Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 789
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nucleic Acids Res ; 49(13): 7680-7694, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34181710

ABSTRACT

The enormous sequence heterogeneity of telomerase RNA (TR) subunits has thus far complicated their characterization in a wider phylogenetic range. Our recent finding that land plant TRs are, similarly to known ciliate TRs, transcribed by RNA polymerase III and under the control of the type-3 promoter, allowed us to design a novel strategy to characterize TRs in early diverging Viridiplantae taxa, as well as in ciliates and other Diaphoretickes lineages. Starting with the characterization of the upstream sequence element of the type 3 promoter that is conserved in a number of small nuclear RNAs, and the expected minimum TR template region as search features, we identified candidate TRs in selected Diaphoretickes genomes. Homologous TRs were then used to build covariance models to identify TRs in more distant species. Transcripts of the identified TRs were confirmed by transcriptomic data, RT-PCR and Northern hybridization. A templating role for one of our candidates was validated in Physcomitrium patens. Analysis of secondary structure demonstrated a deep conservation of motifs (pseudoknot and template boundary element) observed in all published TRs. These results elucidate the evolution of the earliest eukaryotic TRs, linking the common origin of TRs across Diaphoretickes, and underlying evolutionary transitions in telomere repeats.


Subject(s)
Evolution, Molecular , RNA, Plant/chemistry , RNA, Plant/genetics , RNA/chemistry , RNA/genetics , Telomerase/chemistry , Telomerase/genetics , Mutation , Nucleic Acid Conformation , RNA/biosynthesis , RNA Polymerase II/metabolism , RNA Polymerase III/metabolism , RNA, Plant/biosynthesis , Sequence Alignment , Telomerase/biosynthesis , Telomere/chemistry , Transcription, Genetic , Transcriptome , Viridiplantae/genetics
2.
Genes Dev ; 29(11): 1095-105, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-26063571

ABSTRACT

Telomerase is the ribonucleoprotein enzyme that catalyzes the extension of telomeric DNA in eukaryotes. Recent work has begun to reveal key aspects of the assembly of the human telomerase complex, its intracellular trafficking involving Cajal bodies, and its recruitment to telomeres. Once telomerase has been recruited to the telomere, it appears to undergo a separate activation step, which may include an increase in its repeat addition processivity. This review covers human telomerase biogenesis, trafficking, and activation, comparing key aspects with the analogous events in other species.


Subject(s)
Telomerase/physiology , Enzyme Activation , Humans , Protein Multimerization , Protein Transport , Telomerase/biosynthesis , Telomerase/metabolism
3.
J Biol Chem ; 295(29): 10062-10075, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32518154

ABSTRACT

The human telomerase gene (hTERT) is repressed in most somatic cells. How transcription factors activate the hTERT promoter in its repressive chromatin environment is unknown. Here, we report that the ETS family protein ETS variant transcription factor 5 (ETV5) mediates epidermal growth factor (EGF)-induced hTERT expression in MCF10A cells. This activation required MYC proto-oncogene bHLH transcription factor (c-Myc) and depended on the chromatin state of the hTERT promoter. Using chromatinized bacterial artificial chromosome (BAC) reporters in human fibroblasts, we found that ETV5 and c-Myc/MYC-associated factor X (MAX) synergistically activate the hTERT promoter via two identical, but inverted, composite Ets/E-box motifs enclosing the core promoter. Mutations of Ets or E-box sites in either DNA motif abolished the activation and reduced or eliminated the synergism. ETV5 and c-Myc facilitated each other's binding to the hTERT promoter. ETV5 bound to the hTERT promoter in both telomerase-negative and -positive cells, but it activated the repressed hTERT promoter and altered histone modifications only in telomerase-negative cells. The synergistic ETV5/c-Myc activation disappeared when hTERT promoter repression became relieved because of the loss of distal regulatory elements in chimeric human/mouse BAC reporters. Our results suggest that the binding of c-Myc and ETS family proteins to the Ets/E-box motifs derepresses the hTERT promoter by inducing an active promoter configuration, providing a mechanistic insight into hTERT activation during tumorigenesis.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic , Proto-Oncogene Proteins c-myc/metabolism , Response Elements , Telomerase/biosynthesis , Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Line, Tumor , Cells, Cultured , DNA-Binding Proteins/genetics , Humans , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/genetics , Telomerase/genetics , Transcription Factors/genetics
4.
J Immunol ; 200(4): 1283-1294, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29305433

ABSTRACT

Airway smooth muscle (ASM) cells contribute to asthmatic lung pathology with chemokine hypersecretion and increased ASM cell mass. With little recent progress in the development of asthma therapies, a greater understanding of lung inflammation mechanisms has become a priority. Chemokine gene expression in ASM cells is dependent upon NF-κB transcription factor activity. The telomerase/shelterin complex maintains chromosomal telomere ends during cell division. Telomerase is a possible cofactor for NF-κB activity, but its role in NF-κB activity in airway tissue inflammation is not known. In this study, we sought to address two key questions: whether telomerase is involved in inflammation in ASM cells, and whether components of the shelterin complex are also required for an inflammatory response in ASM cells. Telomerase inhibitors and telomerase small interfering RNA (siRNA) reduced TNF-α-induced chemokine expression in ASM cells. Telomerase siRNA and inhibitors reduced NF-κB activity. An siRNA screen of shelterin components identified a requirement for PIN2/TERF1 interacting-telomerase inhibitor 1 (PINX1) in chemokine gene expression. High-level PINX1 overexpression reduced NF-κB reporter activity, but low-level expression amplified NF-κB activity. Coimmunoprecipitation studies showed association of PINX1 and p65. Overexpression of the N terminus (2-252 aa) of PINX1, but not the C-terminal telomerase-inhibitor domain (253-328 aa), amplified TNF-α-induced NF-κB activity. GST pull-downs demonstrated that the N terminus of PINX1 bound more p65 than the C-terminal telomerase-inhibitor domain; these observations were confirmed in whole cells with N-terminal and C-terminal PINX1 immunoprecipitation. We conclude that telomerase and PINX1 are required for chemokine expression in ASM cells and represent significant new targets for future anti-inflammatory therapies for lung diseases, such as asthma.


Subject(s)
Chemokines/biosynthesis , Gene Expression Regulation/physiology , Myocytes, Smooth Muscle/immunology , Telomerase/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Cell Cycle Proteins , Cell Line , Humans , Inflammation/immunology , Inflammation/metabolism , Lung/immunology , Lung/metabolism , Myocytes, Smooth Muscle/metabolism , Telomerase/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Suppressor Proteins/immunology
5.
Mol Biol Rep ; 47(10): 7735-7743, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32959195

ABSTRACT

Telomerase is a cancer promoting ribonucleoprotein complex and is a potential therapeutic target for cancer. In this study, the effects of telomerase downregulation on the whole cell proteome were investigated. Understanding how the effect of downregulation on the whole proteome profile will generate a greater understanding of the possible roles played by telomerase in cancer. Downregulation was achieved by RNA interference (RNAi), targeting the telomerase reverse transcriptase (TERT) subunits of telomerase. Transfection of TERT siRNA downregulates TERT gene expression and induced downregulation of telomerase activity. Investigation of the effect of silencing TERT in telomerase was further validated through proteomic analysis by performing 2-dimension electrophoresis (2DE) coupled with MALDI-TOF/TOF. 12 protein spots in HeLa cells were reported to be significantly differentially expressed with 11 of them were upregulated and 1 downregulated. Through STRING analysis, differentially expressed proteins demonstrated strong associations with endoplasmic reticulum stress marker and mitochondrial energy production marker. In conclusions, the result exhibited novel integrated proteomic response involving endoplasmic reticulum stress and mitochondrial energy production in response to the TERT downregulation in cervical cancer cells.


Subject(s)
Down-Regulation , Endoplasmic Reticulum Stress , Energy Metabolism , Mitochondria/metabolism , RNA Interference , Telomerase/biosynthesis , HeLa Cells , Humans , Telomerase/genetics
6.
Neurosurg Rev ; 43(3): 903-910, 2020 Jun.
Article in English | MEDLINE | ID: mdl-30788677

ABSTRACT

Telomerase activity and (human) Telomerase Reverse Transcriptase (hTERT) expression are considered hallmarks in oncogenesis of neoplasms and are upregulated by alterations of the hTERT promoter. In meningiomas, numerous studies investigated hTERT expression, telomerase activity, promoter mutations, and methylations. Moreover, reports about hTERT-targeted chemotherapy in meningiomas have recently been published. We provide a systematic review of the literature about the role of hTERT in meningiomas. TERT expression and telomerase activity is found in benign and high-grade meningiomas and increase with WHO grade. Remarkably, rates of TERT expression/telomerase activity usually exceed mutation frequency and both telomerase activity and TERT expression have also been found in hTERT promoter wildtype meningiomas, indicating further mechanisms of TERT upregulation. Although hTERT promoter methylation has been reported in the vast majority of meningiomas, correlation with TERT expression remains controversial. Rates of promoter mutations, and methylation were shown to increase with rising WHO grade. Moreover, promoter methylation and mutations strongly correlate with prognosis. Although mutations predicted malignant progression, de novo mutations in high-grade recurrences of former benign lesions were also observed. Retroviral transduction of the TERT gene enabled immortalization in several grade I-III meningioma cell lines. In vitro analyses revealed significant effects on viability in hTERT-mutated meningioma cells after targeted treatment. Alternative mechanisms of telomere lengthening are usually absent in meningiomas. TERT and hTERT promoter alterations play a major role during oncogenesis of meningiomas with implications for prognosis and potentially treatment.


Subject(s)
Brain Neoplasms/genetics , Meningioma/genetics , Telomerase/genetics , Telomerase/metabolism , Telomere Homeostasis/genetics , Telomere/genetics , Brain Neoplasms/ultrastructure , Humans , Meningioma/ultrastructure , Telomerase/biosynthesis , Telomere/ultrastructure
7.
J Cell Biochem ; 120(3): 4444-4450, 2019 03.
Article in English | MEDLINE | ID: mdl-30260020

ABSTRACT

OBJECTIVE: The current study was designed to investigate the inhibitory effects of ginsenoside Rd (Gs-Rd) on human glioma U251 cells in vitro and its possible underlying mechanisms. METHODS: The groups included blank control group, low concentration Gs-Rd treatment group (20 µM), mid concentration Gs-Rd treatment group (40 µM), and high concentration Gs-Rd treatment group (80 µM). The proliferative activity of human glioma U251 cells was detected by the MTT assay. Flow cytometry was performed to measure cell apoptosis of human glioma U251 cells. In addition, the ELISA assay was used to measure the telomerase activities in different groups on 24 hours, 48 hours, and 72 hours. Furthermore, real-time quantitative polymerase chain reaction (RT-PCR) and Western blot analysis were performed to measure the expression of Bcl-2, human telomerase catalytic subunit (hTERT), and caspase-3 in different groups on 48 hours at both messenger RNA (mRNA) and protein levels. RESULTS: The proliferation of U251 cells was inhibited by Gs-Rd with different concentrations in the dose- and time-dependent manners. In addition, Gs-Rd promoted U251 cell apoptosis rate in a dose-dependent manner. Gs-Rd with different concentrations (20 µM, 40 µM, and 80 µM) significantly enhanced the expression of teleomerase on 24 hours and 48 hours. In addition, Gs-Rd with different concentrations significantly increased caspase-3 and decreased Bcl-2 and hTERT expressions at both mRNA and protein levels. CONCLUSION: The Gs-Rd can remarkably inhibit the proliferation and promote cell apoptosis of human glioma U251 cells. The possible underlying mechanisms could be related to inhibiting telomerase activity, downregulating expression of Bcl-2 and hTERT, and upregulating expression of caspase-3 of human glioma U251 cells.


Subject(s)
Caspase 3/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Ginsenosides/pharmacology , Glioma , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Telomerase/biosynthesis , Cell Line, Tumor , Glioma/drug therapy , Glioma/metabolism , Glioma/pathology , Humans
8.
PLoS Pathog ; 13(10): e1006681, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29045464

ABSTRACT

HIV1-TAT interactive protein (TIP60) is a haploinsufficient tumor suppressor. However, the potential mechanisms endowing its tumor suppressor ability remain incompletely understood. It plays a vital role in virus-induced cancers where TIP60 down-regulates the expression of human papillomavirus (HPV) oncoprotein E6 which in turn destabilizes TIP60. This intrigued us to identify the role of TIP60, in the context of a viral infection, where it is targeted by oncoproteins. Through an array of molecular biology techniques such as Chromatin immunoprecipitation, expression analysis and mass spectrometry, we establish the hitherto unknown role of TIP60 in repressing the expression of the catalytic subunit of the human telomerase complex, TERT, a key driver for immortalization. TIP60 acetylates Sp1 at K639, thus inhibiting Sp1 binding to the TERT promoter. We identified that TIP60-mediated growth suppression of HPV-induced cervical cancer is mediated in part due to TERT repression through Sp1 acetylation. In summary, our study has identified a novel substrate for TIP60 catalytic activity and a unique repressive mechanism acting at the TERT promoter in virus-induced malignancies.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Histone Acetyltransferases/metabolism , Neoplasm Proteins/metabolism , Response Elements , Sp1 Transcription Factor/metabolism , Telomerase/biosynthesis , Uterine Cervical Neoplasms/metabolism , Female , HeLa Cells , Histone Acetyltransferases/genetics , Humans , Lysine Acetyltransferase 5 , Neoplasm Proteins/genetics , Sp1 Transcription Factor/genetics , Telomerase/genetics , Uterine Cervical Neoplasms/genetics
9.
Anal Biochem ; 583: 113361, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31306622

ABSTRACT

Phage N15 protelomerase (TelN) cleaves double-stranded circular DNA containing a telomerase-occupancy-site (tos) and rejoins the resulting linear-ends to form closed-hairpin-telomeres in Escherichia coli (E. coli). Continued TelN expression is essential to support resolution of the linear structure. In mammalian cells, no enzyme with TelN-like activities has been found. In this work, we show that phage TelN, expressed transiently and stably in human and mouse cells, recapitulates its native activities in these exogenous environments. We found TelN to accurately resolve tos-DNA in vitro and in vivo within human and mouse cells into linear DNA-containing terminal telomeres that are resistant to RecBCD degradation, a hallmark of protelomerase processing. In stable cells, TelN activity was detectable for at least 60 days, which suggests the possibility of limited silencing of its expression. Correspondingly, linear plasmid containing a 100 kb human ß-globin gene expressed for at least 120 h in non-ß-globin-expressing mouse cells with TelN presence. Our results demonstrate TelN is able to cut and heal DNA as hairpin-telomeres within mammalian cells, providing a tool for creating novel structures by DNA resolution in these hosts. The TelN protelomerase may be useful for exploring novel technologies for genome interrogation and chromosome engineering.


Subject(s)
DNA Replication/physiology , DNA/metabolism , Enzyme Precursors , Telomerase , Viral Proteins , beta-Globins/genetics , Animals , Enzyme Precursors/biosynthesis , Enzyme Precursors/physiology , Escherichia coli , Genetic Engineering/methods , HeLa Cells , Humans , Mice , NIH 3T3 Cells , Telomerase/biosynthesis , Telomerase/physiology , Viral Proteins/biosynthesis , Viral Proteins/physiology
10.
Med J Malaysia ; 74(6): 504-508, 2019 12.
Article in English | MEDLINE | ID: mdl-31929476

ABSTRACT

INTRODUCTION: Choriocarcinoma is malignant cancer originating from placental trophoblast. The incidence of this cancer is estimated at 0.57-1.1 per 1000 births in the United States of America, Australia, Europe, and New Zealand. The rate is much higher in South East Asia and Japan with two occurrences per a thousand births. Telomerase activity is an important part of the apoptotic process. Increased telomerase activity will result in cellular immortality and poor prognosis in cancer. Vitamin A possess an essential role in cell proliferation and differentiation. One of the active metabolites of vitamin A is All-Trans Retinoic Acid (ATRA). METHODS: In this study, we examined the role of ATRA against telomerase activity in choriocarcinoma cell. This cell was derived from BeWo cell line (ATCC CCL-98) and were given different doses of ATRA. RESULTS: From this study, Choriocarcinoma cell that was given ATRA in dosage of 50µg/ml inhibit telomerase activity by extending the cycle time of 39.51±0.09, compared to the control group with a cycle time of 37.62±0.43. Cycle length change consistently with higher dose of ATRA. CONCLUSION: This study has proven that ATRA could inhibit telomerase activity by lengthening the cycle. Changes in the increase of ATRA doses in this experimental test need to be studied further on experimental animals, either administered as a single agent or as an addition to standard treatment of trophoblastic disease.


Subject(s)
Choriocarcinoma/drug therapy , Telomerase/antagonists & inhibitors , Tretinoin/administration & dosage , Uterine Neoplasms/drug therapy , Antineoplastic Agents/administration & dosage , Biomarkers, Tumor/biosynthesis , Cell Differentiation/drug effects , Choriocarcinoma/metabolism , Choriocarcinoma/pathology , Dose-Response Relationship, Drug , Female , Humans , Pregnancy , Telomerase/biosynthesis , Tumor Cells, Cultured , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology
11.
Dokl Biochem Biophys ; 485(1): 150-152, 2019 Mar.
Article in English | MEDLINE | ID: mdl-31201638

ABSTRACT

A hybrid 6XRE-hTERT promoter consisting of the hTERT tumor-specific promoter and six copies of the XRE element from the CYP1A1 human gene promoter was created. Using a human lung cancer cells as a model, we showed that XRE elements in the hybrid promoter greatly increase the activity of the hTERT promoter and ensure the reporter gene transcriptional activation in response to the treatment of the cells with the AhR ligand benzo(a)pyrene. However, similar effects were also observed in normal human bronchial epithelial cells HBEpC, which indicates the loss of the tumor-specific activity by the 6XRE-hTERT hybrid promoter. XRE elements can be used for nonspecific transcription enhancement but are unsuitable for the creation of tumor-specific promoters with enhanced activity.


Subject(s)
Cytochrome P-450 CYP1A1 , Response Elements , Telomerase , Transcriptional Activation/drug effects , A549 Cells , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Benzo(a)pyrene/toxicity , Bronchi/metabolism , Bronchi/pathology , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Telomerase/biosynthesis , Telomerase/genetics
12.
J Cell Physiol ; 233(12): 9763-9776, 2018 12.
Article in English | MEDLINE | ID: mdl-30078190

ABSTRACT

Primary porcine bronchial epithelial cells (PBECs) are an ideal model to study the molecular and pathogenic mechanisms of various porcine respiratory pathogens. However, the short lifespan of primary PBECs greatly limit their application. Here, we isolated and cultured primary PBECs and established immortalized PBECs by transfecting primary PBECs with the pEGFP-hTERT recombinant plasmid containing human telomerase reverse transcriptase (hTERT). Immortalized PBECs (hTERT-PBECs) retained the morphological and functional features of primary PBECs as indicated by cytokeratin 18 expression, telomerase activity assay, proliferation assays, karyotype analysis, and quantitative reverse-transcriptase polymerase chain reaction. Compared to primary PBECs, hTERT-PBECs had higher telomerase activity, extended replicative lifespan, and displayed enhanced proliferative activity. Moreover, this cell line is not transformed in vitro and does not exhibit a malignant phenotype in vivo, suggesting that it can be safely used in further studies. Besides, hTERT-PBECs were susceptible to swine influenza virus of H3N2 subtype and porcine circovirus type 2. In conclusion, the immortalized hTERT-PBECs represent a valuable in vitro model, which can be widely used in the study of porcine respiratory pathogenic infections.


Subject(s)
Bronchi/cytology , Epithelial Cells/enzymology , Primary Cell Culture/methods , Telomerase/genetics , Animals , Bronchi/enzymology , Cell Proliferation/genetics , Circovirus/pathogenicity , Humans , Karyotype , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/virology , Swine , Telomerase/biosynthesis
13.
Int J Cancer ; 142(10): 2028-2039, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29266240

ABSTRACT

Papillary thyroid carcinoma (PTC) is the most common type of thyroid cancer. Tumor recurrence occurs in ∼20% of PTCs and some reach advanced stages. Promoter mutation in the telomerase reverse transcriptase (TERT) gene is identified to be a prognostic marker in PTC. However, the contribution of TERT promoter mutation to cancer progression in PTC patients is still not fully understood. In this study, we investigated the incidence of TERT promoter mutations and TERT protein expression and their association with clinicopathological outcomes in a large cohort of PTC samples using direct sequencing technology and immunohistochemistry. Furthermore, two PTC cell lines were utilized to investigate role of TERT mutations in mediating metastasis. Two promoter hotspot mutations C228T and C250T were identified in 18.0% (167/927) of our cohort and were significantly associated with poor 5 years disease-free survival and distant metastasis of PTC. TERT protein overexpression was noted in 20.1% of our PTC cohort and was significantly associated with poor prognostic markers such as older age, extrathyroidal extension and Stage IV tumors. A significant association was also found between TERT overexpression and epithelial-mesenchymal transition (EMT) markers. Functional analysis showed that TERT inhibition reduced cell growth, invasion, migration and angiogenesis in PTC via suppression of EMT in PTC cells. Our results suggest that TERT promoter mutation is an independent predictor of disease-free survival and might drive the metastasis, and downregulation of TERT could potentiate antitumor and antimetastatic activities in PTC.


Subject(s)
Telomerase/genetics , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/mortality , Thyroid Neoplasms/genetics , Thyroid Neoplasms/mortality , Adult , Aminobenzoates/pharmacology , Animals , Cohort Studies , Disease-Free Survival , Down-Regulation , Epithelial-Mesenchymal Transition , Female , Genetic Testing , Heterografts , Humans , Male , Mice , Mice, Nude , Middle Aged , Mutation , Naphthalenes/pharmacology , Neoplasm Metastasis , Promoter Regions, Genetic , Saudi Arabia/epidemiology , Telomerase/antagonists & inhibitors , Telomerase/biosynthesis , Thyroid Cancer, Papillary/enzymology , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/pathology
14.
Blood ; 127(14): 1770-9, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26903545

ABSTRACT

Aplastic anemia is a fatal bone marrow disorder characterized by peripheral pancytopenia and marrow hypoplasia. The disease can be hereditary or acquired and develops at any stage of life. A subgroup of the inherited form is caused by replicative impairment of hematopoietic stem and progenitor cells due to very short telomeres as a result of mutations in telomerase and other telomere components. Abnormal telomere shortening is also described in cases of acquired aplastic anemia, most likely secondary to increased turnover of bone marrow stem and progenitor cells. Here, we test the therapeutic efficacy of telomerase activation by using adeno-associated virus (AAV)9 gene therapy vectors carrying the telomerase Tert gene in 2 independent mouse models of aplastic anemia due to short telomeres (Trf1- and Tert-deficient mice). We find that a high dose of AAV9-Tert targets the bone marrow compartment, including hematopoietic stem cells. AAV9-Tert treatment after telomere attrition in bone marrow cells rescues aplastic anemia and mouse survival compared with mice treated with the empty vector. Improved survival is associated with a significant increase in telomere length in peripheral blood and bone marrow cells, as well as improved blood counts. These findings indicate that telomerase gene therapy represents a novel therapeutic strategy to treat aplastic anemia provoked or associated with short telomeres.


Subject(s)
Anemia, Aplastic/therapy , Dependovirus , Genetic Therapy/methods , Telomerase/biosynthesis , Telomere Homeostasis , Telomere/metabolism , Transduction, Genetic , Anemia, Aplastic/genetics , Anemia, Aplastic/metabolism , Anemia, Aplastic/pathology , Animals , Disease Models, Animal , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Mice , Mice, Knockout , Telomerase/genetics , Telomere/genetics
15.
Blood ; 127(21): 2587-97, 2016 05 26.
Article in English | MEDLINE | ID: mdl-26907633

ABSTRACT

Multiple myeloma (MM) cell lines and primary tumor cells are addicted to the MYC oncoprotein for survival. Little is known, however, about how MYC expression is upregulated in MM cells. The mucin 1 C-terminal subunit (MUC1-C) is an oncogenic transmembrane protein that is aberrantly expressed in MM cell lines and primary tumor samples. The present studies demonstrate that targeting MUC1-C with silencing by clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein 9 editing or with the GO-203 inhibitor is associated with downregulation of MYC messenger RNA and protein. The results show that MUC1-C occupies the MYC promoter and thereby activates the MYC gene by a ß-catenin/transcription factor 4 (TCF4)-mediated mechanism. In this way, MUC1-C (1) increases ß-catenin occupancy on the MYC promoter, (2) forms a complex with ß-catenin and TCF4, and, in turn, (3) drives MYC transcription. Analysis of MM cells using quantitative real-time reverse transcription polymerase chain reaction arrays further demonstrated that silencing MUC1-C is associated with downregulation of MYC target genes, including CCND2, hTERT, and GCLC Analysis of microarray data sets further demonstrated that MUC1 levels positively correlate with MYC expression in MM progression and in primary cells from over 800 MM patients. These findings collectively provide convincing evidence that MUC1-C drives MYC expression in MM.


Subject(s)
Down-Regulation , Gene Expression Regulation, Neoplastic , Mucin-1/biosynthesis , Multiple Myeloma/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Response Elements , Transcription, Genetic , CRISPR-Cas Systems , Cell Line, Tumor , Cyclin D2/biosynthesis , Cyclin D2/genetics , Glutamate-Cysteine Ligase/biosynthesis , Glutamate-Cysteine Ligase/genetics , Humans , Mucin-1/genetics , Multiple Myeloma/genetics , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-myc/genetics , Telomerase/biosynthesis , Telomerase/genetics , beta Catenin/genetics , beta Catenin/metabolism
16.
Microbiol Immunol ; 62(5): 317-326, 2018 May.
Article in English | MEDLINE | ID: mdl-29577368

ABSTRACT

Mechanisms involved in survival of productively-infected memory CD4+cells after initial antigenic stimulation and their subsequent reversion to the resting state are critical for the development of a predominant replication-competent HIV reservoir. These mechanisms may also counter their elimination after HIV reactivation through latency-reversing agents (LRA). Thus, their evaluation is critical when using an appropriate HIV latency model that recapitulates the predominant replication-competent HIV reservoir to develop strategies for HIV eradication. The model for evaluating the possible survival mechanisms after T cell receptor (TCR) stimulation was developed by infecting memory CD4+cells with an HIV-1C primary isolate and cytokine secretion and gene expression patterns determined. Infected cells showed compromised functionality as evident from 6.8-fold lower secretion of IL-2 than from uninfected control cells. After TCR stimulation, the infected cells showed significantly higher fold increases in CD27 and CCR5 and smaller increases in CD5 mRNA over baseline values. Because CD27 expression may influence telomerase activity through AKT phosphorylation, CD27, human telomerase reverse transcriptase (hTERT) and pAKT expression in productively-infected cells from HIV-infected patients was evaluated by flow cytometry. HIV harbored in memory CD4+ cells was reactivated by HIV-1 envelope peptides, which have been shown to act as effective LRA. P24+CD4+cell showed significantly higher expression of CD27, hTERT and pAKT than P24-CD4+cells. These findings indicate compromised functionality of HIV-infected cells after TCR stimulation, which may interfere with their elimination by the immune system. They also indicate that pAKT and hTERT induction are possible survival mechanisms of productively-infected CD4+cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , HIV Infections/immunology , HIV Infections/virology , HIV-1/pathogenicity , Telomerase/biosynthesis , CD5 Antigens/metabolism , Cytokines , DNA Viruses/genetics , Flow Cytometry , Gene Expression , Gene Expression Profiling , HIV Antigens/metabolism , Humans , RNA, Messenger/biosynthesis , Receptors, Antigen, T-Cell , Receptors, CCR5/metabolism , Telomerase/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Viral Envelope Proteins/metabolism , Virus Activation , Virus Latency , Virus Replication
17.
Nature ; 484(7393): 260-4, 2012 Mar 25.
Article in English | MEDLINE | ID: mdl-22446625

ABSTRACT

In most eukaryotes, the progressive loss of chromosome-terminal DNA sequences is counteracted by the enzyme telomerase, a reverse transcriptase that uses part of an RNA subunit as template to synthesize telomeric repeats. Many cancer cells express high telomerase activity, and mutations in telomerase subunits are associated with degenerative syndromes including dyskeratosis congenita and aplastic anaemia. The therapeutic value of altering telomerase activity thus provides ample impetus to study the biogenesis and regulation of this enzyme in human cells and model systems. We have previously identified a precursor of the fission yeast telomerase RNA subunit (TER1) and demonstrated that the mature 3'-end is generated by the spliceosome in a single cleavage reaction akin to the first step of splicing. Directly upstream and partly overlapping with the spliceosomal cleavage site is a putative binding site for Sm proteins. Sm and like-Sm (LSm) proteins belong to an ancient family of RNA-binding proteins represented in all three domains of life. Members of this family form ring complexes on specific sets of target RNAs and have critical roles in their biogenesis, function and turnover. Here we demonstrate that the canonical Sm ring and the Lsm2-8 complex sequentially associate with fission yeast TER1. The Sm ring binds to the TER1 precursor, stimulates spliceosomal cleavage and promotes the hypermethylation of the 5'-cap by Tgs1. Sm proteins are then replaced by the Lsm2-8 complex, which promotes the association with the catalytic subunit and protects the mature 3'-end of TER1 from exonucleolytic degradation. Our findings define the sequence of events that occur during telomerase biogenesis and characterize roles for Sm and Lsm complexes as well as for the methylase Tgs1.


Subject(s)
Multiprotein Complexes/metabolism , RNA-Binding Proteins/metabolism , RNA/biosynthesis , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Spliceosomes/metabolism , Telomerase/biosynthesis , Base Sequence , Chromosomes, Fungal/genetics , Chromosomes, Fungal/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Methyltransferases/metabolism , Multiprotein Complexes/chemistry , Protein Binding , RNA/genetics , RNA Splicing , RNA, Fungal/genetics , RNA, Fungal/metabolism , Schizosaccharomyces/enzymology , Schizosaccharomyces pombe Proteins/genetics , Telomerase/genetics , Telomere/genetics , Telomere/metabolism , tRNA Methyltransferases/metabolism
18.
J BUON ; 23(1): 229-233, 2018.
Article in English | MEDLINE | ID: mdl-29552789

ABSTRACT

PURPOSE: Distinction of thyroid neoplasms that include papillary carcinoma (PC) and follicular carcinoma (FC) from benign thyroid neoplasms can be performed successfully by histopathologic examination in most of the cases. However, in some cases it may be difficult to distinct PC and FC as well as FC and follicular adenoma (FA) and also FA and the dominant nodule of multinodular goiter (MNG) histopathologically. In this study, we aimed to determine the role of expression of the human telomerase reverse transcriptase (hTERT) in the distinction of thyroid neoplasms and its relation with prognostic factors by immunohistochemical methods. METHODS: This retrospective study included 138 cases histopathologically diagnosed with benign and malignant thyroid neoplasia. Sections obtained from formalin-fixed paraffin- embedded blocks were stained with hTERT antibody. Cases were divided into hTERT-positive and -negative categories according to hTERT expression score that included percentage and intensity of staining in neoplastic cells. RESULTS: hTERT expression was negative in 93 (67.4%) and positive in 45 (32.6%) patients. Twenty-three (46.0%) of 50 PC, 12 (36.0%) of 33 FA, 1 (10.0%) of 10 FC, 4 (13.0%) of 31 MNG, 2 (66.0%) of 3 medullary carcinoma (MC) patients were found hTERT (+), showing that the difference between PC and FC was significant (p=0.034). There was also a significant difference between FA and MNG (p=0.030). There was no difference between FA and FC (p=0.117). CONCLUSION: The high expression of hTERT can be useful for making a differential diagnosis between PC and FC, and between FA and MNG when histopathological findings are equivocal.


Subject(s)
Telomerase/biosynthesis , Thyroid Neoplasms/enzymology , Adenocarcinoma, Follicular/enzymology , Adenocarcinoma, Follicular/genetics , Female , Humans , Immunohistochemistry , Male , Retrospective Studies , Telomerase/genetics , Thyroid Cancer, Papillary/enzymology , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology
19.
J BUON ; 23(4): 1103-1110, 2018.
Article in English | MEDLINE | ID: mdl-30358218

ABSTRACT

PURPOSE: The purpose of this study was to investigate into the expression of cyclin A and telomerase in renal cell carcinoma (RCC) and to analyze the relationship between expression and the clinicopathological characteristics of the tumor and their impact on survival. METHODS: The overall material included 74 samples of RCC and 4 of normal renal tissue. Primary cyclin A antibody from Santa Cruz Biotechnology and TERT MA5-16034 antibody from Thermo Fisher Scientific Inc were used. Staining was performed by streptavidin-biotin technique using DAKO LSAB+ kit. Statistical analyses were performed using of SPSS 23 Statistics software from IBM. RESULTS: No differences in cyclin A and telomerase expression among gender and age groups were found, nor did the tumor dimensions have any significant impact on expression. Also, tumor grades and stages did not differ. However, histological types differed in favor of the papillary type. A significant positive correlation between both markers, as well as between the expression and tumor stage and grade was noticed. Only the tumor stage had negative impact on survival. CONCLUSIONS: Although not affecting survival, the expression of cyclin A and telomerase increased with tumor stage and grade, suggesting that cyclin A and telomerase could be potential proliferative immunohistochemical markers of RCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cyclin A/biosynthesis , Kidney Neoplasms/metabolism , Telomerase/biosynthesis , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/pathology , Cell Proliferation/physiology , Female , Humans , Immunohistochemistry , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Survival Analysis
20.
J Biol Chem ; 291(18): 9690-9, 2016 Apr 29.
Article in English | MEDLINE | ID: mdl-26953344

ABSTRACT

ARID1A is a tumor suppressor gene that belongs to the switch/sucrose non-fermentable chromatin remodeling gene family. It is mutated in many types of human cancer with the highest frequency in endometrium-related ovarian and uterine neoplasms including ovarian clear cell, ovarian endometrioid, and uterine endometrioid carcinomas. We have previously reported that mutations in the promoter of human telomerase reverse transcriptase (TERT) rarely co-occur with the loss of ARID1A protein expression, suggesting a potential role of ARID1A in telomere biology. In this study, we demonstrate that ARID1A negatively regulates TERT transcriptional regulation and activity via binding to the regulatory element of TERT and promotes a repressive histone mode. Induction of ARID1A expression was associated with increased occupancy of SIN3A and H3K9me3, known transcription repressor and histone repressor marks, respectively. Thus, loss of ARID1A protein expression caused by inactivating mutations reactivates TERT transcriptional activity and confers a survival advantage of tumor cells by maintaining their telomeres.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Mutation , Neoplasms/metabolism , Nuclear Proteins/metabolism , Telomerase/biosynthesis , Telomere Homeostasis , Transcription Factors/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/metabolism , Cell Line, Tumor , DNA-Binding Proteins , Histones/genetics , Histones/metabolism , Humans , Neoplasms/genetics , Neoplasms/pathology , Nuclear Proteins/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex , Telomerase/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL