Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Dalton Trans ; 51(21): 8350-8367, 2022 May 31.
Article in English | MEDLINE | ID: mdl-35583248

ABSTRACT

A series of new chiral PCN pincer Pd(II) complexes 3a-l with aryl-based (phosphine)-(imidazoline) ligands were conveniently synthesized from readily available starting materials with the key step being phosphination/C-H palladation reaction. All of the new complexes were characterized by 1H NMR, 13C{1H} NMR, 31P{1H} NMR, and elemental analysis. In addition, the molecular structures of complexes 3c, 3e, 3i, and 3k have been determined by X-ray single crystal diffraction analysis. The potential of the obtained pincer Pd(II) complexes in catalytic enantioselective hydrophosphination reaction was examined. With a loading of 5 mol% and in the presence of a KOAc base, complex 3e acted as an active and stereoselective catalyst for hydrophosphination of various 2-alkenoylpyridines with diphenylphosphine, providing optically active pyridine-containing phosphine derivatives with structural diversity in excellent yields with moderate to good enantioselectivities (up to >99 yield and 87% ee).

2.
Cell Biosci ; 9: 47, 2019.
Article in English | MEDLINE | ID: mdl-31210926

ABSTRACT

BACKGROUND: Telomere maintenance is an important factor in tumorigenesis. PinX1 is a potent telomerase regulator which also involves in telomerase loading to telomeres. Nucleophosmin (NPM) can partially attenuate PinX1 inhibition of telomerase activity and NPM loading to hTERT requires PinX1. However, the role of the PinX1/NPM interaction in telomerase activity is not fully understood. METHOD: The long-term effects of PinX1 and NPM down-regulation on telomere length were investigated by TRF assay. The localization of the PinX1/NPM association and the NPM/PinX1/hTERT complex formation were examined by immunofluorescence studies. RESULTS: Concurrent long-term down-regulation of PinX1 and NPM led to a substantial decrease in telomere length. The interaction with PinX1 was crucial in NPM localization in the nucleolus during the S phase. PinX1 and NPM associated throughout S phase and the NPM/PinX1/hTERT complex formation peaked during the early-S phase. The PinX1/NPM interaction was shown to localize away from Cajal Bodies at the start of S phase. CONCLUSION: PinX1/NPM interaction is important in telomerase regulation during catalysis. NPM is recruited to hTERT by PinX1 and is required in the proposed telomerase modulating unit to activate telomerase when telomere extension occurs during S phase.

3.
Sci Rep ; 7: 43650, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28255170

ABSTRACT

Telomerase activation and telomere maintenance are critical for cellular immortalization and transformation. PIN2/TERF1-interacting telomerase inhibitor 1 (PinX1) is a telomerase regulator and the aberrant expression of PinX1 causes telomere shortening. Identifying PinX1-interacting proteins is important for understanding telomere maintenance. We found that PinX1 directly interacts with nucleophosmin (NPM), a protein that has been shown to positively correlate with telomerase activity. We further showed that PinX1 acts as a linker in the association between NPM and hTERT, the catalytic subunit of telomerase. Additionally, the recruitment of NPM by PinX1 to the telomerase complex could partially attenuate the PinX1-mediated inhibition on telomerase activity. Taken together, our data reveal a novel mechanism that regulates telomerase activation through the interaction between NPM, PinX1 and the telomerase complex.


Subject(s)
Nuclear Proteins/metabolism , Telomerase/metabolism , Tumor Suppressor Proteins/metabolism , Cell Cycle Proteins , Cell Line , Enzyme Activation , Humans , Multiprotein Complexes , Nuclear Proteins/chemistry , Nucleophosmin , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Telomerase/antagonists & inhibitors , Telomerase/chemistry , Tumor Suppressor Proteins/chemistry
4.
Oncotarget ; 7(43): 69688-69702, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27626169

ABSTRACT

Curcumin has shown promise as a safe and specific anticancer agent. The COP9 signalosome (CSN) component CSN5, a known specific target for curcumin, can control p53 stability by increasing its degradation through ubiquitin system. But the correlation of CSN5-controlled p53 to anticancer therapeutic effect of curcumin is currently unknown. Here we showed that CSN5-controlled p53 was transcriptional inactive and responsible for autophagy in human normal BJ cells and cancer HepG2 cells under curcumin treatment. Of note, CSN5-initiated cellular autophagy by curcumin treatment was abolished in p53-null HCT116p53-/- cancer cells, which could be rescued by reconstitution with wild-type p53 or transcription inactive p53 mutant p53R273H. Furthermore, CSN5-controlled p53 conferred a pro-survival autophagy in diverse cancer cells response to curcumin. Genetic p53 deletion, as well as autophagy pharmacological inhibition by chloroquine, significantly enhanced the therapeutic effect of curcumin on cancer cells in vitro and in vivo, but not normal cells. This study identifies a novel CSN5-controlled p53 in autophagy of human cells. The p53 expression state is a useful biomarker for predicting the anticancer therapeutic effect of curcumin. Therefore, the pharmacologic autophagy manipulation may benefit the ongoing anticancer clinical trials of curcumin.


Subject(s)
Autophagy/physiology , COP9 Signalosome Complex/physiology , Curcumin/pharmacology , Intracellular Signaling Peptides and Proteins/physiology , Neoplasms/drug therapy , Peptide Hydrolases/physiology , Tumor Suppressor Protein p53/physiology , Antineoplastic Agents/pharmacology , COP9 Signalosome Complex/genetics , Cell Survival , HeLa Cells , Hep G2 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Peptide Hydrolases/genetics , RNA, Small Interfering/genetics , Transcription, Genetic
5.
Antioxid Redox Signal ; 22(10): 848-70, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25602171

ABSTRACT

AIMS: Increased fructose consumption predisposes the liver to nonalcoholic fatty liver disease (NAFLD), but the mechanisms are elusive. Thioredoxin-interacting protein (TXNIP) links oxidative stress to NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome activation and this signaling axis may be involved in fructose-induced NAFLD. Here, we explore the role of reactive oxygen species (ROS)-induced TXNIP overexpression in fructose-mediated hepatic NLRP3 inflammasome activation, inflammation, and lipid accumulation. RESULTS: Rats were fed a 10% fructose diet for 8 weeks and treated with allopurinol and quercetin during the last 4 weeks. Five millimolars of fructose-exposed hepatocytes (primary rat hepatocytes, rat hepatic parenchymal cells [RHPCs], HLO2, HepG2) were co-incubated with antioxidants or caspase-1 inhibitor or subjected to TXNIP or NLRP3 siRNA interference. Fructose induced NLRP3 inflammasome activation and pro-inflammatory cytokine secretion, janus-activated kinase 2/signal transducers and activators of transcription 3-mediated inflammatory signaling, and expression alteration of lipid metabolism-related genes in cultured hepatocytes and rat livers. NLRP3 silencing and caspase-1 suppression blocked these effects in primary rat hepatocytes and RHPCs, confirming that inflammasome activation alters hepatocyte lipid metabolism. Hepatocellular ROS and TXNIP were increased in animal and cell models. TXNIP silencing blocked NLRP3 inflammasome activation, inflammation, and lipid metabolism perturbations but not ROS induction in fructose-exposed hepatocytes, whereas antioxidants addition abrogated TXNIP induction and diminished the detrimental effects in fructose-exposed hepatocytes and rat livers. INNOVATION AND CONCLUSIONS: This study provides a novel mechanism for fructose-induced NAFLD pathogenesis by which the ROS-TXNIP pathway mediates hepatocellular NLRP3 inflammasome activation, inflammation and lipid accumulation. Antioxidant-based interventions can inhibit the ROS-TXNIP pathway.


Subject(s)
Carrier Proteins/metabolism , Hepatocytes/metabolism , Inflammasomes/metabolism , Inflammation/metabolism , Lipid Metabolism , Allopurinol/pharmacology , Animals , Antioxidants/pharmacology , Cell Cycle Proteins , Cell Line , Cytokines/metabolism , Enzyme Inhibitors/pharmacology , Fructose/metabolism , Liver/metabolism , Male , NLR Family, Pyrin Domain-Containing 3 Protein , Non-alcoholic Fatty Liver Disease/metabolism , Oxidative Stress/drug effects , Quercetin/pharmacology , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
6.
Oncol Lett ; 6(3): 748-752, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24137404

ABSTRACT

A 27-kDa C-terminal fragment of human telomerase reverse transcriptase, hTERTC27, has previously been reported to inhibit the growth and tumorigenicity of HeLa human cervical cancer cells and U87-MG human glioblastoma multiforme cells. However, the antitumor effects of hTERTC27 in hepatoma and its underlying mechanisms are unclear. In the current study, the therapeutic effect of hTERTC27, mediated by recombinant adenovirus, in hepatocellular carcinoma (HCC) was explored in vitro and in vivo to investigate the possible mechanisms. The results indicated that recombinant adenovirus carrying hTERTC27 (rAdv-hTERTC27) effectively inhibited the growth and induced apoptosis of the Hepa 1-6 HCC cells. Dendritic cells transduced with rAdv-hTERTC27 were highly effective at inducing antigen-specific T cell proliferation and increasing the activated cytotoxicity of T cells against Hepa 1-6 cells. HCC was inhibited significantly when a single dose of 5×107 pfu rAdv-hTERTC27 was administered intravenously. In summary, the results of this study demonstrated that rAdv-hTERTC27 may serve as a reagent for intravenous administration when combined with telomerase-based gene therapy and immunotherapy for cancer.

7.
Regul Toxicol Pharmacol ; 67(1): 53-62, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23827714

ABSTRACT

The safety of rAd5-hTERTC27, a replication defective adenovirus vector carrying hTERTC27 for possible use against hepatocellular carcinoma (HCC) was assessed. In single-dose evaluations, intravenous dose levels of up to 2×10(11)VP/kg in rats and 9×10(10)VP/kg in monkeys were well tolerated with no abnormal changes in general signs, body weight and food consumption, and no significant differences in biochemical parameters, urinalysis, ECG, and systemic necropsy observations between the rAd5 groups and solvent control group except that slight hematological change was observed. No hemolytic effect using rabbit blood, local perivasculitis following intravenous injection in rabbits or systemic anaphylaxis in guinea pigs following intravenous dosing was seen. No effects on the central nervous system of mice occurred following intravenous dosing with the exception of an increase in sleep duration at the dose of 1.2×10(11)VP/kg (p<0.05) but not at lower doses of 2×10(10) and 6×10(10)VP/kg in the hypnotic synergism test. These results demonstrate that administration of rAd5-hTERTC27 was well tolerated in an initial set of safety studies as part of an evaluation to allow human trials for the treatment of HCC.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Telomerase/administration & dosage , Telomerase/genetics , Adenoviridae/genetics , Adenoviridae/physiology , Animals , Drug Evaluation, Preclinical , Female , Guinea Pigs , HEK293 Cells , Haplorhini , Humans , Injections, Intravenous , Male , Mice , Rabbits , Random Allocation , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Sleep/drug effects , Virus Replication
8.
J Pathol ; 229(5): 765-74, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23341363

ABSTRACT

Chemoradiotherapy (CRT) is a standard treatment for oesophageal squamous cell carcinoma (ESCC) in its advanced stages. The telomerase/telomere interacting protein PinX1 contributes to telomere maintenance, tumourigenicity, and influences the DNA damage agent-induced apoptotic response in telomerase-positive cancer cells. However, the clinical and biological significance of PinX1 in human ESCCs remains unclear. We examined the expression dynamics of PinX1 by immunohistochemistry in a learning cohort (n = 98) and a validation cohort (n = 59) of ESCC patients treated with definite chemoradiotherapy (CRT). A series of in vivo and in vitro assays were performed to elucidate the effect of PinX1 on ESCC cells' CRT response and underlying mechanisms. Knockdown of PinX1 did not affect ESCC cells' chemosensitivities to 5-fluorouracil and cisplatin, but substantially increased ESCC cells' therapeutic efficacy of radiation both in vitro and in vivo. Ectopic overexpression of PinX1 dramatically enhanced ESCC cells' resistance to radiotherapy. Furthermore, we demonstrated that PinX1 resistance to radiotherapy (RT) was attributed to PinX1 maintaining telomere stability, reducing ESCC cell death by RT-induced mitosis catastrophe (MC). High expression of Pinx1 correlated positively with ESCC's resistance to CRT, and was a strong and independent predictor for short disease-specific survival (DSS) of ESCC patients. Our data suggest that PinX1 could serve as a novel predictor for a CRT response to ESCC patients, and the pathway of PinX1-mediated telomere stability might represent a new target to improve the RT effect of ESCC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/therapy , Chemoradiotherapy , Esophageal Neoplasms/therapy , Tumor Suppressor Proteins/metabolism , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Cell Cycle Proteins , Cell Line, Tumor , Cisplatin/administration & dosage , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Resistance, Neoplasm , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Female , Fluorouracil/administration & dosage , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Middle Aged , Mitosis/drug effects , Mitosis/radiation effects , RNA Interference , Survival Analysis , Telomerase/metabolism , Time Factors , Transfection , Treatment Outcome , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
9.
FEBS Lett ; 586(19): 3166-71, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22749911

ABSTRACT

Telomerase recruitment to telomere is the prerequisite for telomere extension, but the proteins involved in this process are still largely unknown. PinX1 is a telomerase inhibitor and has been implicated in telomere maintenance. Silencing of PinX1 significantly reduced the localization of telomerase to telomere during mid-late S phase, suggesting the involvement of PinX1 in the cell cycle-dependent trafficking of hTERT to telomere. We also revealed that PinX1 mediated the chromosomal localization of hTERT during anaphase. This study revealed the role of PinX1 in telomerase function regulation by mediating its localization inside cells.


Subject(s)
Telomerase/metabolism , Tumor Suppressor Proteins/metabolism , Cell Cycle , Cell Cycle Proteins , Gene Silencing , HEK293 Cells , HeLa Cells , Humans , RNA, Small Interfering/genetics , S Phase , Signal Transduction , Telomere/metabolism , Telomeric Repeat Binding Protein 2/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
10.
Oncol Rep ; 27(4): 1163-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22210010

ABSTRACT

hTERTC27, a 27-kDa hTERT C-terminal polypeptide has been demonstrated to cause hTERT-positive HeLa cell apoptosis and inhibits the growth of mouse melanoma. hTERTC27 has been associated with telomere dysfunction, regulation of gene-regulated apoptosis, the cell cycle and activation of natural killer (NK) cells, but its mechanism of action is not fully understood. Here, we report that dendritic cells (DCs) transduced with hTERTC27 can increase T-cell proliferation, and augment the concentration of interleukin-2 (IL-2) and interferon-γ (IFN-γ) in the supernatants of T cells. It can also induce antigen-specific cytotoxic T lymphocytes (CTL) against glioma cells in vitro. Moreover, hTERTC27 gene-transduced DCs exhibit a very potent cytotoxicity to glioma cells in vivo. It could prolong the survival time and inhibit the growth of glioma-bearing mice. These data suggest that hTERTC27 gene-transduced DCs can efficiently enhance immunity against gliomas in vitro and in vivo.


Subject(s)
Adenoviridae/enzymology , Brain Neoplasms/therapy , Dendritic Cells/transplantation , Genetic Therapy/methods , Genetic Vectors , Glioma/therapy , Peptide Fragments/metabolism , Telomerase/metabolism , Transduction, Genetic , Adaptive Immunity , Adenoviridae/genetics , Animals , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Dendritic Cells/enzymology , Dendritic Cells/immunology , Female , Glioma/enzymology , Glioma/genetics , Glioma/immunology , Glioma/pathology , Humans , Interferon-gamma/metabolism , Interleukin-2/metabolism , Mice , Mice, Inbred C57BL , Peptide Fragments/genetics , Phenotype , T-Lymphocytes/immunology , T-Lymphocytes, Cytotoxic/immunology , Telomerase/genetics , Time Factors , Tumor Burden
11.
Cancer Res ; 69(1): 75-83, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19117989

ABSTRACT

The nucleolar protein PinX1 has been proposed to be a putative tumor suppressor due to its binding to and inhibition of the catalytic activity of telomerase, an enzyme that is highly expressed in most human cancers in which it counteracts telomere shortening-induced senescence to confer cancer cell immortalization. However, the role of PinX1 in telomere regulation, as well as in cancer, is still poorly understood. In this study, we showed that the PinX1 protein is constitutively expressed in various human cells regardless of their telomerase activity and malignant status. Most interestingly, we found that silencing PinX1 expression by a potent short hairpin RNA construct led to a robust telomere length shortening and growth inhibition in telomerase-positive but not in telomerase-negative human cancer cells. We further showed that silencing PinX1 significantly reduced the endogenous association of telomerase with the Pot1-containing telomeric protein complex, and therefore, could account for the phenotypic telomere shortening in the affected telomerase-positive cancer cells. Our results thus reveal a novel positive role for PinX1 in telomerase/telomere regulations and suggest that the constitutive expression of PinX1 attributes to telomere maintenance by telomerase and tumorigenicity in cancer cells.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Telomerase/metabolism , Telomere/metabolism , Tumor Suppressor Proteins/deficiency , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Proteins , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , DNA Damage , Etoposide/pharmacology , Gene Silencing , Humans , Mice , Mice, Inbred BALB C , RNA, Small Interfering/genetics , Shelterin Complex , Telomerase/biosynthesis , Telomere/genetics , Telomere-Binding Proteins/metabolism , Transfection , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics
12.
J Cell Sci ; 121(Pt 13): 2169-76, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18522991

ABSTRACT

Telomerase maintains telomere length and has been implicated in both aging and carcinogenesis of human cells. This enzyme is a specialized ribonucleoprotein (RNP) complex, minimally consisting of two essential components: the protein catalytic subunit TERT (telomerase reverse transcriptase) and the integral RNA moiety TR (telomerase RNA, TERC). Both TERT and TR have been found to localize to nucleoli within the nucleus, leading to the suggestion of nucleoli as the site for telomerase RNP biogenesis in human cells. However, whether this statement is true or not has not yet been convincingly demonstrated. Here, we identify that residues 965-981 of the human TERT polypeptide constitute an active nucleolar-targeting signal (NTS) essential for mediating human TERT nucleolar localization. Mutational inactivation of this NTS completely disrupted TERT nucleolar translocation in both normal and malignant human cells. Most interestingly, such a TERT mutant still retained the capacity to activate telomerase activity, maintain telomere length and extend the life-span of cellular proliferation, as does wild-type TERT, in BJ cells (normal fibroblasts). Therefore, our data suggest that TERT nucleolar localization is unrelated to telomerase function in human cells.


Subject(s)
Telomerase/genetics , Telomerase/metabolism , Amino Acid Sequence , Animals , Catalytic Domain , Cell Line , Cell Nucleolus/metabolism , Humans , Mice , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , RNA/genetics , RNA/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Sequence Homology, Amino Acid , Telomere/genetics , Telomere/metabolism
13.
Cell Mol Immunol ; 3(1): 1-11, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16549043

ABSTRACT

The human telomerase reverse transcriptase (hTERT) is expressed in more than 85% of tumor cells but is usually not found in normal cells, which makes hTERT as an ideal tumor-associate antigen (TAA) to develop potential vaccine specifically destroying cancers without impairing normal tissues in human cancer immunotherapy. Here are reviewed the fundamental advances of studies on immunogenicity of hTERT or its peptides and the early clinical trials using the hTERT vaccine approach in the last decades.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , DNA-Binding Proteins/immunology , Immunotherapy , Neoplasms/therapy , Peptides/immunology , Telomerase/immunology , Antigens, Neoplasm/metabolism , Cancer Vaccines/therapeutic use , Clinical Trials as Topic , DNA-Binding Proteins/metabolism , Humans , Neoplasms/immunology , Peptides/therapeutic use , Telomerase/metabolism
14.
BJU Int ; 96(6): 890-4, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16153224

ABSTRACT

OBJECTIVE: To construct a mutant enhanced green fluorescence protein (pEGFP) human telomerase reverse transcriptase (hTERT) gene expression vector (pEGFP-hTERT), to observe its expression in transfected human bladder cancer cell line T24 and its role in the molecular regulatory mechanisms of telomerase, and to provide a new target gene for bladder cancer therapy. MATERIALS AND METHODS: Polymerase chain reaction (PCR) amplification was performed using primers based on the gene sequence of hTERT. The PCR product was cloned into plasmid pGEMT-T Easy and the sequence of mutant hTERT gene analysed. A recombinant mutant hTERT vector (pEGFP-hTERT) was constructed at the EcoR I and Sal I sites of the pEGFP-C1 vector. After transfecting the fusion gene into T24 cells by the method of calcium phosphate-DNA co-precipitation, we detected steady expression of the GFP-hTERT fusion protein by fluorescent-light microscopy. Changes in the proliferation of T24 cells were detected by light microscopy, and beta-galactosidase staining correlated with senescence. RESULTS: Identification of pEGFP-hTERT by enzyme digestion showed that the mutant hTERT fragment had been cloned into EcoR I and Sal I sites of the pEGFP-C1 vector. Steady expression of GFP-hTERT fusion protein was located in the nucleus of transfected cells. Positive expression senescence-associated beta-galactosidase staining in transfected cells increased gradually with extended cultured time, and their growth was suppressed. CONCLUSION: The recombinant mutant vector (pEGFP-hTERT) was successfully constructed and expressed steadily in T24 cells. The mutant-type hTERT gene suppresses the proliferation of T24 cells by a competitive effect on telomerase activity. This suggests that the hTERT gene might be a suitable gene target for bladder cancer therapy.


Subject(s)
DNA-Binding Proteins/genetics , Telomerase/genetics , Urinary Bladder Neoplasms/therapy , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Genetic Therapy/methods , Genetic Vectors , Green Fluorescent Proteins/metabolism , Humans , Polymerase Chain Reaction/methods , Recombinant Proteins , Transfection , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism
16.
Cancer Res ; 62(11): 3226-32, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12036938

ABSTRACT

The COOH-terminus of telomerase reverse transcriptase (hTERT) has been shown to participatein the nuclear translocation of TERT. Here, we constructed plasmids expressing the COOH-terminal M(r) 27,000 polypeptide of hTERT (hTERTC27) withthe telomerase RNA-binding domains and the reverse transcriptase domains deleted. We showed that ectopic overexpression of this polypeptide caused a defect in telomere maintenance in hTERT-positive HeLa cells, which led to senescence-like growth arrest and apoptosis. The hTERTC27 appears to work by inducing telomere dysfunction, exemplified by significantly increased anaphase chromosome end-to-end fusion events in transfected cells. Significantly, it had no effect on the cellular telomerase enzymatic activity or telomere length. The in vivo effect was further demonstrated as HeLa cells stably expressing hTERTC27 have significantly lower growth rate and reduced tumorigenicity in nude mice xenografts. Results from this study revealed an important function for the COOH terminus of hTERT in maintaining the integrity of telomere structure and chromosome ends, as well as in cell senescence and apoptosis. Furthermore, hTERTC27 provides a new strategy for cancer therapy by inducing telomere dysfunction in cancer cells without affecting the telomerase enzymatic activity.


Subject(s)
Peptide Fragments/physiology , Telomerase/physiology , Telomere/physiology , Animals , Apoptosis/physiology , Cell Division/physiology , DNA-Binding Proteins , HeLa Cells , Humans , Mice , Mice, Nude , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Telomerase/biosynthesis , Telomerase/genetics , Telomerase/metabolism , Transfection , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...