Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 145(3): 435-46, 2011 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-21529715

RESUMO

Chromosomal instability in early cancer stages is caused by stress on DNA replication. The molecular basis for replication perturbation in this context is currently unknown. We studied the replication dynamics in cells in which a regulator of S phase entry and cell proliferation, the Rb-E2F pathway, is aberrantly activated. Aberrant activation of this pathway by HPV-16 E6/E7 or cyclin E oncogenes significantly decreased the cellular nucleotide levels in the newly transformed cells. Exogenously supplied nucleosides rescued the replication stress and DNA damage and dramatically decreased oncogene-induced transformation. Increased transcription of nucleotide biosynthesis genes, mediated by expressing the transcription factor c-myc, increased the nucleotide pool and also rescued the replication-induced DNA damage. Our results suggest a model for early oncogenesis in which uncoordinated activation of factors regulating cell proliferation leads to insufficient nucleotides that fail to support normal replication and genome stability.


Assuntos
Instabilidade Genômica , Neoplasias/genética , Nucleotídeos/biossíntese , Ciclina E/metabolismo , Replicação do DNA , Fatores de Transcrição E2F/metabolismo , Humanos , Perda de Heterozigosidade , Neoplasias/metabolismo , Neoplasias/patologia , Nucleotídeos/metabolismo , Proteínas Oncogênicas Virais/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Proteínas Repressoras/metabolismo , Proteína do Retinoblastoma/metabolismo , Fase S
2.
Am J Pathol ; 182(1): 142-51, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23245831

RESUMO

In normal human cells, oncogene-induced senescence (OIS) depends on induction of DNA damage response. Oxidative stress and hyperreplication of genomic DNA have been proposed as major causes of DNA damage in OIS cells. Here, we report that down-regulation of deoxyribonucleoside pools is another endogenous source of DNA damage in normal human fibroblasts (NHFs) undergoing HRAS(G12V)-induced senescence. NHF-HRAS(G12V) cells underexpressed thymidylate synthase (TS) and ribonucleotide reductase (RR), two enzymes required for the entire de novo deoxyribonucleotide biosynthesis, and possessed low dNTP levels. Chromatin at the promoters of the genes encoding TS and RR was enriched with retinoblastoma tumor suppressor protein and histone H3 tri-methylated at lysine 9. Importantly, ectopic coexpression of TS and RR or addition of deoxyribonucleosides substantially suppressed DNA damage, senescence-associated phenotypes, and proliferation arrest in two types of NHF-expressing HRAS(G12V). Reciprocally, short hairpin RNA-mediated suppression of TS and RR caused DNA damage and senescence in NHFs, although less efficiently than HRAS(G12V). However, overexpression of TS and RR in quiescent NHFs did not overcome proliferation arrest, suggesting that unlike quiescence, OIS requires depletion of dNTP pools and activated DNA replication. Our data identify a previously unknown role of deoxyribonucleotides in regulation of OIS.


Assuntos
Senescência Celular/genética , Dano ao DNA/genética , Desoxirribonucleotídeos/metabolismo , Oncogenes/fisiologia , Proliferação de Células , Células Cultivadas , Senescência Celular/fisiologia , Replicação do DNA/genética , Desoxirribonucleotídeos/genética , Fibroblastos/metabolismo , Fibroblastos/fisiologia , Humanos , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , Ribonucleotídeo Redutases/biossíntese , Ribonucleotídeo Redutases/fisiologia , Timidilato Sintase/biossíntese , Timidilato Sintase/fisiologia
3.
Mutat Res ; 684(1-2): 1-10, 2010 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-20004674

RESUMO

Nucleoside analogs are efficacious cancer chemotherapeutics due to their incorporation into tumor cell DNA. However, they exhibit vastly different antitumor efficacies, suggesting that incorporation produces divergent effects on DNA replication. Here we have evaluated the consequences of incorporation on DNA replication and its fidelity for three structurally related deoxyguanosine analogs: ganciclovir (GCV), currently in clinical trials in a suicide gene therapy approach for cancer, D-carbocyclic 2'-deoxyguanosine (CdG) and penciclovir (PCV). GCV and CdG elicited similar cytotoxicity at low concentrations, whereas PCV was 10-100-fold less cytotoxic in human tumor cells. DNA replication fidelity was evaluated using a supF plasmid-based mutation assay. Only GCV induced a dose-dependent increase in mutation frequency, predominantly GC-->TA transversions, which contributed to cytotoxicity and implicated the ether oxygen in mutagenicity. Activation of mismatch repair with hydroxyurea decreased mutations but failed to repair the GC-->TA transversions. GCV slowed S-phase progression and CdG also induced a G2/M block, but both drugs allowed completion of one cell cycle after drug treatment followed by cell death in the second cell cycle. In contrast, PCV induced a lengthy early S-phase block due to profound suppression of DNA synthesis, with cell death in the first cell cycle after drug treatment. These data suggest that GCV and CdG elicit superior cytotoxicity due to their effects in template DNA, whereas strong inhibition of nascent strand synthesis by PCV may protect against cytotoxicity. Nucleoside analogs based on the carbohydrate structures of GCV and CdG is a promising area for antitumor drug development.


Assuntos
Antineoplásicos/farmacologia , Carboidratos/química , Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Desoxiguanosina/análogos & derivados , Desoxiguanosina/química , Ganciclovir/farmacologia , Aciclovir/análogos & derivados , Aciclovir/farmacologia , Sequência de Bases , Linhagem Celular Tumoral , Reparo de Erro de Pareamento de DNA , Desoxiguanosina/agonistas , Desoxiguanosina/farmacologia , Genes Transgênicos Suicidas , Guanina , Células HCT116 , Humanos , Dados de Sequência Molecular , Mutação
4.
Mol Pharmacol ; 74(3): 863-71, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18535288

RESUMO

The antitumor drug 5-fluoro-2'-deoxyuridine (FdUrd) also sensitizes tumor cells to ionizing radiation in vitro and in vivo. Although radiosensitization with FdUrd requires dTTP depletion and S-phase arrest, the exact mechanism by which these events produce radiosensitization remains unknown. We hypothesized that the depletion of dTTP produces DNA mismatches that, if not repaired before irradiation, would result in radiosensitization. We evaluated this hypothesis in mismatch repair (MMR)-deficient HCT116 0-1 cells that lack the expression of the required MMR protein MLH1 (inactive MLH1), and in MMR-proficient (wild-type MLH1) HCT116 1-2 cells. Although HCT116 0-1 cells were less sensitive to FdUrd (IC(50) = 3.5 microM) versus HCT116 1-2 cells (IC(50) = 0.75 microM), when irradiation followed FdUrd (IC(50)) the MLH1-inactivated cells exhibited greater radiosensitization compared with MMR-wild-type cells [radiation enhancement ratio (RER) = 1.8 +/- 0.28 versus 1.1 +/- 0.1, respectively] and an increase (> or =8-fold) in nucleotide misincorporations. In SW620 cells and HCT116 1-2 MLH1-wild-type cells, FdUrd (IC(50)) did not produce radiosensitization nor did it increase the mutation frequency, but after short hairpin RNA-directed suppression of MLH1 this concentration produced excellent radiosensitization (RER = 1.6 +/- 0.10 and 1.5 +/- 0.06, respectively) and an increase in nucleotide misincorporations (8-fold and 6-fold, respectively). Incubation with higher concentrations of FdUrd (IC(90)) after suppression of MLH1 produced a further increase in ionizing radiation sensitivity in both SW620 and HCT116 1-2 cells (RER = 1.8 +/- 0.03 and 1.7 +/- 0.13, respectively) and nucleotide misincorporations (>10-fold in both cell lines). These results demonstrate an important role for MLH1 and implicate mismatches in radiosensitization by FdUrd.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/deficiência , Pareamento Incorreto de Bases/efeitos dos fármacos , Floxuridina/farmacologia , Proteínas Nucleares/deficiência , Tolerância a Radiação/efeitos dos fármacos , Pareamento Incorreto de Bases/efeitos da radiação , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Morte Celular/efeitos dos fármacos , Morte Celular/efeitos da radiação , Células HCT116 , Humanos , Proteína 1 Homóloga a MutL , Mutação/genética , Nucleotídeos/metabolismo , Plasmídeos/genética , RNA Interferente Pequeno/metabolismo , Tolerância a Radiação/efeitos da radiação , Radiação Ionizante
5.
Cancer Res ; 66(7): 3845-51, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16585212

RESUMO

Suicide gene therapy with the herpes simplex virus thymidine kinase (HSV-TK) cDNA and ganciclovir can elicit cytotoxicity to transgene-expressing and nonexpressing bystander cells via transfer of ganciclovir phosphates through gap junctions. HeLa cells do not exhibit bystander cytotoxicity, although we showed recently that they transfer low levels of ganciclovir phosphates to bystander cells. Here, we attempted to induce bystander cytotoxicity using hydroxyurea, an inhibitor of ribonucleotide reductase, to decrease the endogenous dGTP pool, which should lessen competition with ganciclovir triphosphate for DNA incorporation. Addition of hydroxyurea to cocultures of HSV-TK-expressing and bystander cells synergistically increased ganciclovir-mediated cytotoxicity to both cell populations while producing primarily an additive effect in cultures of 100% HSV-TK-expressing cells. Whereas HSV-TK-expressing cells in coculture were approximately 50-fold less sensitive to ganciclovir compared with cultures of 100% HSV-TK-expressing cells, addition of hydroxyurea restored ganciclovir sensitivity. Quantification of deoxynucleoside triphosphate pools showed that hydroxyurea decreased dGTP pools without significantly affecting ganciclovir triphosphate levels. Although hydroxyurea significantly increased the ganciclovir triphosphate:dGTP value for 12 to 24 hours in HSV-TK-expressing and bystander cells from coculture (1.4- to 4.9-fold), this value was increased for <12 hours (2.5-fold) in 100% HSV-TK-expressing cells. These data suggest that the prolonged increase in the ganciclovir triphosphate:dGTP value in cells in coculture resulted in synergistic cytotoxicity. Compared with enhancement of bystander cytotoxicity through modulation of gap junction intercellular communication, this strategy is superior because it increased cytotoxicity to both HSV-TK-expressing and bystander cells in coculture. This approach may improve clinical efficacy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Ganciclovir/farmacologia , Hidroxiureia/farmacologia , Simplexvirus/genética , Timidina Quinase/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Técnicas de Cocultura , Nucleotídeos de Desoxiguanina/metabolismo , Sinergismo Farmacológico , Ganciclovir/efeitos adversos , Ganciclovir/análogos & derivados , Ganciclovir/metabolismo , Ganciclovir/farmacocinética , Terapia Genética/métodos , Células HeLa , Humanos , Hidroxiureia/efeitos adversos , Nucleotídeos/metabolismo , Simplexvirus/enzimologia , Timidina Quinase/biossíntese , Timidina Quinase/genética
6.
Cancer Res ; 66(6): 3230-7, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16540675

RESUMO

The combination of cytosine deaminase (CD) and herpes simplex virus thymidine kinase (HSV-TK) suicide gene protocols has resulted in enhanced antitumor activity in cultured tumor cells and animal models. In this study, we show that concurrent addition of prodrugs 5-fluorocytosine (5-FC) and ganciclovir (GCV) was less efficacious than sequential treatment in human DU145 prostate carcinoma cells infected with an adenovirus containing a CD/HSV-TK fusion gene. If cells were incubated for 24 hours with 5-FC followed by a 24-hour GCV treatment, GCV triphosphate levels were 2-fold higher, incorporation of GCV monophosphate into DNA was 2.5-fold higher, and growth inhibition was increased 4-fold compared with simultaneous treatment. As expected, cellular dTTP levels were reduced during the 5-FC preincubation. However, dGTP pools also declined parallel to the dTTP decrease. Similar results were obtained when 5-fluorouracil or 5-fluoro-2'-deoxyuridine was used instead of CD/5-FC. These data allowed us to propose a novel hypothesis for the synergistic interaction between CD/5-FC and HSV-TK/GCV treatments. We suggest that the CD/5-FC-mediated reduction of dTTP results in a concurrent decrease of dGTP due to allosteric regulation of ribonucleotide reductase. Because dGTP is the endogenous competitor of GCV triphosphate, depleted dGTP at the time of GCV addition results in increased GCV in DNA and cell kill. In fact, addition of deoxyguanosine during the 5-FC incubation reverses the dGTP depletion, reduces the amount of GCV monophosphate incorporated into DNA, and prevents the CD/5-FC-mediated enhancement of HSV-TK/GCV cytotoxicity. Understanding this mechanistic interaction may help recognize better strategies for creating more efficacious clinical protocols.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Citosina Desaminase/genética , Flucitosina/farmacologia , Ganciclovir/farmacologia , Terapia Genética/métodos , Neoplasias da Próstata/terapia , Simplexvirus/genética , Timidina Quinase/genética , Adenoviridae/genética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Linhagem Celular Tumoral , Citosina Desaminase/biossíntese , Citosina Desaminase/metabolismo , Desoxiguanosina/farmacologia , Desoxirribonucleotídeos/metabolismo , Esquema de Medicação , Sinergismo Farmacológico , Flucitosina/administração & dosagem , Flucitosina/farmacocinética , Fluoruracila/farmacocinética , Fluoruracila/farmacologia , Ganciclovir/administração & dosagem , Ganciclovir/farmacocinética , Vetores Genéticos/genética , Humanos , Masculino , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/genética , Simplexvirus/enzimologia , Timidina Quinase/biossíntese , Timidina Quinase/metabolismo
7.
Mol Cancer Ther ; 6(6): 1858-68, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17575114

RESUMO

Radiation sensitization by 2',2'-difluoro-2'-deoxycytidine (dFdCyd) has correlated with dATP depletion [dFdCDP-mediated inhibition of ribonucleotide reductase (RR)] and S-phase accumulation. We hypothesized that radiosensitization by dFdCyd is due to nucleotide misincorporations in the presence of deoxynucleotide triphosphate pool imbalances, which, if not repaired, augments cell death following irradiation. The ability of dFdCyd to produce misincorporations was measured as pSP189 plasmid mutations in hMLH1-deficient [mismatch repair (MMR) deficient] and hMLH1-expressing (MMR proficient) HCT116 cells. Only MMR-deficient cells showed a significant increase in nucleotide misincorporations (2- to 3-fold increase; P or=5-fold increase; P < 0.05), thus further implicating the inhibition of RR as the mechanism underlying radiosensitization by dFdCyd. These data showed that the presence and persistence of mismatched nucleotides is integral to radiosensitization by dFdCyd and suggest a role for hMLH1 deficiency in eliciting the radiosensitizing effect.


Assuntos
Pareamento Incorreto de Bases , Desoxicitidina/análogos & derivados , Radiossensibilizantes/farmacologia , Linhagem Celular Tumoral , Desoxicitidina/farmacologia , Humanos , Mutação , RNA Interferente Pequeno , Gencitabina
8.
Methods Mol Biol ; 1534: 165-173, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27812878

RESUMO

DNA damage response has been characterized as an important mediator of senescence phenotypes induced by activated oncogenes in normal human cells. Depletion of intracellular deoxyribonucleotide pools has been recently recognized as one of the major causes for DNA damage in these cells. Cells undergoing oncogene-induced senescence display decreased expression of several rate-limiting enzymes involved in the biosynthesis of deoxyribonucleotides, including thymidylate synthase (TS) and ribonucleotide reductase (RR). Individual depletion of these enzymes leads to premature senescence. Reciprocally, ectopic expression of TS and RR or addition of deoxyribonucleosides resulted in suppression of senescence phenotypes in normal or tumor cells caused by overexpression of activated HRAS or depletion of C-MYC, respectively. Therefore, in the current chapter, we will describe a methodology for the quantitative measurement of nucleotide pools in senescent cells.


Assuntos
Senescência Celular , Nucleotídeos/metabolismo , Oncogenes , Linhagem Celular , Senescência Celular/genética , Cromatografia Líquida de Alta Pressão , Dano ao DNA , Desoxirribonucleotídeos/química , Desoxirribonucleotídeos/metabolismo , Fibroblastos/metabolismo , Expressão Gênica , Genes myc , Humanos , Nucleotídeos/química , Oncogenes/genética , Ribonucleotídeo Redutases/metabolismo , Timidilato Sintase/metabolismo
10.
Cancer Res ; 63(20): 6935-41, 2003 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-14583494

RESUMO

Gemcitabine [2',2'-difluoro-2'-deoxycytidine (dFdCyd)] is a potent ionizing radiation sensitizer in solid tumor cells in vitro and in vivo. Previously, we have demonstrated (Shewach et al., Cancer Res., 54: 3218-3223, 1994) a strong correlation between depletion of dATP (caused by dFdCyd diphosphate-mediated inhibition of ribonucleotide reductase) and radiosensitization. In addition, we and others (Latz et al., Int. J. Radiat. Oncol. Biol. Phys., 41: 875-882, 1998; Ostruszka and Shewach, Cancer Res., 60: 6080-6088, 2000) have shown that the accumulation of cells in S phase prior to irradiation is also important for radiosensitization with dFdCyd. This led us to hypothesize that the incorporation of incorrect nucleotides because of the dATP pool imbalance was important for radiosensitization with dFdCyd, and, therefore, cells deficient in mismatch repair (MMR) would exhibit greater radiosensitization. We tested this hypothesis by evaluating the ability of HCT116 colon carcinoma cell lines, which differ in MMR proficiency, to be radiosensitized by dFdCyd. The MMR-proficient cell line (HCT116 + ch3) was more sensitive to dFdCyd alone than were the MMR-deficient cell lines (HCT116, HCT116 + ch2, and HCT116 p53(-/-)). Interestingly, the MMR-proficient cells could not be radiosensitized at concentrations of dFdCyd IC(96)) enhanced cell killing with radiation. In contrast, the MMR-deficient cells were radiosensitized at concentrations of dFdCyd or=80% decrease in dATP within 4 h after drug addition, and this low dATP level was maintained for another 12-20 h. Although the IC(50) of dFdCyd was unable to sustain a >80% decrease in the dATP level in the MMR-proficient cells, the IC(90) did achieve this level of dATP depletion; however, it was unable to radiosensitize the MMR-proficient cells. Similar results were obtained with HCT116 cells, in which the MMR deficiency was corrected by transfection with a vector containing the hMLH1 cDNA. In addition, the deletion of p53 did not increase radiation enhancement ratios. These results demonstrate that MMR deficiency promotes radiosensitization with dFdCyd. We suggest that dATP depletion produces errors of replication in MMR-deficient cells, which, if left unrepaired, enhances cell death by ionizing radiation.


Assuntos
Pareamento Incorreto de Bases/fisiologia , Reparo do DNA/fisiologia , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Radiossensibilizantes/farmacologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , DNA de Neoplasias/metabolismo , Nucleotídeos de Desoxiadenina/metabolismo , Células HCT116 , Humanos , Nucleotídeos/metabolismo , Tolerância a Radiação/genética , Gencitabina
11.
Radiat Res ; 186(5): 466-477, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27740890

RESUMO

Gemcitabine (dFdCyd) shows broad antitumor activity in solid tumors in chemotherapeutic regimens or when combined with ionizing radiation (radiosensitization). While it is known that mismatches in DNA are necessary for dFdCyd radiosensitization, the critical event resulting in radiosensitization has not been identified. Here we hypothesized that late DNA damage (≥24 h after drug washout/irradiation) is a causal event in radiosensitization by dFdCyd, and that homologous recombination repair (HRR) is required for this late DNA damage. Using γ-H2AX as a measurement of DNA damage in MCF-7 breast cancer cells, we demonstrate that 10 or 80 nM dFdCyd alone produced significantly more late DNA damage compared to that observed within 4 h after treatment. The combination of dFdCyd treatment followed by irradiation did not produce a consistent increase in DNA damage in the first 4 h after treatment, however, there was a synergistic increase 24-48 h later relative to treatment with dFdCyd or radiation alone. RNAi suppression of the essential HRR protein, XRCC3, significantly decreased both radiosensitization and late DNA damage. Furthermore, inhibition of HRR with the Rad51 inhibitor B02 prevented radiosensitization when added after, but not during, treatment with dFdCyd and radiation. To our knowledge, this is the first published study to show that radiosensitization with dFdCyd results from a synergistic increase in DNA damage at 24-48 h after drug and radiation treatment, and that this damage and radiosensitization require HRR. These results suggest that tumors that overexpress HRR will be more vulnerable to chemoradiotherapy, and treatments that increase HRR and/or mismatches in DNA will enhance dFdCyd radiosensitization.


Assuntos
Dano ao DNA , Desoxicitidina/análogos & derivados , Radiossensibilizantes/farmacologia , Reparo de DNA por Recombinação/efeitos dos fármacos , Reparo de DNA por Recombinação/efeitos da radiação , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Proteínas de Ligação a DNA/metabolismo , Desoxicitidina/farmacologia , Relação Dose-Resposta a Droga , Raios gama , Histonas/metabolismo , Humanos , Células MCF-7 , Rad51 Recombinase/antagonistas & inibidores , Fatores de Tempo , Gencitabina
12.
Radiat Res ; 183(1): 114-23, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25564718

RESUMO

Gemcitabine (difluorodeoxycytidine; dFdCyd) is a potent radiosensitizer, noted for its ability to enhance cytotoxicity with radiation at noncytotoxic concentrations in vitro and subchemotherapeutic doses in patients. Radiosensitization in human tumor cells requires dFdCyd-mediated accumulation of cells in S phase with inhibition of ribonucleotide reductase, resulting in ≥80% deoxyadenosine triphosphate (dATP) depletion and errors of replication in DNA. Less is known of the role of specific DNA replication and repair pathways in the radiosensitization mechanism. Here the role of homologous recombination (HR) in relationship to the metabolic and cell cycle effects of dFdCyd was investigated using a matched pair of CHO cell lines that are either proficient (AA8 cells) or deficient (irs1SF cells) in HR based on expression of the HR protein XRCC3. The results demonstrated that the characteristics of radiosensitization in the rodent AA8 cells differed significantly from those in human tumor cells. In the AA8 cells, radiosensitization was achieved only under short (≤4 h) cytotoxic incubations, and S-phase accumulation did not appear to be required for radiosensitization. In contrast, human tumor cell lines were radiosensitized using noncytotoxic concentrations of dFdCyd and required early S-phase accumulation. Studies of the metabolic effects of dFdCyd demonstrated low dFdCyd concentrations did not deplete dATP by ≥80% in AA8 and irs1SF cells. However, at higher concentrations of dFdCyd, failure to radiosensitize the HR-deficient irs1SF cells could not be explained by a lack of dATP depletion or lack of S-phase accumulation. Thus, these parameters did not correspond to dFdCyd radiosensitization in the CHO cells. To evaluate directly the role of HR in radiosensitization, XRCC3 expression was suppressed in the AA8 cells with a lentiviral-delivered shRNA. Partial XRCC3 suppression significantly decreased radiosensitization [radiation enhancement ratio (RER) = 1.6 ± 0.15], compared to nontransduced (RER = 2.7 ± 0.27; P = 0.012), and a substantial decrease compared to nonspecific shRNA-transduced (RER = 2.5 ± 0.42; P = 0.056) AA8 cells. Although the results support a role for HR in radiosensitization with dFdCyd in CHO cells, the differences in the underlying metabolic and cell cycle characteristics suggest that dFdCyd radiosensitization in the nontumor-derived CHO cells is mechanistically distinct from that in human tumor cells.


Assuntos
Desoxicitidina/análogos & derivados , Radiossensibilizantes/metabolismo , Radiossensibilizantes/farmacologia , Reparo de DNA por Recombinação/efeitos dos fármacos , Reparo de DNA por Recombinação/efeitos da radiação , Animais , Células CHO , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Proteínas de Ligação a DNA/metabolismo , Desoxicitidina/metabolismo , Desoxicitidina/farmacologia , Humanos , Fosfatos/metabolismo , Gencitabina
13.
EMBO Mol Med ; 7(9): 1138-52, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26197802

RESUMO

Chromosomal instability in early cancer stages is caused by replication stress. One mechanism by which oncogene expression induces replication stress is to drive cell proliferation with insufficient nucleotide levels. Cancer development is driven by alterations in both genetic and environmental factors. Here, we investigated whether replication stress can be modulated by both genetic and non-genetic factors and whether the extent of replication stress affects the probability of neoplastic transformation. To do so, we studied the effect of folate, a micronutrient that is essential for nucleotide biosynthesis, on oncogene-induced tumorigenicity. We show that folate deficiency by itself leads to replication stress in a concentration-dependent manner. Folate deficiency significantly enhances oncogene-induced replication stress, leading to increased DNA damage and tumorigenicity in vitro. Importantly, oncogene-expressing cells, when grown under folate deficiency, exhibit a significantly increased frequency of tumor development in mice. These findings suggest that replication stress is a quantitative trait affected by both genetic and non-genetic factors and that the extent of replication stress plays an important role in cancer development.


Assuntos
Carcinogênese/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Ácido Fólico/metabolismo , Instabilidade Genômica/efeitos dos fármacos , Oncogenes/efeitos dos fármacos , Animais , Camundongos
14.
Hum Gene Ther ; 13(4): 543-51, 2002 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11874632
15.
Semin Oncol ; 31(2 Suppl 5): 2-12, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15199526

RESUMO

Because treatment regimens for breast cancer commonly include gemcitabine, we evaluated two promising combinations in preclinical studies: gemcitabine (Gemzar; Eli Lilly and Company, Indianapolis, IN) with either ionizing radiation or docetaxel (Taxotere; Aventis Pharmaceuticals, Inc, Parsippany, NJ). In breast cancer cell lines that expressed either wild-type p53 (MCF-7) or mutant p53 (MCF-7/Adr), sensitivity to the cytotoxic effects of gemcitabine during a 24-hour incubation was similar (IC(50) values 80 and 60 nmol/L in MCF-7 and MCF-7/Adr, respectively). Both cell lines were well radiosensitized by gemcitabine at the corresponding IC(50), with radiation enhancement ratios of 1.6 to 1.7. Although the MCF-7 cells accumulated nearly twice as much gemcitabine triphosphate compared with the MCF-7/Adr cells, a similar reduction in 2'-deoxyadenosine 5'-triphosphate pools was observed. While the number of dying cells, as measured by sub-G1 DNA content or S-phase cells unable to replicate DNA, differed between the wild-type p53 or mutant p53-expressing cell lines, neither parameter correlated with radiosensitization. Docetaxel was a more potent cytotoxic agent than gemcitabine in MCF-7 cells (IC(50) = 1 nmol/L). Strong synergistic cytotoxicity was observed in cells treated with gemcitabine (24 hours) followed by docetaxel (24 hours) or the reverse sequence. However, simultaneous addition of the two drugs was antagonistic. To determine whether synergy with radiation or docetaxel was mediated by increased DNA damage, DNA double-strand breaks (double-strand breaks) were measured by immunostaining for phosphorylated H2AX. Ionizing radiation produced more double-strand breaks than gemcitabine alone, while no significant double-strand breaks formed with docetaxel alone. The addition of docetaxel or ionizing radiation to gemcitabine-treated cells did not increase H2AX foci formation. These results show that the combination of gemcitabine with ionizing radiation or docetaxel produces strong, schedule-dependent synergy in breast cancer cells that is not mediated through increasing DNA double-strand breaks.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Desoxicitidina/análogos & derivados , Desoxicitidina/administração & dosagem , Neoplasias da Mama/radioterapia , Linhagem Celular Tumoral , DNA de Neoplasias/efeitos dos fármacos , DNA de Neoplasias/efeitos da radiação , Docetaxel , Sinergismo Farmacológico , Histonas , Humanos , Radiossensibilizantes/administração & dosagem , Radioterapia Adjuvante , Taxoides/administração & dosagem , Ensaio Tumoral de Célula-Tronco , Gencitabina
16.
Cancer Chemother Pharmacol ; 52(4): 325-32, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12811514

RESUMO

PURPOSE: Cytotoxicity from the anticancer drug 2',2'-difluoro-2'-deoxycytidine (dFdCyd) has been correlated with its incorporation into DNA. However, cytotoxicity may also result from inhibition of DNA synthesis, due to either (1) dFdCyd diphosphate-mediated inhibition of ribonucleotide reductase, or (2) direct inhibition of DNA polymerases by the 5'-triphosphate of dFdCyd (dFdCTP). To elucidate the role of DNA synthesis inhibition in the cytotoxicity of dFdCyd, we compared dFdCyd to hydroxyurea (HU), a ribonucleotide reductase inhibitor, and aphidicolin, an inhibitor of DNA polymerases, in the U251 and D54 human glioblastoma cell lines. METHODS: Sensitivity to dFdCyd, HU, and aphidicolin were determined using a colony formation assay. The effects of these drugs on DNA synthesis were measured by dual parameter flow cytometry, while the effects on nucleotide pool levels were analyzed by high-performance liquid chromatography. RESULTS: HU and aphidicolin elicited substantially less cytotoxicity than the multi-log killing with dFdCyd. When used at equitoxic concentrations (24-h IC50 values), dFdCyd and HU decreased purine dNTP pools primarily, but dFdCyd was less effective than HU. dFdCyd had decreased dATP by about 80% after 4-12 h, and required 8-24 h to decrease DNA synthesis by 50%. In contrast, HU rapidly depleted dATP by >98% within 2 h, which resulted in >90% inhibition of DNA synthesis. Aphidicolin at a concentration similar to its Ki values for DNA polymerases (1 microM) decreased DNA synthesis by >70% within 2 h. However, this decreased cell survival by only 10% (U251 cells) and 40% (D54 cells). CONCLUSIONS: These results demonstrate that HU and aphidicolin produced a more rapid and profound inhibition of DNA synthesis than dFdCyd, but resulted in significantly less cytotoxicity. This suggests that inhibition of DNA synthesis accounted for less than one log of the multi-log cytotoxicity observed with dFdCyd, whereas incorporation of dFdCTP into DNA is a more lethal event.


Assuntos
Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Inibidores da Síntese de Ácido Nucleico , Antineoplásicos/farmacologia , Afidicolina/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , DNA de Neoplasias/biossíntese , Desoxicitidina/metabolismo , Inibidores Enzimáticos/farmacologia , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Humanos , Hidroxiureia/farmacologia , Nucleosídeos de Pirimidina/metabolismo , Ribonucleotídeo Redutases/antagonistas & inibidores , Gencitabina
17.
DNA Repair (Amst) ; 12(12): 1114-21, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24231389

RESUMO

The nucleoside analog ganciclovir (GCV) elicits cytotoxicity in tumor cells via a novel mechanism in which drug incorporation into DNA produces minimal disruption of replication, but numerous DNA double strand breaks occur during the second S-phase after drug exposure. We propose that homologous recombination (HR), a major repair pathway for DNA double strand breaks, can prevent GCV-induced DNA damage, and that inhibition of HR will enhance cytotoxicity with GCV. Survival after GCV treatment in cells expressing a herpes simplex virus thymidine kinase was strongly dependent on HR (>14-fold decrease in IC50 in HR-deficient vs. HR-proficient CHO cells). In a homologous recombination reporter assay, the histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA; vorinostat), decreased HR repair events up to 85%. SAHA plus GCV produced synergistic cytotoxicity in U251tk human glioblastoma cells. Elucidation of the synergistic mechanism demonstrated that SAHA produced a concentration-dependent decrease in the HR proteins Rad51 and CtIP. GCV alone produced numerous Rad51 foci, demonstrating activation of HR. However, the addition of SAHA blocked GCV-induced Rad51 foci formation completely and increased γH2AX, a marker of DNA double strand breaks. SAHA plus GCV also produced synergistic cytotoxicity in HR-proficient CHO cells, but the combination was antagonistic or additive in HR-deficient CHO cells. Collectively, these data demonstrate that HR promotes survival with GCV and compromise of HR by SAHA results in synergistic cytotoxicity, revealing a new mechanism for enhancing anticancer activity with GCV.


Assuntos
Antineoplásicos/farmacologia , Ganciclovir/farmacologia , Recombinação Homóloga/efeitos dos fármacos , Ácidos Hidroxâmicos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Células CHO , Proteínas de Transporte/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cricetulus , Endodesoxirribonucleases , Células HeLa , Humanos , Proteínas Nucleares/metabolismo , Rad51 Recombinase/metabolismo , Vorinostat
18.
Adv Healthc Mater ; 2(10): 1337-50, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23554387

RESUMO

Poly(amidoamine) (PAMAM) dendrimers are branched water-soluble polymers defined by consecutive generation numbers (Gn) indicating a parallel increase in size, molecular weight, and number of surface groups available for conjugation of bioactive agents. In this article, we compare the biodistribution of N-acetylgalactosamine (NAcGal)-targeted [(14) C]1 -G5-(NH2 )5 -(Ac)108 -(NAcGal)14 particles to non-targeted [(14) C]1 -G5-(NH2 )127 and PEGylated [(14) C]1 -G5-(NH2 )44 -(Ac)73 -(PEG)10 particles in a mouse hepatic cancer model. Results show that both NAcGal-targeted and non-targeted particles are rapidly cleared from the systemic circulation with high distribution to the liver. However, NAcGal-targeted particles exhibited 2.5-fold higher accumulation in tumor tissue compared to non-targeted ones. In comparison, PEGylated particles showed a 16-fold increase in plasma residence time and a 5-fold reduction in liver accumulation. These results motivated us to engineer new PEGylated G5 particles with PEG chains anchored to the G5 surface via acid-labile cis-aconityl linkages where the free PEG tips are functionalized with NAcGal or SP94 peptide to investigate their potential as targeting ligands for hepatic cancer cells as a function of sugar conformation (α versus ß), ligand concentration (100-4000 nM), and incubation time (2 and 24 hours) compared to fluorescently (Fl)-labeled and non-targeted G5-(Fl)6 -(NH2 )122 and G5-(Fl)6 -(Ac)107 -(cPEG)15 particles. Results show G5-(Fl)6 -(Ac)107 -(cPEG[NAcGalß ])14 particles achieve faster uptake and higher intracellular concentrations in HepG2 cancer cells compared to other G5 particles while escaping the non-specific adsorption of serum protein and phagocytosis by Kupffer cells, which make these particles the ideal carrier for selective drug delivery into hepatic cancer cells.


Assuntos
Acetilgalactosamina/química , Acetilglucosamina/análogos & derivados , Dendrímeros/química , Peptídeos/química , Polietilenoglicóis/química , Acetilglucosamina/química , Acetilglucosamina/farmacocinética , Acetilglucosamina/uso terapêutico , Animais , Antígenos CD7/química , Antígenos CD7/metabolismo , Células Cultivadas , Dendrímeros/farmacocinética , Dendrímeros/uso terapêutico , Corantes Fluorescentes/química , Meia-Vida , Células Hep G2 , Humanos , Células de Kupffer/citologia , Células de Kupffer/efeitos dos fármacos , Ligantes , Fígado/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Nus , Tamanho da Partícula , Peptídeos/metabolismo , Fagocitose , Polietilenoglicóis/farmacocinética , Polietilenoglicóis/uso terapêutico , Ratos , Fatores de Tempo , Distribuição Tecidual , Transplante Heterólogo
19.
Cell Rep ; 5(2): 493-507, 2013 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-24139804

RESUMO

Melanoma is one of the most aggressive types of human cancers, and the mechanisms underlying melanoma invasive phenotype are not completely understood. Here, we report that expression of guanosine monophosphate reductase (GMPR), an enzyme involved in de novo biosynthesis of purine nucleotides, was downregulated in the invasive stages of human melanoma. Loss- and gain-of-function experiments revealed that GMPR downregulates the amounts of several GTP-bound (active) Rho-GTPases and suppresses the ability of melanoma cells to form invadopodia, degrade extracellular matrix, invade in vitro, and grow as tumor xenografts in vivo. Mechanistically, we demonstrated that GMPR partially depletes intracellular GTP pools. Pharmacological inhibition of de novo GTP biosynthesis suppressed whereas addition of exogenous guanosine increased invasion of melanoma cells as well as cells from other cancer types. Our data identify GMPR as a melanoma invasion suppressor and establish a link between guanosine metabolism and Rho-GTPase-dependent melanoma cell invasion.


Assuntos
GMP Redutase/metabolismo , Melanoma/enzimologia , Nucleosídeos de Purina/biossíntese , Animais , Linhagem Celular Tumoral , Movimento Celular , Matriz Extracelular/metabolismo , GMP Redutase/antagonistas & inibidores , GMP Redutase/genética , Guanosina Trifosfato/metabolismo , Células HCT116 , Humanos , IMP Desidrogenase/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Fenótipo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transplante Heterólogo , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
20.
Int J Radiat Oncol Biol Phys ; 84(5): e613-20, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22867891

RESUMO

PURPOSE: To determine the effect of short hairpin ribonucleic acid (shRNA)-mediated suppression of thymidylate synthase (TS) on cytotoxicity and radiosensitization and the mechanism by which these events occur. METHODS AND MATERIALS: shRNA suppression of TS was compared with 5-fluoro-2'-deoxyuridine (FdUrd) inactivation of TS with or without ionizing radiation in HCT116 and HT29 colon cancer cells. Cytotoxicity and radiosensitization were measured by clonogenic assay. Cell cycle effects were measured by flow cytometry. The effects of FdUrd or shRNA suppression of TS on dNTP deoxynucleotide triphosphate imbalances and consequent nucleotide misincorporations into deoxyribonucleic acid (DNA) were analyzed by high-pressure liquid chromatography and as pSP189 plasmid mutations, respectively. RESULTS: TS shRNA produced profound (≥ 90%) and prolonged (≥ 8 days) suppression of TS in HCT116 and HT29 cells, whereas FdUrd increased TS expression. TS shRNA also produced more specific and prolonged effects on dNTPs deoxynucleotide triphosphates compared with FdUrd. TS shRNA suppression allowed accumulation of cells in S-phase, although its effects were not as long-lasting as those of FdUrd. Both treatments resulted in phosphorylation of Chk1. TS shRNA alone was less cytotoxic than FdUrd but was equally effective as FdUrd in eliciting radiosensitization (radiation enhancement ratio: TS shRNA, 1.5-1.7; FdUrd, 1.4-1.6). TS shRNA and FdUrd produced a similar increase in the number and type of pSP189 mutations. CONCLUSIONS: TS shRNA produced less cytotoxicity than FdUrd but was equally effective at radiosensitizing tumor cells. Thus, the inhibitory effect of FdUrd on TS alone is sufficient to elicit radiosensitization with FdUrd, but it only partially explains FdUrd-mediated cytotoxicity and cell cycle inhibition. The increase in DNA mismatches after TS shRNA or FdUrd supports a causal and sufficient role for the depletion of dTTP thymidine triphosphate and consequent DNA mismatches underlying radiosensitization. Importantly, shRNA suppression of TS avoids FP-mediated TS elevation and its negative prognostic role. These studies support the further exploration of TS suppression as a novel radiosensitizing strategy.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Reparo de Erro de Pareamento de DNA , Floxuridina/farmacologia , RNA Interferente Pequeno/farmacologia , Tolerância a Radiação/genética , Timidilato Sintase/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem , Citidina Trifosfato/metabolismo , Ativação Enzimática/efeitos dos fármacos , Guanosina Trifosfato/metabolismo , Células HT29 , Humanos , Fosforilação , Proteínas Quinases/metabolismo , Ensaio Tumoral de Célula-Tronco/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA