scholarly journals Downstream E-Box–mediated Regulation of the Human Telomerase Reverse Transcriptase (hTERT) Gene Transcription: Evidence for an Endogenous Mechanism of Transcriptional Repression

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.

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.


1992 ◽  
Vol 12 (10) ◽  
pp. 4486-4495 ◽  
Author(s):  
E F Petricoin ◽  
R H Hackett ◽  
H Akai ◽  
K Igarashi ◽  
D S Finbloom ◽  
...  

Phorbol esters activate the expression of a variety of early-response genes through protein kinase C-dependent pathways. In addition, phorbol esters may promote cell growth by the inhibition of expression of cellular gene products regulated by antiproliferative agents such as interferons (IFN)s. In human diploid fibroblasts, phorbol 12-myristate 13-acetate (PMA) selectively inhibits the IFN-alpha-induced cellular gene ISG54. Using transient transfection assays, we have delineated two elements in the promoter of this gene that are necessary for the inhibitory actions of PMA. These elements include (i) the IFN-stimulated response element (ISRE) which is necessary for IFN-alpha-induced cellular gene expression, and (ii) an element located near the site of transcription initiation. IFN-alpha treatment resulted in the rapid induction of ISGF3, a multisubunit transcription factor which binds to the ISRE. PMA caused a substantial reduction in IFN alpha-induced ISGF3 in both nuclear and cytoplasmic extracts, as determined by electrophoretic mobility shift assays with the ISRE as a probe. In vitro reconstitution experiments revealed that IFN-alpha activation of the ISGF3 alpha component of ISGF3 was not affected by PMA. Further experiments were consistent with the possibility that PMA regulated the activity of a cellular factor which competed with ISGF3 gamma for binding of the activated ISGF3 alpha polypeptides. Electrophoretic mobility shift assays using the cap site of ISG54 as a probe demonstrated the formation of a specific complex whose DNA binding activity was not affected by treatment of cells with PMA or IFN-alpha. Competitive inhibition studies were consistent with the DNA-protein complex at the cap site of ISG54 containing proteins with DNA binding sites in common with those which also interact with the ISRE. These data suggest a unique regulatory mechanism by which phorbol esters can modulate IFN signaling.


1992 ◽  
Vol 12 (10) ◽  
pp. 4486-4495
Author(s):  
E F Petricoin ◽  
R H Hackett ◽  
H Akai ◽  
K Igarashi ◽  
D S Finbloom ◽  
...  

Phorbol esters activate the expression of a variety of early-response genes through protein kinase C-dependent pathways. In addition, phorbol esters may promote cell growth by the inhibition of expression of cellular gene products regulated by antiproliferative agents such as interferons (IFN)s. In human diploid fibroblasts, phorbol 12-myristate 13-acetate (PMA) selectively inhibits the IFN-alpha-induced cellular gene ISG54. Using transient transfection assays, we have delineated two elements in the promoter of this gene that are necessary for the inhibitory actions of PMA. These elements include (i) the IFN-stimulated response element (ISRE) which is necessary for IFN-alpha-induced cellular gene expression, and (ii) an element located near the site of transcription initiation. IFN-alpha treatment resulted in the rapid induction of ISGF3, a multisubunit transcription factor which binds to the ISRE. PMA caused a substantial reduction in IFN alpha-induced ISGF3 in both nuclear and cytoplasmic extracts, as determined by electrophoretic mobility shift assays with the ISRE as a probe. In vitro reconstitution experiments revealed that IFN-alpha activation of the ISGF3 alpha component of ISGF3 was not affected by PMA. Further experiments were consistent with the possibility that PMA regulated the activity of a cellular factor which competed with ISGF3 gamma for binding of the activated ISGF3 alpha polypeptides. Electrophoretic mobility shift assays using the cap site of ISG54 as a probe demonstrated the formation of a specific complex whose DNA binding activity was not affected by treatment of cells with PMA or IFN-alpha. Competitive inhibition studies were consistent with the DNA-protein complex at the cap site of ISG54 containing proteins with DNA binding sites in common with those which also interact with the ISRE. These data suggest a unique regulatory mechanism by which phorbol esters can modulate IFN signaling.


2010 ◽  
Vol 21 (5) ◽  
pp. 821-832 ◽  
Author(s):  
Shuwen Wang ◽  
Chunguang Hu ◽  
Jiyue Zhu

hTERT, the human telomerase reverse transcriptase, is highly expressed in stem cells and embryonic tissues but undetectable in most adult somatic cells. To understand its repression mechanisms in somatic cells, we investigated the endogenous hTERT gene regulation during differentiation of human leukemic HL60 cells. Our study revealed that silencing of the hTERT promoter was a biphasic process. Within 24 h after initiation of differentiation, hTERT mRNA expression decreased dramatically, accompanied by increased expression of Mad1 gene and disappearance of a nucleosome-free region at the hTERT core promoter. Subsequent to this early repression, nucleosomal remodeling continued at the promoter and downstream region for several days, as demonstrated by micrococcal nuclease and restriction enzyme accessibility assays. This later nucleosomal remodeling correlated with stable silencing of the hTERT promoter. Progressive changes of core histone modifications occurred throughout the entire differentiation process. Surprisingly, inhibition of histone deacetylation at the hTERT promoter did not prevent hTERT repression or nucleosomal deposition, indicating that nucleosomal deposition at the core promoter, but not histone deacetylation, was the cause of transcriptional repression. Our data also suggested that succeeding nucleosomal remodeling and histone deacetylation worked in parallel to establish the stable repressive status of hTERT gene in human somatic cells.


2019 ◽  
Vol 2019 ◽  
pp. 1-13 ◽  
Author(s):  
Aaron L. Slusher ◽  
Tiffany M. Zúñiga ◽  
Edmund O. Acevedo

Age-related elevations in proinflammatory cytokines, known as inflamm-aging, are associated with shorter immune cell telomere lengths. Purpose. This study examined the relationship of plasma PTX3 concentrations, a biomarker of appropriate immune function, with telomere length in 15 middle-aged (40-64 years) and 15 young adults (20-31 years). In addition, PBMCs were isolated from middle-aged and young adults to examine their capacity to express a key mechanistic component of telomere length maintenance, human telomerase reverse transcriptase (hTERT), following ex vivo cellular stimulation. Methods. Plasma PTX3 and inflammatory cytokines (i.e., IL-6, IL-10, TGF-β, and TNF-α), PBMC telomere lengths, and PBMC hTERT gene expression and inflammatory protein secretion following exposure to LPS, PTX3, and PTX3+LPS were measured. Results. Aging was accompanied by the accumulation of centrally located visceral adipose tissue, without changes in body weight and BMI, and alterations in the systemic inflammatory milieu (decreased plasma PTX3 and TGF-β; increased TNF-α (p≤0.050)). In addition, shorter telomere lengths in middle-aged compared to young adults (p=0.011) were negatively associated with age, body fat percentages, and plasma TNF-α (r=−0.404, p=0.027; r=−0.427, p=0.019; and r=−0.323, p=0.041, respectively). Finally, the capacity of PBMCs to increase hTERT gene expression following ex vivo stimulation was impaired in middle-aged compared to young adults (p=0.033) and negatively associated with telomere lengths (r=0.353, p=0.028). Conclusions. Proinflammation and the impaired hTERT gene expression capacity of PBMCs may contribute to age-related telomere attrition and disease.


2015 ◽  
Vol 14 (4) ◽  
pp. 485-490 ◽  
Author(s):  
Mohammad Pourhassan-Moghaddam ◽  
Nosratollah Zarghami ◽  
Afshin Mohsenifar ◽  
Mohammad Rahmati-Yamchi ◽  
Hadis Daraee ◽  
...  

2015 ◽  
Vol 35 (2) ◽  
Author(s):  
Hua Li ◽  
Jiwen He ◽  
Huimin Yi ◽  
Guoan Xiang ◽  
Kaiyun Chen ◽  
...  

In the present study, we delivered human telomerase reverse transcriptase (hTERT) siRNA into SMMC-7721 hepatoma cells using a matrix metalloproteinase-2 (MMP2)-activatable cell-penetrating peptide (aCPP). The siRNA subsequently induced down-regulation of the hTERT gene and G1-arrest, implicating the utility of this delivery system in cancer therapy.


1993 ◽  
Vol 13 (7) ◽  
pp. 3860-3871
Author(s):  
P L Sawaya ◽  
B R Stripp ◽  
J A Whitsett ◽  
D S Luse

We have shown that a large fragment (-2339 to +57) from the rat CC10 gene directed lung-specific expression of a reporter construct in transgenic animals. Upon transfection, a smaller fragment (-165 to +57) supported reporter gene expression exclusively in the Clara cell-like NCI-H441 cell line, suggesting that a Clara cell-specific transcriptional element resided on this fragment (B. R. Stripp, P. L. Sawaya, D. S. Luse, K. A. Wikenheiser, S. E. Wert, J. A. Huffman, D. L. Lattier, G. Singh, S. L. Katyal, and J. A. Whitsett, J. Biol. Chem. 267:14703-14712, 1992). The interactions of nuclear proteins with a particular segment of the CC10 promoter which extends from 79 to 128 bp upstream of the CC10 transcription initiation site (CC10 region I) have now been studied. This sequence can stimulate both in vitro transcription in H441 nuclear extract and transient expression of reporter constructs in H441 cells. Electrophoretic mobility shift assays using extracts from H441, HeLa, rat liver, and fetal sheep lung cells were used to demonstrate that members of the AP-1, octamer, and HNF-3 families bind to CC10 region I. Transcription factors from H441 cells which are capable of binding to CC10 region I are either absent in HeLa, rat liver, and fetal sheep lung extracts or enriched in H441 extracts relative to extracts from non-Clara cells.


Sign in / Sign up

Export Citation Format

Share Document