Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Genes (Basel) ; 14(4)2023 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-37107534

RESUMO

The nuclear envelope (NE) in eukaryotic cells is essential to provide a protective compartment for the genome. Beside its role in connecting the nucleus with the cytoplasm, the NE has numerous important functions including chromatin organization, DNA replication and repair. NE alterations have been linked to different human diseases, such as laminopathies, and are a hallmark of cancer cells. Telomeres, the ends of eukaryotic chromosomes, are crucial for preserving genome stability. Their maintenance involves specific telomeric proteins, repair proteins and several additional factors, including NE proteins. Links between telomere maintenance and the NE have been well established in yeast, in which telomere tethering to the NE is critical for their preservation and beyond. For a long time, in mammalian cells, except during meiosis, telomeres were thought to be randomly localized throughout the nucleus, but recent advances have uncovered close ties between mammalian telomeres and the NE that play important roles for maintaining genome integrity. In this review, we will summarize these connections, with a special focus on telomere dynamics and the nuclear lamina, one of the main NE components, and discuss the evolutionary conservation of these mechanisms.


Assuntos
Membrana Nuclear , Telômero , Animais , Humanos , Membrana Nuclear/genética , Membrana Nuclear/metabolismo , Telômero/genética , Telômero/metabolismo , Replicação do DNA/genética , Proteínas de Ligação a Telômeros/genética , Proteínas de Ligação a Telômeros/metabolismo , Meiose , Mamíferos/genética , Mamíferos/metabolismo
2.
Nucleic Acids Res ; 49(17): 9886-9905, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34469544

RESUMO

Telomere maintenance is essential to preserve genomic stability and involves telomere-specific proteins, DNA replication and repair proteins. Lamins are key components of the nuclear envelope and play numerous roles, including maintenance of the nuclear integrity, regulation of transcription, and DNA replication. Elevated levels of lamin B1, one of the major lamins, have been observed in some human pathologies and several cancers. Yet, the effect of lamin B1 dysregulation on telomere maintenance remains unknown. Here, we unveil that lamin B1 overexpression drives telomere instability through the disruption of the shelterin complex. Indeed, lamin B1 dysregulation leads to an increase in telomere dysfunction-induced foci, telomeric fusions and telomere losses in human cells. Telomere aberrations were preceded by mislocalizations of TRF2 and its binding partner RAP1. Interestingly, we identified new interactions between lamin B1 and these shelterin proteins, which are strongly enhanced at the nuclear periphery upon lamin B1 overexpression. Importantly, chromosomal fusions induced by lamin B1 in excess were rescued by TRF2 overexpression. These data indicated that lamin B1 overexpression triggers telomere instability through a mislocalization of TRF2. Altogether our results point to lamin B1 as a new interacting partner of TRF2, that is involved in telomere stability.


Assuntos
Lamina Tipo B/metabolismo , Complexo Shelterina/metabolismo , Telômero/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Células Cultivadas , Humanos , Lamina Tipo B/química , Proteínas de Ligação a Telômeros/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/química
3.
Sci Adv ; 7(35)2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34452908

RESUMO

Double-strand breaks (DSBs) are harmful lesions and a major cause of genome instability. Studies have suggested a link between the nuclear envelope and the DNA damage response. Here, we show that lamin B1, a major component of the nuclear envelope, interacts directly with 53BP1 protein, which plays a pivotal role in the DSB repair. This interaction is dissociated after DNA damage. Lamin B1 overexpression impedes 53BP1 recruitment to DNA damage sites and leads to a persistence of DNA damage, a defect in nonhomologous end joining and an increased sensitivity to DSBs. The identification of interactions domains between lamin B1 and 53BP1 allows us to demonstrate that the defect of 53BP1 recruitment and the DSB persistence upon lamin B1 overexpression are due to sequestration of 53BP1 by lamin B1. This study highlights lamin B1 as a factor controlling the recruitment of 53BP1 to DNA damage sites upon injury.


Assuntos
Quebras de DNA de Cadeia Dupla , Lamina Tipo B , Dano ao DNA , Reparo do DNA por Junção de Extremidades , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo
4.
J Exp Med ; 213(6): 1011-28, 2016 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-27185855

RESUMO

Inherited bone marrow failure syndromes are human conditions in which one or several cell lineages of the hemopoietic system are affected. They are present at birth or may develop progressively. They are sometimes accompanied by other developmental anomalies. Three main molecular causes have been recognized to result in bone marrow failure syndromes: (1) defects in the Fanconi anemia (FA)/BRCA DNA repair pathway, (2) defects in telomere maintenance, and (3) abnormal ribosome biogenesis. We analyzed a patient with mild bone marrow failure and microcephaly who did not present with the typical FA phenotype. Cells from this patient showed increased sensitivity to ionizing radiations and phleomycin, attesting to a probable DNA double strand break (dsb) repair defect. Linkage analysis and whole exome sequencing revealed a homozygous nonsense mutation in the ERCC6L2 gene. We identified a new ERCC6L2 alternative transcript encoding the DNA repair factor Hebo, which is critical for complementation of the patient's DNAdsb repair defect. Sequence analysis revealed three structured regions within Hebo: a TUDOR domain, an adenosine triphosphatase domain, and a new domain, HEBO, specifically present in Hebo direct orthologues. Hebo is ubiquitously expressed, localized in the nucleus, and rapidly recruited to DNAdsb's in an NBS1-dependent manner.


Assuntos
Doenças da Medula Óssea , Núcleo Celular , Códon sem Sentido , DNA Helicases , Homozigoto , Microcefalia , Adolescente , Doenças da Medula Óssea/genética , Doenças da Medula Óssea/metabolismo , Doenças da Medula Óssea/patologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Células Cultivadas , Quebras de DNA de Cadeia Dupla , DNA Helicases/biossíntese , DNA Helicases/genética , Feminino , Regulação da Expressão Gênica , Ligação Genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Microcefalia/genética , Microcefalia/metabolismo , Microcefalia/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Domínios Proteicos
5.
Cell Rep ; 5(1): 21-8, 2013 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-24095737

RESUMO

The choice of the appropriate double-strand break (DSB) repair pathway is essential for the maintenance of genomic stability. Here, we show that the Bloom syndrome gene product, BLM, counteracts CtIP/MRE11-dependent long-range deletions (>200 bp) generated by alternative end-joining (A-EJ). BLM represses A-EJ in an epistatic manner with 53BP1 and RIF1 and is required for ionizing-radiation-induced 53BP1 focus assembly. Conversely, in the absence of 53BP1 or RIF1, BLM promotes formation of A-EJ long deletions, consistent with a role for BLM in DSB end resection. These data highlight a dual role for BLM that influences the DSB repair pathway choice: (1) protection against CtIP/MRE11 long-range deletions associated with A-EJ and (2) promotion of DNA resection. These antagonist roles can be regulated, according to cell-cycle stage, by interacting partners such as 53BP1 and TopIII, to avoid unscheduled resection that might jeopardize genome integrity.


Assuntos
Proteínas de Transporte/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Proteínas de Ligação a DNA/genética , Proteínas Nucleares/genética , RecQ Helicases/genética , RecQ Helicases/metabolismo , Sequência de Aminoácidos , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/metabolismo , Endodesoxirribonucleases , Humanos , Proteína Homóloga a MRE11 , Proteínas Nucleares/metabolismo , Transfecção
6.
Nucleus ; 3(5): 411-7, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22895091

RESUMO

Progeroid phenotypes are mainly encountered in 2 types of syndromes: in laminopathies, which are characterized by nuclear shape abnormalities due to lamin A alteration, and in DNA damage response defect syndromes. Because lamin A dysregulation leads to DNA damages, it has been proposed that senescence occurs in both types of syndromes through the accumulation of damages. We recently showed that elevated oxidative stress is responsible for lamin B1 accumulation, nuclear shape alteration and senescence in the DDR syndrome, ataxia telangiectasia (A-T). Interestingly, overexpression of lamin B1 in wild type cells is sufficient to induce senescence without the induction of DNA damages. Here, we will discuss the importance of controlling the lamins level in order for maintenance nuclear architecture and we will comment the relationships of lamins with other senescence mechanisms. Finally, we will describe emerging data reporting redox control by lamins, leading us to propose a general mechanism by which reactive oxygen species can induce senescence through lamin dysregulation and NSA.


Assuntos
Núcleo Celular/metabolismo , Senescência Celular , Lamina Tipo A/metabolismo , Lamina Tipo B/metabolismo , Estresse Oxidativo , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/metabolismo , Forma do Núcleo Celular , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Humanos , Camundongos , Oxirredução , Progéria/metabolismo , Progéria/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Telômero/metabolismo , Proteínas Supressoras de Tumor/metabolismo
7.
EMBO J ; 31(5): 1080-94, 2012 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-22246186

RESUMO

We report crosstalk between three senescence-inducing conditions, DNA damage response (DDR) defects, oxidative stress (OS) and nuclear shape alterations. The recessive autosomal genetic disorder Ataxia telangiectasia (A-T) is associated with DDR defects, endogenous OS and premature ageing. Here, we find frequent nuclear shape alterations in A-T cells, as well as accumulation of the key nuclear architecture component lamin B1. Lamin B1 overexpression is sufficient to induce nuclear shape alterations and senescence in wild-type cells, and normalizing lamin B1 levels in A-T cells reciprocally reduces both nuclear shape alterations and senescence. We further show that OS increases lamin B1 levels through p38 Mitogen Activated Protein kinase activation. Lamin B1 accumulation and nuclear shape alterations also occur during stress-induced senescence and oncogene-induced senescence (OIS), two canonical senescence situations. These data reveal lamin B1 as a general molecular mediator that controls OS-induced senescence, independent of established Ataxia Telangiectasia Mutated (ATM) roles in OIS.


Assuntos
Envelhecimento , Lamina Tipo B/metabolismo , Estresse Oxidativo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Células Cultivadas , Humanos
8.
Cancer Res ; 71(10): 3590-602, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21444675

RESUMO

Genetic stability requires coordination of a network of pathways including DNA repair/recombination and apoptosis. In addition to its canonical anti-apoptotic role, Bcl-2 negatively impacts genome stability. In this study, we identified the breast cancer tumor suppressor BRCA1, which plays an essential role in homologous recombination (HR), as a target for Bcl-2 in the repression of HR. Indeed, ionizing radiation-induced BRCA1 foci assembly was repressed when Bcl-2 was expressed ectopically, in human SV40 fibroblasts, or spontaneously, in lymphoma t(14:18) cells and in HeLa and H460 cancer cell lines. Moreover, we showed that the transmembrane (TM) domain of Bcl-2 was required for both inhibition of BRCA1 foci assembly and the inhibition of HR induced by a double-strand break targeted into an intrachromosomal HR substrate by the meganuclease I-SceI. Fluorescence confocal microscopy, proximity ligation assay, and electron microscopy analyses as well as Western blot analysis of subcellular fractions showed that Bcl-2 and BRCA1 colocalized to mitochondria and endoplasmic reticulum in a process requiring the TM domain of Bcl-2. Targeting BRCA1 to the endomembranes depletes BRCA1 from the nucleus and, thus, accounts for the inhibition of HR. Furthermore, our findings support an apoptosis-stimulatory role for the cytosolic form of BRCA1, suggesting a new tumor suppressor function of BRCA1. Together, our results reveal a new mode of BRCA1 regulation and for HR in the maintenance of genome stability.


Assuntos
Membrana Celular/metabolismo , Regulação Neoplásica da Expressão Gênica , Genes BRCA1 , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Recombinação Genética , Linhagem Celular Tumoral , Citoplasma/metabolismo , Retículo Endoplasmático/metabolismo , Células HeLa , Humanos , Microscopia Confocal/métodos , Microscopia de Fluorescência/métodos , Mitocôndrias/metabolismo , Modelos Genéticos , Frações Subcelulares/metabolismo
9.
Nucleic Acids Res ; 36(5): 1741-54, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18263609

RESUMO

Telomeres are known to prevent chromosome ends from being recognized as DNA double-strand breaks. Conversely, many DNA damage response proteins, including ATM, are thought to participate to telomere maintenance. However, the precise roles of ATM at telomeres remain unclear due to its multiple functions in cell checkpoints and apoptosis. To gain more insights into the role of ATM in telomere maintenance, we determined the effects of the G-quadruplex ligand 360A in various cell lines lacking functional ATM. We showed, by using Fluorescence in situ hybridization (FISH) and Chromosome Orientation-FISH using telomere PNA probes, that 360A induced specific telomere aberrations occurring during or after replication, mainly consisting in sister telomere fusions and also recombinations that involved preferentially the lagging strand telomeres. We demonstrate that ATM reduced telomere instability independently of apoptosis induction. Our results suggest thus that ATM has a direct role in preventing inappropriate DNA repair at telomeres, which could be related to its possible participation to the formation of protected structures at telomeres.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Reparo do DNA , Proteínas de Ligação a DNA/fisiologia , Quadruplex G/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/fisiologia , Piridinas/toxicidade , Quinolinas/toxicidade , Telômero/química , Proteínas Supressoras de Tumor/fisiologia , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia , Ciclo Celular , Proteínas de Ciclo Celular/antagonistas & inibidores , Aberrações Cromossômicas , Dano ao DNA , Proteínas de Ligação a DNA/antagonistas & inibidores , Células HeLa , Humanos , Ligantes , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Transdução de Sinais , Troca de Cromátide Irmã/efeitos dos fármacos , Telômero/efeitos dos fármacos , Proteínas Supressoras de Tumor/antagonistas & inibidores
10.
Am J Respir Crit Care Med ; 174(8): 858-66, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16858015

RESUMO

RATIONALE: Primary ciliary dyskinesia (PCD) is a rare, usually autosomal recessive, genetic disorder characterized by ciliary dysfunction, sino-pulmonary disease, and situs inversus. Disease-causing mutations have been reported in DNAI1 and DNAH5 encoding outer dynein arm (ODA) proteins of cilia. OBJECTIVES: We analyzed DNAI1 to identify disease-causing mutations in PCD and to determine if the previously reported IVS1+2_3insT (219+3insT) mutation represents a "founder" or "hot spot" mutation. METHODS: Patients with PCD from 179 unrelated families were studied. Exclusion mapping showed no linkage to DNAI1 for 13 families; the entire coding region was sequenced in a patient from the remaining 166 families. Reverse transcriptase-polymerase chain reaction (RT-PCR) was performed on nasal epithelial RNA in 14 families. RESULTS: Mutations in DNAI1 including 12 novel mutations were identified in 16 of 179 (9%) families; 14 harbored biallelic mutations. Deep intronic splice mutations were not identified by reverse transcriptase-polymerase chain reaction. The prevalence of mutations in families with defined ODA defect was 13%; no mutations were found in patients without a defined ODA defect. The previously reported IVS1+2_3insT mutation accounted for 57% (17/30) of mutant alleles, and marker analysis indicates a common founder for this mutation. Seven mutations occurred in three exons (13, 16, and 17); taken together with previous reports, these three exons are emerging as mutation clusters harboring 29% (12/42) of mutant alleles. CONCLUSIONS: A total of 10% of patients with PCD are estimated to harbor mutations in DNAI1; most occur as a common founder IVS1+2_3insT or in exons 13, 16, and 17. This information is useful for establishing a clinical molecular genetic test for PCD.


Assuntos
DNA/genética , Dineínas/genética , Síndrome de Kartagener/genética , Mutação , Adolescente , Adulto , Idoso , Dineínas do Axonema , Criança , Pré-Escolar , Éxons , Feminino , Efeito Fundador , Frequência do Gene , Genótipo , Humanos , Lactente , Masculino , Linhagem , Fenótipo , Reação em Cadeia da Polimerase
11.
Nucleic Acids Res ; 33(13): 4182-90, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16052031

RESUMO

The G-overhangs of telomeres are thought to adopt particular conformations, such as T-loops or G-quadruplexes. It has been suggested that G-quadruplex structures could be stabilized by specific ligands in a new approach to cancer treatment consisting in inhibition of telomerase, an enzyme involved in telomere maintenance and cell immortality. Although the formation of G-quadruplexes was demonstrated in vitro many years ago, it has not been definitively demonstrated in living human cells. We therefore investigated the chromosomal binding of a tritiated G-quadruplex ligand, 3H-360A (2,6-N,N'-methyl-quinolinio-3-yl)-pyridine dicarboxamide [methyl-3H]. We verified the in vitro selectivity of 3H-360A for G-quadruplex structures by equilibrium dialysis. We then showed by binding experiments with human genomic DNA that 3H-360A has a very potent selectivity toward G-quadruplex structures of the telomeric 3'-overhang. Finally, we performed autoradiography of metaphase spreads from cells cultured with 3H-360A. We found that 3H-360A was preferentially bound to chromosome terminal regions of both human normal (peripheral blood lymphocytes) and tumor cells (T98G and CEM1301). In conclusion, our results provide evidence that a specific G-quadruplex ligand interacts with the terminal ends of human chromosomes. They support the hypothesis that G-quadruplex ligands induce and/or stabilize G-quadruplex structures at telomeres of human cells.


Assuntos
Cromossomos Humanos/química , DNA/metabolismo , Piridinas/metabolismo , Quinolinas/metabolismo , Telômero/química , Sítios de Ligação , Linhagem Celular Tumoral , Células Cultivadas , Cromossomos Humanos/metabolismo , DNA/química , Quadruplex G , Guanina/química , Humanos , Ligantes , Linfócitos/ultraestrutura , Metáfase , Piridinas/química , Quinolinas/química , Telômero/metabolismo
12.
Oncogene ; 24(18): 2917-28, 2005 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-15735722

RESUMO

Telomerase represents a relevant target for cancer therapy. Molecules able to stabilize the G-quadruplex (G4), a structure adopted by the 3'-overhang of telomeres, are thought to inhibit telomerase by blocking its access to telomeres. We investigated the cellular effects of four new 2,6-pyridine-dicarboxamide derivatives displaying strong selectivity for G4 structures and strong inhibition of telomerase in in vitro assays. These compounds inhibited cell proliferation at very low concentrations and then induced a massive apoptosis within a few days in a dose-dependent manner in cultures of three telomerase-positive glioma cell lines, T98G, CB193 and U118-MG. They had also antiproliferative effects in SAOS-2, a cell line in which telomere maintenance involves an alternative lengthening of telomeres (ALT) mechanism. We show that apoptosis was preceded by multiple alterations of the cell cycle: activation of S-phase checkpoints, dramatic increase of metaphase duration and cytokinesis defects. These effects were not associated with telomere shortening, but they were directly related to telomere instability involving telomere end fusion and anaphase bridge formation. Pyridine-based G-quadruplex ligands are therefore promising agents for the treatment of various tumors including malignant gliomas.


Assuntos
Apoptose/fisiologia , Instabilidade Genômica/fisiologia , Telômero/genética , Telômero/metabolismo , Ciclo Celular , Divisão Celular/genética , Divisão Celular/imunologia , Glioma/tratamento farmacológico , Humanos , Ligantes , Piridinas/farmacologia , Telomerase/antagonistas & inibidores
13.
Oncogene ; 24(4): 541-51, 2005 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-15608689

RESUMO

Hepatoblasts are bipotent progenitors of both hepatocytes and cholangiocytes. The lack of stable in vitro culture systems for such cells makes it necessary to generate liver progenitor cell lines by means of immortalization. In this study, we describe the long-term behaviour of a clone of simian foetal hepatic progenitor cells immortalized by Simian virus 40 (SV40) large T-antigen (T-Ag) flanked by loxP sites. Immortalization was associated with the re-expression of telomerase activity, which decreased at late passages (population doubling 120) after more than a year in culture. This decrease was concomitant to telomere shortening and karyotypic instability. However, the chromosomes carrying the p53 gene remained intact and long-term immortalized progenitor cells maintained contact inhibition and proliferative properties. They also displayed the features of a normal bipotent phenotype. We constructed a retroviral vector expressing an inducible Cre recombinase and transferred it into the immortalized progenitors. Activation of the Cre recombinase by 4-hydroxy-tamoxifen induced SV40 T-Ag excision, leading to the death of cells expressing Cre recombinase. Immortalized progenitors at late passages stopped growing and eventually disappeared after transplantation into the livers of immunocompromised mice. These cells provide a novel model to study hepatic differentiation and carcinogenesis.


Assuntos
Antígenos Virais de Tumores/genética , Antígenos Virais de Tumores/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Vírus 40 dos Símios/genética , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/genética , Linhagem Celular Transformada , Proliferação de Células , Células Cultivadas , Cromossomos de Mamíferos/metabolismo , Haplorrinos , Cariotipagem , Camundongos , Camundongos Knockout , Telomerase/metabolismo , Telômero/metabolismo , Fatores de Tempo
14.
J Cell Biochem ; 93(5): 968-79, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15389875

RESUMO

TRF1 and Pin2 play an essential role in telomere homeostasis, by regulating telomere maintenance. They are generated from the same gene, TRF1/Pin2, by alternative splicing but no functional differences between these proteins have been demonstrated. We report here the detection of new alternative transcripts of the TRF1/Pin2 gene in peripheral blood lymphocytes resulting from a 76 nt insertion. Real-time RT-PCR showed that these transcripts were also produced in various normal human cells and tissues and in immortalized cell lines, but at levels lower (by a factor of 8-111) than those for the TRF1 and Pin2 transcripts. These new transcripts are predicted to encode polypeptides identical to TRF1/Pin2 at the C-terminal end but entirely lacking the acid domain and the amino-terminal part of the homodimerization domain of TRF1/Pin2. These proteins, fused at their N-terminal ends to enhanced green fluorescent protein (EGFP), were found to be located at telomeres and to induce apoptosis in cell lines with short telomeres, thereby displaying similar activity to TRF1/Pin2. However, these putative proteins lack regions important for interactions with other proteins and for homodimerization. Unlike TRF1/Pin2, they were unable to interact with tankyrase 1, suggesting that these proteins may play a role in telomere homeostasis different from those of TRF1/Pin2. The production of these alternative transcripts was down-regulated in peripheral blood lymphocytes following PHA-p activation, suggesting a possible role in resting lymphocytes.


Assuntos
Processamento Alternativo , Telômero/metabolismo , Proteína 1 de Ligação a Repetições Teloméricas/genética , Proteína 1 de Ligação a Repetições Teloméricas/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Humanos , Linfócitos/metabolismo , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Tanquirases/metabolismo , Distribuição Tecidual
15.
Cytotechnology ; 45(1-2): 75-90, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19003245

RESUMO

The enzyme telomerase is involved in the replication of telomeres, specialized structures that cap and protect the ends of chromosomes. Its activity is required for maintenance of telomeres and for unlimited lifespan, a hallmark of cancer cells. Telomerase is overexpressed in the vast majority of human cancer cells and therefore represents an attractive target for therapy. Several approaches have been developed to inhibit this enzyme through the targeting of its RNA or catalytic components as well as its DNA substrate, the single-stranded 3'-telomeric overhang. Telomerase inhibitors are chemically diverse and include modified oligonucleotides as well as small diffusable molecules, both natural and synthetic. This review presents an update of recent investigations pertaining to these agents and discusses their biological properties in the context of the initial paradigm that the exposure of cancer cells to these agents should lead to progressive telomere shortening followed by a delayed growth arrest response.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA