Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Chem Biol ; 30(12): 1652-1665.e6, 2023 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-38065101

RESUMO

The TRF2 shelterin component is an essential regulator of telomere homeostasis and genomic stability. Mutations in the TRF2TRFH domain physically impair t-loop formation and prevent the recruitment of several factors that promote efficient telomere replication, causing telomeric DNA damage. Here, we design, synthesize, and biologically test covalent cyclic peptides that irreversibly target the TRF2TRFH domain. We identify APOD53 as our most promising compound, as it consistently induces a telomeric DNA damage response in cancer cell lines. APOD53 forms a covalent adduct with a reactive cysteine residue present in the TRF2TRFH domain and induces phenotypes consistent with TRF2TRFH domain mutants. These include induction of a telomeric DNA damage response, increased telomeric replication stress, and impaired recruitment of RTEL1 and SLX4 to telomeres. We demonstrate that APOD53 impairs cancer cell growth and find that co-treatment with APOD53 can exacerbate telomere replication stress caused by the G4 stabilizer RHPS4 and low dose aphidicolin (APH).


Assuntos
Peptídeos Cíclicos , Proteína 2 de Ligação a Repetições Teloméricas , Dano ao DNA , Peptídeos Cíclicos/farmacologia , Telômero , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Proteína 2 de Ligação a Repetições Teloméricas/química , Proteína 2 de Ligação a Repetições Teloméricas/genética , Domínios Proteicos , Linhagem Celular Tumoral
2.
J Cell Sci ; 135(13)2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35660868

RESUMO

We investigated the role of telomerase and telomere repeat-binding factor 2 (TRF2 or TERF2) in T-cell dysfunction in chronic viral infection. We found that the expression and activity of telomerase in CD4+ T (CD4T) cells from patients with hepatitis C virus (HCV) infections or people living with HIV (PLWH) were intact, but TRF2 expression was significantly inhibited at the post-transcriptional level, suggesting that TRF2 inhibition is responsible for the CD4T cell dysfunction observed during chronic viral infection. Silencing TRF2 expression in CD4T cells derived from healthy subjects induced telomeric DNA damage and CD4T cell dysfunction without affecting telomerase activity or translocation - similar to what we observed in CD4T cells from HCV patients and PLWH. These findings indicate that premature T-cell aging and dysfunction during chronic HCV or HIV infection are primarily caused by chronic immune stimulation and T-cell overactivation and/or proliferation that induce telomeric DNA damage due to TRF2 inhibition, rather than telomerase disruption. This study suggests that restoring TRF2 presents a novel approach to prevent telomeric DNA damage and premature T-cell aging, thus rejuvenating T-cell functions during chronic viral infection.


Assuntos
Linfócitos T CD4-Positivos , Infecções por HIV , Telomerase , Proteína 2 de Ligação a Repetições Teloméricas , Linfócitos T CD4-Positivos/imunologia , Dano ao DNA , Infecções por HIV/genética , Infecções por HIV/imunologia , Hepacivirus , Hepatite C Crônica/genética , Hepatite C Crônica/imunologia , Humanos , Telomerase/genética , Telomerase/metabolismo , Telômero , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Proteína 2 de Ligação a Repetições Teloméricas/genética , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
3.
Bioorg Med Chem Lett ; 30(21): 127401, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32871539

RESUMO

Telomeric repeat binding factor 2 (TRF2) plays an important role in protecting telomeres from being recognized as DNA breaks. TRF2 performs its telomere protecting functions partially by recruiting a number of accessory proteins to telomeres through its TRF homology (TFRH) domain. Identification of small molecular compounds which can bind to the TRFH domain of TRF2 and block the interactions between TRF2 and its associated proteins is crucial for elucidating the molecular mechanisms of these protein-protein interactions. Using a previously identified peptidic mimetic of ApolloTBM as a lead compound, we designed and synthesized a series of novel TRF2 inhibitors by non-peptidic modifications of the N-terminal residues. These compounds can maintain the binding affinities to TRF2 but have much reduced peptidic characteristics compared to the lead compound.


Assuntos
Peptídeos Cíclicos/farmacologia , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Relação Dose-Resposta a Droga , Humanos , Modelos Moleculares , Estrutura Molecular , Peptídeos Cíclicos/síntese química , Peptídeos Cíclicos/química , Relação Estrutura-Atividade
4.
Anal Biochem ; 602: 113796, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32485162

RESUMO

TRF2 is a telomere associated protein which plays an important role in telomere maintenance. Knockdown of TRF2 can cause chromosomal end to end fusions and induce DNA damage responses. TRF2 exerts its functions partially by recruiting a number of accessory proteins through its TRF homology domain (TRFH), therefore identification of small molecular compounds which can bind to the TRFH domain of TRF2 and block the interactions of TRF2 with its associated proteins is important to elucidate the molecular mechanism of these protein-protein interactions. Development of robust and sensitive screening and evaluation assays is critical to the identification of TRF2 inhibitors, in this paper we reported the development and optimization of a cascade of screening and binding affinity evaluation assays, including a competitive FP (Fluorescence Polarization) assay utilized in our previous research, and two novel label-free DSF (Differential Scanning Fluorescence) and BLI (Biolayer Interferometry) assays. A previously identified TRF2 inhibitor TRF2-27 was used as an internal reference compound and evaluated in all of these assays. According to the results, DSF assay is not suitable for TRF2 screening because of the low ΔTm, while the optimized labeled-free BLI assay was demonstrated to be an accurate and reproducible assay for TRF2 inhibitor screening and characterization.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Peptídeos/farmacologia , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Polarização de Fluorescência , Humanos , Conformação Molecular , Peptídeos/química , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
5.
Cell Death Dis ; 9(9): 900, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-30185784

RESUMO

T cells play a crucial role in viral clearance and vaccine responses; however, the mechanisms that regulate their homeostasis during viral infections remain unclear. In this study, we investigated the machineries of T-cell homeostasis and telomeric DNA damage using a human model of hepatitis C virus (HCV) infection. We found that naïve CD4 T cells in chronically HCV-infected patients (HCV T cells) were significantly reduced due to apoptosis compared with age-matched healthy subjects (HSs). These HCV T cells were not only senescent, as demonstrated by overexpression of aging markers and particularly shortened telomeres; but also DNA damaged, as evidenced by increased dysfunctional telomere-induced foci (TIF). Mechanistically, the telomere shelterin protein, in particular telomeric repeat binding factor 2 (TRF2) that functions to protect telomeres from DNA damage, was significantly inhibited posttranscriptionally via the p53-dependent Siah-1a ubiquitination. Importantly, knockdown of TRF2 in healthy T cells resulted in increases in telomeric DNA damage and T-cell apoptosis, whereas overexpression of TRF2 in HCV T cells alleviated telomeric DNA damage and T-cell apoptosis. To the best of our knowledge, this is the first report revealing that inhibition of TRF2 promotes T-cell telomere attrition and telomeric DNA damage that accelerates T-cell senescent and apoptotic programs, which contribute to naïve T-cell loss during viral infection. Thus, restoring the impaired T-cell telomeric shelterin machinery may offer a new strategy to improve immunotherapy and vaccine response against human viral diseases.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Dano ao DNA/fisiologia , Hepatite C/metabolismo , Telômero/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Apoptose/fisiologia , Linfócitos T CD4-Positivos/virologia , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Células HEK293 , Hepacivirus/patogenicidade , Hepatite C/virologia , Humanos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/virologia , Proteínas Supressoras de Tumor/metabolismo
6.
Cell Death Dis ; 9(5): 518, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29725012

RESUMO

The nucleolus is an important organelle that is responsible for the biogenesis of ribosome RNA (rRNA) and ribosomal subunits assembly. It is also deemed to be the center of metabolic control, considering the critical role of ribosomes in protein translation. Perturbations of rRNA synthesis are closely related to cell proliferation and tumor progression. Telomeric repeat-binding factor 2 (TRF2) is a member of shelterin complex that is responsible for telomere DNA protection. Interestingly, it was recently reported to localize in the nucleolus of human cells in a cell-cycle-dependent manner, while the underlying mechanism and its role on the nucleolus remained unclear. In this study, we found that nucleolar and coiled-body phosphoprotein 1 (NOLC1), a nucleolar protein that is responsible for the nucleolus construction and rRNA synthesis, interacted with TRF2 and mediated the shuttle of TRF2 between the nucleolus and nucleus. Abating the expression of NOLC1 decreased the nucleolar-resident TRF2. Besides, the nucleolar TRF2 could bind rDNA and promoted rRNA transcription. Furthermore, in hepatocellular carcinoma (HCC) cell lines HepG2 and SMMC7721, TRF2 overexpression participated in the nucleolus stress-induced rRNA inhibition and cell-cycle arrest.


Assuntos
Antineoplásicos/farmacologia , Nucléolo Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Proteínas Nucleares/genética , Fosfoproteínas/genética , RNA Ribossômico/genética , Proteína 2 de Ligação a Repetições Teloméricas/genética , Sequência de Aminoácidos , Camptotecina/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Nucléolo Celular/metabolismo , Nucléolo Celular/ultraestrutura , Proliferação de Células/efeitos dos fármacos , Dactinomicina/farmacologia , Etoposídeo/farmacologia , Células HEK293 , Células Hep G2 , Humanos , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Fosfoproteínas/antagonistas & inibidores , Fosfoproteínas/metabolismo , Ligação Proteica , Transporte Proteico , RNA Ribossômico/biossíntese , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ribossomos/efeitos dos fármacos , Ribossomos/genética , Ribossomos/metabolismo , Transdução de Sinais , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
7.
Cell Biochem Biophys ; 76(1-2): 311-319, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29134494

RESUMO

Telomere DNA can form specialized nucleoprotein structure with telomere-associated proteins to hide free DNA ends or G-quadruplex structures under certain conditions especially in presence of G-quadruplex ligand. Telomere DNA is transcribed to form non-coding telomere repeat-containing RNA (TERRA) whose biogenesis and function is poorly understood. Our aim was to find the role of telomere-associated proteins and telomere structures in TERRA transcription. We silenced four [two shelterin (TRF1, TRF2) and two non-shelterin (PARP-1, SLX4)] telomere-associated genes using siRNA and verified depletion in protein level. Knocking down of one gene modulated expression of other telomere-associated genes and increased TERRA from 10q, 15q, XpYp and XqYq chromosomes in A549 cells. Telomere was destabilized or damaged by G-quadruplex ligand pyridostatin (PDS) and bleomycin. Telomere dysfunction-induced foci (TIFs) were observed for each case of depletion of proteins, treatment with PDS or bleomycin. TERRA level was elevated by PDS and bleomycin treatment alone or in combination with depletion of telomere-associated proteins.


Assuntos
RNA Longo não Codificante/metabolismo , Telômero/metabolismo , Proteína 1 de Ligação a Repetições Teloméricas/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Células A549 , Bleomicina/farmacologia , Quadruplex G , Humanos , Hibridização in Situ Fluorescente , Microscopia de Fluorescência , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Poli(ADP-Ribose) Polimerase-1/genética , Poli(ADP-Ribose) Polimerase-1/metabolismo , Interferência de RNA , RNA Longo não Codificante/genética , RNA Interferente Pequeno/metabolismo , Recombinases/antagonistas & inibidores , Recombinases/genética , Recombinases/metabolismo , Telômero/química , Proteína 1 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Proteína 1 de Ligação a Repetições Teloméricas/genética , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Proteína 2 de Ligação a Repetições Teloméricas/genética , Regulação para Cima/efeitos dos fármacos
8.
Biochim Biophys Acta Gen Subj ; 1861(12): 3246-3256, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28943299

RESUMO

BACKGROUND: Telomeric repeat-containing RNA (TERRA) is a large non-coding RNA in mammalian cells, which forms an integral component of telomeric heterochromatin. TERRA can bind to an allosteric site of telomeric repeat factor 2 (TRF2), a key component of Shelterin that protect chromosome termini. Both TERRA and TRF2 have been recognized as promising new therapeutic targets for cancer treatment. METHODS: Our methods include FRET assay, SPR, CD, microscale thermophoresis (MST), enzyme-linked immunosorbent assay (ELISA), chromatin immunoprecipitation (ChIP), colony formation assays, Western blot, immunofluorescence, cell cycle arrest and apoptosis detection, and xCELLigence real-time cell analysis (RTCA). RESULTS: In our routine screening of small molecule libraries, we found that a Quindoline derivative, CK1-14 could bind to and stabilize TERRA G-quadruplex structure, which could bind more tightly with an allosteric site of a telomeric binding protein TRF2, resulting in dissociation of TRF2 from telomeric DNA. Further in cellular studies indicated that the above effect of CK1-14 on TERRA G-quadruplex could activate DNA-damage response and cause cell cycle arrest, resulting in inhibition of U2OS cell proliferation and causing cell apoptosis. CONCLUSIONS: Our mechanistic studies indicated that interaction of CK1-14 with TERRA induces telomeric DNA-damage response in U2OS cancer cells through inhibition of TRF2. CK1-14 could be further developed as a promising lead compound targeting telomere for cancer treatment. GENERAL SIGNIFICANCE: Our present study provides the first evidence that allosteric modulation of TRF2 by TERRA G-quadruplex with a binding ligand could become a promising new strategy for cancer treatment especially for ALT tumor cells.


Assuntos
Alcaloides/farmacologia , Dano ao DNA , Indóis/farmacologia , Neoplasias/tratamento farmacológico , Quinolinas/farmacologia , RNA Longo não Codificante/metabolismo , Telômero , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Alcaloides/metabolismo , Regulação Alostérica , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Quadruplex G , Humanos , Indóis/metabolismo , Neoplasias/genética , Neoplasias/patologia , Quinolinas/metabolismo
9.
Biochim Biophys Acta Proteins Proteom ; 1865(11 Pt A): 1372-1382, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28870734

RESUMO

Telomeric repeat factor 2 (known as TRF2 or TERF2) is a key component of telomere protection protein complex named as Shelterin. TRF2 helps the folding of telomere to form T-loop structure and the suppression of ATM-dependent DNA damage response activation. TRF2 has been recognized as a potentially new therapeutic target for cancer treatment. In our routine screening of small molecule libraries, we found that Curcusone C had significant effect in disrupting the binding between TRF2 and telomeric DNA, with potent antitumor activity against cancer cells. Our result showed that Curcusone C could bind with TRF2 without binding interaction with TRF1 (telomeric repeat factor 1) although these two proteins share high sequence homology, indicating that their binding conformations and biological functions in telomere could be different. Our mechanistic studies showed that Curcusone C bound with TRF2 possibly through its DNA binding site causing blockage of its interaction with telomeric DNA. Further in cellular studies indicated that the interaction of TRF2 with Curcusone C could activate DNA-damage response, inhibit tumor cell proliferation, and cause cell cycle arrest, resulting in tumor cell apoptosis. Our studies showed that Curcusone C could become a promising lead compound for further development for cancer treatment. Here, TRF2 was firstly identified as a target of Curcusone C. It is likely that the anti-cancer activity of some other terpenes and terpenoids are related with their possible effect for telomere protection proteins.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , DNA de Neoplasias/genética , Diterpenos/farmacologia , Regulação Neoplásica da Expressão Gênica , Telômero/efeitos dos fármacos , Proteína 2 de Ligação a Repetições Teloméricas/genética , Antineoplásicos Fitogênicos/isolamento & purificação , Sítios de Ligação , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , DNA de Neoplasias/metabolismo , Diterpenos/isolamento & purificação , Relação Dose-Resposta a Droga , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células HeLa , Humanos , Especificidade de Órgãos , Ligação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Telômero/química , Proteína 1 de Ligação a Repetições Teloméricas/genética , Proteína 1 de Ligação a Repetições Teloméricas/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
10.
PLoS One ; 10(3): e0115651, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25730259

RESUMO

Telomere binding factors viz. TRF1 and TRF2 are a part of sheltrin complex that are present exclusively at the ends of chromosomes. These factors play an important role in maintaining chromosomal integrity at the ends. However, their status and role are not clear in renal cell carcinoma (RCC). Therefore, the present study was conducted to evaluate TRF1 and TRF2 expressions in RCC tissues. Further, the role of these factors involved in tumorigenesis was elucidated by gene silencing using siRNA in RCC cell line (A498). The present study documented a significant over-expression of TRF1 (P = 0.005) and TRF2 (P = 0.0048) mRNAs by real time PCR in RCC tissues as compared with adjacent normal kidney tissues. Immunohistochemistry studies also revealed higher expression of TRF1 and TRF2 proteins in RCC. Moreover, TRF1 or TRF2 gene silencing using siRNA showed marked reduction in proliferation of RCC cells (P = 0.000). Further, significantly induced cell cycle arrest (P = 0.000) and apoptosis of RCC cells (P = 0.000) was documented upon TRF1 or TRF2 gene silencing. Henceforth, the results deduce that TRF1 or TRF2 inhibitions play an important role in the induction of apoptosis in A498 cells, which may serve as a potential therapeutic target in RCC.


Assuntos
Carcinoma de Células Renais/patologia , Neoplasias Renais/patologia , RNA Interferente Pequeno/metabolismo , Proteína 1 de Ligação a Repetições Teloméricas/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Adulto , Idoso , Apoptose , Carcinoma de Células Renais/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular , Humanos , Imuno-Histoquímica , Rim/metabolismo , Neoplasias Renais/metabolismo , Masculino , Pessoa de Meia-Idade , Pontos de Checagem da Fase S do Ciclo Celular , Proteína 1 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Proteína 1 de Ligação a Repetições Teloméricas/genética , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Proteína 2 de Ligação a Repetições Teloméricas/genética , Regulação para Cima
11.
Am J Physiol Cell Physiol ; 308(5): C372-7, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25518961

RESUMO

The increased level of chromosome instability in cancer cells is not only a driving force for oncogenesis but also can be the Achille's heel of the disease since many chemotherapies kill cells by inducing a nontolerable rate of DNA damage. A wealth of published evidence showed that telomere stability can be more affected than the bulk of the genome by several conventional antineoplastic drugs. In the present study, HT1080 cell lines compromised for either telomere repeats binding factor 2 (TRF2) or POT1 were treated with ICRF-193 (3 µM, 24 h) or bleomycin (1 µM, 24 h). DNA damage was assayed by combining telomeric DNA staining of a (CCCTAA)n PNA probe with immunofluorescence of 53BP1 to score the rate of telomere colocalization with 53BP1 foci. We found that ICRF-193, but not bleomycin, leads to DNA damage preferentially at telomeres, which can be rescued by TRF2 inhibition. POT1 inhibition exacerbates telomere dysfunction induced by ICRF-193. Thus, ICRF-193 induces damage at telomeres properly capped by TRF2 but not by POT1. These findings are expected to broaden our view on the mechanism by which conventional therapeutic molecules act to eliminate cancer cells and how to use TRF2 and POT1 levels as surrogate markers for anti-topoisomerase II sensitivity.


Assuntos
Sistemas de Liberação de Medicamentos , Piperazinas/administração & dosagem , Telômero/efeitos dos fármacos , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Inibidores da Topoisomerase II/administração & dosagem , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/fisiologia , Dicetopiperazinas , Sistemas de Liberação de Medicamentos/métodos , Células HEK293 , Humanos , Telômero/metabolismo , Telômero/patologia , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
12.
J Am Chem Soc ; 136(48): 16708-11, 2014 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-25393214

RESUMO

The shelterin protein TRF2 has come to the limelight for its role in telomere maintenance and tumorigenesis. Herein, the application of rational design and synthesis allowed identifying the first TRF2TRFH binder able to elicit a marked DNA damage response in cancer cells. This work paves the way for the unprecedented employment of a chemical tool to finely tune specific mechanisms underlying telomere maintenance.


Assuntos
Desenho de Fármacos , Peptídeos Cíclicos/farmacologia , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Humanos , Modelos Moleculares , Peptídeos Cíclicos/síntese química , Peptídeos Cíclicos/química , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
13.
Exp Cell Res ; 322(2): 324-34, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24485912

RESUMO

CARF is an ARF-binding protein that has been shown to regulate the p53-p21-HDM2 pathway. CARF overexpression was shown to cause growth arrest of human cancer cells and premature senescence of normal cells through activation of the p53 pathway. Because replicative senescence involves permanent withdrawal from the cell cycle in response to DNA damage response-mediated signaling, in the present study we investigated the relationship between CARF and the cell cycle and whether it is involved in the DNA damage response. We demonstrate that the half-life of CARF protein is less than 60 min, and that in cycling cells CARF levels are highest in G2 and early prophase. Serially passaged normal human skin and stromal fibroblasts showed upregulation of CARF during replicative senescence. Induction of G1 growth arrest and senescence by a variety of drugs was associated with increase in CARF expression at the transcriptional and translational level and was seen to correlate with increase in DNA damage response and checkpoint proteins, ATM, ATR, CHK1, CHK2, γH2AX, p53 and p21. Induction of growth arrest by oncogenic RAS and shRNA-mediated knockdown of TRF2 in cancer cells also caused upregulation of CARF. We conclude that CARF is associated with DNA damage response and checkpoint signaling pathways.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Pontos de Checagem do Ciclo Celular/fisiologia , Dano ao DNA/fisiologia , Fibroblastos/metabolismo , Proteínas de Ligação a RNA/metabolismo , Pele/metabolismo , Células Estromais/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Western Blotting , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Senescência Celular/fisiologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Imunofluorescência , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Pele/citologia , Pele/efeitos dos fármacos , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Proteína 2 de Ligação a Repetições Teloméricas/genética
14.
Nat Cell Biol ; 15(7): 818-28, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23792691

RESUMO

Dysfunctional telomeres suppress tumour progression by activating cell-intrinsic programs that lead to growth arrest. Increased levels of TRF2, a key factor in telomere protection, are observed in various human malignancies and contribute to oncogenesis. We demonstrate here that a high level of TRF2 in tumour cells decreased their ability to recruit and activate natural killer (NK) cells. Conversely, a reduced dose of TRF2 enabled tumour cells to be more easily eliminated by NK cells. Consistent with these results, a progressive upregulation of TRF2 correlated with decreased NK cell density during the early development of human colon cancer. By screening for TRF2-bound genes, we found that HS3ST4--a gene encoding for the heparan sulphate (glucosamine) 3-O-sulphotransferase 4--was regulated by TRF2 and inhibited the recruitment of NK cells in an epistatic relationship with TRF2. Overall, these results reveal a TRF2-dependent pathway that is tumour-cell extrinsic and regulates NK cell immunity.


Assuntos
Neoplasias da Mama/prevenção & controle , Neoplasias do Colo/prevenção & controle , Células Matadoras Naturais/imunologia , Linfócitos do Interstício Tumoral/imunologia , Melanoma Experimental/prevenção & controle , Sulfotransferases/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Animais , Apoptose , Western Blotting , Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Adesão Celular , Proliferação de Células , Neoplasias do Colo/imunologia , Neoplasias do Colo/metabolismo , Primers do DNA/química , Receptor com Domínio Discoidina 1 , Feminino , Citometria de Fluxo , Células HeLa , Humanos , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Linfócitos do Interstício Tumoral/patologia , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Camundongos , Camundongos Nus , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sulfotransferases/genética , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Proteína 2 de Ligação a Repetições Teloméricas/genética , Células Tumorais Cultivadas
15.
Cell Res ; 23(5): 705-19, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23459151

RESUMO

Human single-strand (ss) DNA binding proteins 1 and 2 (hSSB1 and 2) are components of the hSSB1/2-INTS3-C9orf80 heterotrimeric protein complex shown to participate in DNA damage response and maintenance of genome stability. However, their roles at telomeres remain unknown. Here, we generated murine SSB1 conditional knockout mice and cells and found that mSSB1 plays a critical role in telomere end protection. Both mSSB1 and mSSB2 localize to a subset of telomeres and are required to repair TRF2-deficient telomeres. Deletion of mSSB1 resulted in increased chromatid-type fusions involving both leading- and lagging-strand telomeric DNA, suggesting that it is required for the protection of G-overhangs. mSSB1's interaction with INTS3 is required for its localization to damaged DNA. mSSB1 interacts with Pot1a, but not Pot1b, and its association with telomeric ssDNA requires Pot1a. mSSB1(Δ/Δ) mice die at birth with developmental abnormalities, while mice with the hypomorphic mSSB1(F/F) allele are born alive and display increased sensitivity to ionizing radiation (IR). Our results suggest that mSSB1 is required to maintain genome stability, and document a previously unrecognized role for mSSB1/2 in the protection of newly replicated leading- and lagging-strand telomeres.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas Mitocondriais/metabolismo , Telômero/metabolismo , Alelos , Animais , Linhagem Celular , Cromátides/metabolismo , Dano ao DNA/efeitos da radiação , Reparo do DNA , DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Instabilidade Genômica , Humanos , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/antagonistas & inibidores , Proteínas Mitocondriais/genética , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Radiação Ionizante , Complexo Shelterina , Proteínas de Ligação a Telômeros , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Proteína 2 de Ligação a Repetições Teloméricas/genética , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
16.
J Vasc Res ; 50(2): 109-23, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23172421

RESUMO

The anthracycline doxorubicin is a widely used effective anti-cancer drug. However, its application and dosage are severely limited due to its cardiotoxicity. The exact mechanisms of doxorubicin-induced cardiotoxic side effects remain poorly understood. Even less is known about the impact of doxorubicin treatment on vascular damage. We found that low doses of doxorubicin induced a senescent response in human primary vascular smooth muscle cells (VSMC). We observed that expression of urokinase receptor (uPAR) was upregulated in response to doxorubicin. Furthermore, the level of uPAR expression played a decisive role in developing doxorubicin-induced senescence. uPAR silencing in human VSMC by means of RNA interference as well as uPAR knockout in mouse VSMC resulted in abrogation of doxorubicin-induced cellular senescence. On the contrary, uPAR overexpression promoted VSMC senescence. We further found that proteasomal degradation of telomeric repeat binding factor 2 (TRF2) mediates doxorubicin-induced VSMC senescence. Our results demonstrate that uPAR controls the ubiquitin-proteasome system in VSMC and regulates doxorubicin-induced TRF2 ubiquitination and proteasomal degradation via this mechanism. Therefore, VSMC senescence induced by low doses of doxorubicin may contribute to vascular damage upon doxorubicin treatment. uPAR-mediated TRF2 ubiquitination and proteasomal degradation are further identified as a molecular mechanism underlying this process.


Assuntos
Senescência Celular/efeitos dos fármacos , Doxorrubicina/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Animais , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Doxorrubicina/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/metabolismo , Cultura Primária de Células , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/deficiência , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Proteínas Recombinantes de Fusão/fisiologia , Telômero/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Proteína 2 de Ligação a Repetições Teloméricas/genética , Transfecção , Ubiquitinação/efeitos dos fármacos , Artérias Umbilicais/citologia
17.
Expert Opin Ther Targets ; 15(5): 579-93, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21288186

RESUMO

INTRODUCTION: The limitless replicative potential of cancer cells relies on telomere integrity (which is guaranteed by a complex interaction between several specialized proteins and telomeric DNA) and the activation of specific mechanisms for telomere length maintenance. Two mechanisms are currently known in human cancer, namely telomerase activity and the alternative lengthening of telomere pathway. EXPERT OPINION: In this review, we summarize the available data concerning the therapeutic strategies proposed thus far and the current challenges posed for the development of innovative telomere-based therapeutic approaches with broad-spectrum anticancer activity and for their translation into the clinical setting. AREAS COVERED: Due to their essential role in tumor cell proliferation, telomere maintenance mechanisms have become extremely attractive targets for the development of new anticancer interventions. Although numerous efforts have been made to identify specific approaches to interfere with telomere maintenance mechanisms in human cancers, the only molecule currently tested in clinical trials is the oligonucleotide GRN163L. However, a growing body of evidence suggests that interfering with telomeres, through the direct targeting of telomeric G-quadruplex structures, may be a valuable antitumor therapeutic strategy, independent of the specific telomere maintenance mechanism operating in the tumor.


Assuntos
Neoplasias/tratamento farmacológico , Neoplasias/genética , Telômero/efeitos dos fármacos , Animais , Quadruplex G/efeitos dos fármacos , Humanos , Telomerase/antagonistas & inibidores , Telômero/fisiologia , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores
18.
Cancer Biol Ther ; 8(22): 2166-74, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19783902

RESUMO

EXPERIMENTAL DESIGN: Telomere repeat binding factor 2 (TRF2) plays a key role in the protective activity of telomere and is overexpression in several kinds of solid cancer cells. However, the role of overexpressed TRF2 in colorectal carcinoma remains unclear. The aim of this study was to determine the expression of TRF2, address the mechanism of TRF2 overexpression in human colorectal carcinoma. In present study, we examined the expression of TRF2 in colorectal cancer tissues from 39 patients, peritumoral normal tissues from 21 patients, and colon carcinoma SW480 cell line by quantitative PCR, immunohistochemistry and western blot. After siRNA silencing TRF2 expression in SW480, tumorigenesis of TRF2 was tested by cell proliferation, soft agar assay, cytofluorimetric analysis and cytogenetic analysis. To discover transcription factor that mediated TRF2 expression, Chromatin Immunoprecipitation (Chip) Assay and Electrophoretic mobility shift assays (EMSA) were employed. RESULTS: Overexpression of TRF2 protein was detected in SW480 cells and 19 of 39 colorectal carcinoma tissues (49%), no overexpression was observed in 21 of 21 adjacent peritumoral normal colorectal tissues. After siRNA silencing TRF2 expression, the proliferation and colony formation of SW480 cells were significantly inhibited. Defective TRF2 induced apoptosis and increased chromosomal instability in SW480 cells, in which there were more end-to-end fusions and ring chromosomes. Chip assay and EMSA showed that transcription factor Sp1 is involved in upregulation of TRF2. These results indicate that TRF2 is overexpressed in colorectal carcinoma, Sp1 upregulates TRF2 expression, TRF2 inhibition reduces tumorigenesis of colorectal cancer, which suggests that TRF2 and SP1 may become new targets for the development of anti-cancer therapy in colorectal carcinoma.


Assuntos
Adenocarcinoma/patologia , Neoplasias Colorretais/patologia , Proteínas de Neoplasias/fisiologia , Fator de Transcrição Sp1/fisiologia , Proteína 2 de Ligação a Repetições Teloméricas/fisiologia , Adenocarcinoma/química , Adenocarcinoma/genética , Apoptose , Divisão Celular , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Neoplasias Colorretais/química , Neoplasias Colorretais/genética , Ensaio de Desvio de Mobilidade Eletroforética , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Telômero/metabolismo , Telômero/ultraestrutura , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Proteína 2 de Ligação a Repetições Teloméricas/biossíntese , Proteína 2 de Ligação a Repetições Teloméricas/genética , Ensaio Tumoral de Célula-Tronco , Regulação para Cima
19.
Cancer Biol Ther ; 8(12): 1136-45, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19363303

RESUMO

Hydroxyurea (HU) is a chemotherapeutic agent commonly used for various malignancies and hematological disorders, including chronic myelogenous leukemia and sickle cell anemia. We show here that chronic, low-level treatment with HU induces a variety of defects in telomere replication and maintenance. HU treatment preferentially decreased the rate of telomere DNA synthesis and altered the cell cycle timing of telomere replication. HU reduced the expression levels of telomere repeat RNA (TERRA). In some cells, HU caused a rapid loss of telomere restriction fragment length. Chromatin immunoprecipitation (ChIP) assay indicated that telomere repeat binding factors TRF1 and TRF2 dissociate from telomere DNA after HU treatment. TRF2 protein purified from HU treated cells showed a modest reduction in DNA binding activity and a change in isoelectric point as measured by 2D gel electrophoresis. However, chronic low level HU treatment did not evoke a DNA replication checkpoint response, suggesting that the mechanism of action is distinct from the well-characterized S-phase checkpoint pathway. We conclude that therapeutic doses of HU preferentially effects telomere replication and maintenance, through a mechanism that may involve the direct modification of TRF2. These findings provide new insight into the potential mechanisms of action of HU at telomeres and in cancer chemotherapies.


Assuntos
Antineoplásicos/farmacologia , Hidroxiureia/farmacologia , Neoplasias/terapia , Telômero/efeitos dos fármacos , Telômero/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Técnicas de Cultura de Células , Imunoprecipitação da Cromatina , Células HCT116 , Células HeLa , Humanos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas de Ligação a Telômeros/genética , Proteínas de Ligação a Telômeros/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/genética , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Transfecção
20.
Cancer Biol Ther ; 6(8): 1186-92, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17643074

RESUMO

Overexpression of human telomere repeat binding factor 2 (TRF2), which may play an important role in the fate of cancer cells, has been observed in adult T-cell leukemia. Previous reports have shown that the inhibition of TRF2 results in the apoptosis of cancer cells. In this study, we demonstrated that arsenic trioxide (As2O3) induced in vitro growth inhibition and/or apoptosis of human T-cell leukemia cell line Molt-4 in a caspase-independent manner. Telomerase activity was not inhibited, although the level of the reverse transcriptase subunit of the human telomerase gene (hTERT) mRNA expression was down regulated during the early times and then recovered to the level found in untreated controls about 48 hours after treatment with As2O3. Furthermore, a remarkable telomere shortening related to exposure of As2O3 was observed in 50 population doubling. Inc ontrast, the alteration of telomere length did not occur after exposure to higher concentration of As2O3 (10 microM) for 24 hours and 48 hours, respectively, suggesting that the shortening of telomeres induced by As2O3 is dependent of a series of cell division cycles. Chromosomal analysis showed that As2O3 exposure caused chromosomal end-to-end fusion in human T-cell leukemia cells while downregulation of TRF2 was observed. Finally, the inhibition of TRF2 protein expression and the sensitivity to As2O3 in a panel of leukemia cell lines were checked. The data revealed that inhibition of TRF2 rendered leukemia cells more susceptible to As2O3. In conclusion, the downregulation of TRF2 by As2O3 contribute to chromosomal end-to-end fusion, and apoptosis in leukemia cells, suggesting that TRF2 could be an attractive target for new therapies of leukemia.


Assuntos
Antineoplásicos/farmacologia , Arsenicais/farmacologia , Instabilidade Cromossômica , Leucemia-Linfoma de Células T do Adulto/metabolismo , Óxidos/farmacologia , Telômero/efeitos dos fármacos , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Apoptose , Trióxido de Arsênio , Caspases/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Leucemia-Linfoma de Células T do Adulto/genética , Telomerase/antagonistas & inibidores , Telomerase/genética , Telomerase/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...