Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Dermatol ; 23(3): 344-9, 2013.
Article in English | MEDLINE | ID: mdl-23816528

ABSTRACT

BACKGROUND: The pituitary homeobox 1 (PITX1) protein is a member of the bicoid-related homeobox transcription factors and has essential roles in human development. Recently, the PITX1 gene has been considered as a tumor suppressor gene in various human cancers. OBJECTIVE: This study examined the expression of PITX1 in the development and progression of human cutaneous malignant melanoma. MATERIALS & METHODS: Immunohistochemical and/or immunofluorescence analyses were performed to examine the histological expression of PITX1 in healthy skin and 40 cutaneous malignant melanoma cases, including 10 melanoma in situ cases. RESULTS: Expression of PITX1 was shown in nuclei of melanocytes in normal skin. PITX1 expression was positive (labeling index: ≥10%) in 21 (52.5%) cases and negative (labeling index: <10%) in 19 (47.5%) of 40 cases of primary cutaneous malignant melanoma. The mean tumor thickness in PITX1-negative cases (7.11 ± 10.3 mm) was significantly higher than that in the positive cases (1.90 ± 3.19 mm) (P<0.01). The numbers of cases showing metastasis were 1 (4.76%) of 21 cases in PITX1-positive cases and 7 (36.8%) of 19 cases in PITX1-negative cases; the frequency was significantly higher in PITX1-negative cases than the positive cases (P = 0.012). Moreover, the reduction in PITX1 expression correlated significantly with clinical stage (P<0.001). Interestingly, PITX1 expression was inversely correlated with cell proliferation of cutaneous malignant melanoma (P<0.001). CONCLUSIONS: Down-regulation of PITX1 expression might contribute to the progression of cutaneous malignant melanoma via promoting cell proliferative activity.


Subject(s)
Gene Expression Regulation, Neoplastic , Melanoma/genetics , Paired Box Transcription Factors/genetics , Aged , Disease Progression , Female , Humans , Male , Prognosis , Skin Neoplasms , Melanoma, Cutaneous Malignant
2.
Biochim Biophys Acta ; 1823(4): 889-99, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22343000

ABSTRACT

Down-regulation of hMSH3 is associated with elevated microsatellite alterations at selected tetranucleotide repeats and low levels of microsatellite instability in colorectal cancer (CRC). However, the mechanism that down-regulates hMSH3 in CRC is not known. In this study, a significant association between over-expression of glucose transporter 1, a marker for hypoxia, and down-regulation of hMSH3 in CRC tissues was observed. Therefore, we examined the effect of hypoxia on the expression of hMSH3 in human cell lines. When cells with wild type p53 (wt-p53) were exposed to hypoxia, rapid down-regulation of both hMSH2 and hMSH3 occurred. In contrast, when null or mutated p53 (null/mut-p53) cells were exposed to hypoxia, only hMSH3 was down-regulated, and at slower rate than wt-p53 cells. Using a reporter assay, we found that disruption of the two putative hypoxia response elements (HREs) located within the promoter region of the hMSH3 abrogated the suppressive effect of hypoxia on reporter activity regardless of p53 status. In an EMSA, two different forms of HIF-1α complexes that specifically bind to these HREs were detected. A larger complex containing HIF-1α predominantly bound to the HREs in hypoxic null/mut-p53 cells whereas a smaller complex predominated in wt-p53 cells. Finally, HIF-1α knockdown by siRNA significantly inhibited down-regulation of hMSH3 by hypoxia in both wt-p53 and mut-p53 cells. Taken together, our results suggest that the binding of HIF-1α complexes to HRE sites is necessary for down-regulation of hMSH3 in both wt-p53 and mut-p53 cells.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA-Binding Proteins/genetics , Down-Regulation/genetics , Base Sequence , Cell Hypoxia/genetics , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Genes, Reporter , Glucose Transporter Type 1/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Molecular Sequence Data , MutS Homolog 3 Protein , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Response Elements/genetics , Tumor Suppressor Protein p53/metabolism
3.
Mol Cell Biol ; 31(8): 1624-36, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21300782

ABSTRACT

Telomerase, a ribonucleoprotein enzyme that maintains telomere length, is crucial for cellular immortalization and cancer progression. Telomerase activity is attributed primarily to the expression of telomerase reverse transcriptase (TERT). Using microcell-mediated chromosome transfer (MMCT) into the mouse melanoma cell line B16F10, we previously found that human chromosome 5 carries a gene, or genes, that can negatively regulate TERT expression (H. Kugoh, K. Shigenami, K. Funaki, J. Barrett, and M. Oshimura, Genes Chromosome Cancer 36:37-47, 2003). To identify the gene responsible for the regulation of TERT transcription, we performed cDNA microarray analysis using parental B16F10 cells, telomerase-negative B16F10 microcell hybrids with a human chromosome 5 (B16F10MH5), and its revertant clones (MH5R) with reactivated telomerase. Here, we report the identification of PITX1, whose expression leads to the downregulation of mouse tert (mtert) transcription, as a TERT suppressor gene. Additionally, both human TERT (hTERT) and mouse TERT (mtert) promoter activity can be suppressed by PITX1. We show that three and one binding site within the hTERT and mtert promoters, respectively, that express a unique conserved region are responsible for the transcriptional activation of TERT. Furthermore, we showed that PITX1 binds to the TERT promoter both in vitro and in vivo. Thus, PITX1 suppresses TERT transcription through direct binding to the TERT promoter, which ultimately regulates telomerase activity.


Subject(s)
Chromosomes, Human, Pair 5 , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Telomerase/metabolism , Animals , Cell Line, Tumor , Humans , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , Sequence Alignment , Sequence Homology, Nucleic Acid , Telomerase/genetics , Transcription, Genetic
4.
Genome Integr ; 1(1): 6, 2010 May 26.
Article in English | MEDLINE | ID: mdl-20678252

ABSTRACT

Telomerase is a ribonucleoprotein enzyme that synthesizes telomeric DNA. The reactivation of telomerase activity by aberrant upregulation/expression of its catalytic subunit hTERT is a major pathway in human tumorigenesis. However, regulatory mechanisms that control hTERT expression are largely unknown. Previously, we and others have demonstrated that the introduction of human chromosome 3, via microcell-mediated chromosome transfer (MMCT), repressed transcription of the hTERT gene. These results suggested that human chromosome 3 contains a regulatory factor(s) involved in the repression of hTERT. To further localize this putative hTERT repressor(s), we have developed a unique experimental approach by introducing various truncated chromosome 3 regions produced by a novel chromosomal engineering technology into the renal cell carcinoma cell line (RCC23 cells). These cells autonomously express ectopic hTERT (exohTERT) promoted by a retroviral LTR promoter in order to permit cellular division after repression of endogenous hTERT. We found a telomerase repressor region located within a 7-Mb interval on chromosome 3p21.3. These results provide important information regarding hTERT regulation and a unique method to identify hTERT repressor elements.

5.
Biochem Biophys Res Commun ; 398(4): 695-701, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20621064

ABSTRACT

Telomerase activation is crucial for cells that tend to be immortalized. Increased telomerase activity is correlated with upregulation of telomerase reverse transcriptase (TERT) expression. In most human somatic cells, hTERT expression is suppressed by multiple factors. We have previously shown that human chromosome 5 carries a possible suppressor of mouse tert mtert expression in a mouse melanoma cell line, B16-F10 cells. However, the function of the transcriptional regulator of TERT on this chromosome remains unclear. To examine the functional role of a putative hTERT regulator(s) on this chromosome, we transferred human chromosome 5 in a human melanoma cell line, A2058 cells by microcell-mediated chromosome transfer (MMCT). Microcell hybrid clones with an introduced chromosome 5, but not chromosome 10, showed a remarkable decrease in the growth rate with an obvious cellular morphological alteration and eventually cellular senescence. Moreover, this phenomenon was accompanied by a reduction of hTERT expression and telomerase activity. Most importantly, we found that transcriptional suppression of hTERT by the introduction of chromosome 5 is largely mediated by regulating hTERT promoter activity. Furthermore, the hTERT promoter region between -1623 and -1047 was responsible for this function. These results provide evidence that transcriptional regulator(s) of the hTERT is carried on human chromosome 5 as an endogenous mechanism of hTERT suppression.


Subject(s)
Chromosomes, Human, Pair 5/genetics , Gene Expression Regulation, Enzymologic , Repressor Proteins/genetics , Telomerase/genetics , Animals , Cell Line, Tumor , Cellular Senescence/genetics , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...