the hCP peptidome in bone marrow blasts derived from patients with de novo AML with hTERT transcriptional repression but not in normal bone marrow mononuclear cells derived from donors. In conclusion our proteomic analysis of hTERT transcriptional regulation allowed the detection of specific preleukemic factors and provided evidence that in proliferating HeLa cells 15% of the hTERT promoter peptidome depends on the absence (9%) or the presence (6%) of Tax. The Tax-specific factor DEK physically interacts with Tax, is recruited to the hTERT promoter in Tax+ cells in which it represses hTERT transcription synergistically with Tax, through the same downstream E-box involved in Tax-dependent hTERT repression, and in a phosphorylation-dependent manner. The interplay between Tax and DEK in repressing the transcription of additional genes suggests that previous scenarios of Tax-mediated transcriptional repression in HTLV-1 should be revisited and updated to a new model in which DEK connects tax expression and gene repression. Finally, DEK and hTERT interactions appear to pertain to other malignancies than ATLL, such as AML.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 791-791
Author(s):  
Lili Yang ◽  
Adam W Mailloux ◽  
Dana E Rollison ◽  
Jong Park ◽  
Jeffrey S. Painter ◽  
...  

Abstract Abstract 791 Background: Myelodysplastic syndromes (MDS) are characterized by dysregulated myelopoiesis and peripheral cytopenias with enormous disease heterogeneity owing to diverse molecular pathobiology. The early manifestations of MDS, however, are relatively well conserved and include increased apoptosis coupled to excessive proliferation of myeloid progenitors. In addition to myeloid abnormalities, repertoire contraction and memory expansion is demonstrable in T cells. The notion that apoptosis of hematopoetic cells may be triggered through an immune–mediated mechanism arose from similarities with aplastic anemia (AA). Our recent data showed that MDS responsive to immunosuppressive therapy has accelerated naïve T cell turnover (ie, high proliferative index plus excessive cell death) which led us to hypothesize the presence of an inherent T cell abnormality impairing homeostatic regulation. AA can be caused by somatic mutations within telomere repair components. T-cells are one of a few somatic cells that retain telomerase function to control naïve T-cell survival, replication potential, and antigenic diversity. To this end, we examined telomere function and replicative burst capacity of MDS T cells as a possible mechanism for immune dysregulation. Methods: Primary specimens from MDS (n=37), AA (n=8), and controls (n=42) were investigated. Peripheral blood mononuclear cells were isolated from patient blood or buffy coats by Ficoll-Hypaque gradient centrifugation. Purified CD3+ T cells were isolated using negative selection and then stimulated with anti-CD3/anti-CD28 T cell activator beads (Dynabead®) for 3 days. Telomere length was assessed by quantitative PCR (q-PCR) and telomerase enzymatic function measured by Telomere Repeat Amplification Protocol (TRAP) assays. Results: Mean telomere length in purified T cells was significantly shorter among MDS patients compared to controls after adjusting for age and sex (p<0.0001). To assess telomerase repair function in MDS T-cells, we performed TRAP assays with purified T cells after stimulation and found that inducible telomerase activity is severely suppressed in MDS compare to controls. In comparison to controls, the inducible telomerase activity fell below the 95% confidence internal in all cases (MDS median 18.70, 95% CI, 15.93–20.54 vs control median 45.0, 95% CI, 45.79 – 64.5, p<0.0001) and the amount of telomerase activity was unrelated to risk stratification by the International Prognostic Scoring System (IPSS), World Health Organization (WHO) classification, and age indicating that it is a frequent abnormality in the disease. Analysis of telomerase function and telomere length in T cells from patients with AA showed a similar deficiency in telomerase repair function. The mechanism responsible for telomerase insufficiency in MDS was mediated by defective induction of telomerase reverse transcriptase (hTERT) transcription; the key enzyme involved in telomere maintenance. Next, to determine the functional consequences of the disturbance in telomere repair in MDS, the ability of T cells to enter S-phase and to undergo an antigen-induced proliferative burst were examined. TCR signaling was shown to be preserved, evidenced by induction of an early activation antigen CD69. Although some cells were capable of entering S-phase, the replicative burst potential was severely impaired in T cells form all patients. Telomere repair is exclusively present in naïve T cells and progressively declines after memory transition. TCR triggered telomerase activity was measured in sorted naïve (CD45RA+, CD45RO-) and memory (CD45RO+, CD45RA-) T cells. The telomere length in naïve cells was shorter in MDS patients compared to controls (p=0.018) and the telomerase activity was suppressed in naïve MDS T cells (p=0.0207) indicating that telomere dysfunction underlies the altered homeostasis of naïve T cells in MDS, a feature mechanistically akin to AA and other telomere repair disorders. Conclusion: Results of this study indicate that there is loss of telomere maintenance in naïve T cells due to a defect in hTERT transcription is associated with impaired replicative potential. This abnormality in naïve T cell homeostasis represents an inherent defect that contributes to a memory cell growth advantage and repertoire contraction associated with autoimmunity in AA and MDS. Disclosures: No relevant conflicts of interest to declare.


2016 ◽  
Vol 160 (5) ◽  
pp. 309-313 ◽  
Author(s):  
Shuntaro Yamashita ◽  
Kaoru Fujii ◽  
Chong Zhao ◽  
Hiroshi Takagi ◽  
Yoshinori Katakura

Oncogene ◽  
2003 ◽  
Vol 22 (24) ◽  
pp. 3734-3741 ◽  
Author(s):  
Anne-Sophie Gabet ◽  
Franck Mortreux ◽  
Pierre Charneau ◽  
Patrice Riou ◽  
Madeleine Duc-Dodon ◽  
...  
Keyword(s):  

Sign in / Sign up

Export Citation Format

Share Document