Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Virus Res ; 145(1): 112-20, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19576936

RESUMO

The Adenovirus (Ad) dl309 mutant, which lacks several E3 region genes, has been used as the backbone for a number of replication selective cytopathic Ads designed to treat tumours. We report that dl309 has enhanced cytopathogenicity in a range of different cell lines when compared with Ad5. The E3 region modifications found in dl309 contributed to reduced late gene expression in both cocksackie-adenovirus receptor (CAR) positive and negative cells. We show that completion of the dl309 viral lifecycle was less efficient and apoptosis was triggered in the CAR negative K1 thyroid cancer-derived cell line. There was increased E1A expression in dl309-infected K1 cells, compared with Ad5, and significantly, whereas E1A in Ad5-infected cells was distributed both in the nuclear and cytoplasmic compartments, E1A was predominantly nuclear in dl309-infected K1 cells. From these results we conclude that the regions of dl309 that are deleted or otherwise modified can contribute to viral replication and inhibition of apoptosis, possibly indirectly by regulating E1A. These data have implications in the development of dl309-based Ads for the treatment of tumours in vivo.


Assuntos
Adenoviridae/patogenicidade , Proteínas E3 de Adenovirus/deficiência , Apoptose , Terapia Viral Oncolítica , Vírus Oncolíticos/patogenicidade , Neoplasias da Glândula Tireoide/terapia , Replicação Viral , Adenoviridae/fisiologia , Proteínas E1A de Adenovirus/biossíntese , Proteínas E3 de Adenovirus/genética , Linhagem Celular Tumoral , Receptor Constitutivo de Androstano , Regulação Neoplásica da Expressão Gênica , Regulação Viral da Expressão Gênica , Genoma Viral , Interações Hospedeiro-Patógeno , Humanos , Vírus Oncolíticos/fisiologia , Receptores Citoplasmáticos e Nucleares/biossíntese , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/virologia , Virulência
2.
Int J Cancer ; 121(7): 1445-54, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17582609

RESUMO

We previously showed that phorbol esters are cytotoxic to human thyroid epithelial cells expressing a mutant RAS oncogene. Here we explore the generality of this finding using cells derived from pancreatic cancer, which, like thyroid, shows a high frequency of RAS mutation, but is a much greater cause of cancer mortality. The response to phorbol myristate acetate (PMA) and related agents was assessed on a panel of 9 pancreatic cancer cell lines, using a range of assays for cell growth and death in vitro and in vivo. In most lines, PMA induced non-apoptotic cell death which was, surprisingly, independent of its "classic" target, protein kinase C. With 24 hr exposure, the IC(50) in the most sensitive line (Aspc-1) was <1 ng/ml (1.6 nM), with survival undetectable at concentrations >/=>/=16 nM, and after only 1 hr exposure the IC(50) was still

Assuntos
Neoplasias Pancreáticas/tratamento farmacológico , Ésteres de Forbol/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Humanos , Indóis/farmacologia , Concentração Inibidora 50 , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Maleimidas/farmacologia , Proteínas de Membrana/metabolismo , Camundongos , Substrato Quinase C Rico em Alanina Miristoilada , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Dibutirato de 12,13-Forbol/farmacologia , Fosforilação/efeitos dos fármacos , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
3.
Eur J Cancer ; 43(1): 194-201, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17046239

RESUMO

Normal human thyroid follicular epithelial cells exhibit a very low proliferative rate which in vitro is dramatically increased by RAS oncogene activation, resulting in clones displaying a phenotype consistent with that of a ras-induced follicular adenoma in vivo. Eventual spontaneous cessation of growth of these clones is closely correlated with increasing expression of the tumour suppressor gene p16(INK4a), suggesting that p16 may limit clonal expansion in this tumour model. We therefore hypothesised that p16 expression would also increase in vivo in follicular adenomas, and further that escape from growth control in follicular cancers would be accompanied by loss of p16 expression. This was tested using tissue microarrays, representing multiple stages of thyroid tumourigenesis. Whereas the majority of normal thyroids showed no immunostaining, p16 protein was readily detectable in follicular adenomas. Unexpectedly, however, p16 expression was also observed in follicular and papillary carcinomas. Poorly differentiated (insular) carcinomas showed either very intense staining, or a complete loss of staining. We conclude that loss of p16 is not necessary for malignant transformation in thyroid follicular cells, but that it may form one of two or more events needed for progression to more aggressive forms of thyroid cancer.


Assuntos
Adenoma/metabolismo , Transformação Celular Neoplásica/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclina A/metabolismo , Células Epiteliais/metabolismo , Humanos , Imuno-Histoquímica/métodos , Componente 2 do Complexo de Manutenção de Minicromossomo , Proteínas Nucleares/metabolismo
4.
Methods Enzymol ; 407: 648-60, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16757359

RESUMO

Activation of Ras oncogene by point mutations is an early frequent event in thyroid tumorigenesis. In this chapter, we describe the use of human primary thyroid follicular epithelial cells expressing oncogenic mutant Ras by means of retroviral transduction as a biological model of human cancer initiation that provides powerful insights into thyroid tumorigenesis. We describe protocols for manipulating primary epithelial cells and describe the use of this model to dissect the signaling pathways required for Ras-induced proliferation in these cells. We also highlight the importance of studying Ras signaling in an appropriate cell context, summarizing some of the key differences identified between more widespread experimental models based on fibroblasts or rodent cell lines and primary epithelial cells.


Assuntos
Transformação Celular Neoplásica , Glândula Tireoide/citologia , Proteínas ras/fisiologia , Células Epiteliais/patologia , Vetores Genéticos , Humanos , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Modelos Biológicos , Proteína Oncogênica p21(ras)/genética , Proteína Oncogênica p21(ras)/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Retroviridae/genética , Glândula Tireoide/patologia , Quinases raf/fisiologia , Fator ral de Troca do Nucleotídeo Guanina/fisiologia , Proteínas ras/genética
5.
Hum Pathol ; 37(7): 824-30, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16784981

RESUMO

Although the follicular variant of papillary thyroid carcinoma (FVPTC) has been classified as a papillary cancer based on nuclear features, its follicular growth pattern and potential for hematogenous spread are more characteristic of follicular carcinoma. To gain insight into the biologic nature of FVPTC, we compared genetic alterations characteristic of papillary and follicular thyroid carcinomas in 24 FVPTCs and 26 classic PTC (CPTCs). In FVPTCs, we observed ras mutation in 6 of 24 cases (25%), BRAF mutation in 1 of 13 cases (7.6%), and ret rearrangement in 5 of 12 cases (41.7%). In CPTCs, we found ras mutation in no case, BRAF mutation in 3 of 10 cases (30%), and ret rearrangement in 5 of 11 cases (45%). One FVPTC exhibited simultaneous ras mutation and ret/PTC1 rearrangement, and one CPTC harbored simultaneous BRAF mutation and ret/PTC3 rearrangement. Based on these findings, we concluded that ras mutation correlates with follicular differentiation of thyroid tumors whereas ret activation is associated with papillary nuclei but not with papillary architecture. ret activation is not exclusive of ras or BRAF mutation, whereas ras and BRAF mutations are mutually exclusive. The implications of these results for follicular and papillary carcinogenesis are discussed.


Assuntos
Adenocarcinoma Folicular/genética , Adenocarcinoma Papilar/genética , Neoplasias da Glândula Tireoide/genética , Adenocarcinoma Folicular/patologia , Adenocarcinoma Papilar/patologia , Adulto , Análise Mutacional de DNA , Primers do DNA , Feminino , Genes ras/genética , Humanos , Masculino , Biologia Molecular , Mutação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias da Glândula Tireoide/patologia
6.
Biochim Biophys Acta ; 1763(3): 263-71, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16545470

RESUMO

Both autonomously functioning thyroid nodules (AFTNs) and cold thyroid nodules (CTNs) are characterized by an increased proliferation, however, they have opposite functional activities. Therefore, with the aim to further understand the distinct molecular pathology of each entity and to discover common mechanisms like those leading to increased proliferation in both, AFTNs and CTNs, we now compared gene expression of AFTNs and CTNs with in vitro model systems (TSH-stimulated and ras-transfected primary cultures (PC)) whose gene expression patterns can be attributed to specific molecular alterations. Since combinations of co-regulated genes are more likely to reveal molecular mechanisms, we used a procedure which groups co-regulated genes within "gene sets". We found a co-regulated gene set in the AFTNs that overlaps with differential expression in TSH-stimulated PCs but not in CTNs or ras-transfected PCs. In addition to thyroid peroxidase and sialyltransferase 1, this set of co-regulated genes comprises metallothioneins and the G-protein-coupled receptor 56. Although their role in the thyroid is unknown so far, their appearance in one group indicates a functional relevance in TSH-TSH receptor-stimulated mechanisms. Furthermore, we identified down-regulated gene sets with concordant expression patterns in AFTNs, CTNs and ras-transfected PCs. However, these expression patterns are not of relevance in the TSH-stimulated PCs. These findings suggest that TSH-stimulated PCs can be used as a model of increased thyroid function (AFTNs), whereas the ras-transfected PCs better reflect the increased proliferation of both AFTNs and CTNs.


Assuntos
Regulação para Baixo/genética , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Perfilação da Expressão Gênica , Nódulo da Glândula Tireoide/genética , Regulação para Cima/genética , Algoritmos , Células Cultivadas , Humanos , Temperatura
7.
J Hum Genet ; 50(3): 151-154, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15744458

RESUMO

We report that a subject with Cockayne syndrome type A (CS3BE) was a compound heterozygote for mutations in CKN1, the gene encoding the CSA protein (MIM 216400). CS3BE displayed a novel missense mutation (A160V) and a previously described nonsense mutation (E13X). Although residing between the second and third WD-40 repeats characteristic of the CSA protein, A160 is completely conserved in all species that possess a CKN1 homologue. We also describe a mutation in a previously uncharacterised xeroderma pigmentosum group C subject (XP8CA) in the XPC gene (MIM 278720). XP8CA was homozygous for a 2 bp TG deletion in codon 547 resulting in premature termination at codon 572. Immunoblotting of XP8CA extracts confirmed the absence of full-length XPC protein that was present in unaffected cell lines.


Assuntos
Síndrome de Cockayne/genética , Proteínas de Ligação a DNA/genética , Mutação/genética , Proteínas/genética , Xeroderma Pigmentoso/genética , Linhagem Celular , Primers do DNA , Enzimas Reparadoras do DNA , Humanos , Immunoblotting , Análise de Sequência de DNA , Fatores de Transcrição
8.
Oncogene ; 23(47): 7839-45, 2004 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-15361839

RESUMO

Ras mutations occur as an early event in many human tumours of epithelial origin, including thyroid. Using primary human thyroid epithelial cells to model tumour initiation by Ras, we have shown previously that activation of both the MAP kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K) effector pathways are necessary, but even when activated together are not sufficient, for Ras-induced proliferation. Here, we show that a third effector, RalGEF, is also activated by Ras in these cells, that this activation is necessary for Ras-induced proliferation, and furthermore that in combination with the MAPK and PI3K effectors, it is able to reproduce the proliferative effect of activated Ras. The requirement for three effector pathways indicates a more robust control of cell proliferation in this normal human epithelial cell type than has been displayed in previous similar studies using rodent and human cell lines. Our findings highlight the importance of the appropriate cellular context in models of Ras-induced tumour development.


Assuntos
Divisão Celular/genética , Células Epiteliais/citologia , Genes ras/genética , Glândula Tireoide/citologia , Proteínas ras/genética , Linhagem Celular , Transformação Celular Neoplásica/genética , Humanos , Proteínas Recombinantes de Fusão/metabolismo , Retroviridae/genética , Transfecção , Proteínas ras/metabolismo
9.
Oncogene ; 23(35): 5994-9, 2004 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-15208687

RESUMO

Using microinjection of recombinant protein to directly control 'expression' levels, we have compared the proliferative response to ras oncogene activation in two normal cell types--fibroblast and thyroid epithelial cell--which give rise to human tumours with very low and high frequencies of ras mutation respectively. A concentration-dependent stimulation of DNA synthesis was observed in thyrocytes, matched by an almost perfectly reciprocal inhibition in fibroblasts. A concentration-dependent induction of the cyclin-dependent kinase (CDK) inhibitor p21WAF1 was observed in both cell types, but p16Ink4a was induced by ras only in fibroblasts. This difference could not account for the fibroblast specificity of the growth-inhibitory response, however, since proliferation of p16-deficient fibroblasts was also inhibited by mutant ras. We conclude that the striking contrast in proliferative response to ras between fibroblasts and thyroid epithelial cells cannot readily be explained by differential induction of either of the two key CDK inhibitors, p16Ink4a and p21WAF1, but is consistent with a differential ability of p21WAF1 to antagonize ras-induced mitogenic signals in the two cell types. Such tissue-specific differences provide an attractive explanation for the observed specificity of ras mutation for particular human tumour types, and emphasize the inappropriateness of fibroblasts as a model for ras-induced tumorigenesis.


Assuntos
Genes ras , Mutação , Neoplasias/etiologia , Divisão Celular , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Células Epiteliais/citologia , Fibroblastos/citologia , Genes p16 , Humanos , Neoplasias/patologia
10.
EMBO J ; 23(13): 2554-63, 2004 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-15192702

RESUMO

Telomere shortening in normal human cells causes replicative senescence, a p53-dependent growth arrest state, which is thought to represent an innate defence against tumour progression. However, although it has been postulated that critical telomere loss generates a 'DNA damage' signal, the signalling pathway(s) that alerts cells to short dysfunctional telomeres remains only partially defined. We show that senescence in human fibroblasts is associated with focal accumulation of gamma-H2AX and phosphorylation of Chk2, known mediators of the ataxia-telangiectasia mutated regulated signalling pathway activated by DNA double-strand breaks. Both these responses increased in cells grown beyond senescence through inactivation of p53 and pRb, indicating that they are driven by continued cell division and not a consequence of senescence. gamma-H2AX (though not Chk2) was shown to associate directly with telomeric DNA. Furthermore, inactivation of Chk2 in human fibroblasts led to a fall in p21(waf1) expression and an extension of proliferative lifespan, consistent with failure to activate p53. Thus, Chk2 forms an essential component of a common pathway signalling cell cycle arrest in response to both telomere erosion and DNA damage.


Assuntos
Ciclo Celular , Senescência Celular , Dano ao DNA , Replicação do DNA , DNA/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Proliferação de Células , Quinase do Ponto de Checagem 2 , Imunoprecipitação da Cromatina , Inibidor de Quinase Dependente de Ciclina p21 , Fibroblastos/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Corantes Fluorescentes , Genes Supressores de Tumor , Histonas/metabolismo , Humanos , Pulmão/citologia , Masculino , Microscopia Confocal , Modelos Biológicos , Fosforilação , Proteínas Serina-Treonina Quinases/genética , RNA Interferente Pequeno/metabolismo , Proteína do Retinoblastoma/metabolismo , Retroviridae/genética , Telomerase/metabolismo , Telômero/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Xantenos
11.
Oncogene ; 23(23): 4136-45, 2004 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-15064743

RESUMO

Telomerase activity in tumours is often associated with p53 mutation. Many antitelomerase therapies take advantage of the inability of cells expressing mutant p53 to undergo replicative senescence, since this allows telomere erosion to continue until 'crisis', hence providing the desired cytotoxic effect. However, some tumour types, including breast, melanomas and thyroid, retain wild-type p53 function and the effectiveness of antitelomerase therapies in such tumour cells have not been adequately addressed. To explore this, we made use of two thyroid cancer cell lines K1 and K2, which retain wt p53. Telomere erosion induced by the expression of a dominant-negative (DN) hTERT resulted in delayed onset of growth arrest in K1 and K2 cells, reminiscent of replicative senescence, with low levels of BrdU labelling and apoptosis, associated with high p21(WAF1) and senescence-associated beta galactosidase expression. In contrast, abrogation of p53 function by the expression of HPV16 E6 in K1 and K2 cells either at the same time as DNhTERT or just prior to the onset of senescence allowed cells to continue growing until 'crisis'. Likewise, microinjection of a p53 neutralizing antibody into 'senescent' K1 DNhTERT cells permitted re-entry into the cell cycle. We conclude that thyroid tumour cells with wild-type p53 retain an intact p53-mediated growth arrest response to telomere erosion. This raises the intriguing question of why, therefore, p53 mutation is not selected for in such cancers, and also calls into question the therapeutic value of telomerase inhibitors in such cases.


Assuntos
Telomerase/metabolismo , Telômero/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Apoptose/fisiologia , Proteínas de Ligação a DNA , Humanos , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/terapia
12.
Exp Cell Res ; 292(1): 151-6, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14720514

RESUMO

The selective pressure for disruption of the cyclin-dependent kinase inhibitor p16(INK4a) in human cancer has been postulated to reflect its role in mediating growth arrest, both in response to telomere erosion (replicative senescence) and to oncogene-induced and other "stress" signals. Given the known species-specific differences in regulation of senescence, we have tested this hypothesis in human, as opposed to rodent, cells by designing a small interfering RNA (siRNA) to knock down p16(INK4a) expression. Transfection of this siRNA into late-passage normal human diploid fibroblasts allowed at least temporary escape from entry into replicative senescence. Furthermore, in our in vitro model of early-stage, RAS-induced thyroid tumorigenesis, sequential transfections with this siRNA allowed outgrowth of small clusters of proliferating epithelial cells, consistent with escape from the spontaneous "senescence", which normally curtails their proliferative response to mutant RAS. These data provide the first direct evidence that p16(INK4a) is necessary for the initiation of both telomere-dependent and telomere-independent senescence in human cells.


Assuntos
Divisão Celular , Senescência Celular/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas ras/metabolismo , Células Cultivadas , DNA/biossíntese , Diploide , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Fibroblastos/citologia , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Mutação , Retroviridae/genética , Glândula Tireoide/citologia , Transfecção , Proteínas ras/genética
13.
Cancer Res ; 63(16): 4854-61, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12941806

RESUMO

An in vitro model, based on normal (primary) human astrocytes (NHAs), was used to investigate the nature of the selection pressures for events that occur during the progression of astrocyte-derived tumors and, in particular, the potential role of proliferative life span barriers (PLBs). As with fibroblasts, NHAs senesced with elevated p21(WAF1) and senescence-associated beta-galactosidase activities. Unlike fibroblasts, replicative senescence (M1) occurred much earlier, after approximately 20 pd and was not bypassed by hTERT expression. Abrogation of p53 function, by expression of human papillomavirus type 16 E6, led to an extension of life span, implying that replicative senescence in NHAs was p53-dependent but telomere-independent. human papillomavirus type16 E6 expression promoted additional growth of up to 12 pd, until a second telomere-independent PLB (termed M(INT)) was imposed associated with elevated p16(INK4A) levels. A proportion of cells escaped from M(INT) lost p16(INK4A) expression and achieved approximately an additional 25 pd until a crisis-like third PLB (M2) was reached. Expression of hTERT in post-M(INT) cells allowed these cells to become immortal and bypass this third PLB. The in vitro PLBs appear, in order of occurrence, dependent upon p53, p16(INK4A), and telomere erosion, a situation that mirrors an equivalent order of mutational events during tumor progression in vivo. This study describes a model that provides a plausible explanation for the selective pressures driving mutational events in this tumor type and provides direct evidence of a p53-dependent, telomere-independent PLB.


Assuntos
Astrócitos/fisiologia , Glioma/etiologia , Proteínas Repressoras , Telômero , Proteína Supressora de Tumor p53/fisiologia , Divisão Celular , Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina/análise , Proteínas de Ligação a DNA , Glioma/genética , Humanos , Proteínas Oncogênicas Virais/análise , Telomerase/análise
14.
J Natl Cancer Inst ; 95(10): 723-32, 2003 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-12759390

RESUMO

BACKGROUND: The melanoma susceptibility locus cyclin-dependent kinase inhibitor 2A encodes two unrelated cell growth inhibitors, p16 and alternative reading frame (ARF). In fibroblasts, both proteins are implicated in cellular senescence, a key barrier to tumor development. The p16 coding sequence is more often mutated in melanoma families than is the ARF sequence. To investigate the role of p16 in melanocytes, we assessed aspects of growth, apoptosis, and immortalization in melanocytes cultured from two melanoma patients, both of whom had two inactive p16 alleles but functional ARF. METHODS: Growth and senescence were evaluated by cumulative population-doubling curves, and apoptosis by terminal deoxytransferase labeling. Expression of p53 and p21, which are associated with fibroblast senescence, was assessed by immunoblotting. Amphotropic retroviruses were used to transfer exogenous gene sequences into the melanocytes. RESULTS: Both melanocyte cultures showed high rates of apoptosis, which were reduced when the cells were grown in the presence of keratinocyte feeder cells or human stem cell factor plus endothelin 1. With these growth factors, both cultures proliferated for 45-55 net population doublings, markedly longer than the maximum of 10 net population doublings of normal adult human melanocytes in similar media, indicating impaired senescence. One of the cultures developed chromosomal aberrations, with numerous dicentric chromosomes at senescence, consistent with telomere dysfunction. p53 and p21 levels were not elevated in senescent normal melanocytes but were elevated in senescent p16-deficient melanocytes. Interference with p53 function by transfer of human papillomavirus 16-E6 further extended the lifespan of p16-deficient melanocytes. Human telomerase reverse transcriptase was sufficient to immortalize both these cell strains but not normal melanocytes. CONCLUSION: Normal senescence in human melanocytes requires p16 activity. p53 contributes to a delayed form of senescence that requires telomere shortening, in p16-deficient melanocytes. These findings provide some basis for the role of p16 in melanoma susceptibility.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/deficiência , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Melanócitos/metabolismo , Melanoma/metabolismo , Telomerase/metabolismo , Apoptose , Linhagem Celular Transformada , Senescência Celular , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Proteínas de Ligação a DNA , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Immunoblotting , Cariotipagem , Cinética , Fenótipo
15.
Exp Cell Res ; 285(2): 236-42, 2003 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12706118

RESUMO

Functional wild-type p53 is required for human diploid fibroblasts (HDF) to enter an irreversible growth arrest known as replicative senescence. Experimentally, abrogation of p53 function by expression of human papillomavirus type 16 E6 or disruption of a key downstream effector p21 by homologous recombination both extended HDF life span. However, although sufficient to extend life span, p21 down-regulation is not necessary, because expression of a dominant-negative mutant p53 (143(ala)) extends life span without apparently decreasing p21 expression. Given the importance of p53 in cellular senescence and the general assumption that p21 may be the sole mediator of its action in this process, we have investigated how abrogation of p53 function can overcome senescence without lowering expression of p21. We have found up-regulated levels of the cyclin-dependent kinase 2 (cdk2) protein in HDF expressing 143(ala) mutant p53 as compared to senescent controls, together with an increase in p21-free cdk2 which, in conjunction with cyclin E, is able to form an active kinase which can phosphorylate the retinoblastoma protein. However, forced overexpression of cdk2 in near-senescent HDF failed to restore cdk2-associated kinase activity. Our data suggest that p53-mediated senescence depends on factor(s) other than p21 which modulate formation of cyclin E-cdk2 complexes.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Senescência Celular , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Fibroblastos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteína Supressora de Tumor p53/fisiologia , Proteínas de Ciclo Celular/análise , Divisão Celular , Linhagem Celular , Ciclina D1/metabolismo , Ciclina E/metabolismo , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , Ciclinas/análise , Fibroblastos/citologia , Humanos , Cinética , Substâncias Macromoleculares , Mutação , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/análise
16.
J Cell Sci ; 116(Pt 7): 1349-57, 2003 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-12615976

RESUMO

Werner-syndrome fibroblasts have a reduced in vitro life span before entering replicative senescence. Although this has been thought to be causal in the accelerated ageing of this disease, controversy remains as to whether Werner syndrome is showing the acceleration of a normal cellular ageing mechanism or the occurrence of a novel Werner-syndrome-specific process. Here, we analyse the signalling pathways responsible for senescence in Werner-syndrome fibroblasts. Cultured Werner-syndrome (AG05229) fibroblasts senesced after approximately 20 population doublings with most of the cells having a 2N content of DNA. This was associated with hypophosphorylated pRb and high levels of p16(Ink4a) and p21(Waf1). Senescent AG05229 cells re-entered the cell cycle following microinjection of a p53-neutralizing antibody. Similarly, production of the human papilloma virus 16 E6 oncoprotein in presenescent AG05229 cells resulted in senescence being bypassed and extended cellular life span. Werner-syndrome fibroblasts expressing E6 did not proliferate indefinitely but reached a second proliferative lifespan barrier, termed M(int), that could be bypassed by forced production of telomerase in post-M1 E6-producing cells. The conclusions from these studies are that: (1) replicative senescence in Werner-syndrome fibroblasts is a telomere-induced p53-dependent event; and (2) the intermediate lifespan barrier M(int) is also a telomere-induced event, although it appears to be independent of p53. Werner-syndrome fibroblasts resemble normal human fibroblasts for both these proliferative lifespan barriers, with the strong similarity between the signalling pathway linking telomeres to cell-cycle arrest in Werner-syndrome and normal fibroblasts providing further support for the defect in Werner syndrome causing the acceleration of a normal ageing mechanism.


Assuntos
Senescência Celular/genética , Proteínas de Ligação a DNA , Fibroblastos/metabolismo , Telômero/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Síndrome de Werner/genética , Envelhecimento/genética , Envelhecimento/metabolismo , Anticorpos/farmacologia , Divisão Celular/genética , Linhagem Celular , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , DNA/genética , Fibroblastos/patologia , Genes cdc/fisiologia , Humanos , Longevidade/genética , Mitose/genética , Proteínas Oncogênicas Virais/metabolismo , Ploidias , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais/genética , Telomerase/genética , Telomerase/metabolismo , Telômero/genética , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética , Síndrome de Werner/metabolismo , Síndrome de Werner/patologia
17.
Nat Genet ; 33(2): 203-7, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12539050

RESUMO

By imposing a limit on the proliferative lifespan of most somatic cells, telomere erosion represents an innate mechanism for tumor suppression and may contribute to age-related disease. A detailed understanding of the pathways that link shortened telomeres to replicative senescence has been severely hindered by the inability of current methods to analyze telomere dynamics in detail. Here we describe single telomere length analysis (STELA), a PCR-based approach that accurately measures the full spectrum of telomere lengths from individual chromosomes. STELA analysis of human XpYp telomeres in fibroblasts identifies several features of telomere biology. We observe bimodal distributions of telomeres in normal fibroblasts; these distributions result from inter-allelic differences of up to 6.5 kb, indicating that unexpectedly large-scale differences in zygotic telomere length are maintained throughout development. Most telomeres shorten in a gradual fashion consistent with simple losses through end replication, and the rates of erosion are independent of allele size. Superimposed on this are occasional, more substantial changes in length, which may be the consequence of additional mutational mechanisms. Notably, some alleles show almost complete loss of TTAGGG repeats at senescence.


Assuntos
Alelos , Senescência Celular/fisiologia , Mapeamento Cromossômico , Cromossomos/genética , Fibroblastos/citologia , Telômero/ultraestrutura , Neoplasias da Glândula Tireoide/genética , Divisão Celular , DNA/análise , Primers do DNA/química , Replicação do DNA , Fibroblastos/ultraestrutura , Variação Genética , Humanos , Hibridização in Situ Fluorescente , Metáfase , Mutação , Reação em Cadeia da Polimerase , Telomerase , Telômero/fisiologia , Células Tumorais Cultivadas
18.
Oncogene ; 21(27): 4277-88, 2002 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-12082615

RESUMO

Replicative senescence is thought to be a significant barrier to human tumorigenesis, which in human fibroblasts, and many other cell types, can be overcome experimentally by combined loss of function of p53 and Rb 'pathways'. To avoid the confounding pleiotropic effects of HPVE7 frequently used in such studies, here we have employed retroviral vectors over-expressing CDK4 or CDK6 as a more representative model of naturally-occurring mutations targeting the Rb pathway. We show that these can extend fibroblast lifespan by approximately 10 population doublings, ending in a viable senescence-like state which contrasts with the apoptotic end-stage seen with E7. Compared with 'normal' senescence, this growth arrest was, in most cases, not accompanied by any further increase in p21(Waf1) levels but with up to a 19-fold increase in p16(Ink4a). Surprisingly however, this could not explain arrest, since expression of mutant CDK4 and/or CDK6, incapable of binding p16(Ink4a), did not confer any greater lifespan extension than the wild-type CDKs. Subsequent abrogation of p53 function by a second vector, encoding HPVE6, downregulated p21(Waf1) and conferred a second lifespan extension, ending in a crisis-like state, consistent with full escape from senescence. These data: (i) point to a back-up 'senescence' mechanism distinct from induction of p21(Waf1) or p16(Ink4a); and (ii) provide an in vitro model of clonal evolution through successive dysfunction of Rb and p53 pathways in a relevant human cell context.


Assuntos
Senescência Celular/genética , Quinases Ciclina-Dependentes/fisiologia , Fibroblastos/citologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas , Proteínas Repressoras , Proteína do Retinoblastoma/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Apoptose , Ciclo Celular/genética , Divisão Celular/genética , Transformação Celular Viral/genética , Células Clonais/citologia , Células Clonais/metabolismo , Quinase 4 Dependente de Ciclina , Quinase 6 Dependente de Ciclina , Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Inibidor de Quinase Dependente de Ciclina p21 , Quinases Ciclina-Dependentes/genética , Ciclinas/biossíntese , Ciclinas/genética , Fibroblastos/metabolismo , Genes do Retinoblastoma , Genes p16 , Genes p53 , Vetores Genéticos/genética , Vetores Genéticos/farmacologia , Humanos , Modelos Biológicos , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/fisiologia , Proteínas E7 de Papillomavirus , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas Recombinantes de Fusão/fisiologia , Proteína Supressora de Tumor p53/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...