Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Mol Cancer Res ; 19(9): 1571-1582, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34045311

RESUMO

Programmed death-ligand 1 (PD-L1) promotes tumor immune evasion by engaging the PD-1 receptor and inhibiting T-cell activity. While the regulation of PD-L1 expression is not fully understood, its expression is associated with tumor mutational burden and response to immune checkpoint therapy. Here, we report that Apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like 3A (APOBEC3A) is an important regulator of PD-L1 expression. Using an APOBEC3A inducible expression system as well as siRNA against endogenous APOBEC3A, we found that APOBEC3A regulates PD-L1 mRNA and protein levels as well as PD-L1 cell surface expression in cancer. Mechanistically, APOBEC3A-induced PD-L1 expression was dependent on APOBEC3A catalytic activity as catalytically dead APOBEC3A mutant (E72A) failed to induce PD-L1 expression. Furthermore, APOBEC3A-induced PD-L1 expression was dependent on replication-associated DNA damage and JNK/c-JUN signaling but not interferon signaling. In addition, we confirmed the relevance of these finding in patient tumors as APOBEC3A expression and mutational signature correlated with PD-L1 expression in multiple patient cancer types. These data provide a novel link between APOBEC3A, its DNA mutagenic activity and PD-L1-mediated antitumoral immunity. This work nominates APOBEC3A as a mechanism of immune evasion and a potential biomarker for the therapeutic efficacy of immune checkpoint blockade. IMPLICATIONS: APOBEC3A catalytic activity induces replication-associated DNA damage to promote PD-L1 expression implying that APOBEC3A-driven mutagenesis represents both a mechanism of tumor immune evasion and a therapeutically targetable vulnerability in cancer cells.


Assuntos
Antígeno B7-H1/metabolismo , Biomarcadores Tumorais/metabolismo , Citidina Desaminase/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Neoplasias/patologia , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Apoptose , Antígeno B7-H1/genética , Biomarcadores Tumorais/genética , Proliferação de Células , Citidina Desaminase/genética , Humanos , Proteína Quinase 8 Ativada por Mitógeno/genética , Neoplasias/genética , Neoplasias/metabolismo , Prognóstico , Proteínas/genética , Proteínas Proto-Oncogênicas c-jun/genética , Células Tumorais Cultivadas
2.
Mol Cancer Ther ; 20(2): 263-273, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33268569

RESUMO

PARP inhibitor monotherapy (olaparib) was recently FDA approved for the treatment of BRCA1/2-mutant, homologous recombination (HR) repair-deficient pancreatic cancer. Most pancreatic cancers, however, are HR proficient and thus resistant to PARP inhibitor monotherapy. We tested the hypothesis that combined therapy with radiation and ataxia telangiectasia and Rad3-related (ATR) inhibitor (AZD6738) would extend the therapeutic indication of olaparib to HR-proficient pancreatic cancers. We show that olaparib combined with AZD6738 significantly reduced radiation survival relative to either agent alone, regardless of HR status. Whereas catalytic inhibition of PARP with low concentrations of olaparib radiosensitized HR-deficient models, maximal sensitization in HR-proficient models required concentrations of olaparib that induce formation of PARP1-DNA complexes. Furthermore, CRISPR-Cas9-mediated PARP1 deletion failed to recapitulate the effects of olaparib on radiosensitivity and negated the combinatorial efficacy of olaparib and AZD6738 on radiosensitization, suggesting that PARP1-DNA complexes, rather than PARP catalytic inhibition, were responsible for radiosensitization. Mechanistically, therapeutic concentrations of olaparib in combination with radiation and AZD6738 increased DNA double-strand breaks. DNA fiber combing revealed that high concentrations of olaparib did not stall replication forks but instead accelerated replication fork progression in association with an ATR-mediated replication stress response that was antagonized by AZD6738. Finally, in HR-proficient tumor xenografts, the combination of olaparib, radiation, and AZD6738 significantly delayed tumor growth compared with all other treatments. These findings suggest that PARP1-DNA complexes are required for the therapeutic activity of olaparib combined with radiation and ATR inhibitor in HR-proficient pancreatic cancer and support the clinical development of this combination for tumors intrinsically resistant to PARP inhibitors.


Assuntos
Terapia Combinada/métodos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/radioterapia , Ftalazinas/uso terapêutico , Piperazinas/uso terapêutico , Poli(ADP-Ribose) Polimerase-1/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/patologia , Ftalazinas/farmacologia , Piperazinas/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Neoplasias Pancreáticas
3.
Proc Natl Acad Sci U S A ; 114(31): 8241-8246, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28716944

RESUMO

Human ribonucleotide reductase (hRR) is crucial for DNA replication and maintenance of a balanced dNTP pool, and is an established cancer target. Nucleoside analogs such as gemcitabine diphosphate and clofarabine nucleotides target the large subunit (hRRM1) of hRR. These drugs have a poor therapeutic index due to toxicity caused by additional effects, including DNA chain termination. The discovery of nonnucleoside, reversible, small-molecule inhibitors with greater specificity against hRRM1 is a key step in the development of more effective treatments for cancer. Here, we report the identification and characterization of a unique nonnucleoside small-molecule hRR inhibitor, naphthyl salicylic acyl hydrazone (NSAH), using virtual screening, binding affinity, inhibition, and cell toxicity assays. NSAH binds to hRRM1 with an apparent dissociation constant of 37 µM, and steady-state kinetics reveal a competitive mode of inhibition. A 2.66-Å resolution crystal structure of NSAH in complex with hRRM1 demonstrates that NSAH functions by binding at the catalytic site (C-site) where it makes both common and unique contacts with the enzyme compared with NDP substrates. Importantly, the IC50 for NSAH is within twofold of gemcitabine for growth inhibition of multiple cancer cell lines, while demonstrating little cytotoxicity against normal mobilized peripheral blood progenitor cells. NSAH depresses dGTP and dATP levels in the dNTP pool causing S-phase arrest, providing evidence for RR inhibition in cells. This report of a nonnucleoside reversible inhibitor binding at the catalytic site of hRRM1 provides a starting point for the design of a unique class of hRR inhibitors.


Assuntos
Hidrazonas/farmacologia , Naftalenos/farmacologia , Ribonucleotídeo Redutases/antagonistas & inibidores , Salicilatos/farmacologia , Domínio Catalítico , Ciclo Celular/efeitos dos fármacos , Cristalografia por Raios X , Nucleotídeos de Desoxiadenina/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Humanos , Hidrazonas/química , Naftalenos/química , Ribonucleosídeo Difosfato Redutase , Ribonucleotídeo Redutases/química , Ribonucleotídeo Redutases/metabolismo , Salicilatos/química , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/metabolismo
4.
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
5.
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
6.
Nurs Times ; 110(38): 20-2, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-26012012

RESUMO

Student nurses need a variety of high-quality practice placements to prepare them for qualification yet, in reality, this can be difficult to achieve. A practice placement allocation model has enabled one university and its partner healthcare organisations to shift from a traditional, process-led system to a robust, proactive, student-focused approach. The model is based on partnership concepts including advance planning of student placements and clear lines of communication. It has resulted in 100% of first-year students taking part in a new fundamentals of care placement and received positive feedback from students and mentors.


Assuntos
Estudantes de Enfermagem , Inglaterra , Modelos Educacionais
7.
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
8.
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
9.
Aging (Albany NY) ; 4(12): 917-22, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23249808

RESUMO

The down-regulation of dominant oncogenes, including C-MYC, in tumor cells often leads to the induction of senescence via mechanisms that are not completely identified. In the current study, we demonstrate that MYC-depleted melanoma cells undergo extensive DNA damage that is caused by the underexpression of thymidylate synthase (TS) and ribonucleotide reductase (RR) and subsequent depletion of deoxyribonucleoside triphosphate pools. Simultaneous genetic inhibition of TS and RR in melanoma cells induced DNA damage and senescence phenotypes very similar to the ones caused by MYC-depletion. Reciprocally, overexpression of TS and RR in melanoma cells or addition of deoxyribo-nucleosides to culture media substantially inhibited DNA damage and senescence-associated phenotypes caused by C-MYC depletion. Our data demonstrate the essential role of TS and RR in C-MYC-dependent suppression of senescence in melanoma cells.


Assuntos
Senescência Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Desoxirribonucleosídeos/farmacologia , Melanoma/enzimologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Ribonucleotídeo Redutases/metabolismo , Neoplasias Cutâneas/enzimologia , Timidilato Sintase/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Genótipo , Humanos , Melanoma/genética , Melanoma/patologia , Fenótipo , Proteínas Proto-Oncogênicas c-myc/genética , Interferência de RNA , Ribonucleosídeo Difosfato Redutase/metabolismo , Ribonucleotídeo Redutases/genética , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Timidilato Sintase/genética , Fatores de Tempo , Transfecção , Proteínas Supressoras de Tumor/metabolismo
10.
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
11.
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
12.
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
13.
Leuk Lymphoma ; 48(9): 1816-27, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17786719

RESUMO

Guanosine initiated apoptosis in NB4 cells in a transport-dependent manner. Apoptosis was partially attributed to an imbalance in nucleosides with some protection upon the addition of pyrimidines. The effect of guanosine on cell proliferation and viability was biphasic whereby cells were able to recover from an initial cell cycle arrest and re-enter the cell cycle upon removal of guanosine in a time-dependent fashion. However, exposure to guanosine beyond 24 h prevented recovery and ultimately led to death. Death occurred with a decrease in bcl-2 protein expression, thus suggesting that the pathway to apoptosis involved change(s) in the intracellular environment that were ultimately sensed by the mitochondria. Expression of the unique guanosine-specific nucleoside transporter csg in NB4 cells may provide an opportunity to harness guanosine-mediated cell death in the treatment of APL and related malignancies while sparing normal cells.


Assuntos
Apoptose/efeitos dos fármacos , Guanosina/farmacologia , Leucemia Promielocítica Aguda/tratamento farmacológico , Transporte Biológico , Caspase 3/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , DNA/análise , Fragmentação do DNA , Dipiridamol/farmacologia , Guanosina/metabolismo , Humanos , Leucemia Promielocítica Aguda/patologia , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/análise , Pirimidinas/farmacologia
14.
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
15.
Leuk Res ; 31(7): 955-68, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17045336

RESUMO

NB4 cells express multiple nucleoside transporters (NTs), including: hENT1 (es), and hENT2 (ei), and the CNT subtype referred to as, csg; a concentrative sensitive guanosine specific transporter. csg activity is a distinguishing feature of the NB4 cell line and its presence suggests a particular requirement of these cells for guanosine salvage. Proliferation and differentiation pathways determine, in part, the number of NTs in cells and tissues. In this study, all-trans-retinoic acid (ATRA)-induced granulocytic differentiation of NB4 cells resulted in biphasic changes in guanosine transport. Transient increases in csg and es activity, the result of an increase in V(max) (pmol/muls) of both transporter systems, served as early markers of differentiation while expression of a fully differentiated phenotype was accompanied by a selective loss of csg activity and the return of es activity to that of proliferating cells. Intracellular incorporation of [(3)H]-guanosine decreased as cells matured despite increased transport rates and suggested a reduced intracellular requirement of NB4-granulocytes compared to their proliferating counterparts. Whether a loss of csg activity could serve to assess clinical response to differentiation therapies is not known. Nitrobenzylthioinosine (NBMPR) binding sites within nuclear membrane (NM) preparations, suggested the presence of functional intracellular NTs. An increase in plasma membrane (PM) associated transporters coincided with the early increase in guanosine transport and a decrease in NBMPR binding to NM fractions and suggests that intracellular NTs may serve as a reserve pool for translocation to the (PM) when additional transport capacity is required. The modulation of transporters during differentiation could potentially regulate drug bioavailability and cytotoxicity and should be evaluated prior to combining differentiating agents with traditional nucleoside analogs in the treatment of APL.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Granulócitos/patologia , Guanosina/metabolismo , Leucemia Promielocítica Aguda/patologia , Proteínas de Transporte de Nucleosídeos/metabolismo , Tretinoína/farmacologia , Marcadores de Afinidade/metabolismo , Transporte Biológico , Membrana Celular , Humanos , Cinética , Leucemia Promielocítica Aguda/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Frações Subcelulares , Tioinosina/análogos & derivados , Tioinosina/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo , Uridina/metabolismo
16.
Biochem Pharmacol ; 66(5): 733-7, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12948853

RESUMO

We studied acceptance of various deoxyguanosine analogues by the unique guanosine preferring nucleoside transport system exhibited by NB4 cells, csg. Indirect assessment of acceptance using transport inhibition assays revealed that both 1-beta-D-arabinofuranosylguanine (ara-G) and 4'-thio-beta-D-xylofuranosylguanine (thio-xyl-G) compete with guanosine for the csg system, inhibiting guanosine flux by approximately 50%. Direct examination of [3H]-ara-G transport revealed total transport was equally allocated to csg, and es systems and a total transport rate similar to that determined for guanosine [Flanagan and Meckling-Gill, J Biol Chem 1997;272:18026-32]. Cytotoxicity assays revealed that although both ara-G and thio-xyl-G were capable of competing with guanosine for the csg system, neither analogue elicited cytotoxic effects at physiologically relevant concentrations. The analog, 4'-thio-beta-D-arabinofuranosylguanine does not gain entry to NB4 cells via the csg transport system. Competition assays revealed that this analogue potentiated the inward flux of guanosine and was capable of killing NB4 cells with potency similar to the conventional leukemia drug, ara-C.


Assuntos
Antineoplásicos/farmacocinética , Arabinonucleosídeos/farmacocinética , Desoxiguanosina/farmacocinética , Antineoplásicos/farmacologia , Arabinonucleosídeos/farmacologia , Transporte Biológico , Divisão Celular/efeitos dos fármacos , Desoxiguanosina/farmacologia , Guanosina/metabolismo , Humanos , Trítio , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA