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.
PLoS One ; 8(9): e72169, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24086258

RESUMO

Amyloid beta peptide (Aß) causes neurodegeneration by several mechanisms including oxidative stress, which is known to induce DNA damage with the consequent activation of poly (ADP-ribose) polymerase (PARP-1). To elucidate the role of PARP-1 in the neurodegenerative process, SH-SY5Y neuroblastoma cells were treated with Aß25-35 fragment in the presence or absence of MC2050, a new PARP-1 inhibitor. Aß25-35 induces an enhancement of PARP activity which is prevented by cell pre-treatment with MC2050. These data were confirmed by measuring PARP-1 activity in CHO cells transfected with amylod precursor protein and in vivo in brains specimens of TgCRND8 transgenic mice overproducing the amyloid peptide. Following Aß25-35 exposure a significant increase in intracellular ROS was observed. These data were supported by the finding that Aß25-35 induces DNA damage which in turn activates PARP-1. Challenge with Aß25-35 is also able to activate NF-kB via PARP-1, as demonstrated by NF-kB impairment upon MC2050 treatment. Moreover, Aß25-35 via PARP-1 induces a significant increase in the p53 protein level and a parallel decrease in the anti-apoptotic Bcl-2 protein. These overall data support the hypothesis of PARP-1 involvment in cellular responses induced by Aß and hence a possible rationale for the implication of PARP-1 in neurodegeneration is discussed.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , Poli(ADP-Ribose) Polimerases/fisiologia , Animais , Sequência de Bases , Células CHO , Linhagem Celular , Ensaio Cometa , Cricetinae , Cricetulus , Dano ao DNA , Primers do DNA , Ensaio de Desvio de Mobilidade Eletroforética , Camundongos , Camundongos Transgênicos , Poli(ADP-Ribose) Polimerase-1 , Espécies Reativas de Oxigênio/metabolismo
2.
Cancer Chemother Pharmacol ; 72(1): 117-25, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23636450

RESUMO

PURPOSE: Defective expression of the mismatch repair protein MSH3 is frequently detected in colon cancer, and down-regulation of its expression was found to decrease sensitivity to platinum compounds or poly(ADP-ribose) polymerase inhibitors (PARPi) monotherapy. We have investigated whether MSH3 transfection in MSH3-deficient colon cancer cells confers resistance to oxaliplatin or PARPi and whether their combination restores chemosensitivity. METHODS: MSH3-deficient/MLH1-proficient colon cancer HCT116(MLH1) cells were transfected with the MSH3 cDNA cloned into the pcDNA3.1(-) vector. MSH3/MLH1-deficient HCT116, carrying MLH1 and MSH3 mutations on chromosome 3 and 5, respectively, and HCT116 in which wild-type MLH1 (HCT116+3), MSH3 (HCT116+5) or both genes (HCT116+3+5) were introduced by chromosome transfer were also tested. Sensitivity to oxaliplatin and to PARPi was evaluated by analysis of clonogenic survival, cell proliferation, apoptosis and cell cycle. RESULTS: MSH3 transfection in HCT116 cells did not confer resistance to oxaliplatin or PARPi monotherapy. MSH3-proficient HCT116+5 or HCT116+3+5 cells, which were more resistant to oxaliplatin and PARPi in comparison with their MSH3-deficient counterparts, expressed higher levels of the nucleotide excision repair ERCC1 and XPF proteins, involved in the resistance to platinum compounds, and lower PARP-1 levels. In all cases, PARPi increased sensitivity to oxaliplatin. CONCLUSIONS: Restoring of MSH3 expression by cDNA transfection, rather than by chromosome transfer, did not affect colon cancer sensitivity to oxaliplatin or PARPi monotherapy; PARP-1 levels seemed to be more crucial for the outcome of PARPi monotherapy.


Assuntos
Antineoplásicos/farmacologia , Neoplasias do Colo/tratamento farmacológico , Proteínas de Ligação a DNA/metabolismo , Inibidores Enzimáticos/farmacologia , Proteínas de Neoplasias/metabolismo , Compostos Organoplatínicos/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Códon sem Sentido , Neoplasias do Colo/enzimologia , Neoplasias do Colo/metabolismo , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Mutação da Fase de Leitura , Células HCT116 , Humanos , Concentração Inibidora 50 , Proteína 1 Homóloga a MutL , Proteína 3 Homóloga a MutS , Proteínas Mutantes/biossíntese , Proteínas Mutantes/metabolismo , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oxaliplatina , Poli(ADP-Ribose) Polimerase-1 , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Transfecção
3.
Int J Oncol ; 43(1): 210-8, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23653048

RESUMO

Poly(ADP-ribose) polymerase inhibitors (PARPi) are currently evaluated in clinical trials in combination with topoisomerase I (Top1) inhibitors against a variety of cancers, including colon carcinoma. Since the mismatch repair component MLH1 is defective in 10-15% of colorectal cancers we have investigated whether MLH1 affects response to the Top1 inhibitor irinotecan, alone or in combination with PARPi. To this end, the colon cancer cell lines HCT116, carrying MLH1 mutations on chromosome 3 and HCT116 in which the wild-type MLH1 gene was replaced via chromosomal transfer (HCT116+3) or by transfection of the corresponding MLH1 cDNA (HCT116 1-2) were used. HCT116 cells or HCT116+3 cells stably silenced for PARP-1 expression were also analysed. The results of in vitro and in vivo experiments indicated that MLH1, together with low levels of Top1, contributed to colon cancer resistance to irinotecan. In the MLH1-proficient cells SN-38, the active metabolite of irinotecan, induced lower levels of DNA damage than in MLH1-deficient cells, as shown by the weaker induction of γ-H2AX and p53 phosphorylation. The presence of MLH1 contributed to induce of prompt Chk1 phosphorylation, restoring G2/M cell cycle checkpoint and repair of DNA damage. On the contrary, in the absence of MLH1, HCT116 cells showed minor Chk1 phosphorylation and underwent apoptosis. Remarkably, inhibition of PARP function by PARPi or by PARP-1 gene silencing always increased the antitumor activity of irinotecan, even in the presence of low PARP-1 expression.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Camptotecina/análogos & derivados , Neoplasias do Colo/tratamento farmacológico , Proteínas Nucleares/genética , Inibidores de Poli(ADP-Ribose) Polimerases , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Apoptose/genética , Camptotecina/administração & dosagem , Quinase 1 do Ponto de Checagem , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Humanos , Irinotecano , Proteína 1 Homóloga a MutL , Proteínas Nucleares/antagonistas & inibidores , Fosforilação/efeitos dos fármacos , Proteínas Quinases/metabolismo , Inibidores da Topoisomerase I/administração & dosagem
4.
Mutat Res ; 712(1-2): 40-8, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21570414

RESUMO

Common fragile sites (CFS) are specific chromosomal areas prone to form gaps and breaks when cells are exposed to stresses that affect DNA synthesis, such as exposure to aphidicolin (APC), an inhibitor of DNA polymerases. The APC-induced DNA damage is repaired primarily by homologous recombination (HR), and RAD51, one of the key players in HR, participates to CFS stability. Since another DNA repair pathway, the mismatch repair (MMR), is known to control HR, we examined the influence of both the MMR and HR DNA repair pathways on the extent of chromosomal damage and distribution of CFS provoked by APC and/or by RAD51 silencing in MMR-deficient and -proficient colon cancer cell lines (i.e., HCT-15 and HCT-15 transfected with hMSH6, or HCT-116 and HCT-116/3+6, in which a part of a chromosome 3 containing the wild-type hMLH1 allele was inserted). Here, we show that MMR-deficient cells are more sensitive to APC-induced chromosomal damage particularly at the CFS as compared to MMR-proficient cells, indicating an involvement of MMR in the control of CFS stability. The most expressed CFS is FRA16D in 16q23, an area containing the tumour suppressor gene WWOX often mutated in colon cancer. We also show that silencing of RAD51 provokes a higher number of breaks in MMR-proficient cells with respect to their MMR-deficient counterparts, likely as a consequence of the combined inhibitory effects of RAD51 silencing on HR and MMR-mediated suppression of HR. The RAD51 silencing causes a broader distribution of breaks at CFS than that observed with APC. Treatment with APC of RAD51-silenced cells further increases DNA breaks in MMR-proficient cells. The RNAi-mediated silencing of PARP-1 does not cause chromosomal breaks or affect the expression/distribution of CFS induced by APC. Our results indicate that MMR modulates colon cancer sensitivity to chromosomal breaks and CFS induced by APC and RAD51 silencing.


Assuntos
Sítios Frágeis do Cromossomo , Neoplasias do Colo/genética , Reparo de Erro de Pareamento de DNA , Poli(ADP-Ribose) Polimerases/genética , Rad51 Recombinase/genética , Linhagem Celular Tumoral , Quebra Cromossômica , Inativação Gênica , Humanos
5.
Eur J Cancer ; 47(8): 1219-30, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21269821

RESUMO

First line treatment of metastatic melanoma includes the methylating agent dacarbazine or its analogue temozolomide (TMZ) with improved pharmacokinetics and tolerability. However, the prognosis of the metastatic disease is poor and several trials are evaluating TMZ in polychemotherapy protocols. The novel glutathione transferase P1-1 (GSTP1-1) inhibitor 6-(7-nitro-2,1,3-benzoxadiazol-4-ylthio)hexanol (NBDHEX) has recently shown activity against melanoma through c-Jun N-terminal kinase activation. In this study we have investigated the in vitro and in vivo efficacy of NBDHEX and TMZ combination against melanoma. The results indicated that NBDHEX and TMZ exerted in vitro synergistic anti-proliferative effects in murine B16 and human A375 melanoma cells. In B16 cells TMZ as single agent caused cell accumulation at the G(2)/M phase of cell cycle, whereas NBDHEX induced mainly apoptotic effects. NBDHEX provoked a higher level of p53 phosphorylation with respect to TMZ and the drug combination caused a more than additive increase of p53 activation. The in vivo efficacy of NBDHEX and TMZ has been investigated in an orthotopic B16 model. Treatment with NBDHEX provoked a reduction of tumour growth comparable to that obtained with TMZ, whereas the drug combination significantly increased tumour growth inhibition with respect to the single agents, without worsening TMZ myelotoxicity. Immunohistochemical analysis of tumour grafts revealed a profound reduction of Cyclin D1 and CD31 in all treatment groups; VEGF expression was, instead, markedly decreased only in NBDHEX or NBDHEX and TMZ treated samples. These findings indicate that NBDHEX represents a good candidate for combination therapies including TMZ, offering new perspectives for the treatment of melanoma.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Dacarbazina/análogos & derivados , Melanoma/tratamento farmacológico , Oxidiazóis/administração & dosagem , Animais , Linhagem Celular Tumoral , Proliferação de Células , Metilação de DNA , Dacarbazina/administração & dosagem , Sinergismo Farmacológico , Glutationa Transferase/antagonistas & inibidores , Humanos , Concentração Inibidora 50 , Masculino , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Temozolomida
6.
Mutat Res ; 664(1-2): 39-47, 2009 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-19428379

RESUMO

Mismatch repair (MMR) has been shown to control homologous recombination (HR) by aborting strand exchange between divergent sequences. We previously demonstrated that MMR-deficient tumour cells are more resistant to chromosomal damage induced by bleomycin (BLM) during the G(2) phase, likely due to the lack of the MMR inhibitory effect on HR. Aim of this study was to investigate whether inhibition of HR by the nucleoside analogue BVDU [(E)-5(2-bromovinyl)-2'-deoxyuridine, brivudin], or silencing of genes involved in HR function, might affect sensitivity of MMR-deficient tumour cells to DNA damage induced by BLM in G(2). The results indicated that BVDU increased chromatid damage and DNA double strand breaks induced by BLM only in MMR-deficient MT-1, HL-60R, HCT116 cells, which are more resistant to BLM with respect to MMR-proficient TK-6, HL-60S and HCT116/3-6 lines. Silencing of RAD51, a key component of HR, increased sensitivity of MMR-deficient HCT-15 cells to BLM clastogenicity; in this case combined treatment with BVDU had no additional effect. Similarly, treatment with BVDU did not affect BLM clastogenicity in CAPAN-1 cells, characterized by a defective HR due to BRCA2 mutations. Conversely, BVDU increased chromatid breaks induced by BLM in HCT-15 cells transiently silenced for DNA-PK catalytic subunit, which plays a key role in non-homologous end joining. The BVDU-mediated increase of chromatid breaks in MMR-deficient cells did not depend on its previously reported inhibitory effect on poly(ADP-ribose) polymerase (PARP). In fact, it was observed also in cells stably silenced for PARP-1, which is responsible for most of cellular PARP activity. These data support the suggestion that the higher sensitivity of MMR-proficient versus MMR-deficient cells to BLM-induced chromatid breaks in the G(2) phase is a consequence of the inhibition of HR by MMR. In MMR-deficient cells, BVDU attenuates the repair of BLM-induced DSBs and this is likely to occur via inhibition of HR.


Assuntos
Bleomicina/farmacologia , Bromodesoxiuridina/análogos & derivados , Dano ao DNA , Rad51 Recombinase/antagonistas & inibidores , Rad51 Recombinase/genética , Recombinação Genética/efeitos dos fármacos , Sequência de Bases , Bromodesoxiuridina/farmacologia , Linhagem Celular Tumoral , Cromátides/efeitos dos fármacos , Cromossomos Humanos/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Reparo de Erro de Pareamento de DNA/genética , Reparo de Erro de Pareamento de DNA/fisiologia , Primers do DNA/genética , Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Células HL-60 , Humanos , Modelos Genéticos , Inibidores de Poli(ADP-Ribose) Polimerases , RNA Interferente Pequeno/genética , Rad51 Recombinase/metabolismo
7.
Cell Cycle ; 8(3): 421-9, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19182530

RESUMO

Aneuploidy is a characteristic of most solid tumors, often associated with negative prognosis. It can arise from two principal mechanisms: from a tetraploid intermediate state, or directly from errors at cell division. The control of cell division, crucial to maintain genomic stability, is still poorly understood in its relationship to aneuploidy. Here we show that the TAp73alpha isoform induces polyploidy when overexpressed. This is possibly due to the interaction of TAp73alpha with kinetochore-related proteins leading to the alteration of mitotic checkpoint abilities. TAp73alpha but not p53 or any of the other p73 isoforms binds Bub1 and Bub3. Since TAp73alpha is frequently overexpressed in cancer, this interaction may contribute to the aneuploidy observed in cancer progression. Our results suggest a novel molecular mechanism leading to aneuploidy involving interference of TAp73alpha with Bub1 and Bub3 resulting in an altered mitotic checkpoint.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Cinetocoros/metabolismo , Proteínas Nucleares/metabolismo , Poliploidia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Humanos , Hibridização in Situ Fluorescente , Proteínas Nucleares/genética , Proteínas de Ligação a Poli-ADP-Ribose , Proteínas Serina-Treonina Quinases/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/genética
8.
Cell Cycle ; 7(15): 2357-67, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18677100

RESUMO

Cajal Bodies are one of many specialised organelles contained in the eukaryotic cell nucleus, and are involved in a number of functions, including regulation of replication-dependent histone gene transcription. In normal diploid cells their number varies between 0 and 4 depending on the cell cycle phase, although in cancer cell lines their number is extremely variable and it has been suggested that it correlates with cell ploidy. Here we show that in mammalian cells, as in Drosophila, two distinct though functionally related bodies exist: a histone gene locus body and a Cajal Body. The first one can be detected using FLASH or NPAT as markers while the second is labelled using antibodies against Coilin. Only the number of FLASH/NPAT histone gene locus bodies correlates with ploidy and only these organelles appear to be regulated during the cell cycle. Finally, we show that the two organelles completely co-localize during the S phase of the cell cycle.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Corpos Enovelados/metabolismo , Proteínas Nucleares/metabolismo , Ploidias , Aneuploidia , Ciclo Celular/fisiologia , Células Cultivadas , Análise Citogenética , Células HCT116 , Células HeLa , Humanos
9.
Mutat Res ; 594(1-2): 63-77, 2006 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-16139849

RESUMO

The mismatch repair (MMR) system and p53 protein play a pivotal role in maintaining genomic stability and modulate cell chemosensitivity. Aim of this study was to examine the effects of either MMR-deficiency or p53 inactivation, or both, on cellular responses to bleomycin. The MMR-deficient colon carcinoma cell line HCT116 and its MMR-proficient subline HCT116/3-6, both expressing wild-type p53, were transfected with an expression vector encoding a dominant-negative p53 mutant, or with the empty vector. Four transfected clones, having the following phenotypes, MMR-proficient/p53 wild-type, MMR-proficient/p53 mutant, MMR-deficient/p53 wild-type, MMR-deficient/p53 mutant, were subjected to treatment with bleomycin. Loss of MMR function alone was associated with increased resistance to apoptosis, chromosomal damage and inhibition of colony formation caused by bleomycin. Loss of p53 alone resulted in abrogation of G1 arrest and increased sensitivity to apoptosis and chromosomal damage induced by the drug, but did not affect clonogenic survival after bleomycin treatment. Disabling both p53 and MMR function led to abrogation of G1 arrest and to a moderate impairment of drug-induced apoptosis. Chromosomal damage was reduced in the MMR-deficient/p53 mutant clone with respect to the MMR-proficient/p53 wild-type one, when evaluated 48 h after bleomycin treatment, but was comparable in both clones 96 h after drug exposure. Clonogenic survival of the MMR-deficient/p53 mutant clone was similar to that of the MMR-deficient/p53 wild-type one. The effects of MMR-deficiency on cellular responses to bleomycin were confirmed using the MMR-proficient lymphoblastoid cell line TK6 and its MMR-deficient subline MT1, both expressing wild-type p53. In conclusion, our data show that loss of MMR and p53 function exerts opposite and independent effects on apoptosis and chromosomal damage induced by bleomycin. Moreover, inactivation of MMR confers resistance to the cytotoxic activity of the anticancer agent in cells expressing either wild-type or mutant p53.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Pareamento Incorreto de Bases/genética , Bleomicina/toxicidade , Reparo do DNA/efeitos dos fármacos , Mutagênicos/toxicidade , Proteína Supressora de Tumor p53/fisiologia , Pareamento Incorreto de Bases/efeitos dos fármacos , Linhagem Celular Transformada , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Inibidor de Quinase Dependente de Ciclina p21/genética , Dano ao DNA/efeitos dos fármacos , Células HCT116 , Humanos , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética
10.
Int J Oncol ; 27(2): 525-35, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16010436

RESUMO

Assessment of chemosensitivity of neovessel endo-thelium associated to tumor mass is hindered by the limited availability of experimental models of actively proliferating endothelial cells. In fact, primary endothelial cells possess a limited lifespan and replicative senescence represents a major limit to their long-term culture. Moreover, non-dividing senescent cells undergo a gradual loss of phenotypic markers and become unable to respond to mitogenic stimuli. We report the generation of an immortalized human endothelial cell line by transfection of human umbilical vein endothelial cells (HUVEC) with both SV40 large/small T antigens and the catalytic subunit of human telomerase. This cell line (HUV-ST) possesses stabilized telomere length and increased proliferation rate with respect to parental cells or to cells transfected with SV40 T antigens only (HUV-S). Nevertheless, even at PD > 100 it is not tumorigenic and displays all major endothelial phenotypic markers, such as von Willebrand factor, CD31, vascular endothelial growth factor (VEGF) receptors (VEGFR1/Flt-1, VEGR2/KDR) and CD105/endoglin. HUV-ST cells are capable of organizing into tubule-like networks with branching morphology in response to appropriate stimuli and migrate upon exposure to VEGF. Interestingly, HUV-ST cells over-express the tumor endothelial marker-1/endosialin which is regarded as the most differentially expressed molecule in tumor-derived endothelium versus normal-derived endothelium. Analysis of chemosensitivity to the wide spectrum methylating agent temozolomide (TMZ), an anticancer drug more effective against actively dividing cells than against resting or slowing proliferating cells, indicated that HUV-ST cells are more susceptible to the drug with respect to HUVEC or HUV-S cells. Abrogation of poly(ADP-ribose) polymerase activity significantly enhances growth inhibition induced by TMZ. In conclusion, the immortalized human endothelial line HUV-ST represents a suitable model for studying the efficacy of anti-neovascular therapy, mimicking proliferating neovascular endothelial cells associated to the tumor mass.


Assuntos
Proliferação de Células/efeitos dos fármacos , Dacarbazina/análogos & derivados , Células Endoteliais/metabolismo , Animais , Antígenos CD , Antígenos de Neoplasias , Antígenos Transformantes de Poliomavirus/genética , Antineoplásicos Alquilantes/farmacologia , Western Blotting , Linhagem Celular , Linhagem Celular Transformada , Movimento Celular/genética , Transformação Celular Neoplásica/genética , Proteínas de Ligação a DNA/genética , Dacarbazina/farmacologia , Células Endoteliais/citologia , Citometria de Fluxo , Expressão Gênica , Células HT29 , Humanos , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Plasmídeos/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Telomerase/genética , Telômero/genética , Telômero/metabolismo , Temozolomida , Transfecção , Transplante Heterólogo , Ensaio Tumoral de Célula-Tronco , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fator de von Willebrand/genética , Fator de von Willebrand/metabolismo
11.
Mutagenesis ; 19(2): 99-104, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14981156

RESUMO

The activation of telomerase in phytohemagglutinin-stimulated peripheral lymphocytes is thought to play a role in telomere maintenance and DNA repair. Considering the importance of this enzyme in both cancer and senescence, we studied the effects of the telomerase inhibitor 3'-azido-3'-deoxythymidine on the frequency of chromosomal aberrations and micronuclei induced in peripheral blood lymphocytes (PBL) of elderly monozygotic and dizygotic twins, evaluated with respect to the genotoxic effects induced in unrelated young subjects. Our results show that the cytogenetic damage induced by 3'-azido-3'-deoxythymidine in human PBL was mainly regulated by genetic factors and allowed the identification of hypersensitive subjects. Ageing, which did not modify the individual susceptibility to 3'-azido-3'-deoxythymidine induction of chromosome aberrations and micronuclei, nevertheless determined an overall increase in nuclear damage.


Assuntos
Instabilidade Cromossômica/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Telomerase/antagonistas & inibidores , Zidovudina/farmacologia , Fatores Etários , Idoso , Dano ao DNA , Humanos , Masculino , Gêmeos
12.
Mutat Res ; 546(1-2): 55-64, 2004 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-14757193

RESUMO

The induction of chromosome damage in cultured human lymphocytes by in vitro treatments with aphidicolin (APC) and bleomycin (BLM) has been proposed as test of sensitivity to mutagens. To assess their validity, we have investigated whether the individual expression of induced chromosome damage has a genetic rather than an environmental basis. Metaphase analysis for chromosomal aberrations (CA) and micronucleus (MN) assay in cytokinesis-blocked cells have been performed in peripheral blood lymphocytes from 19 healthy male twins (9 monozygotic and 10 dizygotic pairs), aged 70-78 years, after APC, BLM and APC+BLM treatments. Concordance between twins revealed a high genetic component in the sensitivity towards clastogenic action of APC both as percentages of CA and MN. The micronucleus assay demonstrated a genetic basis also in the expression of chromosome damage induced by BLM and APC+BLM treatments. Since twins were elderly people, to investigate the possible role of age, CA and MN frequencies were compared with those found in lymphocytes from 11 young male donors. Basal and APC-induced chromosome damage were clearly increased in the former. Following BLM and APC+BLM treatments, age significantly increased mitotic delay, as shown by the mitotic indexes (MI) and by the ratios between binucleated and mononucleated (B/M) cells.


Assuntos
Afidicolina/toxicidade , Biomarcadores , Bleomicina/toxicidade , Aberrações Cromossômicas , Mutagênicos/toxicidade , Idoso , Humanos , Masculino
13.
Cancer Chemother Pharmacol ; 52(3): 185-92, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12827294

RESUMO

PURPOSE: The mismatch repair (MMR) system plays a major role in mediating the cytotoxicity and clastogenicity of agents generating O(6)-methylguanine in DNA. Loss of MMR has also been associated with tumor cell resistance to the cytotoxic effects of 6-thioguanine and cisplatin and with hypersensitivity to N-(2-chloroethyl)- N'-cyclohexyl- N-nitrosourea (CCNU). The aim of the present investigation was to elucidate the role played by the MMR system in the generation of chromosomal damage in cells exposed to 6-thioguanine, cisplatin or CCNU. METHODS: The MMR-proficient cell lines TK6 and HCT116/3-6, and their MMR-deficient counterparts MT1 and HCT116, were treated with 6-thioguanine, cisplatin or CCNU, and analyzed for cell growth inhibition and chromosomal damage. As a control, similar experiments were performed with the methylating agent temozolomide. RESULTS: Cytotoxicity, chromosomal aberrations and sister chromatid exchanges induced by 6-thioguanine and temozolomide were significantly reduced in the MMR-deficient cell lines with respect to their MMR-proficient counterparts. In contrast, although conferring some protection against cytotoxicity, the loss of MMR did not affect cytogenetic damage induced by cisplatin. CCNU produced comparable levels of cytotoxicity, chromosomal aberrations and sister chromatid exchanges in both MMR-proficient and MMR-deficient cell lines. CONCLUSIONS: The MMR system is involved in the generation of chromosomal damage in cells exposed to 6-thioguanine. The system does not play a relevant role in the generation of chromosomal damage in cells treated with CDDP and does not confer protection against the clastogenic effects of CCNU, at least in the cell lines investigated.


Assuntos
Antineoplásicos/efeitos adversos , Pareamento Incorreto de Bases/efeitos dos fármacos , Aberrações Cromossômicas/induzido quimicamente , Troca de Cromátide Irmã/efeitos dos fármacos , Células Tumorais Cultivadas/efeitos dos fármacos , Pareamento Incorreto de Bases/fisiologia , Humanos
14.
Genes Chromosomes Cancer ; 37(2): 165-75, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12696065

RESUMO

Inactivation of mismatch repair (MMR) genes has been linked to the hereditary nonpolyposis colon cancer syndrome and to a subset of sporadic cancers. A phenotypic characteristic of tumors with defective MMR is microsatellite instability (MSI). Although MSI has been reported in a proportion of cutaneous melanomas, inactivation of MMR genes in this tumor type has not been detected thus far. We recently described a human melanoma cell line, PR-Mel, and a cutaneous metastasis from the same patient, which displayed a MMR defect, and showed high MSI. Here we report that in the PR-Mel cell line both MLH1 alleles are somatically inactivated. One allele is lost through a chromosomal deletion of the region 3p21-24, whereas the remaining allele harbors a G --> A transition at position -1 of the acceptor splice site of intron 15, leading to the in-frame skipping of exon 16. The primary melanoma of the PR patient shows loss of heterozygosity at the BAT21 microsatellite marker, located in the MLH1 gene, and does not express the MLH1 and PMS2 proteins. Moreover, it harbors the same mutation detected in the PR-Mel cells. These results demonstrate that biallelic inactivation of MLH1 had occurred in the primary melanoma of the PR patient and suggest that disruption of MMR might have had a role in the development of the melanoma. This is the first report in which genetic defects leading to disruption of MMR function in a human melanoma have been identified.


Assuntos
Alelos , Pareamento Incorreto de Bases/genética , Enzimas Reparadoras do DNA , Reparo do DNA/genética , Melanoma/genética , Proteínas de Neoplasias/genética , Neoplasias Cutâneas/genética , Proteínas Adaptadoras de Transdução de Sinal , Adenosina Trifosfatases/biossíntese , Adenosina Trifosfatases/deficiência , Adenosina Trifosfatases/genética , Proteínas de Transporte , Deleção Cromossômica , Cromossomos Humanos Par 3/genética , Neoplasias do Colo/patologia , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Feminino , Marcadores Genéticos/genética , Humanos , Perna (Membro)/patologia , Perna (Membro)/cirurgia , Perda de Heterozigosidade/genética , Melanoma/patologia , Melanoma/secundário , Melanoma/cirurgia , Pessoa de Meia-Idade , Endonuclease PMS2 de Reparo de Erro de Pareamento , Proteína 1 Homóloga a MutL , Mutação/genética , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/deficiência , Proteínas Nucleares , RNA Mensageiro/genética , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/secundário , Neoplasias Cutâneas/cirurgia , Células Tumorais Cultivadas
15.
Cancer Chemother Pharmacol ; 49(3): 217-24, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11935214

RESUMO

UNLABELLED: Selective N3-adenine methylation represents a novel strategy for tumors with a phenotype of poor responsiveness to a number of anticancer agents currently used in the clinic. Resistance to N3-methyladenine-inducing agents, such as MeOSO(2)(CH(2))(2)-lexitropsin (Me-Lex), is due to high levels of N-methylpurine glycosylase (MPG). However, tumor cells with high MPG activity can be rendered susceptible to Me-Lex using poly(ADP-ribose) polymerase-1 (PARP-1) inhibitors. PURPOSE: To evaluate the potential toxicity of Me-Lex, used as single agent or combined with PARP-1 inhibitors, in normal peripheral blood lymphocytes (PBL). METHODS: PBL either resting or activated with phytohemagglutinin (PHA), obtained from healthy donors, were treated with graded concentrations of Me-Lex with or without PARP-1 inhibitor (3-aminobenzamide, AB, or NU1025, NU). MPG activity, apoptosis and sister chromatid exchanges (SCE) were evaluated. RESULTS: (a) Me-Lex was cytotoxic mainly in PHA-activated PBL with low MPG activity; (b) combined treatment with Me-Lex and AB induced apoptotic effects as early as 24 h after drug exposure both in non-stimulated and PHA-activated PBL. When concentrations of PARP-1 inhibitors (25 microM NU and 4 m M AB) that produced a twofold increase in Me-Lex cytotoxicity in tumor cells were compared, NU induced a less-pronounced increase in apoptosis in PBL treated with Me-Lex; (c) Me-Lex at concentrations that allowed cytogenetic analysis did not induce a significant number of SCE; (d) PARP-1 inhibitors provoked a dose-dependent increase in SCE, but 25 microM NU was devoid of genotoxic effects and did not significantly increase SCE in PBL treated with Me-Lex. CONCLUSIONS: Me-Lex showed preferential cytotoxicity against mitogen-activated PBL. Our results also indicated that for each PARP-1 inhibitor it is necessary to define the concentration devoid of genotoxic effects in normal cells, but still capable of enhancing the efficacy of DNA-damaging agents in tumor cells.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/toxicidade , Linfócitos/efeitos dos fármacos , Mutagênicos/toxicidade , Netropsina/análogos & derivados , Netropsina/toxicidade , Inibidores de Poli(ADP-Ribose) Polimerases , Alquilantes/toxicidade , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citometria de Fluxo , Humanos , Células Jurkat , Ativação Linfocitária/efeitos dos fármacos , Linfócitos/citologia , Linfócitos/imunologia , Valores de Referência , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...