Cargando…

Receptor C(k)-dependent signaling regulates hTERT gene transcription

BACKGROUND: Available evidence suggests that the regulation of telomerase activity primarily depends on the transcriptional control of the human telomerase reverse transcriptase (hTERT) gene. Although several activators and repressors of hTERT gene transcription have been identified, the exact mecha...

Descripción completa

Detalles Bibliográficos
Autores principales: Sikand, Kavleen, Kaul, Deepak, Varma, Neelam
Formato: Texto
Lenguaje:English
Publicado: BioMed Central 2006
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1351175/
https://www.ncbi.nlm.nih.gov/pubmed/16405739
http://dx.doi.org/10.1186/1471-2121-7-2
_version_ 1782126647454990336
author Sikand, Kavleen
Kaul, Deepak
Varma, Neelam
author_facet Sikand, Kavleen
Kaul, Deepak
Varma, Neelam
author_sort Sikand, Kavleen
collection PubMed
description BACKGROUND: Available evidence suggests that the regulation of telomerase activity primarily depends on the transcriptional control of the human telomerase reverse transcriptase (hTERT) gene. Although several activators and repressors of hTERT gene transcription have been identified, the exact mechanism by which hTERT transcription is repressed in normal cells and activated in cancer cells remains largely unknown. In an attempt to identify possible novel mechanisms involved in the regulation of hTERT transcription, the present study examined the role of Receptor C(k), a cell surface receptor specific for cholesterol, in the transcription of hTERT gene in normal human peripheral blood mononuclear cells. RESULTS: Activated Receptor C(k )was found to down-regulate hTERT mRNA expression by repressing the transcription of c-myc gene. Receptor C(k)-dependent signaling was also found to down-regulate the mRNA expression of the gene coding for the ligand inducible transcription factor, peroxisome proliferator-activated receptor γ (PPARγ). The ligand activation of PPARγ resulted in the down-regulation of c-myc and hTERT mRNA expression. By using specific activator and inhibitor of protein kinase C (PKC), it was demonstrated that Receptor C(k )dependent down-regulation of hTERT gene transcription involved inhibition of PKC. In addition, 25-hydroxycholesterol was found to contribute to the transcriptional regulation of hTERT gene. CONCLUSION: Taken together, the findings of this study present evidence for a molecular link between cholesterol-activated Receptor C(k )and hTERT transcription, and provide new insights into the regulation of hTERT expression in normal human peripheral blood mononuclear cells.
format Text
id pubmed-1351175
institution National Center for Biotechnology Information
language English
publishDate 2006
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-13511752006-01-26 Receptor C(k)-dependent signaling regulates hTERT gene transcription Sikand, Kavleen Kaul, Deepak Varma, Neelam BMC Cell Biol Research Article BACKGROUND: Available evidence suggests that the regulation of telomerase activity primarily depends on the transcriptional control of the human telomerase reverse transcriptase (hTERT) gene. Although several activators and repressors of hTERT gene transcription have been identified, the exact mechanism by which hTERT transcription is repressed in normal cells and activated in cancer cells remains largely unknown. In an attempt to identify possible novel mechanisms involved in the regulation of hTERT transcription, the present study examined the role of Receptor C(k), a cell surface receptor specific for cholesterol, in the transcription of hTERT gene in normal human peripheral blood mononuclear cells. RESULTS: Activated Receptor C(k )was found to down-regulate hTERT mRNA expression by repressing the transcription of c-myc gene. Receptor C(k)-dependent signaling was also found to down-regulate the mRNA expression of the gene coding for the ligand inducible transcription factor, peroxisome proliferator-activated receptor γ (PPARγ). The ligand activation of PPARγ resulted in the down-regulation of c-myc and hTERT mRNA expression. By using specific activator and inhibitor of protein kinase C (PKC), it was demonstrated that Receptor C(k )dependent down-regulation of hTERT gene transcription involved inhibition of PKC. In addition, 25-hydroxycholesterol was found to contribute to the transcriptional regulation of hTERT gene. CONCLUSION: Taken together, the findings of this study present evidence for a molecular link between cholesterol-activated Receptor C(k )and hTERT transcription, and provide new insights into the regulation of hTERT expression in normal human peripheral blood mononuclear cells. BioMed Central 2006-01-12 /pmc/articles/PMC1351175/ /pubmed/16405739 http://dx.doi.org/10.1186/1471-2121-7-2 Text en Copyright © 2006 Sikand et al; licensee BioMed Central Ltd. http://creativecommons.org/licenses/by/2.0 This is an Open Access article distributed under the terms of the Creative Commons Attribution License ( (http://creativecommons.org/licenses/by/2.0) ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Article
Sikand, Kavleen
Kaul, Deepak
Varma, Neelam
Receptor C(k)-dependent signaling regulates hTERT gene transcription
title Receptor C(k)-dependent signaling regulates hTERT gene transcription
title_full Receptor C(k)-dependent signaling regulates hTERT gene transcription
title_fullStr Receptor C(k)-dependent signaling regulates hTERT gene transcription
title_full_unstemmed Receptor C(k)-dependent signaling regulates hTERT gene transcription
title_short Receptor C(k)-dependent signaling regulates hTERT gene transcription
title_sort receptor c(k)-dependent signaling regulates htert gene transcription
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1351175/
https://www.ncbi.nlm.nih.gov/pubmed/16405739
http://dx.doi.org/10.1186/1471-2121-7-2
work_keys_str_mv AT sikandkavleen receptorckdependentsignalingregulateshtertgenetranscription
AT kauldeepak receptorckdependentsignalingregulateshtertgenetranscription
AT varmaneelam receptorckdependentsignalingregulateshtertgenetranscription