Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
NAR Cancer ; 5(2): zcad015, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36992846

RESUMO

DNA-methylating environmental carcinogens such as N-nitrosodimethylamine (NDMA) and certain alkylators used in chemotherapy form O 6-methylguanine (m6G) as a functionally critical intermediate. NDMA is a multi-organ carcinogen found in contaminated water, polluted air, preserved foods, tobacco products, and many pharmaceuticals. Only ten weeks after exposure to NDMA, neonatally-treated mice experienced elevated mutation frequencies in liver, lung and kidney of ∼35-fold, 4-fold and 2-fold, respectively. High-resolution mutational spectra (HRMS) of liver and lung revealed distinctive patterns dominated by GC→AT mutations in 5'-Pu-G-3' contexts, very similar to human COSMIC mutational signature SBS11. Commonly associated with alkylation damage, SBS11 appears in cancers treated with the DNA alkylator temozolomide (TMZ). When cells derived from the mice were treated with TMZ, N-methyl-N-nitrosourea, and streptozotocin (two other therapeutic methylating agents), all displayed NDMA-like HRMS, indicating mechanistically convergent mutational processes. The role of m6G in shaping the mutational spectrum of NDMA was probed by removing MGMT, the main cellular defense against m6G. MGMT-deficient mice displayed a strikingly enhanced mutant frequency, but identical HRMS, indicating that the mutational properties of these alkylators is likely owed to sequence-specific DNA binding. In sum, the HRMS of m6G-forming agents constitute an early-onset biomarker of exposure to DNA methylating carcinogens and drugs.

2.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35197283

RESUMO

Alkylating agents damage DNA and proteins and are widely used in cancer chemotherapy. While cellular responses to alkylation-induced DNA damage have been explored, knowledge of how alkylation affects global cellular stress responses is sparse. Here, we examined the effects of the alkylating agent methylmethane sulfonate (MMS) on gene expression in mouse liver, using mice deficient in alkyladenine DNA glycosylase (Aag), the enzyme that initiates the repair of alkylated DNA bases. MMS induced a robust transcriptional response in wild-type liver that included markers of the endoplasmic reticulum (ER) stress/unfolded protein response (UPR) known to be controlled by XBP1, a key UPR effector. Importantly, this response is significantly reduced in the Aag knockout. To investigate how AAG affects alkylation-induced UPR, the expression of UPR markers after MMS treatment was interrogated in human glioblastoma cells expressing different AAG levels. Alkylation induced the UPR in cells expressing AAG; conversely, AAG knockdown compromised UPR induction and led to a defect in XBP1 activation. To verify the requirements for the DNA repair activity of AAG in this response, AAG knockdown cells were complemented with wild-type Aag or with an Aag variant producing a glycosylase-deficient AAG protein. As expected, the glycosylase-defective Aag does not fully protect AAG knockdown cells against MMS-induced cytotoxicity. Remarkably, however, alkylation-induced XBP1 activation is fully complemented by the catalytically inactive AAG enzyme. This work establishes that, besides its enzymatic activity, AAG has noncanonical functions in alkylation-induced UPR that contribute to cellular responses to alkylation.


Assuntos
DNA Glicosilases/metabolismo , Reparo do DNA , Desdobramento de Proteína , Alquilação , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Estresse do Retículo Endoplasmático , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Camundongos , Proteína 1 de Ligação a X-Box/metabolismo
3.
DNA Repair (Amst) ; 106: 103176, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34365116

RESUMO

DNA damage can be cytotoxic and mutagenic, and it is directly linked to aging, cancer, and other diseases. To counteract the deleterious effects of DNA damage, cells have evolved highly conserved DNA repair pathways. Many commonly used DNA repair assays are relatively low throughput and are limited to analysis of one protein or one pathway. Here, we have explored the capacity of the CometChip platform for parallel analysis of multiple DNA repair activities. Taking advantage of the versatility of the traditional comet assay and leveraging micropatterning techniques, the CometChip platform offers increased throughput and sensitivity compared to the traditional comet assay. By exposing cells to DNA damaging agents that create substrates of Base Excision Repair, Nucleotide Excision Repair, and Non-Homologous End Joining, we show that the CometChip is an effective method for assessing repair deficiencies in all three pathways. With these applications of the CometChip platform, we expand the utility of the comet assay for precise, high-throughput, parallel analysis of multiple DNA repair activities.


Assuntos
Ensaio Cometa/métodos , Dano ao DNA , Reparo do DNA , Ensaios de Triagem em Larga Escala/métodos , Linhagem Celular , Linhagem Celular Tumoral , DNA/efeitos dos fármacos , DNA/metabolismo , DNA/efeitos da radiação , Reparo do DNA por Junção de Extremidades , Humanos , Mutagênicos/toxicidade
4.
Free Radic Biol Med ; 174: 89-99, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34324980

RESUMO

Although DNA repair is known to impact susceptibility to cancer and other diseases, relatively few population studies have been performed to evaluate DNA repair kinetics in people due to the difficulty of assessing DNA repair in a high-throughput manner. Here we use the CometChip, a high-throughput comet assay, to explore inter-individual variation in repair of oxidative damage to DNA, a known risk factor for aging, cancer and other diseases. DNA repair capacity after H2O2-induced DNA oxidation damage was quantified in peripheral blood mononuclear cells (PBMCs). For 10 individuals, blood was drawn at several times over the course of 4-6 weeks. In addition, blood was drawn once from each of 56 individuals. DNA damage levels were quantified prior to exposure to H2O2 and at 0, 15, 30, 60, and 120-min post exposure. We found that there is significant variability in DNA repair efficiency among individuals. When subdivided into quartiles by DNA repair efficiency, we found that the average t1/2 is 81 min for the slowest group and 24 min for the fastest group. This work shows that the CometChip can be used to uncover significant differences in repair kinetics among people, pointing to its utility in future epidemiological and clinical studies.


Assuntos
Peróxido de Hidrogênio , Leucócitos Mononucleares , Ensaio Cometa , Dano ao DNA , Reparo do DNA , Humanos , Individualidade , Cinética , Linfócitos , Estresse Oxidativo/genética
5.
Cell Rep ; 34(11): 108864, 2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-33730582

RESUMO

N-Nitrosodimethylamine (NDMA) is a DNA-methylating agent that has been discovered to contaminate water, food, and drugs. The alkyladenine DNA glycosylase (AAG) removes methylated bases to initiate the base excision repair (BER) pathway. To understand how gene-environment interactions impact disease susceptibility, we study Aag-knockout (Aag-/-) and Aag-overexpressing mice that harbor increased levels of either replication-blocking lesions (3-methyladenine [3MeA]) or strand breaks (BER intermediates), respectively. Remarkably, the disease outcome switches from cancer to lethality simply by changing AAG levels. To understand the underlying basis for this observation, we integrate a suite of molecular, cellular, and physiological analyses. We find that unrepaired 3MeA is somewhat toxic, but highly mutagenic (promoting cancer), whereas excess strand breaks are poorly mutagenic and highly toxic (suppressing cancer and promoting lethality). We demonstrate that the levels of a single DNA repair protein tip the balance between blocks and breaks and thus dictate the disease consequences of DNA damage.


Assuntos
Replicação do DNA/genética , Mutagênese/genética , Neoplasias/genética , Neoplasias/patologia , Animais , Biomarcadores Tumorais/metabolismo , Morte Celular , Instabilidade Cromossômica/genética , Dano ao DNA/genética , DNA Glicosilases/deficiência , DNA Glicosilases/metabolismo , Reparo do DNA/genética , Dietilnitrosamina , Suscetibilidade a Doenças , Histonas/metabolismo , Recombinação Homóloga/genética , Fígado/patologia , Neoplasias Hepáticas/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Micronúcleos com Defeito Cromossômico , Nitrosaminas , Fenótipo , Fosfoproteínas/metabolismo , Fosforilação
6.
Nucleic Acids Res ; 48(3): e13, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-31822921

RESUMO

Genotoxicity testing is critical for predicting adverse effects of pharmaceutical, industrial, and environmental chemicals. The alkaline comet assay is an established method for detecting DNA strand breaks, however, the assay does not detect potentially carcinogenic bulky adducts that can arise when metabolic enzymes convert pro-carcinogens into a highly DNA reactive products. To overcome this, we use DNA synthesis inhibitors (hydroxyurea and 1-ß-d-arabinofuranosyl cytosine) to trap single strand breaks that are formed during nucleotide excision repair, which primarily removes bulky lesions. In this way, comet-undetectable bulky lesions are converted into comet-detectable single strand breaks. Moreover, we use HepaRG™ cells to recapitulate in vivo metabolic capacity, and leverage the CometChip platform (a higher throughput more sensitive comet assay) to create the 'HepaCometChip', enabling the detection of bulky genotoxic lesions that are missed by current genotoxicity screens. The HepaCometChip thus provides a broadly effective approach for detection of bulky DNA adducts.


Assuntos
Ensaio Cometa/métodos , Adutos de DNA/análise , Carcinogênese , Linhagem Celular , Quebras de DNA de Cadeia Simples , Reparo do DNA , Humanos , Análise em Microsséries/métodos , Sensibilidade e Especificidade
7.
Nat Commun ; 10(1): 5460, 2019 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-31784530

RESUMO

Base excision repair (BER) initiated by alkyladenine DNA glycosylase (AAG) is essential for removal of aberrantly methylated DNA bases. Genome instability and accumulation of aberrant bases accompany multiple diseases, including cancer and neurological disorders. While BER is well studied on naked DNA, it remains unclear how BER efficiently operates on chromatin. Here, we show that AAG binds to chromatin and forms complex with RNA polymerase (pol) II. This occurs through direct interaction with Elongator and results in transcriptional co-regulation. Importantly, at co-regulated genes, aberrantly methylated bases accumulate towards the 3'end in regions enriched for BER enzymes AAG and APE1, Elongator and active RNA pol II. Active transcription and functional Elongator are further crucial to ensure efficient BER, by promoting AAG and APE1 chromatin recruitment. Our findings provide insights into genome stability maintenance in actively transcribing chromatin and reveal roles of aberrantly methylated bases in regulation of gene expression.


Assuntos
Cromatina/metabolismo , DNA Glicosilases/metabolismo , Reparo do DNA/genética , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Regulação da Expressão Gênica/genética , RNA Polimerase II/metabolismo , Cromatina/genética , Metilação de DNA , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , Expressão Gênica , Instabilidade Genômica , Células HEK293 , Humanos , RNA Polimerase II/genética , Elongação da Transcrição Genética , Fatores de Elongação da Transcrição/genética , Fatores de Elongação da Transcrição/metabolismo
8.
Environ Health ; 18(1): 51, 2019 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-31174534

RESUMO

BACKGROUND: Growing evidence indicates that in utero arsenic exposures in humans may increase the risk of adverse health effects and development of diseases later in life. This study aimed to evaluate potential health risks of in utero arsenic exposure on genetic damage in newborns in relation to maternal arsenic exposure. METHODS: A total of 205 pregnant women residing in arsenic-contaminated areas in Hanam province, Vietnam, were recruited. Prenatal arsenic exposure was determined by arsenic concentration in mother's toenails and urine during pregnancy and in umbilical cord blood collected at delivery. Genetic damage in newborns was assessed by various biomarkers of early genetic effects including oxidative/nitrative DNA damage (8-hydroxydeoxyguanosine, 8-OHdG, and 8-nitroguanine), DNA strand breaks and micronuclei (MN) in cord blood. RESULTS: Maternal arsenic exposure, measured by arsenic levels in toenails and urine, was significantly increased (p <  0.05) in subjects residing in areas with high levels of arsenic contamination in drinking water. Cord blood arsenic level was significantly increased in accordance with maternal arsenic exposure (p <  0.001). Arsenic exposure in utero is associated with genotoxic effects in newborns indicated as increased levels of 8-OHdG, 8-nitroguanine, DNA strand breaks and MN frequency in cord blood with increasing levels of maternal arsenic exposure. Maternal toenail arsenic level was significantly associated with all biomarkers of early genetic effects, while cord blood arsenic levels associated with DNA strand breaks and MN frequency. CONCLUSIONS: In utero arsenic exposure is associated with various types of genetic damage in newborns potentially contributing to the development of diseases, including cancer, later in life.


Assuntos
Arsênio/toxicidade , Dano ao DNA/efeitos dos fármacos , Sangue Fetal/química , Exposição Materna/efeitos adversos , Micronúcleos com Defeito Cromossômico/efeitos dos fármacos , Adulto , Biomarcadores/sangue , Feminino , Humanos , Recém-Nascido , Unhas/química , Gravidez , Vietnã , Adulto Jovem
9.
Cell Rep ; 26(6): 1668-1678.e4, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30726746

RESUMO

Cell survival is a critical and ubiquitous endpoint in biology. The broadly accepted colony formation assay (CFA) directly measures a cell's ability to divide; however, it takes weeks to perform and is incompatible with high-throughput screening (HTS) technologies. Here, we describe the MicroColonyChip, which exploits microwell array technology to create an array of colonies. Unlike the CFA, where visible colonies are counted by eye, using fluorescence microscopy, microcolonies can be analyzed in days rather than weeks. Using automated analysis of microcolony size distributions, the MicroColonyChip achieves comparable sensitivity to the CFA (and greater sensitivity than the 2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide [XTT] assay). Compared to CellTiter-Glo, the MicroColonyChip is as sensitive and also robust to artifacts caused by differences in initial cell seeding density. We demonstrate efficacy via studies of radiosensitivity and chemosensitivity and show that the approach is amenable to multiplexing. We conclude that the MicroColonyChip is a rapid and automated alternative for cell survival quantitation.


Assuntos
Aflatoxina B1/toxicidade , Antineoplásicos Alquilantes/farmacologia , Bioensaio/instrumentação , Carmustina/farmacologia , Raios gama/efeitos adversos , Procedimentos Analíticos em Microchip , Contagem de Células , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Fibroblastos/efeitos da radiação , Células HeLa , Células Hep G2 , Humanos , Linfócitos/citologia , Linfócitos/efeitos dos fármacos , Linfócitos/fisiologia , Linfócitos/efeitos da radiação
10.
Sci Signal ; 12(568)2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30755477

RESUMO

DNA-alkylating agents are commonly used to kill cancer cells, but the base excision repair (BER) pathway they trigger can also produce toxic intermediates that cause tissue damage, such as retinal degeneration (RD). Apoptosis, a process of programmed cell death, is assumed to be the main mechanism of this alkylation-induced photoreceptor (PR) cell death in RD. Here, we studied the involvement of necroptosis (another programmed cell death process) and inflammation in alkylation-induced RD. Male mice exposed to a methylating agent exhibited a reduced number of PR cell rows, active gliosis, and cytokine induction and macrophage infiltration in the retina. Dying PRs exhibited a necrotic morphology, increased 8-hydroxyguanosine abundance (an oxidative damage marker), and overexpression of the necroptosis-associated genes Rip1 and Rip3 The activity of PARP1, which mediates BER, cell death, and inflammation, was increased in PR cells and associated with the release of proinflammatory chemokine HMGB1 from PR nuclei. Mice lacking the anti-inflammatory cytokine IL-10 exhibited more severe RD, whereas deficiency of RIP3 (also known as RIPK3) conferred partial protection. Female mice were partially protected from alkylation-induced RD, showing reduced necroptosis and inflammation compared to males. PRs in mice lacking the BER-initiating DNA glycosylase AAG did not exhibit alkylation-induced necroptosis or inflammation. Our findings show that AAG-initiated BER at alkylated DNA bases induces sex-dependent RD primarily by triggering necroptosis and activating an inflammatory response that amplifies the original damage and, furthermore, reveal new potential targets to prevent this side effect of chemotherapy.


Assuntos
DNA Glicosilases/metabolismo , Reparo do DNA , Inflamação/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Degeneração Retiniana/metabolismo , Animais , Antineoplásicos Alquilantes/efeitos adversos , Apoptose/efeitos dos fármacos , Apoptose/genética , Morte Celular/efeitos dos fármacos , Morte Celular/genética , DNA Glicosilases/genética , Feminino , Inflamação/genética , Inflamação/patologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Necrose , Poli(ADP-Ribose) Polimerase-1/genética , Poli(ADP-Ribose) Polimerase-1/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Degeneração Retiniana/induzido quimicamente , Degeneração Retiniana/genética
11.
PLoS One ; 14(2): e0208341, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30811507

RESUMO

The DNA repair protein O6-methylguanine DNA methyltransferase (MGMT) strongly influences the effectiveness of cancer treatment with chemotherapeutic alkylating agents, and MGMT status in cancer cells could potentially contribute to tailored therapies for individual patients. However, the promoter methylation and immunohistochemical assays presently used for measuring MGMT in clinical samples are indirect, cumbersome and sometimes do not accurately report MGMT activity. Here we directly compare the accuracy of 6 analytical methods, including two fluorescent reporter assays, against the in vitro MGMT activity assay that is considered the gold standard for measuring MGMT DNA repair capacity. We discuss the relative advantages of each method. Our data indicate that two recently developed fluorescence-based assays measure MGMT activity accurately and efficiently, and could provide a functional dimension to clinical efforts to identify patients who are likely to benefit from alkylating chemotherapy.


Assuntos
Bioensaio/métodos , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Corantes Fluorescentes/administração & dosagem , Proteínas Supressoras de Tumor/genética , Antineoplásicos Alquilantes/farmacologia , Linhagem Celular , Metilação de DNA/efeitos dos fármacos , Reparo do DNA/genética , Humanos , Regiões Promotoras Genéticas/genética
12.
Science ; 363(6428)2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30765538

RESUMO

Certain Escherichia coli strains residing in the human gut produce colibactin, a small-molecule genotoxin implicated in colorectal cancer pathogenesis. However, colibactin's chemical structure and the molecular mechanism underlying its genotoxic effects have remained unknown for more than a decade. Here we combine an untargeted DNA adductomics approach with chemical synthesis to identify and characterize a covalent DNA modification from human cell lines treated with colibactin-producing E. coli Our data establish that colibactin alkylates DNA with an unusual electrophilic cyclopropane. We show that this metabolite is formed in mice colonized by colibactin-producing E. coli and is likely derived from an initially formed, unstable colibactin-DNA adduct. Our findings reveal a potential biomarker for colibactin exposure and provide mechanistic insights into how a gut microbe may contribute to colorectal carcinogenesis.


Assuntos
Carcinogênese/metabolismo , Neoplasias Colorretais/microbiologia , Ciclopropanos/metabolismo , Adutos de DNA/metabolismo , Dano ao DNA , Escherichia coli/metabolismo , Microbioma Gastrointestinal , Mutagênicos/metabolismo , Peptídeos/metabolismo , Policetídeos/metabolismo , Alquilantes , Alquilação , Animais , Carcinogênese/genética , Neoplasias Colorretais/genética , Ciclopropanos/química , Escherichia coli/patogenicidade , Vida Livre de Germes , Células HT29 , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mutagênicos/toxicidade , Peptídeos/química , Peptídeos/toxicidade , Policetídeos/química , Policetídeos/toxicidade
13.
Nat Med ; 24(5): 556-562, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29736026

RESUMO

ARID1A (the AT-rich interaction domain 1A, also known as BAF250a) is one of the most commonly mutated genes in cancer1,2. The majority of ARID1A mutations are inactivating mutations and lead to loss of ARID1A expression 3 , which makes ARID1A a poor therapeutic target. Therefore, it is of clinical importance to identify molecular consequences of ARID1A deficiency that create therapeutic vulnerabilities in ARID1A-mutant tumors. In a proteomic screen, we found that ARID1A interacts with mismatch repair (MMR) protein MSH2. ARID1A recruited MSH2 to chromatin during DNA replication and promoted MMR. Conversely, ARID1A inactivation compromised MMR and increased mutagenesis. ARID1A deficiency correlated with microsatellite instability genomic signature and a predominant C>T mutation pattern and increased mutation load across multiple human cancer types. Tumors formed by an ARID1A-deficient ovarian cancer cell line in syngeneic mice displayed increased mutation load, elevated numbers of tumor-infiltrating lymphocytes, and PD-L1 expression. Notably, treatment with anti-PD-L1 antibody reduced tumor burden and prolonged survival of mice bearing ARID1A-deficient but not ARID1A-wild-type ovarian tumors. Together, these results suggest ARID1A deficiency contributes to impaired MMR and mutator phenotype in cancer, and may cooperate with immune checkpoint blockade therapy.


Assuntos
Imunoterapia , Mutação/genética , Neoplasias/genética , Neoplasias/imunologia , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Animais , Linhagem Celular Tumoral , Reparo de Erro de Pareamento de DNA , Proteínas de Ligação a DNA , Feminino , Humanos , Linfócitos do Interstício Tumoral/imunologia , Camundongos Endogâmicos C57BL , Proteína 2 Homóloga a MutS/metabolismo , Ligação Proteica
14.
Proc Natl Acad Sci U S A ; 114(48): E10379-E10388, 2017 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-29122935

RESUMO

The integrity of our DNA is challenged with at least 100,000 lesions per cell on a daily basis. Failure to repair DNA damage efficiently can lead to cancer, immunodeficiency, and neurodegenerative disease. Base excision repair (BER) recognizes and repairs minimally helix-distorting DNA base lesions induced by both endogenous and exogenous DNA damaging agents. Levels of BER-initiating DNA glycosylases can vary between individuals, suggesting that quantitating and understanding interindividual differences in DNA repair capacity (DRC) may enable us to predict and prevent disease in a personalized manner. However, population studies of BER capacity have been limited because most methods used to measure BER activity are cumbersome, time consuming and, for the most part, only allow for the analysis of one DNA glycosylase at a time. We have developed a fluorescence-based multiplex flow-cytometric host cell reactivation assay wherein the activity of several enzymes [four BER-initiating DNA glycosylases and the downstream processing apurinic/apyrimidinic endonuclease 1 (APE1)] can be tested simultaneously, at single-cell resolution, in vivo. Taking advantage of the transcriptional properties of several DNA lesions, we have engineered specific fluorescent reporter plasmids for quantitative measurements of 8-oxoguanine DNA glycosylase, alkyl-adenine DNA glycosylase, MutY DNA glycosylase, uracil DNA glycosylase, and APE1 activity. We have used these reporters to measure differences in BER capacity across a panel of cell lines collected from healthy individuals, and to generate mathematical models that predict cellular sensitivity to methylmethane sulfonate, H2O2, and 5-FU from DRC. Moreover, we demonstrate the suitability of these reporters to measure differences in DRC in multiple pathways using primary lymphocytes from two individuals.


Assuntos
Variação Biológica da População/fisiologia , Dano ao DNA/fisiologia , DNA Glicosilases/metabolismo , Reparo do DNA/fisiologia , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Linhagem Celular , DNA/genética , DNA/metabolismo , Dano ao DNA/efeitos dos fármacos , DNA Glicosilases/genética , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , Citometria de Fluxo/métodos , Fluoruracila/toxicidade , Técnicas de Silenciamento de Genes , Voluntários Saudáveis , Humanos , Peróxido de Hidrogênio/toxicidade , Metanossulfonato de Metila/toxicidade , Modelos Biológicos , Mutagênese/efeitos dos fármacos , Mutagênese/fisiologia , Mutagênicos/toxicidade , Medicina de Precisão/métodos , Cultura Primária de Células , RNA Interferente Pequeno/metabolismo , Análise de Célula Única/métodos , Linfócitos T
15.
Oncotarget ; 8(40): 68707-68720, 2017 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-28978150

RESUMO

Alkylating agents are commonly used to treat cancer. Although base excision repair (BER) is a major pathway for repairing DNA alkylation damage, under certain conditions, the initiation of BER produces toxic repair intermediates that damage healthy tissues. The initiation of BER by the alkyladenine DNA glycosylase (AAG, a.k.a. MPG) can mediate alkylation-induced cytotoxicity in specific cells in the retina and cerebellum of male mice. Cytotoxicity in both wild-type and Aag-transgenic (AagTg) mice is abrogated in the absence of Poly(ADP-ribose) polymerase-1 (PARP1). Here, we tested whether PARP inhibitors can also prevent alkylation-induced retinal and cerebellar degeneration in male and female WT and AagTg mice. Importantly, we found that WT mice display sex-dependent alkylation-induced retinal damage (but not cerebellar damage), with WT males being more sensitive than females. Accordingly, estradiol treatment protects males against alkylation-induced retinal degeneration. In AagTg male and female mice, the alkylation-induced tissue damage in both the retina and cerebellum is exacerbated and the sex difference in the retina is abolished. PARP inhibitors, much like Parp1 gene deletion, protect against alkylation-induced AAG-dependent neuronal degeneration in WT and AagTg mice, regardless of the gender, but their efficacy in preventing alkylation-induced neuronal degeneration depends on PARP inhibitor characteristics and doses. The recent surge in the use of PARP inhibitors in combination with cancer chemotherapeutic alkylating agents might represent a powerful tool for obtaining increased therapeutic efficacy while avoiding the collateral effects of alkylating agents in healthy tissues.

16.
PLoS One ; 12(9): e0184619, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28886188

RESUMO

Alkylating agents are ubiquitous in our internal and external environments, causing DNA damage that contributes to mutations and cell death that can result in aging, tissue degeneration and cancer. Repair of methylated DNA bases occurs primarily through the base excision repair (BER) pathway, a multi-enzyme pathway initiated by the alkyladenine DNA glycosylase (Aag, also known as Mpg). Previous work demonstrated that mice treated with the alkylating agent methyl methanesulfonate (MMS) undergo cerebellar degeneration in an Aag-dependent manner, whereby increased BER initiation by Aag causes increased tissue damage that is dependent on activation of poly (ADP-ribose) polymerase 1 (Parp1). Here, we dissect the molecular mechanism of cerebellar granule neuron (CGN) sensitivity to MMS using primary ex vivo neuronal cultures. We first established a high-throughput fluorescent imaging method to assess primary neuron sensitivity to treatment with DNA damaging agents. Next, we verified that the alkylation sensitivity of CGNs is an intrinsic phenotype that accurately recapitulates the in vivo dependency of alkylation-induced CGN cell death on Aag and Parp1 activity. Finally, we show that MMS-induced CGN toxicity is independent of all the cellular events that have previously been associated with Parp-mediated toxicity, including mitochondrial depolarization, AIF translocation, calcium fluxes, and NAD+ consumption. We therefore believe that further investigation is needed to adequately describe all varieties of Parp-mediated cell death.


Assuntos
Cerebelo/citologia , DNA Glicosilases/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Poli(ADP-Ribose) Polimerase-1/metabolismo , Alquilantes/farmacologia , Alquilação/efeitos dos fármacos , Animais , Morte Celular/efeitos dos fármacos , Células Cultivadas , DNA Glicosilases/genética , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/genética , Hibridização in Situ Fluorescente , Metanossulfonato de Metila/farmacologia , Camundongos , Poli(ADP-Ribose) Polimerase-1/genética
17.
Nucleic Acids Res ; 45(6): 3242-3252, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-28115629

RESUMO

Etheno (ε) DNA base adducts are highly mutagenic lesions produced endogenously via reactions with lipid peroxidation (LPO) products. Cancer-promoting conditions, such as inflammation, can induce persistent oxidative stress and increased LPO, resulting in the accumulation of ε-adducts in different tissues. Using a recently described fluorescence multiplexed host cell reactivation assay, we show that a plasmid reporter bearing a site-specific 3,N4-ethenocytosine (εC) causes transcriptional blockage. Notably, this blockage is exacerbated in Cockayne Syndrome and xeroderma pigmentosum patient-derived lymphoblastoid and fibroblast cells. Parallel RNA-Seq expression analysis of the plasmid reporter identifies novel transcriptional mutagenesis properties of εC. Our studies reveal that beyond the known pathways, such as base excision repair, the process of transcription-coupled nucleotide excision repair plays a role in the removal of εC from the genome, and thus in the protection of cells and tissues from collateral damage induced by inflammatory responses.


Assuntos
Citosina/análogos & derivados , Adutos de DNA/metabolismo , Reparo do DNA , Transcrição Gênica , Adenina/análogos & derivados , Adenina/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Síndrome de Cockayne/genética , Citosina/metabolismo , Enzimas Reparadoras do DNA/genética , Humanos , Camundongos , Camundongos Knockout , Mutagênese , RNA Polimerase II/metabolismo , Xeroderma Pigmentoso/genética
18.
Cancer Res ; 77(1): 198-206, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27793847

RESUMO

Cancer cells can resist the effects of DNA-damaging therapeutic agents via utilization of DNA repair pathways, suggesting that DNA repair capacity (DRC) measurements in cancer cells could be used to identify patients most likely to respond to treatment. However, the limitations of available technologies have so far precluded adoption of this approach in the clinic. We recently developed fluorescence-based multiplexed host cell reactivation (FM-HCR) assays to measure DRC in multiple pathways. Here we apply a mathematical model that uses DRC in multiple pathways to predict cellular resistance to killing by DNA-damaging agents. This model, developed using FM-HCR and drug sensitivity measurements in 24 human lymphoblastoid cell lines, was applied to a panel of 12 patient-derived xenograft (PDX) models of glioblastoma to predict glioblastoma response to treatment with the chemotherapeutic DNA-damaging agent temozolomide. This work showed that, in addition to changes in O6-methylguanine DNA methyltransferase (MGMT) activity, small changes in mismatch repair (MMR), nucleotide excision repair (NER), and homologous recombination (HR) capacity contributed to acquired temozolomide resistance in PDX models and led to reduced relative survival prolongation following temozolomide treatment of orthotopic mouse models in vivo Our data indicate that measuring the combined status of MMR, HR, NER, and MGMT provided a more robust prediction of temozolomide resistance than assessments of MGMT activity alone. Cancer Res; 77(1); 198-206. ©2016 AACR.


Assuntos
Neoplasias Encefálicas/genética , Reparo do DNA/fisiologia , Resistencia a Medicamentos Antineoplásicos/genética , Glioblastoma/genética , Modelos Teóricos , Animais , Antineoplásicos/farmacologia , Área Sob a Curva , Linhagem Celular Tumoral , Reparo do DNA/efeitos dos fármacos , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Humanos , Camundongos , Curva ROC , Temozolomida , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Oncotarget ; 7(29): 44950-44965, 2016 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-27391435

RESUMO

Nephrotoxicity is a common toxic side-effect of chemotherapeutic alkylating agents. Although the base excision repair (BER) pathway is essential in repairing DNA alkylation damage, under certain conditions the initiation of BER produces toxic repair intermediates that damage healthy tissues. We have shown that the alkyladenine DNA glycosylase, Aag (a.k.a. Mpg), an enzyme that initiates BER, mediates alkylation-induced whole-animal lethality and cytotoxicity in the pancreas, spleen, retina, and cerebellum, but not in the kidney. Cytotoxicity in both wild-type and Aag-transgenic mice (AagTg) was abrogated in the absence of Poly(ADP-ribose) polymerase-1 (Parp1). Here we report that Parp1-deficient mice expressing increased Aag (AagTg/Parp1-/-) develop sex-dependent kidney failure upon exposure to the alkylating agent, methyl methanesulfonate (MMS), and suffer increased whole-animal lethality compared to AagTg and wild-type mice. Macroscopic, histological, electron microscopic and immunohistochemical analyses revealed morphological kidney damage including dilated tubules, proteinaceous casts, vacuolation, collapse of the glomerular tuft, and deterioration of podocyte structure. Moreover, mice exhibited clinical signs of kidney disease indicating functional damage, including elevated blood nitrogen urea and creatinine, hypoproteinemia and proteinuria. Pharmacological Parp inhibition in AagTg mice also resulted in sensitivity to MMS-induced nephrotoxicity. These findings provide in vivo evidence that Parp1 modulates Aag-dependent MMS-induced nephrotoxicity in a sex-dependent manner and highlight the critical roles that Aag-initiated BER and Parp1 may play in determining the side-effects of chemotherapeutic alkylating agents.


Assuntos
Antineoplásicos Alquilantes/efeitos adversos , DNA Glicosilases/metabolismo , Rim/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1/metabolismo , Alquilação , Animais , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Caracteres Sexuais
20.
PLoS One ; 11(4): e0152684, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27035132

RESUMO

Common alkylating antitumor drugs, such as temozolomide, trigger their cytotoxicity by methylating the O6-position of guanosine in DNA. However, the therapeutic effect of these drugs is dampened by elevated levels of the DNA repair enzyme, O6-methylguanine DNA methyltransferase (MGMT), which directly reverses this alkylation. As a result, assessing MGMT levels in patient samples provides an important predictor of therapeutic response; however, current methods available to measure this protein are indirect, complex and slow. Here we describe the design and synthesis of fluorescent chemosensors that report directly on MGMT activity in a single step within minutes. The chemosensors incorporate a fluorophore and quencher pair, which become separated by the MGMT dealkylation reaction, yielding light-up responses of up to 55-fold, directly reflecting repair activity. Experiments show that the best-performing probe retains near-native activity at mid-nanomolar concentrations. A nuclease-protected probe, NR-1, was prepared and tested in tumor cell lysates, demonstrating an ability to evaluate relative levels of MGMT repair activity in twenty minutes. In addition, a probe was employed to evaluate inhibitors of MGMT, suggesting utility for discovering new inhibitors in a high-throughput manner. Probe designs such as that of NR-1 may prove valuable to clinicians in selection of patients for alkylating drug therapies and in assessing resistance that arises during treatment.


Assuntos
Antineoplásicos/uso terapêutico , Reparo do DNA , Genes Reporter , Neoplasias/tratamento farmacológico , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Corantes Fluorescentes , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA