Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Nucleic Acids Res ; 50(3): 1484-1500, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35037045

RESUMO

The SARS-CoV-2 coronavirus is the causal agent of the current global pandemic. SARS-CoV-2 belongs to an order, Nidovirales, with very large RNA genomes. It is proposed that the fidelity of coronavirus (CoV) genome replication is aided by an RNA nuclease complex, comprising the non-structural proteins 14 and 10 (nsp14-nsp10), an attractive target for antiviral inhibition. Our results validate reports that the SARS-CoV-2 nsp14-nsp10 complex has RNase activity. Detailed functional characterization reveals nsp14-nsp10 is a versatile nuclease capable of digesting a wide variety of RNA structures, including those with a blocked 3'-terminus. Consistent with a role in maintaining viral genome integrity during replication, we find that nsp14-nsp10 activity is enhanced by the viral RNA-dependent RNA polymerase complex (RdRp) consisting of nsp12-nsp7-nsp8 (nsp12-7-8) and demonstrate that this stimulation is mediated by nsp8. We propose that the role of nsp14-nsp10 in maintaining replication fidelity goes beyond classical proofreading by purging the nascent replicating RNA strand of a range of potentially replication-terminating aberrations. Using our developed assays, we identify drug and drug-like molecules that inhibit nsp14-nsp10, including the known SARS-CoV-2 major protease (Mpro) inhibitor ebselen and the HIV integrase inhibitor raltegravir, revealing the potential for multifunctional inhibitors in COVID-19 treatment.


Assuntos
Antivirais/farmacologia , Avaliação Pré-Clínica de Medicamentos , Exorribonucleases/metabolismo , Genoma Viral/genética , Instabilidade Genômica , SARS-CoV-2/enzimologia , SARS-CoV-2/genética , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , RNA-Polimerase RNA-Dependente de Coronavírus/metabolismo , Exorribonucleases/antagonistas & inibidores , Genoma Viral/efeitos dos fármacos , Instabilidade Genômica/efeitos dos fármacos , Instabilidade Genômica/genética , Inibidores de Integrase de HIV/farmacologia , Isoindóis/farmacologia , Complexos Multienzimáticos/antagonistas & inibidores , Complexos Multienzimáticos/metabolismo , Compostos Organosselênicos/farmacologia , RNA Viral/biossíntese , RNA Viral/genética , Raltegravir Potássico/farmacologia , SARS-CoV-2/efeitos dos fármacos , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas Virais Reguladoras e Acessórias/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
2.
Genes Dev ; 25(17): 1859-70, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21896658

RESUMO

One of the major DNA interstrand cross-link (ICL) repair pathways in mammalian cells is coupled to replication, but the mechanistic roles of the critical factors involved remain largely elusive. Here, we show that purified human SNM1A (hSNM1A), which exhibits a 5'-3' exonuclease activity, can load from a single DNA nick and digest past an ICL on its substrate strand. hSNM1A-depleted cells are ICL-sensitive and accumulate replication-associated DNA double-strand breaks (DSBs), akin to ERCC1-depleted cells. These DSBs are Mus81-induced, indicating that replication fork cleavage by Mus81 results from the failure of the hSNM1A- and XPF-ERCC1-dependent ICL repair pathway. Our results reveal how collaboration between hSNM1A and XPF-ERCC1 is necessary to initiate ICL repair in replicating human cells.


Assuntos
Enzimas Reparadoras do DNA/metabolismo , Reparo do DNA/genética , Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Endonucleases/metabolismo , Proteínas Nucleares/metabolismo , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , Quebras de DNA de Cadeia Simples , Enzimas Reparadoras do DNA/genética , Proteínas de Ligação a DNA/genética , Endonucleases/genética , Exodesoxirribonucleases , Células HeLa , Humanos , Proteínas Nucleares/genética
3.
Int J Cancer ; 134(6): 1495-503, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23982883

RESUMO

As the options for systemic treatment of malignant melanoma (MM) increase, the need to develop biomarkers to identify patients who might benefit from cytotoxic chemotherapy becomes more apparent. In preclinical models, oxaliplatin has activity in cisplatin-resistant cells. In this study, we have shown that oxaliplatin forms interstrand crosslinks (ICLs) in cellular DNA and that loss of the heterodimeric structure-specific endonuclease XPF-ERCC1 causes hypersensitivity to oxaliplatin in mammalian cells. XPF deficiency resulted in late S-phase arrest and persistence of double-strand breaks following oxaliplatin treatment. In a panel of 12 MM cell lines, oxaliplatin sensitivity correlated with XPF and ERCC1 protein levels. The knockdown of ERCC1 and XPF protein levels by RNA interference increased sensitivity of cancer cells to oxaliplatin; overexpression of exogenous ERCC1 significantly decreased drug sensitivity. Following immunohistochemical optimization, XPF protein levels were quantified in MM tissue samples from 183 patients, showing variation in expression and no correlation with prognosis. In 57 patients with MM treated with cisplatin or carboplatin, XPF protein levels did not predict the likelihood of clinical response. We propose that oxaliplatin should not be discarded as a potential treatment for MM on the basis of the limited activity of cisplatin in unselected patients. Moreover, we show that XPF-ERCC1 protein levels are a key determinant of the sensitivity of melanoma cells to oxaliplatin in vitro. Immunohistochemical detection of XPF appears suitable for development as a tissue biomarker for potentially selecting patients for oxaliplatin treatment in a prospective clinical trial.


Assuntos
Antineoplásicos/farmacologia , Biomarcadores Tumorais/metabolismo , Proteínas de Ligação a DNA/metabolismo , Endonucleases/metabolismo , Melanoma/tratamento farmacológico , Compostos Organoplatínicos/farmacologia , Fatores de Transcrição/metabolismo , Estudos de Coortes , Dano ao DNA/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Humanos , Técnicas Imunoenzimáticas , Melanoma/metabolismo , Melanoma/patologia , Pessoa de Meia-Idade , Oxaliplatina , Seleção de Pacientes , Fase S/efeitos dos fármacos , Neoplasias Cutâneas , Análise Serial de Tecidos , Células Tumorais Cultivadas , Melanoma Maligno Cutâneo
4.
Nat Commun ; 15(1): 5392, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38918391

RESUMO

DNA double-strand breaks (DSBs), such as those produced by radiation and radiomimetics, are amongst the most toxic forms of cellular damage, in part because they involve extensive oxidative modifications at the break termini. Prior to completion of DSB repair, the chemically modified termini must be removed. Various DNA processing enzymes have been implicated in the processing of these dirty ends, but molecular knowledge of this process is limited. Here, we demonstrate a role for the metallo-ß-lactamase fold 5'-3' exonuclease SNM1A in this vital process. Cells disrupted for SNM1A manifest increased sensitivity to radiation and radiomimetic agents and show defects in DSB damage repair. SNM1A is recruited and is retained at the sites of DSB damage via the concerted action of its three highly conserved PBZ, PIP box and UBZ interaction domains, which mediate interactions with poly-ADP-ribose chains, PCNA and the ubiquitinated form of PCNA, respectively. SNM1A can resect DNA containing oxidative lesions induced by radiation damage at break termini. The combined results reveal a crucial role for SNM1A to digest chemically modified DNA during the repair of DSBs and imply that the catalytic domain of SNM1A is an attractive target for potentiation of radiotherapy.


Assuntos
Quebras de DNA de Cadeia Dupla , Enzimas Reparadoras do DNA , Reparo do DNA , Exodesoxirribonucleases , Humanos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Exodesoxirribonucleases/metabolismo , Exodesoxirribonucleases/genética , Enzimas Reparadoras do DNA/metabolismo , Enzimas Reparadoras do DNA/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Antígeno Nuclear de Célula em Proliferação/genética , DNA/metabolismo , DNA/genética , Ubiquitinação , Proteínas de Ciclo Celular
5.
Chem Sci ; 15(21): 8227-8241, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38817593

RESUMO

The three human SNM1 metallo-ß-lactamase fold nucleases (SNM1A-C) play key roles in DNA damage repair and in maintaining telomere integrity. Genetic studies indicate that they are attractive targets for cancer treatment and to potentiate chemo- and radiation-therapy. A high-throughput screen for SNM1A inhibitors identified diverse pharmacophores, some of which were shown by crystallography to coordinate to the di-metal ion centre at the SNM1A active site. Structure and turnover assay-guided optimization enabled the identification of potent quinazoline-hydroxamic acid containing inhibitors, which bind in a manner where the hydroxamic acid displaces the hydrolytic water and the quinazoline ring occupies a substrate nucleobase binding site. Cellular assays reveal that SNM1A inhibitors cause sensitisation to, and defects in the resolution of, cisplatin-induced DNA damage, validating the tractability of MBL fold nucleases as cancer drug targets.

6.
Chem Sci ; 14(44): 12498-12505, 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-38020377

RESUMO

Formaldehyde is a pollutant and human metabolite that is toxic at high concentrations. Biological studies on formaldehyde are hindered by its high reactivity and volatility, which make it challenging to deliver quantitatively to cells. Here, we describe the development and validation of a set of N-acyloxymethyl-phthalimides as cell-relevant formaldehyde delivery agents. These esterase-sensitive compounds were similarly or less inhibitory to human cancer cell growth than free formaldehyde but the lead compound increased intracellular formaldehyde concentrations, increased cellular levels of thymidine derivatives (implying increased formaldehyde-mediated carbon metabolism), induced formation of cellular DNA-protein cross-links and induced cell death in pancreatic cancer cells. Overall, our N-acyloxymethyl-phthalimides and control compounds provide an accessible and broadly applicable chemical toolkit for formaldehyde biological research and have potential as cancer therapeutics.

7.
Methods Mol Biol ; 2119: 79-88, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31989516

RESUMO

DNA interstrand cross-links (ICLs) are an extremely toxic form of DNA damage that cells experience upon exposure to natural metabolites. Moreover, ICLs are cytotoxic lesions produced by a range of clinically important anticancer agents. Therefore, improving our understanding of ICL induction and processing has important implications in biology and medicine. The sensitive detection of ICLs in mammalian cells is challenging but has been aided by the development of a modified form of the single-cell gel electrophoresis (SCGE) assay, also known as the "comet assay." Here we describe this method and how it can be used to sensitively monitor the induction and removal of ICLs in single mammalian cells.


Assuntos
Antineoplásicos/farmacologia , Ensaio Cometa , Dano ao DNA , Reparo do DNA/efeitos dos fármacos , DNA , Animais , Antineoplásicos/farmacocinética , Linhagem Celular , DNA/análise , DNA/metabolismo , Humanos
8.
Cancer Chemother Pharmacol ; 61(5): 739-49, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17594094

RESUMO

PURPOSE: The importance of understanding the mechanism of action of anticancer agents is sometimes overlooked in the pursuit of new and therapeutically advantageous compounds. Doxorubicin has long been identified as an inhibitor of the DNA-decatenating enzyme topoisomerase II, this being believed to be the major mechanism of action of this drug. However, the complex nature of cytotoxicity induced by doxorubicin suggests that more than one mechanism of action is responsible for cell kill. Investigation into various other cellular effects has shown that doxorubicin can, in the presence of formaldehyde, form doxorubicin-DNA adducts, resulting in enhanced cell death. METHODS: We have used six catalytic inhibitors of topoisomerase II (aclarubicin, merbarone, suramin, staurosporine, maleimide and sobuzoxane) to investigate the role of topoisomerase II mediated cell effects in doxorubicin-DNA adduct inducing treatments. Adduct levels were determined by scintillation counting of [14C]doxorubicin-DNA lesions and DNA damage responses by Comet analysis and flow cytometry (apoptosis). RESULTS: Here we show that sobuzoxane inhibits topoisomerase II but in the presence of doxorubicin also enhances the production of doxorubicin-DNA adducts resulting in an enhanced cytotoxic response. We show that the formation of doxorubicin-DNA adducts is mediated by formaldehyde released from sobuzoxane when it is metabolised. CONCLUSIONS: Sobuzoxane has also been shown to decrease the normally dose limiting cardiotoxicity commonly exhibited with clinical use of doxorubicin. The potential combination of doxorubicin and sobuzoxane in cancer chemotherapy has two advantages. First, the mechanism of doxorubicin toxicity is shifted away from topoisomerase II inhibition and towards drug-DNA adduct formation which may allow for a lower drug dose to be used and circumvent some drug resistance problems. Second, the addition of a cardioprotecting agent will counteract the commonly dose limiting side effect of cardiac damage resulting from doxorubicin treatment. The importance of the potentiation of cell kill of doxorubicin and sobuzoxane provides a rationalisation of a mechanistic-based combination of anticancer drugs for an improved clinical outcome.


Assuntos
Antineoplásicos/farmacologia , Adutos de DNA/farmacologia , Doxorrubicina/farmacologia , Piperazinas/farmacologia , Inibidores da Topoisomerase II , Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Ensaio Cometa , Dano ao DNA/efeitos dos fármacos , DNA Topoisomerases Tipo II/metabolismo , Sinergismo Farmacológico , Citometria de Fluxo , Formaldeído/metabolismo , Células HL-60 , Humanos , Piperazinas/farmacocinética , Contagem de Cintilação
9.
Cancer Chemother Pharmacol ; 62(3): 471-82, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18030472

RESUMO

Histone deacetylase inhibitory prodrugs that are metabolized to carboxylic acid(s) and aldehyde(s) possess antineoplastic properties. Formaldehyde-releasing prodrugs were shown to be the most potent. The objective of this study was to gain understanding on the mode of action of these prodrugs in cancer cells. HL-60 and MCF-7 cells in the presence of N-acetylcysteine or glutathione were protected from death induced by formaldehyde-releasing prodrugs but not from death caused by the homologous acetaldehyde-releasing ones. Cell death induced by the former was accompanied by depletion of intracellular glutathione and increased reactive oxygen species that were attenuated by N-acetylcysteine. At fourfold higher concentration, acetaldehyde-releasing prodrugs increased reactive oxygen species that were further augmented by N-acetylcysteine. In HL-60 cells, formaldehyde-releasing prodrugs dissipated the mitochondrial membrane potential and glutathione or N-acetylcysteine restored it. Although acetaldehyde-releasing prodrugs dissipated mitochondrial membrane potential, it occurred at 20-fold greater concentration and was unaffected by the antioxidants. Formaldehyde-releasing prodrugs abrogated c-myc protein expression and elevated c-Jun and H2AX phosphorylation, N-acetylcysteine partially reversed these changes. Herein, we show that formaldehyde-releasing prodrugs diminish the level of glutathione most likely by forming S-formylglutathione adducts resulting in increase of reactive oxygen species followed by signaling events that lead to cancer cells death.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Formaldeído/farmacologia , Glutationa/metabolismo , Pró-Fármacos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Acetaldeído/química , Acetaldeído/farmacologia , Acetilcisteína/farmacologia , Antineoplásicos/química , Western Blotting , Linhagem Celular Tumoral , Formaldeído/química , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Estrutura Molecular , Pró-Fármacos/química , Relação Estrutura-Atividade
10.
Cancer Res ; 66(9): 4863-71, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16651442

RESUMO

Doxorubicin (Adriamycin) is one of the most commonly used chemotherapeutic drugs and exhibits a wide spectrum of activity against solid tumors, lymphomas, and leukemias. Doxorubicin is classified as a topoisomerase II poison, although other mechanisms of action have been characterized. Here, we show that doxorubicin-DNA adducts (formed by the coadministration of doxorubicin with non-toxic doses of formaldehyde-releasing prodrugs) induce a more cytotoxic response in HL-60 cells than doxorubicin as a single agent. Doxorubicin-DNA adducts seem to be independent of classic topoisomerase II-mediated cellular responses (as observed by employing topoisomerase II catalytic inhibitors and HL-60/MX2 cells). Apoptosis induced by doxorubicin-DNA adducts initiates a caspase cascade that can be blocked by overexpressed Bcl-2, suggesting that adducts induce a classic mode of apoptosis. A reduction in the level of topoisomerase II-mediated double-strand-breaks was also observed with increasing levels of doxorubicin-DNA adducts and increased levels of apoptosis, further confirming that adducts exhibit a separate mechanism of action compared with the classic topoisomerase II poison mode of cell death by doxorubicin alone. Collectively, these results indicate that the presence of formaldehyde transfers doxorubicin from topoisomerase II-mediated cellular damage to the formation of doxorubicin-DNA adducts, and that these adducts are more cytotoxic than topoisomerase II-mediated lesions. These results also show that doxorubicin can induce apoptosis by a non-topoisomerase II-dependent mechanism, and this provides exciting new prospects for enhancing the clinical use of this agent and for the development of new derivatives and new tumor-targeted therapies.


Assuntos
Adutos de DNA/biossíntese , Dano ao DNA/fisiologia , Doxorrubicina/farmacologia , Inibidores da Topoisomerase II , Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Ciclo Celular , Processos de Crescimento Celular , DNA Topoisomerases Tipo II/metabolismo , DNA de Neoplasias/efeitos dos fármacos , DNA de Neoplasias/metabolismo , Doxorrubicina/biossíntese , Células HL-60 , Humanos
11.
Mol Cancer Ther ; 6(4): 1450-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17431124

RESUMO

The anthracycline group of compounds is extensively used in current cancer chemotherapy regimens and is classified as topoisomerase II inhibitor. However, previous work has shown that doxorubicin can be activated to form DNA adducts in the presence of formaldehyde-releasing prodrugs and that this leads to apoptosis independently of topoisomerase II-mediated damage. To determine which anthracyclines would be useful in combination with formaldehyde-releasing prodrugs, a series of clinically relevant anthracyclines (doxorubicin, daunorubicin, idarubicin, and epirubicin) were examined for their capacity to form DNA adducts in MCF7 and MCF7/Dx (P-glycoprotein overexpressing) cells in the presence of the formaldehyde-releasing drug pivaloyloxymethyl butyrate (AN-9). All anthracyclines, with the exception of epirubicin, efficiently yielded adducts in both sensitive and resistant cell lines, and levels of adducts were similar in mitochondrial and nuclear genomes. Idarubicin was the most active compound in both sensitive and resistant cell lines, whereas adducts formed by doxorubicin and daunorubicin were consistently lower in the resistant compared with sensitive cells. The adducts formed by doxorubicin, daunorubicin, and idarubicin showed the same DNA sequence specificity in sensitive and resistant cells as assessed by lambda-exonuclease-based sequencing of alpha-satellite DNA extracted from drug-treated cells. Growth inhibition assays were used to show that doxorubicin, daunorubicin, and idarubicin were all synergistic in combination with AN-9, whereas the combination of epirubicin with AN-9 was additive. Although apoptosis assays indicated a greater than additive effect for epirubicin/AN-9 combinations, this effect was much more pronounced for doxorubicin/AN-9 combinations.


Assuntos
Antraciclinas/metabolismo , Butiratos/farmacologia , Formaldeído/metabolismo , Pró-Fármacos/farmacologia , Antraciclinas/química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Pareamento de Bases/efeitos dos fármacos , Sequência de Bases , Southern Blotting , Linhagem Celular Tumoral , Reagentes de Ligações Cruzadas/farmacologia , Adutos de DNA/efeitos dos fármacos , Interações Medicamentosas , Humanos
12.
Mol Cancer Ther ; 2(2): 189-98, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12589036

RESUMO

Previous studies have shown that Adriamycin can react with formaldehyde to yield an activated form of Adriamycin that can further react with DNA to yield Adriamycin-DNA adducts. Because hexamethylenetetramine (HMTA) is known to hydrolyze under cellular conditions and release six molecules of formaldehyde in a pH-dependent manner, we examined this clinical agent for its potential as a formaldehyde-releasing prodrug for the activation of Adriamycin. In IMR-32 neuroblastoma cells in culture, increasing levels of HMTA resulted in enhanced levels of Adriamycin-DNA adducts. These adducts were formed in a pH-dependent manner, with 4-fold more detected at pH 6.5 compared with pH 7.4, consistent with the known acid lability of HMTA. The resulting drug-DNA lesion was shown to be cytotoxic, with combined Adriamycin and prodrug treatment resulting in a 3-fold lower IC(50) value compared with that of Adriamycin alone. Given the acidic nature of solid tumors and the preferential release of formaldehyde from HMTA in acidic environments, HMTA therefore has some potential for localized activation of Adriamycin in solid tumors.


Assuntos
Antineoplásicos/farmacocinética , Doxorrubicina/farmacocinética , Metenamina/farmacologia , Neuroblastoma/tratamento farmacológico , Pró-Fármacos/farmacologia , Biotransformação , Adutos de DNA/biossíntese , DNA de Neoplasias/metabolismo , Formaldeído/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Neuroblastoma/genética , Neuroblastoma/metabolismo , Células Tumorais Cultivadas
13.
Mol Cancer Ther ; 2(7): 661-70, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12883039

RESUMO

The anticancer anthracycline compound Adriamycin is a known topoisomerase II inhibitor but is also capable of exerting other cellular consequences. After intercalation, Adriamycin can form covalent adducts with DNA, and the magnitude of these adducts appears to be limited by the cellular availability of formaldehyde. Adducts produced by Adriamycin in the presence of formaldehyde have been well characterized in cell-free systems but not in cells. In this study, we show that when Adriamycin is used in conjunction with the formaldehyde-releasing prodrug AN-9 in IMR-32 tumor cells, this allows the formation of sufficiently high levels of adducts in genomic DNA to enable detection of their DNA sequence specificity for the first time. The 340-bp alpha-satellite EcoRI repeat sequence was isolated from drug-treated cells and digested with lambda-exonuclease to determine adduct sites at which exonuclease digestion was blocked. The Adriamycin adducts were formed predominantly at 5'-GC and GG sequences and unstable with respect to elevated temperatures and extended times at 37 degrees C. The use of three anthracycline derivatives lacking a 3'amino group demonstrated that this amino portion is critical for the formation of anthracycline adducts in cells. The structure of these drug-DNA adducts can therefore be considered to be identical to the Adriamycin adducts, which have been characterized rigorously in cell-free systems by X-ray crystallography, two-dimensional nuclear magnetic resonance, and mass spectrometry.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Adutos de DNA/metabolismo , DNA/química , Doxorrubicina/farmacologia , Neoplasias/genética , Sequência de Bases , Butiratos/farmacologia , DNA/efeitos dos fármacos , DNA/metabolismo , Adutos de DNA/química , Formaldeído/química , Formaldeído/metabolismo , Humanos , Dados de Sequência Molecular , Neoplasias/metabolismo , Células Tumorais Cultivadas
14.
Cancer Chemother Pharmacol ; 71(3): 809-16, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23263186

RESUMO

Epirubicin was developed as a semi-synthetic anthracycline derivative to circumvent the cardiotoxic limitations associated with the use of doxorubicin in the clinic. Anthracycline compounds have been demonstrated to form covalent drug-DNA adducts utilising endogenous and exogenous sources of formaldehyde; however, previous investigations of the formation of epirubicin-DNA adducts provide conflicting evidence for adduct formation. This work provides evidence that epirubicin acts to form drug-DNA adducts at physiologically relevant concentrations and demonstrates that the rate of formation of epirubicin-DNA adducts is slower than that observed for other anthracycline compounds, explaining why they are only detectable under defined experimental conditions. Formation of covalent epirubicin-DNA adducts improves the apoptotic profile of epirubicin and provides opportunities to overcome drug resistance and cardiotoxic limitations.


Assuntos
Antraciclinas/metabolismo , Antibióticos Antineoplásicos/farmacologia , Citotoxinas/metabolismo , Adutos de DNA/efeitos dos fármacos , Epirubicina/análogos & derivados , Epirubicina/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Carboidratos/química , Linhagem Celular Tumoral , Corantes , Reagentes de Ligações Cruzadas , Feminino , Formaldeído/química , Humanos , Hidroxilação , Propídio , Rodaminas , Transcrição Gênica/efeitos dos fármacos
15.
Biochem Pharmacol ; 83(12): 1602-12, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22414726

RESUMO

The cytotoxicity of doxorubicin, a clinically used anti-neoplastic drug, can be enhanced by formaldehyde (either endogenous or exogenous) to promote the formation of doxorubicin-DNA adducts. Formaldehyde supplies the carbon required for the covalent linkage of doxorubicin to one strand of DNA, with hydrogen bonds stabilising the doxorubicin mono-adduct to the other strand of DNA, to act much like an interstrand crosslink. Interstrand crosslinks present a major challenge for cellular repair processes, requiring the activation of numerous DNA damage response proteins for resolution of the resulting DNA intermediates and damage. This work investigates DNA damage response proteins activated by doxorubicin-DNA adducts. Although p53 was phosphorylated at Serine 15 in response to adducts, long term growth inhibition of mammalian cells was not affected by p53 status. Using siRNA technology and kinase inhibitors we observed enhanced cellular sensitivity to doxorubicin-DNA adducts when the activity of the signalling protein kinases ATM and ATR were lost. Cells synchronised using a double thymidine block were sensitised to adduct-initiated cell death upon ATR knockdown, but relatively unaffected by ATM knockdown. Loss of ATR was associated with abrogation of a drug-induced G(2)/M block and induction of mitotic catastrophe, while loss of ATM was associated with drug-induced apoptosis in non-synchronised cells. These proteins may therefore be potential drug targets to achieve synergistic cytotoxic responses to doxorubicin-DNA adduct forming therapies. The analysis of these protein kinases with respect to cell cycle progression indicates that ATR is required for G(2)/M checkpoint responses while ATM appears to function in G(1) mediated responses to anthracycline adducts.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Adutos de DNA/metabolismo , Dano ao DNA , DNA/efeitos dos fármacos , Doxorrubicina/toxicidade , Proteínas Mutadas de Ataxia Telangiectasia , Western Blotting , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/metabolismo , Imunofluorescência , Humanos , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA