Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.572
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 174(1): 218-230.e13, 2018 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-29804836

RESUMO

Ribonucleoprotein enzymes require dynamic conformations of their RNA constituents for regulated catalysis. Human telomerase employs a non-coding RNA (hTR) with a bipartite arrangement of domains-a template-containing core and a distal three-way junction (CR4/5) that stimulates catalysis through unknown means. Here, we show that telomerase activity unexpectedly depends upon the holoenzyme protein TCAB1, which in turn controls conformation of CR4/5. Cells lacking TCAB1 exhibit a marked reduction in telomerase catalysis without affecting enzyme assembly. Instead, TCAB1 inactivation causes unfolding of CR4/5 helices that are required for catalysis and for association with the telomerase reverse-transcriptase (TERT). CR4/5 mutations derived from patients with telomere biology disorders provoke defects in catalysis and TERT binding similar to TCAB1 inactivation. These findings reveal a conformational "activity switch" in human telomerase RNA controlling catalysis and TERT engagement. The identification of two discrete catalytic states for telomerase suggests an intramolecular means for controlling telomerase in cancers and progenitor cells.


Assuntos
RNA não Traduzido/química , Telomerase/metabolismo , Biocatálise , Linhagem Celular , Células HeLa , Humanos , Chaperonas Moleculares , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Conformação de Ácido Nucleico , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , RNA não Traduzido/metabolismo , Telomerase/antagonistas & inibidores , Telomerase/química , Telomerase/genética , Telômero/metabolismo
2.
Cell ; 148(4): 651-63, 2012 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-22341440

RESUMO

To assess telomerase as a cancer therapeutic target and determine adaptive mechanisms to telomerase inhibition, we modeled telomerase reactivation and subsequent extinction in T cell lymphomas arising in Atm(-/-) mice engineered with an inducible telomerase reverse transcriptase allele. Telomerase reactivation in the setting of telomere dysfunction enabled full malignant progression with alleviation of telomere dysfunction-induced checkpoints. These cancers possessed copy number alterations targeting key loci in human T cell lymphomagenesis. Upon telomerase extinction, tumor growth eventually slowed with reinstatement of telomere dysfunction-induced checkpoints, yet growth subsequently resumed as tumors acquired alternative lengthening of telomeres (ALT) and aberrant transcriptional networks centering on mitochondrial biology and oxidative defense. ALT+ tumors acquired amplification/overexpression of PGC-1ß, a master regulator of mitochondrial biogenesis and function, and they showed marked sensitivity to PGC-1ß or SOD2 knockdown. Genetic modeling of telomerase extinction reveals vulnerabilities that motivate coincidental inhibition of mitochondrial maintenance and oxidative defense mechanisms to enhance antitelomerase cancer therapy.


Assuntos
Mitocôndrias , Telomerase/antagonistas & inibidores , Homeostase do Telômero , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/genética , Proteínas de Ligação a DNA/genética , Técnicas de Silenciamento de Genes , Genes cdc , Humanos , Linfoma de Células T/genética , Linfoma de Células T/metabolismo , Linfoma de Células T/patologia , Camundongos , Mitocôndrias/metabolismo , Invasividade Neoplásica/patologia , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinases/genética , Espécies Reativas de Oxigênio/metabolismo , Receptores de Estrogênio/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Telomerase/genética , Telomerase/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/genética
3.
RNA ; 30(9): 1213-1226, 2024 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-38918043

RESUMO

Telomere replication is essential for continued proliferation of human cells, such as stem cells and cancer cells. Telomerase lengthens the telomeric G-strand, while C-strand replication is accomplished by CST-polymerase α-primase (CST-PP). Replication of both strands is inhibited by formation of G-quadruplex (GQ) structures in the G-rich single-stranded DNA. TMPyP4 and pyridostatin (PDS), which stabilize GQ structures in both DNA and RNA, inhibit telomerase in vitro, and in human cells they cause telomere shortening that has been attributed to telomerase inhibition. Here, we show that TMPyP4 and PDS also inhibit C-strand synthesis by stabilizing DNA secondary structures and thereby preventing CST-PP from binding to telomeric DNA. We also show that these small molecules inhibit CST-PP binding to a DNA sequence containing no consecutive guanine residues, which is unlikely to form GQs. Thus, while these "telomerase inhibitors" indeed inhibit telomerase, they are also robust inhibitors of telomeric C-strand synthesis. Furthermore, given their binding to GQ RNA and their limited specificity for GQ structures, they may disrupt many other protein-nucleic acid interactions in human cells.


Assuntos
Inibidores Enzimáticos , Quadruplex G , Telomerase , Telômero , Telomerase/antagonistas & inibidores , Telomerase/metabolismo , Telomerase/genética , Humanos , Telômero/metabolismo , Quadruplex G/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/química , Inibidores Enzimáticos/síntese química , Ácidos Picolínicos/farmacologia , Ácidos Picolínicos/química , Replicação do DNA/efeitos dos fármacos , DNA Polimerase I/antagonistas & inibidores , DNA Polimerase I/metabolismo , DNA/metabolismo , Aminoquinolinas , Porfirinas , DNA Primase
4.
Genes Dev ; 32(9-10): 658-669, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29773556

RESUMO

Telomerase counteracts telomere shortening and cellular senescence in germ, stem, and cancer cells by adding repetitive DNA sequences to the ends of chromosomes. Telomeres are susceptible to damage by reactive oxygen species (ROS), but the consequences of oxidation of telomeres on telomere length and the mechanisms that protect from ROS-mediated telomere damage are not well understood. In particular, 8-oxoguanine nucleotides at 3' ends of telomeric substrates inhibit telomerase in vitro, whereas, at internal positions, they suppress G-quadruplex formation and were therefore proposed to promote telomerase activity. Here, we disrupt the peroxiredoxin 1 (PRDX1) and 7,8-dihydro-8-oxoguanine triphosphatase (MTH1) genes in cancer cells and demonstrate that PRDX1 and MTH1 cooperate to prevent accumulation of oxidized guanine in the genome. Concomitant disruption of PRDX1 and MTH1 leads to ROS concentration-dependent continuous shortening of telomeres, which is due to efficient inhibition of telomere extension by telomerase. Our results identify antioxidant systems that are required to protect telomeres from oxidation and are necessary to allow telomere maintenance by telomerase conferring immortality to cancer cells.


Assuntos
Enzimas Reparadoras do DNA/metabolismo , Peroxirredoxinas/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Telomerase/metabolismo , Encurtamento do Telômero/genética , Dano ao DNA/genética , Enzimas Reparadoras do DNA/genética , Ativação Enzimática/genética , Técnicas de Inativação de Genes , Genoma , Guanina/metabolismo , Células HCT116 , Humanos , Oxirredução , Estresse Oxidativo/genética , Monoéster Fosfórico Hidrolases/genética , Telomerase/antagonistas & inibidores , Homeostase do Telômero/genética
5.
Nucleic Acids Res ; 51(1): 1-16, 2023 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35697349

RESUMO

Transcriptional reactivation of hTERT is the limiting step in tumorigenesis. While mutations in hTERT promoter present in 19% of cancers are recognized as key drivers of hTERT reactivation, mechanisms by which wildtype hTERT (WT-hTERT) promoter is reactivated, in majority of human cancers, remain unknown. Using primary colorectal cancers (CRC) we identified Tert INTeracting region 2 (T-INT2), the critical chromatin region essential for reactivating WT-hTERT promoter in CRCs. Elevated ß-catenin and JunD level in CRC facilitates chromatin interaction between hTERT promoter and T-INT2 that is necessary to turn on hTERTexpression. Pharmacological screens uncovered salinomycin, which inhibits JunD mediated hTERT-T-INT2 interaction that is required for the formation of a stable transcription complex on the hTERT promoter. Our results showed for the first time how known CRC alterations, such as APC, lead to WT-hTERT promoter reactivation during stepwise-tumorigenesis and provide a new perspective for developing cancer-specific drugs.


Healthy and cancer cells harbor the same DNA sequence, but reactivation of the Human Telomerase Reverse Transcriptase (hTERT) gene is observed only in cancer cells. How does that happen was not known for over three decades of research? This study identifies a specific DNA structure that forms only in cancer cells and brings the necessary molecular machinery into the correct position to activate the hTERT gene. The detailed mechanism of hTERT activation provided in this study will be instrumental in designing cancer cell-specific hTERT inhibitors, especially since all the other ways of inhibiting telomerase failed in the clinic.


Assuntos
Neoplasias Colorretais , Telomerase , Humanos , Carcinogênese , Cromatina/genética , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Regiões Promotoras Genéticas , Telomerase/antagonistas & inibidores , Telomerase/genética , Transcrição Gênica
6.
Nano Lett ; 24(27): 8351-8360, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38916238

RESUMO

Targeting telomere maintenance has emerged as a promising strategy for hepatocellular carcinoma (HCC) treatment. However, given the duality of the telomere-telomerase axis in telomere maintenance, a comprehensive strategy is urgently needed. Herein, we develop a poly(amino acid) (D-PAAs)-based strategy for spatiotemporal codelivery of telomerase inhibitor, BIBR1523, and AKT inhibitor, isobavachalcone. By leveraging D-PAAs' modifiability, we synthesize polymer-inhibitor conjugates (PB and PI) and a folic acid-decorated tumor-targeting vector (PF). These building blocks undergo micellization to fabricate a codelivery nanomedicine (P-BI@P-FA) by exploiting D-PAAs' noncovalent assembly. P-BI@P-FA improves the pharmacokinetics, tumor selectivity, and bioavailability of small molecule inhibitors and initiates a dual telomere-specific inhibition by combining telomerase deactivation with telomere disruption. Furthermore, a hybrid tumor-targeting magnetic nanosystem is designed using D-PAAs and manganese dioxide to showcase magnetic resonance imaging capacities. Our D-PAAs-based strategy addresses the pressing need for telomere-specific HCC treatment while allowing for diagnostic application, presenting a promising avenue for nanomedicine design.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Imageamento por Ressonância Magnética , Nanomedicina , Telomerase , Telômero , Carcinoma Hepatocelular/diagnóstico por imagem , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Telomerase/antagonistas & inibidores , Neoplasias Hepáticas/diagnóstico por imagem , Neoplasias Hepáticas/tratamento farmacológico , Humanos , Nanomedicina/métodos , Telômero/metabolismo , Imageamento por Ressonância Magnética/métodos , Animais , Camundongos , Linhagem Celular Tumoral , Aminoácidos/química , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/uso terapêutico
7.
Biochem Soc Trans ; 52(4): 1957-1968, 2024 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-39194999

RESUMO

Telomerase, crucial for maintaining telomere length, is an attractive target for cancer therapy due to its role in cellular immortality. Despite three decades of research efforts, no small-molecule telomerase inhibitors have been clinically approved, highlighting the extensive challenges in developing effective telomerase-based therapeutics. This review examines conventional and emerging methods to measure telomerase activity and discusses existing inhibitors, including oligonucleotides and small molecules. Furthermore, this review highlights recent breakthroughs in structural studies of telomerase using cryo-electron microscopy, which can facilitate improved structure-based drug design. Altogether, advancements in structural methodologies and high-throughput screening offer promising prospects for telomerase-based cancer therapeutic development.


Assuntos
Descoberta de Drogas , Inibidores Enzimáticos , Telomerase , Telomerase/antagonistas & inibidores , Telomerase/metabolismo , Humanos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Descoberta de Drogas/métodos , Microscopia Crioeletrônica , Neoplasias/tratamento farmacológico , Desenho de Fármacos , Telômero/metabolismo , Oligonucleotídeos/química , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico
8.
Mol Biol Rep ; 51(1): 889, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-39105852

RESUMO

BACKGROUND: Ceranib-2, an acid ceramidase (AC) inhibitor, can inhibit cancer cell proliferation and tumor development. However, poor water solubility and low cellular bioavailability limit its efficacy in cancer treatment. METHODS AND RESULTS: This study aimed to investigate the cell death induced by ceranib-2 and its solid lipid nanoformulation (ceranib-2-SLN) produced by the hot homogenization technique and the synergistic relationship between ceramide and telomerase in vitro and in silico. Furthermore, this study proved the possible mechanism of ceranib-2-induced AC inhibition by in silico studies. The effective cytotoxic concentrations of ceranib-2, telomerase level, and changes in ceramide levels were measured by MTT colorimetric cytotoxicity assay, ELISA, and LC/MS/MS methods, respectively. TEM results showed that ceranib-2-SLN was 13-fold smaller than the size of ceranib-2. Ceranib-2 and ceranib-2-SLN had IC50 concentrations of 31.62 (± 2.1) and 27.69 (± 1.75) µM in A549, and 48.79 (± 1.56) and 67.98 (± 2.33) in Beas-2B cells. These compounds simultaneously increased ceramide levels and decreased telomerase levels in A549 cells. Ceranib-2 increased telomerase levels while decreasing ceramide levels in Beas-2B cells. It was shown how the synergistic impact of ceranib-2-induced ceramide production and ceramide-induced telomerase level reduction on cytotoxicity in A549 cells. CONCLUSIONS: Ceranib-2-SLN was discovered to be more cytotoxic on cancer cells than ceranib-2, suggesting that it could be a promising option for the development of a new anti-cancer agent.


Assuntos
Telomerase , Humanos , Telomerase/metabolismo , Telomerase/antagonistas & inibidores , Linhagem Celular Tumoral , Células A549 , Proliferação de Células/efeitos dos fármacos , Antineoplásicos/farmacologia , Ceramidas/metabolismo , Nanopartículas/química , Sobrevivência Celular/efeitos dos fármacos
9.
Int J Mol Sci ; 25(16)2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-39201386

RESUMO

Glioblastoma (GBM) is a primary CNS tumor that is highly lethal in adults and has limited treatment options. Despite advancements in understanding the GBM biology, the standard treatment for GBM has remained unchanged for more than a decade. Only 6.8% of patients survive beyond five years. Telomerase, particularly the hTERT promoter mutations present in up to 80% of GBM cases, represents a promising therapeutic target due to its role in sustaining telomere length and cancer cell proliferation. This review examines the biology of telomerase in GBM and explores potential telomerase-targeted therapies. We conducted a systematic review following the PRISMA-P guidelines in the MEDLINE/PubMed and Scopus databases, from January 1995 to April 2024. We searched for suitable articles by utilizing the terms "GBM", "high-grade gliomas", "hTERT" and "telomerase". We incorporated studies addressing telomerase-targeted therapies into GBM studies, excluding non-English articles, reviews, and meta-analyses. We evaluated a total of 777 records and 46 full texts, including 36 studies in the final review. Several compounds aimed at inhibiting hTERT transcription demonstrated promising preclinical outcomes; however, they were unsuccessful in clinical trials owing to intricate regulatory pathways and inadequate pharmacokinetics. Direct hTERT inhibitors encountered numerous obstacles, including a prolonged latency for telomere shortening and the activation of the alternative lengthening of telomeres (ALT). The G-quadruplex DNA stabilizers appeared to be potential indirect inhibitors, but further clinical studies are required. Imetelstat, the only telomerase inhibitor that has undergone clinical trials, has demonstrated efficacy in various cancers, but its efficacy in GBM has been limited. Telomerase-targeted therapies in GBM is challenging due to complex hTERT regulation and inadequate inhibitor pharmacokinetics. Our study demonstrates that, despite promising preclinical results, no Telomerase inhibitors have been approved for GBM, and clinical trials have been largely unsuccessful. Future strategies may include Telomerase-based vaccines and multi-target inhibitors, which may provide more effective treatments when combined with a better understanding of telomere dynamics and tumor biology. These treatments have the potential to be integrated with existing ones and to improve the outcomes for patients with GBM.


Assuntos
Glioblastoma , Telomerase , Telomerase/antagonistas & inibidores , Telomerase/metabolismo , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Glioblastoma/genética , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/genética , Terapia de Alvo Molecular , Antineoplásicos/uso terapêutico , Antineoplásicos/farmacologia , Telômero/metabolismo , Telômero/efeitos dos fármacos , Animais
10.
Int J Mol Sci ; 25(11)2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38892179

RESUMO

IMP dehydrogenase (IMPDH) inhibition has emerged as a new target therapy for glioblastoma multiforme (GBM), which remains one of the most refractory tumors to date. TCGA analyses revealed distinct expression profiles of IMPDH isoenzymes in various subtypes of GBM and low-grade glioma (LGG). To dissect the mechanism(s) underlying the anti-tumor effect of IMPDH inhibition in adult GBM, we investigated how mycophenolic acid (MPA, an IMPDH inhibitor) treatment affected key oncogenic drivers in glioblastoma cells. Our results showed that MPA decreased the expression of telomerase reverse transcriptase (TERT) in both U87 and U251 cells, and the expression of O6-methylguanine-DNA methyltransferase (MGMT) in U251 cells. In support, MPA treatment reduced the amount of telomere repeats in U87 and U251 cells. TERT downregulation by MPA was associated with a significant decrease in c-Myc (a TERT transcription activator) in U87 but not U251 cells, and a dose-dependent increase in p53 and CCCTC-binding factor (CTCF) (TERT repressors) in both U87 and U251 cells. In U251 cells, MPA displayed strong cytotoxic synergy with BCNU and moderate synergy with irinotecan, oxaliplatin, paclitaxel, or temozolomide (TMZ). In U87 cells, MPA displayed strong cytotoxic synergy with all except TMZ, acting primarily through the apoptotic pathway. Our work expands the mechanistic potential of IMPDH inhibition to TERT/telomere regulation and reveals a synthetic lethality between MPA and anti-GBM drugs.


Assuntos
Glioblastoma , IMP Desidrogenase , Telomerase , Humanos , Telomerase/metabolismo , Telomerase/antagonistas & inibidores , Telomerase/genética , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Glioblastoma/genética , Glioblastoma/patologia , Linhagem Celular Tumoral , IMP Desidrogenase/antagonistas & inibidores , IMP Desidrogenase/metabolismo , IMP Desidrogenase/genética , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/genética , Apoptose/efeitos dos fármacos
11.
Biochem Biophys Res Commun ; 640: 50-55, 2023 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-36502631

RESUMO

Triple-negative breast cancer (TNBC) is a subtype of breast tumor with the highest breast cancer stem cells (BCSCs) content and resistance to conventional treatment. Due to the immunosuppressive tumor microenvironment and immunogenicity of breast cancer cells, the use of immune cells, especially natural killer cells (NK) in the treatment of solid tumors, including breast cancer, has been unsatisfactory. Therefore, identifying novel therapies is requisite for breast cancer treatment. Furthermore, the combination of cancer therapies is an effective strategy to improve therapeutic effectiveness. In this study, we inhibited telomerase (hTERT) with BIBR1532, in stimulating NK cell cytotoxicity against breast cancer cells. The MDA-MB-231 cell line was cured with IC50 level of BIBR1532 for 24 h. Afterward, cells were washed with PBS and were co-cultured with peripheral blood NK cell for 5h. Finally, we assessed the impact of telomerase inhibition on the cytotoxicity of NK cells and apoptosis of breast cancer. Also, the expression of hTERT and apoptotic-related genes were evaluated. The data revealed that inhibition of telomerase increases NK cell cytotoxicity against breast cancer. Furthermore, telomerase inhibition and NK cell synergistically enhanced cell death in breast cancer cells by suppressing hTERT, upregulation of bax, and bad expression. In conclusion, telomerase suppression makes breast cancer cells more sensitive to NK cell therapy. Consequently, the combination of telomerase inhibition and NK cells can be useful in the treatment of breast cancer cells.


Assuntos
Apoptose , Células Matadoras Naturais , Telomerase , Neoplasias de Mama Triplo Negativas , Feminino , Humanos , Linhagem Celular Tumoral , Proliferação de Células , Telomerase/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/terapia , Microambiente Tumoral , Células Matadoras Naturais/transplante , Terapia Baseada em Transplante de Células e Tecidos
12.
Curr Opin Oncol ; 35(2): 100-106, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36700456

RESUMO

PURPOSE OF REVIEW: Checkpoint inhibitors (CPIs) have revolutionized treatment outcomes for patients with malignant melanoma. Long-term follow-up shows that a substantial subset of patients who exhibit clinical responses achieve extended overall survival. Nevertheless, most patients do not achieve durable benefit from CPIs, and improvements are urgently needed. The clinical efficacy of CPIs depends on highly variable preexisting spontaneous T-cell immune responses. Cancer vaccines represent an independent treatment modality uniquely capable of expanding the repertoire of tumor-specific T cells in cancer patients and thus have the capacity to compensate for the variability in spontaneous T-cell responses. Vaccines are, therefore, considered attractive components in a CPI-combination strategy. RECENT FINDINGS: Here we discuss recent results obtained through therapeutic vaccination against telomerase human telomerase reverse transcriptase (hTERT). Recent publications on translational research and clinical results from phase I trials indicate that vaccination against telomerase in combination with CPIs provides relevant immune responses, negligible added toxicity, and signals of clinical efficacy. CONCLUSION: In the near future, randomized data from clinical trials involving therapeutic cancer vaccines and checkpoint inhibitors will be available. Positive readout may spark broad development and allow cancer vaccines to find their place in the clinic as an important component in multiple future CPI combinations.


Assuntos
Vacinas Anticâncer , Melanoma , Neoplasias Cutâneas , Telomerase , Humanos , Vacinas Anticâncer/uso terapêutico , Melanoma/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Telomerase/antagonistas & inibidores , Telomerase/metabolismo , Vacinação
13.
Nucleic Acids Res ; 49(13): 7280-7291, 2021 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-34233000

RESUMO

Rational design of aptamers to incorporate unnatural nucleotides and special chemical moieties can expand their functional complexity and diversity. Spiegelmer (L-RNA aptamer) is a unique class of aptamer that is composed of unnatural L-RNA nucleotides, and so far there are limited L-RNA aptamer candidates and applications being reported. Moreover, the target binding properties of current L-RNA aptamers require significant improvement. Here, using L-Apt.4-1c as an example, we develop a simple and robust strategy to generate the first circular L-RNA aptamer, cycL-Apt.4-1c, quantitatively, demonstrate substantial enhancement in binding affinity and selectivity toward its target, and notably report novel applications of circular L-RNA aptamer in controlling RNA-protein interaction, and gene activity including telomerase activity and gene expression. Our approach and findings will be applicable to any L-RNA aptamers and open up a new avenue for diverse applications.


Assuntos
Aptâmeros de Nucleotídeos/química , RNA Circular/química , Química Click , Ciclização , RNA Helicases DEAD-box/metabolismo , Regulação da Expressão Gênica , Conformação de Ácido Nucleico , RNA , Estabilidade de RNA , Telomerase/antagonistas & inibidores
14.
PLoS Biol ; 17(4): e3000204, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30951520

RESUMO

Telomerase, a unique reverse transcriptase that specifically extends the ends of linear chromosomes, is up-regulated in the vast majority of cancer cells. Here, we show that an indole nucleotide analog, 5-methylcarboxyl-indolyl-2'-deoxyriboside 5'-triphosphate (5-MeCITP), functions as an inhibitor of telomerase activity. The crystal structure of 5-MeCITP bound to the Tribolium castaneum telomerase reverse transcriptase reveals an atypical interaction, in which the nucleobase is flipped in the active site. In this orientation, the methoxy group of 5-MeCITP extends out of the canonical active site to interact with a telomerase-specific hydrophobic pocket formed by motifs 1 and 2 in the fingers domain and T-motif in the RNA-binding domain of the telomerase reverse transcriptase. In vitro data show that 5-MeCITP inhibits telomerase with a similar potency as the clinically administered nucleoside analog reverse transcriptase inhibitor azidothymidine (AZT). In addition, cell-based studies show that treatment with the cell-permeable nucleoside counterpart of 5-MeCITP leads to telomere shortening in telomerase-positive cancer cells, while resulting in significantly lower cytotoxic effects in telomerase-negative cell lines when compared with AZT treatment.


Assuntos
Nucleosídeos/metabolismo , Telomerase/antagonistas & inibidores , Telomerase/fisiologia , Animais , Domínio Catalítico/efeitos dos fármacos , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Modelos Moleculares , Nucleosídeos/síntese química , Nucleosídeos/fisiologia , Nucleotídeos/síntese química , Nucleotídeos/metabolismo , RNA/metabolismo , Inibidores da Transcriptase Reversa/farmacologia , Telômero , Tribolium/genética , Tribolium/metabolismo , Zidovudina/metabolismo , Zidovudina/farmacologia
15.
Bioorg Med Chem Lett ; 57: 128503, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34922028

RESUMO

In this study, a series of novel shikonin N-benzyl matrinic acid ester derivatives (PMMB-299-PMMB-310) were synthesized and tested for their ability to inhibit the proliferation of cancer cells. Compared with shikonin and matrine, some of the ester derivatives were found to exhibit better anti-proliferative activity against seven different cancer cell lines, with less cytotoxicity toward non-cancerous cells. The strongest anti-proliferative activity was exhibited by PMMB-302, which had an IC50 value of 2.71 µM against A549 cells. The compound caused cell cycle arrest in the G2/M phase and induced apoptosis. Effects on the expression of apoptosis-related molecules such as Bcl2, Bcl-XL, caspase-3, caspase-9 and FADD suggested that PMMB-302 has tumor suppressive roles in lung cancer cells. In addition, PMMB-302 inhibited expression of telomerase core proteins, dyskerin and NHP2, and telomerase reverse transcriptase RNA. Moreover, molecular docking of PMMB-302 was subsequently conducted to determine the probable binding mode with telomerase. Taken together, the results indicate that PMMB-302 acts as a tumor suppressor in lung cancer cells by negatively regulating telomerase expression.


Assuntos
Alcaloides/farmacologia , Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Naftoquinonas/farmacologia , Quinolizinas/farmacologia , Telomerase/antagonistas & inibidores , Alcaloides/síntese química , Alcaloides/metabolismo , Antineoplásicos/síntese química , Antineoplásicos/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Simulação de Acoplamento Molecular , Naftoquinonas/síntese química , Naftoquinonas/metabolismo , Ligação Proteica , Quinolizinas/síntese química , Quinolizinas/metabolismo , Telomerase/metabolismo , Matrinas
16.
Cell Mol Biol (Noisy-le-grand) ; 67(4): 115-120, 2022 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-35809296

RESUMO

High-dose chemotherapy and stem cell transplantation are the best treatment options in patients with multiple myeloma. Numerous medicines have been studied as a maintenance treatment after transplantation. Still, the use of medications that, in addition to their maintenance properties, eliminate or delay relapse of the disease has always been researchers' purpose. Therefore, this study was performed to evaluate the efficacy of MST-312 after stem cell transplantation in patients with multiple myeloma. For this purpose, 73 patients with multiple myeloma after stem cell transplantation were studied. Thirty-five patients were in the case group, and 37 patients were in the control group. The case group was treated with 100 mg/day MST-312. Stem cell survival was evaluated in the two groups. Also, the expression of TNFα and IL-6 genes were evaluated by the Real-time PCR technique. The results showed no significant difference between the two groups in terms of stem cell survival in the first year (P=0.72) and second years of treatment (P=0.66). But there was a significant difference between the two groups regarding progression-free survival (PFS) in the first year (P=0.041) and the second year (P=0.029). These results indicate that MST-312 inhibits the progress of the disease by inhibiting the telomerase activity of myeloma cells. Genetic evaluations also showed that IL-6 and TNF-α genes were significantly reduced in the case group. Therefore, it could be suggested that MST-312 has a selective inhibitory effect on myeloma cell growth and can be indicated as a suitable candidate for treating multiple myeloma.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Mieloma Múltiplo , Telomerase , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Benzamidas/farmacologia , Inibidores Enzimáticos , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Interleucina-6/genética , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/etiologia , Transplante de Células-Tronco , Telomerase/antagonistas & inibidores , Telomerase/genética , Transplante Autólogo/métodos
17.
Mol Ther ; 29(3): 1199-1213, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33212301

RESUMO

T cell receptor (TCR)-engineered T cell therapy is a promising cancer treatment approach. Human telomerase reverse transcriptase (hTERT) is overexpressed in the majority of tumors and a potential target for adoptive cell therapy. We isolated a novel hTERT-specific TCR sequence, named Radium-4, from a clinically responding pancreatic cancer patient vaccinated with a long hTERT peptide. Radium-4 TCR-redirected primary CD4+ and CD8+ T cells demonstrated in vitro efficacy, producing inflammatory cytokines and killing hTERT+ melanoma cells in both 2D and 3D settings, as well as malignant, patient-derived ascites cells. Importantly, T cells expressing Radium-4 TCR displayed no toxicity against bone marrow stem cells or mature hematopoietic cells. Notably, Radium-4 TCR+ T cells also significantly reduced tumor growth and improved survival in a xenograft mouse model. Since hTERT is a universal cancer antigen, and the very frequently expressed HLA class II molecules presenting the hTERT peptide to this TCR provide a very high (>75%) population coverage, this TCR represents an attractive candidate for immunotherapy of solid tumors.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Imunoterapia/métodos , Melanoma/terapia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T Citotóxicos/imunologia , Telomerase/antagonistas & inibidores , Animais , Apoptose , Proliferação de Células , Humanos , Melanoma/imunologia , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
18.
PLoS Genet ; 15(6): e1008188, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31237867

RESUMO

Telomerase, particularly its main subunit, the reverse transcriptase, TERT, prevents DNA erosion during eukaryotic chromosomal replication, but also has poorly understood non-canonical functions. Here, in the model social amoeba Dictyostelium discoideum, we show that the protein encoded by tert has telomerase-like motifs, and regulates, non-canonically, important developmental processes. Expression levels of wild-type (WT) tert were biphasic, peaking at 8 and 12 h post-starvation, aligning with developmental events, such as the initiation of streaming (~7 h) and mound formation (~10 h). In tert KO mutants, however, aggregation was delayed until 16 h. Large, irregular streams formed, then broke up, forming small mounds. The mound-size defect was not induced when a KO mutant of countin (a master size-regulating gene) was treated with TERT inhibitors, but anti-countin antibodies did rescue size in the tert KO. Although, conditioned medium (CM) from countin mutants failed to rescue size in the tert KO, tert KO CM rescued the countin KO phenotype. These and additional observations indicate that TERT acts upstream of smlA/countin: (i) the observed expression levels of smlA and countin, being respectively lower and higher (than WT) in the tert KO; (ii) the levels of known size-regulation intermediates, glucose (low) and adenosine (high), in the tert mutant, and the size defect's rescue by supplemented glucose or the adenosine-antagonist, caffeine; (iii) the induction of the size defect in the WT by tert KO CM and TERT inhibitors. The tert KO's other defects (delayed aggregation, irregular streaming) were associated with changes to cAMP-regulated processes (e.g. chemotaxis, cAMP pulsing) and their regulatory factors (e.g. cAMP; acaA, carA expression). Overexpression of WT tert in the tert KO rescued these defects (and size), and restored a single cAMP signaling centre. Our results indicate that TERT acts in novel, non-canonical and upstream ways, regulating key developmental events in Dictyostelium.


Assuntos
Agregação Celular/genética , Dictyostelium/genética , Morfogênese/genética , Telomerase/genética , Adenosina/genética , Animais , Agregação Celular/efeitos dos fármacos , Quimiotaxia/genética , AMP Cíclico/genética , Dictyostelium/crescimento & desenvolvimento , Inibidores Enzimáticos/farmacologia , Técnicas de Inativação de Genes , Glucose/genética , Transdução de Sinais/genética , Telomerase/antagonistas & inibidores
19.
J Neurochem ; 159(4): 690-709, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34532857

RESUMO

After a sublethal ischemic preconditioning (IPC) stimulus, the brain has a remarkable capability of acquiring tolerance to subsequent ischemic insult by establishing precautionary self-protective mechanism. Understanding this endogenous mechanism would reveal novel and effective neuroprotective targets for ischemic brain injury. Our previous study has implied that telomerase reverse transcriptase (TERT) is associated with IPC-induced tolerance. Here, we investigated the mechanism of TERT-mediated ischemic tolerance. Preconditioning was modeled by oxygen-glucose deprivation (OGD) and by TERT inhibitor BIBR1532 in primary neurons. We found that ischemic tolerance was conferred by BIBR1532 preconditioning. We used the Cleavage-Under-Targets-And-Tagmentation approach, a recently developed method with superior signal-to-noise ratio, to comprehensively map the genomic binding sites of TERT in primary neurons, and showed that more than 50% of TERT-binding sites were located at the promoter regions. Mechanistically, we demonstrated that under normal conditions TERT physically bound to many previously unknown genomic loci in neurons, whereas BIBR1532 preconditioning significantly altered TERT-chromatin-binding profile. Intriguingly, we found that BIBR1532-preconditioned neurons showed significant up-regulation of promoter binding of TERT to the mitochondrial anti-oxidant genes, which were correlated with their elevated expression. Functional analysis further indicated that BIBR1532-preconditioning significantly reduced ROS levels and enhanced tolerance to severe ischemia-induced mitochondrial oxidative stress in neurons in a TERT-dependent manner. Together, these results demonstrate that BIBR1532 confers neuronal ischemic tolerance through TERT-mediated transcriptional reprogramming for up-regulation of mitochondrial anti-oxidation gene expression, suggesting the translational potential of BIBR1532 as a therapeutic agent for the treatment of cerebral ischemic injury and oxidative stress-induced neurological disorders.


Assuntos
Aminobenzoatos/uso terapêutico , Isquemia Encefálica/tratamento farmacológico , Naftalenos/uso terapêutico , Neurônios , Inibidores da Transcriptase Reversa/farmacologia , Telomerase/metabolismo , Animais , Antioxidantes/metabolismo , Sítios de Ligação/genética , Isquemia Encefálica/patologia , Cromatina/metabolismo , Mapeamento Cromossômico , Feminino , Técnicas de Silenciamento de Genes , Glucose/deficiência , Hipóxia , Precondicionamento Isquêmico , Camundongos , Camundongos Endogâmicos C57BL , Neuroproteção , Gravidez , Cultura Primária de Células , Espécies Reativas de Oxigênio , Razão Sinal-Ruído , Telomerase/antagonistas & inibidores , Telomerase/genética , Ativação Transcricional
20.
Biochem Biophys Res Commun ; 582: 118-124, 2021 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-34710826

RESUMO

Telomerase is a reverse transcriptase that catalyzes the addition of telomeric repeated DNA onto the 3' ends of linear chromosomes. Telomerase inhibition was broadly used for cancer therapeutics. Here, six antisense oligonucleotides were designed to regulate TERT mRNA alternative splicing and protein translation. To pursue a better stability in vitro, we chemically modified the oligonucleotides into phosphorothioate (PS) backbone and 2'-O-methoxyethyl (2'-MOE PS) version and phosphoroamidate morpholino oligomer (PMO) version. The oligonucleotides were transfected into HEK 293T cells and HeLa cells, and the mRNA expression, protein level and catalytic activity of telomerase were determined. We found the Int8 notably promoted hTERT mRNA exon 7-8 skipping, which greatly reduced telomerase activity, and the 5'-UTR treatment led to an obvious protein translation barrier and telomerase inhibition. These results demonstrate the potential of antisense oligonucleotide drugs targeting hTERT for antitumor therapy. Moreover, two specific antisense oligonucleotides were identified to be effective in reducing telomerase activity.


Assuntos
Morfolinos/genética , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Fosforotioatos/genética , RNA Mensageiro/genética , Telomerase/genética , Processamento Alternativo/efeitos dos fármacos , Antineoplásicos/farmacologia , Células HEK293 , Células HeLa , Humanos , Morfolinos/síntese química , Morfolinos/metabolismo , Oligonucleotídeos Antissenso/síntese química , Oligonucleotídeos Antissenso/metabolismo , Oligonucleotídeos Fosforotioatos/síntese química , Oligonucleotídeos Fosforotioatos/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/metabolismo , Telomerase/antagonistas & inibidores , Telomerase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA