Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 47(22): 11956-11962, 2019 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-31713635

RESUMO

There is increasing demand for single-stranded DNA (ssDNA) of lengths >200 nucleotides (nt) in synthetic biology, biological imaging and bionanotechnology. Existing methods to produce high-purity long ssDNA face limitations in scalability, complexity of protocol steps and/or yield. We present a rapid, high-yielding and user-friendly method for in vitro production of high-purity ssDNA with lengths up to at least seven kilobases. Polymerase chain reaction (PCR) with a forward primer bearing a methanol-responsive polymer generates a tagged amplicon that enables selective precipitation of the modified strand under denaturing conditions. We demonstrate that ssDNA is recoverable in ∼40-50 min (time after PCR) with >70% yield with respect to the input PCR amplicon, or up to 70 pmol per 100 µl PCR reaction. We demonstrate that the recovered ssDNA can be used for CRISPR/Cas9 homology directed repair in human cells, DNA-origami folding and fluorescent in-situ hybridization.


Assuntos
DNA de Cadeia Simples/síntese química , Reação em Cadeia da Polimerase/métodos , Sequência de Bases , Proteína 9 Associada à CRISPR/metabolismo , Reparo do DNA/efeitos dos fármacos , DNA de Cadeia Simples/química , Marcação de Genes/métodos , Células HEK293 , Humanos , Metanol/química , Metanol/farmacologia , Mutagênese Sítio-Dirigida/métodos , Polímeros/química , Fatores de Tempo
2.
Biomed Microdevices ; 20(3): 57, 2018 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-29974243

RESUMO

Inhibition of DNA damage response pathway in combination with DNA alkylating agents may enhance the selective killing of cancer cells leading to better therapeutic effects. MDM2 binding protein (MTBP) in human has a role in G1 phase (interphase of cell cycle) and its overexpression leads to breast and ovarian cancers. Sld7 is an uncharacterized protein in budding yeast and a potential functional homologue of MTBP. To investigate the role of Sld7 as a therapeutic target, the behavior of the wild-type cells and sld7∆ mutants were monitored in 0.5 nL microbioreactors. The brightfield microscopy images were used to analyze the change in the cell size and to determine the durations of G1 and S/G2/M phases of wild type cells and mutants. With the administration of the alkylating agent, the cell size decreased and the duration of cell cycle increased. The replacement of the medium with the fresh one enabled the cells to repair their DNA. The application of calorie restriction together with DNA alkylating agent to mutant cells resulted in smaller cell size and longer G1 phase compared to those in control environment. For therapeutic purposes, the potential of MTBP in humans or Sld7 in yeast as a drug target deserves further exploration. The fabrication simplicity, robustness and low-cost of this microfluidic bioreactor made of polystyrene allowed us to perform yeast culturing experiments and show a potential for further cell culturing studies. The device can successfully be used for therapeutic applications including the discovery of new anti-microbial, anti-inflammatory, anti-cancer drugs.


Assuntos
Ciclo Celular/efeitos dos fármacos , Dispositivos Lab-On-A-Chip , Alquilantes/farmacologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Divisão Celular , Linhagem Celular Tumoral , Meios de Cultura/química , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Marcação de Genes , Humanos , Neoplasias/terapia , Poliestirenos/química , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
3.
Environ Sci Technol ; 50(7): 3838-45, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-26919662

RESUMO

A total of five nonionic surfactants (Brij 30, Span 20, Ecosurf EH-3, polyoxyethylene sorbitol hexaoleate, and R-95 rhamnolipid) were evaluated for their ability to enhance PAH desorption and biodegradation in contaminated soil after treatment in an aerobic bioreactor. Surfactant doses corresponded to aqueous-phase concentrations below the critical micelle concentration in the soil-slurry system. The effect of surfactant amendment on soil (geno)toxicity was also evaluated for Brij 30, Span 20, and POESH using the DT40 B-lymphocyte cell line and two of its DNA-repair-deficient mutants. Compared to the results from no-surfactant controls, incubation of the bioreactor-treated soil with all surfactants increased PAH desorption, and all except R-95 substantially increased PAH biodegradation. POESH had the greatest effect, removing 50% of total measured PAHs. Brij 30, Span 20, and POESH were particularly effective at enhancing biodegradation of four- and five-ring PAHs, including five of the seven carcinogenic PAHs, with removals up to 80%. Surfactant amendment also significantly enhanced the removal of alkyl-PAHs. Most treatments significantly increased soil toxicity. Only the no-surfactant control and Brij 30 at the optimum dose significantly decreased soil genotoxicity, as evaluated with either mutant cell line. Overall, these findings have implications for the feasibility of bioremediation to achieve cleanup levels for PAHs in soil.


Assuntos
Biodegradação Ambiental , Hidrocarbonetos Policíclicos Aromáticos/metabolismo , Poluentes do Solo/metabolismo , Tensoativos/química , Animais , Linfócitos B/efeitos dos fármacos , Reatores Biológicos , Linhagem Celular , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/genética , Micelas , North Carolina , Polidocanol , Hidrocarbonetos Policíclicos Aromáticos/química , Polietilenoglicóis/química , Solo , Poluentes do Solo/química , Poluentes do Solo/toxicidade , Testes de Toxicidade/métodos , Água
4.
Arch Toxicol ; 88(1): 145-60, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23963510

RESUMO

Although carbon nanotubes (CNTs) are promising nanomaterials, their potential carcinogenicity is a major concern. We previously established a genetic method of analyzing genotoxicity of chemical compounds, where we evaluated their cytotoxic effect on the DT40 lymphoid cell line comparing DNA-repair-deficient isogenic clones with parental wild-type cells. However, application of our DT40 system for the cytotoxic and genotoxic evaluation of nanomaterials seemed to be difficult, because DT40 cells only poorly internalized nanoparticles. To solve this problem, we have constructed a chimeric gene encoding a trans-membrane receptor consisting of the 5' region of the transferrin receptor (TR) gene (to facilitate internalization of nanoparticles) and the 3' region of the macrophage receptor with collagenous structure (MARCO) gene (which is a receptor for environmental particles). We expressed the resulting MARCO-TR chimeric receptor on DNA-repair-proficient wild-type cells and mutants deficient in base excision repair (FEN1 (-/-)) and translesion DNA synthesis (REV3 (-/-)). We demonstrated that the chimera mediates uptake of particles such as fluorescence-tagged polystyrene particles and multi-walled carbon nanotubes (MWCNTs), with very poor uptake of those particles by DT40 cells not expressing the chimera. MWCNTs were cytotoxic and this effect was greater in FEN1 (-/-)and REV3 (-/-) cells than in wild-type cells. Furthermore, MWCNTs induced greater oxidative damage (measured as 8-OH-dG formation) and a larger number of mitotic chromosomal aberrations in repair-deficient cells compared to repair-proficient cells. Taken together, our novel assay system using the chimeric receptor-expressing DT40 cells provides a sensitive method to screen for genotoxicity of CNTs and possibly other nanomaterials.


Assuntos
Linfócitos B/efeitos dos fármacos , Testes de Mutagenicidade/métodos , Nanotubos de Carbono/toxicidade , Receptores Imunológicos/metabolismo , 8-Hidroxi-2'-Desoxiguanosina , Animais , Linhagem Celular/efeitos dos fármacos , Galinhas , Aberrações Cromossômicas , Reparo do DNA/efeitos dos fármacos , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Poliestirenos/farmacocinética , Receptores Imunológicos/genética , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
5.
J Exp Clin Cancer Res ; 43(1): 159, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38840237

RESUMO

BACKGROUND: Renal cell carcinoma (RCC) was historically considered to be less responsive to radiation therapy (RT) compared to other cancer indications. However, advancements in precision high-dose radiation delivery through single-fraction and multi-fraction stereotactic ablative radiotherapy (SABR) have led to better outcomes and reduced treatment-related toxicities, sparking renewed interest in using RT to treat RCC. Moreover, numerous studies have revealed that certain therapeutic agents including chemotherapies can increase the sensitivity of tumors to RT, leading to a growing interest in combining these treatments. Here, we developed a rational combination of two radiosensitizers in a tumor-targeted liposomal formulation for augmenting RT in RCC. The objective of this study is to assess the efficacy of a tumor-targeted liposomal formulation combining the mTOR inhibitor everolimus (E) with the survivin inhibitor YM155 (Y) in enhancing the sensitivity of RCC tumors to radiation. EXPERIMENTAL DESIGN: We slightly modified our previously published tumor-targeted liposomal formulation to develop a rational combination of E and Y in a single liposomal formulation (EY-L) and assessed its efficacy in RCC cell lines in vitro and in RCC tumors in vivo. We further investigated how well EY-L sensitizes RCC cell lines and tumors toward radiation and explored the underlying mechanism of radiosensitization. RESULTS: EY-L outperformed the corresponding single drug-loaded formulations E-L and Y-L in terms of containing primary tumor growth and improving survival in an immunocompetent syngeneic mouse model of RCC. EY-L also exhibited significantly higher sensitization of RCC cells towards radiation in vitro than E-L and Y-L. Additionally, EY-L sensitized RCC tumors towards radiation therapy in xenograft and murine RCC models. EY-L mediated induction of mitotic catastrophe via downregulation of multiple cell cycle checkpoints and DNA damage repair pathways could be responsible for the augmentation of radiation therapy. CONCLUSION: Taken together, our study demonstrated the efficacy of a strategic combination therapy in sensitizing RCC to radiation therapy via inhibition of DNA damage repair and a substantial increase in mitotic catastrophe. This combination therapy may find its use in the augmentation of radiation therapy during the treatment of RCC patients.


Assuntos
Carcinoma de Células Renais , Reparo do DNA , Neoplasias Renais , Survivina , Serina-Treonina Quinases TOR , Ensaios Antitumorais Modelo de Xenoenxerto , Carcinoma de Células Renais/patologia , Carcinoma de Células Renais/radioterapia , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/metabolismo , Animais , Survivina/metabolismo , Humanos , Camundongos , Linhagem Celular Tumoral , Neoplasias Renais/patologia , Neoplasias Renais/radioterapia , Neoplasias Renais/tratamento farmacológico , Reparo do DNA/efeitos dos fármacos , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Mitose/efeitos dos fármacos , Mitose/efeitos da radiação , Imidazóis/farmacologia , Dano ao DNA , Everolimo/farmacologia , Naftoquinonas/farmacologia , Naftoquinonas/uso terapêutico , Radiossensibilizantes/farmacologia , Radiossensibilizantes/uso terapêutico , Lipossomos/farmacologia , Inibidores de MTOR/farmacologia , Inibidores de MTOR/uso terapêutico
6.
Toxicol Appl Pharmacol ; 273(3): 569-79, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24128852

RESUMO

The present study aimed to assess arsenic exposure and its effect on oxidative DNA damage and repair in young children exposed in utero and continued to live in arsenic-contaminated areas. To address the need for biological specimens that can be acquired with minimal discomfort to children, we used non-invasive urinary and salivary-based assays for assessing arsenic exposure and early biological effects that have potentially serious health implications. Levels of arsenic in nails showed the greatest magnitude of difference between exposed and control groups, followed by arsenic concentrations in saliva and urine. Arsenic levels in saliva showed significant positive correlations with other biomarkers of arsenic exposure, including arsenic accumulation in nails (r=0.56, P<0.001) and arsenic concentration in urine (r=0.50, P<0.05). Exposed children had a significant reduction in arsenic methylation capacity indicated by decreased primary methylation index and secondary methylation index in both urine and saliva samples. Levels of salivary 8-OHdG in exposed children were significantly higher (~4-fold, P<0.01), whereas levels of urinary 8-OHdG excretion and salivary hOGG1 expression were significantly lower in exposed children (~3-fold, P<0.05), suggesting a defect in hOGG1 that resulted in ineffective cleavage of 8-OHdG. Multiple regression analysis results showed that levels of inorganic arsenic (iAs) in saliva and urine had a significant positive association with salivary 8-OHdG and a significant negative association with salivary hOGG1 expression.


Assuntos
Arsênio/toxicidade , Arsênio/urina , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal , 8-Hidroxi-2'-Desoxiguanosina , Biomarcadores/urina , Criança , Pré-Escolar , DNA Glicosilases/genética , DNA Glicosilases/metabolismo , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Desoxiguanosina/urina , Água Potável/química , Exposição Ambiental , Feminino , Humanos , Masculino , Unhas/química , Gravidez , Saliva/química , Inquéritos e Questionários
7.
J Periodontal Res ; 48(3): 322-30, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23030661

RESUMO

BACKGROUND AND OBJECTIVE: Hydrogen sulfide ( H2S ) is a volatile sulfur compound responsible for physiological halitosis. H2S was also reported as having periodontal pathologic activities. Gingival crevicular epithelium is the first barrier against periodontal pathogens and their products; oral keratinocyte stem cells OKSCs play key roles in maintaining this barrier. The p53 pathway is responsible for regulating key biological events. Increased apoptosis and cell-cycle arrest of DNA repair can affect keratinocyte stem cells, having a direct impact on the architecture of the oral epithelial tissue. However, the link between H2S , p53 activity and OKSCs has not yet been fully explored. The main objective of the present study was to explore the implications of the p53 pathway in OKSCs following exposure to H2S. MATERIAL AND METHODS: OKSCs were isolated from human gingival epithelium and incubated with physiological levels of H2S for 24 and 48 h. Apoptosis and the mitochondrial membrane potential were detected using flow cytometry. Cytochrome c, total p53, phosphorylated p53 and caspase activity were assessed using specific ELISAs. p53 Pathway gene activity was assayed using quantitative RT-PCR. RESULTS: The levels of apoptosis were significantly increased following incubation in the presence of H2S, especially after 48 h (36.95 ± 1.91% vs. 4.77 ± 0.74%). Caspases 9 and 3 were activated, whereas caspase-8 activity remained low. Total p53 activity and particularly phosphorylated p53 at serine 46, were significantly enhanced compared with controls (47.11 ± 9.84 units/mL vs. 1.5 ± 0 units/mL and 32.22 ± 10.23 units/mL vs. 0.15 ± 0 units/mL, respectively, at 48 h). Among p53 pathway genes, apoptosis-related genes [i.e. phosphatase and tensin homolog ( PTEN ), B-cell CLL/lymphoma 2 ( BCL2), sirtuin 3 ( SIRT3) and caspases]) were dramatically increased when compared with controls. Moreover, cell-cycle progression genes [i.e. E2F transcription factor (E2F) family and histone deacetylase ( HDAC )] and DNA-repair genes [i.e. growth arrest and DNA-damage-inducible, gamma ( GADD45G ) family and serine/threonine-protein kinase Chk2 ( CHEK2)] were also increased. CONCLUSION: Following incubation with H2 S , OKSCs express multiple p53-associated genes, including programmed cell death, cell-cycle control and DNA-repair genes.


Assuntos
Apoptose/efeitos dos fármacos , Caspases/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Gengiva/efeitos dos fármacos , Sulfeto de Hidrogênio/toxicidade , Células-Tronco/efeitos dos fármacos , Proteína Supressora de Tumor p53/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Caspases/metabolismo , Pontos de Checagem do Ciclo Celular , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/genética , Gengiva/citologia , Gengiva/metabolismo , Humanos , Queratinócitos/citologia , Queratinócitos/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Reação em Cadeia da Polimerase em Tempo Real , Proteína Supressora de Tumor p53/genética
8.
Mol Biol Rep ; 39(7): 7487-96, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22327778

RESUMO

Methacrylate monomers used in dentistry have been shown to induce DNA double strand breaks (DSBs), one of the most serious DNA damage. In the present work we show that a model dental adhesive consisting of 45% 2-hydroxyethyl methacrylate (HEMA) and 55% bisphenol A-diglycidyl dimethacrylate (Bis-GMA) at concentrations up to 0.25 mM Bis-GMA induced oxidative DNA in cultured primary human gingival fibroblasts (HGFs) as evaluated by the comet assay and probed with human 8-hydroxyguanine DNA-glycosylase 1. HEMA/Bis-GMA induced DSBs in HGFs as assessed by the neutral comet assay and phosphorylation of the H2AX histone and sodium ascorbate or melatonin (5-methoxy-N-acetyltryptamine) both at 50 µM reduced the DSBs, they also inhibited apoptosis induced by HEMA/Bis-GMA. The adhesive slowed the kinetics of the repair of DNA damage induced by hydrogen peroxide in HGFs, while sodium ascorbate or melatonin improved the efficacy of H(2)O(2)-induced damage in the presence of the methacrylates. The adhesive induced a rise in the G2/M cell population, accompanied by a reduction in the S cell population and an increase in G0/G1 cell population. Sodium ascorbate or melatonin elevated the S population and reduced the G2/M population. In conclusion, HEMA/Bis-GMA induce DSBs through, at least in part, oxidative mechanisms, and these compounds may interfere with DSBs repair. Vitamin C or melatonin may reduce the detrimental effects induced by methacrylates applied in dentistry.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA/efeitos dos fármacos , Cimentos Dentários/toxicidade , Metacrilatos/toxicidade , Mutagênicos/toxicidade , Apoptose/efeitos dos fármacos , Ácido Ascórbico/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Resinas Compostas/toxicidade , Gengiva/citologia , Gengiva/efeitos dos fármacos , Humanos , Melatonina/farmacologia
9.
Chem Res Toxicol ; 24(6): 877-86, 2011 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-21574630

RESUMO

The polyphenolics in green tea are believed to be the bioactive components. However, poor bioavailability following ingestion limits their efficacy in vivo. In this study, polyphenon E (poly E), a standardized green tea extract, was administered by sustained-release polycaprolactone implants (two, 2-cm implants; 20% drug load) grafted subcutaneously or via drinking water (0.8% w/v) to female S/D rats. Animals were treated with continuous low dose of benzo[a]pyrene (BP) via subcutaneous polymeric implants (2 cm; 10% load) and euthanized after 1 and 4 weeks. Analysis of lung DNA by (32)P-postlabeling resulted in a statistically significant reduction (50%; p = 0.023) of BP-induced DNA adducts in the implant group; however, only a modest (34%) but statistically insignificant reduction occurred in the drinking water group at 1 week. The implant delivery system also showed significant reduction (35%; p = 0.044) of the known BP diolepoxide-derived DNA adduct after 4 weeks. Notably, the total dose of poly E administered was >100-fold lower in the implant group than the drinking water group (15.7 versus 1,632 mg, respectively). Analysis of selected phase I, phase II, and nucleotide excision repair enzymes at both mRNA and protein levels showed no significant modulation by poly E, suggesting that the reduction in the BP-induced DNA adducts occurred presumably due to known scavenging of the antidiolepoxide of BP by the poly E catechins. In conclusion, our study demonstrated that sustained systemic delivery of poly E significantly reduced BP-induced DNA adducts in spite of its poor bioavailability following oral administration.


Assuntos
Benzo(a)pireno/toxicidade , Carcinógenos Ambientais/toxicidade , Catequina/análogos & derivados , Adutos de DNA/metabolismo , Implantes de Medicamento , Chá/química , Animais , Benzo(a)pireno/antagonistas & inibidores , Benzo(a)pireno/metabolismo , Catequina/administração & dosagem , Catequina/farmacologia , Adutos de DNA/antagonistas & inibidores , Reparo do DNA/efeitos dos fármacos , Implantes de Medicamento/química , Feminino , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Pulmão/metabolismo , Poliésteres/química , Ratos , Ratos Sprague-Dawley
10.
Arch Toxicol ; 85(12): 1575-88, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21881955

RESUMO

Although researchers have expended considerable effort on studying the cytotoxicity of nanomaterials, it is possible that there has been insufficient attention paid to their genotoxic potential. Here, we describe a test model that we have developed to evaluate the DNA-damaging effects of negatively charged nanoparticles of different sizes. We compared the DNA damaging effect induced by nanoparticles of various sizes and found that the effect is closely associated with the internalization pattern of the particles. Macrophage cell line RAW 264.7 cells were incubated with carboxylated polystyrene beads (COOH-PBs) ranging in size from 30 to 500 nm. Size-dependent DNA damage was detected, and the lesion induced by two carboxylated fullerene particles confirmed this observation. Confocal microscopy revealed that the entry pathways of these COOH-PBs shifted from direct penetration to endocytosis with increasing particle size, followed by changes in subcellular localization. Subsequent deposition of 30-nm COOH-PBs in the cytosol led to a reduction of Zn²âº and Mg²âº content in the nucleus and an increased p53 level in the whole cell rather than in nucleus, while localization of 50- and 100-nm COOH-PBs in acidic vesicles induced p53 accumulation in both types of extracts. Based on these results, we assume that the damage resulted from a disruption of the balance between DNA damage and repair.


Assuntos
Dano ao DNA/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Nanopartículas , Poliestirenos/química , Animais , Linhagem Celular , Citosol/metabolismo , Reparo do DNA/efeitos dos fármacos , Endocitose , Fulerenos/química , Macrófagos/patologia , Camundongos , Microscopia Confocal , Permeabilidade , Poliestirenos/farmacocinética , Proteína Supressora de Tumor p53/metabolismo
11.
Cancer Res ; 81(4): 1076-1086, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33323380

RESUMO

PARP inhibitors are approved for treatment of cancers with BRCA1 or BRCA2 defects. In this study, we prepared and characterized a very long-acting PARP inhibitor. Synthesis of a macromolecular prodrug of talazoparib (TLZ) was achieved by covalent conjugation to a PEG40kDa carrier via a ß-eliminative releasable linker. A single injection of the PEG∼TLZ conjugate was as effective as ∼30 daily oral doses of TLZ in growth suppression of homologous recombination-defective tumors in mouse xenografts. These included the KT-10 Wilms' tumor with a PALB2 mutation, the BRCA1-deficient MX-1 triple-negative breast cancer, and the BRCA2-deficient DLD-1 colon cancer; the prodrug did not inhibit an isogenic DLD-1 tumor with wild-type BRCA2. Although the half-life of PEG∼TLZ and released TLZ in the mouse was only ∼1 day, the exposure of released TLZ from a single safe, effective dose of the prodrug exceeded that of oral TLZ given daily over one month. µPET/CT imaging showed high uptake and prolonged retention of an 89Zr-labeled surrogate of PEG∼TLZ in the MX-1 BRCA1-deficient tumor. These data suggest that the long-lasting antitumor effect of the prodrug is due to a combination of its long t 1/2, the high exposure of TLZ released from the prodrug, increased tumor sensitivity upon continued exposure, and tumor accumulation. Using pharmacokinetic parameters of TLZ in humans, we designed a long-acting PEG∼TLZ for humans that may be superior in efficacy to daily oral TLZ and would be useful for treatment of PARP inhibitor-sensitive cancers in which oral medications are not tolerated. SIGNIFICANCE: These findings demonstrate that a single injection of a long-acting prodrug of the PARP inhibitor talazoparib in murine xenografts provides tumor suppression equivalent to a month of daily dosing of talazoparib.


Assuntos
Distúrbios no Reparo do DNA/patologia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Ftalazinas/uso terapêutico , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/genética , Distúrbios no Reparo do DNA/tratamento farmacológico , Distúrbios no Reparo do DNA/genética , Preparações de Ação Retardada/uso terapêutico , Feminino , Genes BRCA2 , Genes do Tumor de Wilms , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Camundongos SCID , Neoplasias/genética , Ftalazinas/química , Polietilenoglicóis/química , Polietilenoglicóis/uso terapêutico , Pró-Fármacos/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto , Zircônio/química , Zircônio/uso terapêutico
12.
Birth Defects Res B Dev Reprod Toxicol ; 89(2): 124-32, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20437471

RESUMO

BACKGROUND: Valproic acid (VPA) is a frequently used antiepileptic agent and known teratogen. Previous research suggests that inhibition of histone deacetylases (HDACs) may play a role in VPA-induced teratogenicity. We have also shown that VPA exposure leads to both an increase in reactive oxygen species (ROS) production and increased frequency of homologous recombination (HR). METHODS: In the present study, we evaluated the role of HDAC inhibition in VPA-initiated HR to determine if HDAC inhibition could alter repair activity and/or cause DNA double-strand breaks (DSBs), which would then initiate repair. Histone acetylation status was assessed to determine if VPA exposure led to HDAC inhibition in CHO 33 cells. RESULTS: Our results demonstrate that VPA (5 mM) exposure leads to increased acetylated histone H3 and H4 protein levels after 10 to 24 hr. Secondly, in our recombination assay where an artificial DNA DSB was induced in CHO 33 cells to assess repair activity, VPA exposure did not affect the repair activity of VPA-initiated HR. Subsequently, to determine if VPA could increase susceptibility to DNA DSBs, the number of gamma-H2AX foci was assessed using immunocytochemistry and results revealed an increase in gamma-H2AX foci after 10- to 24-hr exposure to VPA. CONCLUSIONS: Although we demonstrated the protective effect of polyethylene glycol-catalase against VPA-induced HR and the generation of intracellular ROS within 24 hr, we did not observed an increase in DNA oxidation. These studies suggest that HDAC inhibition and ROS signaling may play roles in DNA maintenance and cell-cycle arrest in initiating DNA damage and repair.


Assuntos
Anticonvulsivantes/farmacologia , Células CHO/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Recombinação Genética/efeitos dos fármacos , Teratogênicos/farmacologia , Acetilação , Animais , Células CHO/metabolismo , Catalase/farmacologia , Cricetinae , Cricetulus , DNA/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Dano ao DNA , Reparo do DNA/efeitos dos fármacos , Histonas/metabolismo , Estresse Oxidativo , Polietilenoglicóis/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Ácido Valproico/farmacologia
13.
Chem Res Toxicol ; 22(8): 1415-26, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19630433

RESUMO

Nickel oxide (NiO) is one of the important industrial materials used in electronic substrates and for ceramic engineering. Advancements in industrial technology have enabled the manufacture of ultrafine NiO particles. On the other hand, it is well-known that nickel compounds exert toxic effects. The toxicity of nickel compounds is mainly caused by nickel ions (Ni(2+)). However, the ion release properties of ultrafine NiO particles are still unclear. In the present study, the influences of ultrafine NiO particles on cell viability were examined in vitro to obtain fundamental data for the biological effects of ultrafine green NiO and ultrafine black NiO. Ultrafine NiO particles showed higher cytotoxicities toward human keratinocyte HaCaT cells and human lung carcinoma A549 cells than fine NiO particles and also showed higher solubilities in culture medium (Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum) than fine NiO particles. In particular, the concentration of Ni(2+) released into the culture medium by ultrafine green NiO was 150-fold higher than that released by fine green NiO. The concentrations of Ni(2+) released by both types of NiO particles in an aqueous solution containing amino acids were remarkably higher than those released by NiO particles in water. Moreover, we prepared a uniform and stable dispersion of ultrafine black NiO in culture medium and examined its influence on cell viability in comparison with that of NiCl(2), a soluble nickel compound. A medium exchange after 6 h of exposure resulted in a loss of cytotoxicity in the cells exposed to NiCl(2), whereas cytotoxicity was retained in the cells exposed to NiO. Transmission electron microscope observations revealed uptake of both ultrafine and fine NiO particles into HaCaT cells. Taken together, the present results suggest that the intracellular Ni(2+) release could be an important factor that determines the cytotoxicity of NiO. Ultrafine NiO is more cytotoxic than fine NiO in vitro.


Assuntos
Células/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Níquel/toxicidade , Silicones/química , Animais , Líquido da Lavagem Broncoalveolar , Carcinógenos , Bovinos , Células/metabolismo , Humanos , Masculino , Níquel/química , Níquel/farmacologia , Tamanho da Partícula , Material Particulado , Alvéolos Pulmonares , Solubilidade
14.
Int J Biol Macromol ; 125: 459-468, 2019 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-30529553

RESUMO

Currently no effective treatment is available to combat infections caused by Corynebacterium pseudotuberculosis in livestock. Survival of this Gram-positive bacterium in rapidly-growing pathogens in hostile environments is strongly dependent on the existence of a robust DNA repair system to prevent DNA mutations and contribute to bacterial colonization and virulence. The adenine/guanine-specific DNA glycosylase (MutY) is evolutionarily conserved and has been well characterized due to its central role in the prevention of mutagenesis and DNA repair. The aim of this study was the characterization of the target protein interaction with free adenine, suramin, and heparin, as well as the binding competition characterization between the molecules. The dissociation constant for free adenine interaction with Corynebacterium pseudotuberculosis MutY (Cp-MutY) was determined, 86 ±â€¯2.5 µM. NMR competition experiments demonstrated, that the polyanions heparin and suramin compete with adenine for the protein active site. The determined dissociation constant for the heparin/Cp-MutY interaction was 5.9 ±â€¯1.0 µM and for suramin was 16 ±â€¯1.5 µM. Docking of both polyanions with Cp-MutY revealed a possible mode of interaction and indicates that these molecules can interfere with the protein interaction with damaged DNA or prevent the binding of the adenine base in the enzyme active site.


Assuntos
Adenina/metabolismo , Corynebacterium pseudotuberculosis/efeitos dos fármacos , DNA/metabolismo , Heparina/farmacologia , Polímeros/farmacologia , Suramina/farmacologia , Domínio Catalítico/efeitos dos fármacos , DNA Glicosilases/metabolismo , Reparo do DNA/efeitos dos fármacos , Guanina/metabolismo , Cinética , Mutagênese/efeitos dos fármacos , Mutação/efeitos dos fármacos , N-Glicosil Hidrolases/metabolismo , Polieletrólitos
15.
PLoS One ; 13(5): e0196803, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29715291

RESUMO

Pyrrole-imidazole (Py-Im) polyamides are synthetic non-genotoxic minor groove-binding small molecules. We hypothesized that Py-Im polyamides can modulate the cellular response to ionizing radiation. Pre-treatment of cells with a Py-Im polyamide prior to exposure to ionizing radiation resulted in a delay in resolution of phosphorylated γ-H2AX foci, increase in XRCC1 foci, and reduced cellular replication potential. RNA-sequencing of cell lines exposed to the polyamide showed induction of genes related to the ultraviolet radiation response. We observed that the polyamide is almost 10-fold more toxic to a cell line deficient in DNA ligase 3 as compared to the parental cell line. Alkaline single cell gel electrophoresis reveals that the polyamide induces genomic fragmentation in the ligase 3 deficient cell line but not the corresponding parental line. The polyamide interferes directly with DNA ligation in vitro. We conclude that Py-Im polyamides may be further explored as sensitizers to genotoxic therapies.


Assuntos
Reparo do DNA/efeitos dos fármacos , DNA/efeitos dos fármacos , Imidazóis/farmacologia , Nylons/farmacologia , Pirróis/farmacologia , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , DNA Ligase Dependente de ATP/metabolismo , Humanos , Radiação Ionizante , Bibliotecas de Moléculas Pequenas/farmacologia , Proteína 1 Complementadora Cruzada de Reparo de Raio-X/metabolismo
16.
Mol Cancer Ther ; 17(12): 2586-2597, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30242094

RESUMO

The triple-negative breast cancer (TNBC) subtype, regardless of their BRCA1 status, has the poorest outcome compared with other breast cancer subtypes, and currently there are no approved targeted therapies for TNBC. We have previously demonstrated the importance of RAD6-mediated translesion synthesis pathway in TNBC development/progression and chemoresistance, and the potential therapeutic benefit of targeting RAD6 with a RAD6-selective small-molecule inhibitor, SMI#9. To overcome SMI#9 solubility limitations, we recently developed a gold nanoparticle (GNP)-based platform for conjugation and intracellular release of SMI#9, and demonstrated its in vitro cytotoxic activity toward TNBC cells. Here, we characterized the in vivo pharmacokinetic and therapeutic properties of PEGylated GNP-conjugated SMI#9 in BRCA1 wild-type and BRCA1-mutant TNBC xenograft models, and investigated the impact of RAD6 inhibition on TNBC metabolism by 1H-NMR spectroscopy. GNP conjugation allowed the released SMI#9 to achieve higher systemic exposure and longer retention as compared with the unconjugated drug. Systemically administered SMI#9-GNP inhibited the TNBC growth as effectively as intratumorally injected unconjugated SMI#9. Inductively coupled mass spectrometry analysis showed highest GNP concentrations in tumors and liver of SMI#9-GNP and blank-GNP-treated mice; however, tumor growth inhibition occurred only in the SMI#9-GNP-treated group. SMI#9-GNP was tolerated without overt signs of toxicity. SMI#9-induced sensitization was associated with perturbation of a common set of glycolytic pathways in BRCA1 wild-type and BRCA1-mutant TNBC cells. These data reveal novel SMI#9 sensitive markers of metabolic vulnerability for TNBC management and suggest that nanotherapy-mediated RAD6 inhibition offers a promising strategy for TNBC treatment.


Assuntos
Reparo do DNA , Tiazinas/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/patologia , Enzimas de Conjugação de Ubiquitina/metabolismo , Animais , Proteína BRCA1/metabolismo , Linhagem Celular Tumoral , Reparo do DNA/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Feminino , Ouro/química , Ouro/farmacocinética , Humanos , Redes e Vias Metabólicas/efeitos dos fármacos , Nanopartículas Metálicas/química , Camundongos Nus , Mutação/genética , Polietilenoglicóis/química , Tiazinas/farmacologia , Distribuição Tecidual/efeitos dos fármacos
17.
Toxicol In Vitro ; 47: 8-17, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29107684

RESUMO

Although methacrylic acid derivatives in their polymeric form are considered to be safe, insufficient polymerization and the release of monomers due to either mechanical or enzymatical factors can lead to their reaching millimolar concentrations in local tissue. The present study evaluates the effect of two methacrylate monomers - ethylene glycol dimethacrylate (EGDMA) and diethylene glycol dimethacrylate (DEGDMA) - on human gingival fibroblasts (HGFs). Both monomers were found to reduce cells viability in MTT assay, increase apoptosis and cause cell cycle arrest in G1/G0 phase. They also increased intracellular reactive oxygen species (ROS) production as measured by DCFH-DA and DHE probes and increased expression of GPx4 and SOD2. Both monomers increased DNA damage in comet assay. Moreover, HGFs were not able to repair those lesions within 120min of repair incubation. However, the monomers were not found to have any effect on the integrity of isolated plasmids. We postulate that EGDMA and DEGDMA exhibit their cytotoxic and genotoxic properties via increased production of ROS, which cause DNA damage, affect apoptosis, viability and cell cycle. Further studies are needed to better understand the properties of methacrylic acid monomers and to evaluate the risk that they cause for patients, dentists and dental technicians.


Assuntos
Apoptose/efeitos dos fármacos , Reagentes de Ligações Cruzadas/toxicidade , Reparo do DNA/efeitos dos fármacos , Etilenoglicóis/toxicidade , Gengiva/efeitos dos fármacos , Metacrilatos/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ensaio Cometa , Dano ao DNA , Materiais Dentários/toxicidade , Indução Enzimática/efeitos dos fármacos , Fase G1/efeitos dos fármacos , Gengiva/citologia , Gengiva/metabolismo , Glutationa Peroxidase/química , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Humanos , Concentração Osmolar , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Plasmídeos/efeitos dos fármacos , Espécies Reativas de Oxigênio/agonistas , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/química , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo
18.
Anticancer Res ; 27(4A): 2091-8, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17649827

RESUMO

Several mouthwash brands were investigated for the evaluation of mutagenicity and cell-transforming activity in short-term mutagenicity test systems. The tests were: the Salmonella typhimurium mutagenicity assay (Ames tests), the unscheduled DNA-repair induction assay in primary rat hepatocytes (UDS-test), and the V79-HGPRT mutagenicity assay. Mouthwash, with and without an external metabolic activation system (S9), consistently showed no mutagenic or cytotoxic activity in the Ames test, UDS assay or the V79-HGPRT-assay. These results indicate that the tested mouthwash brands are unlikely to present a mutagenic or carcinogenic hazard. However, these findings derived from an in vitro study cannot imitate the situation in situ, where a plethora of pharmacologically or bio-chemically active agents pass the oral cavity, thereby reacting with each other and the oral mucosa, in particular when the use of such rinses occurs regularly over a long-term period.


Assuntos
Antissépticos Bucais/efeitos adversos , Mutagênese/efeitos dos fármacos , Animais , Reparo do DNA/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Testes de Mutagenicidade , Ratos , Salmonella typhimurium/efeitos dos fármacos
19.
Biomed Pharmacother ; 94: 944-954, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28810532

RESUMO

Triple-negative breast cancers (TNBC) are aggressive cancers, which do not control by hormonal therapy or therapies that target HER-2 receptors. Curcumin (Cur) has shown cytotoxic effects in multiple cancer cell lines. However, its medical uses remain limited due to low aqueous solubility and poor bioavailability. Therefore, present study was aimed to fabricate the small positive charge curcumin nanoparticles (CN) by nanoprecipitation methods using PLGA and CTAB, and to evaluate its anticancer efficacy and underlying the mechanism in triple negative breast cancer cell lines (MDA-MB-231 cells). In in-vitro drug release assay, Cur was released from CN by flicking diffusion and anomalous transport process. CN showed a higher cellular incorporation than free Cur resulted in higher cytotoxicity. Checking the anticancer activity at the molecular level, Cur has shown to induce the reactive oxygen species production that subsequently causes the DNA damage and resulting in p38-MAPK activation. The p38-MAPK induce the expression of p16/INKK4a, p21/waf1/cip1 and p53 resulting in a reduction in the level of CDK2, CDK4, cyclin D1 and cyclin E and subsequently cell cycle arrest at G1/S and G2/M phase. It also reduces the expression of DNA repair gene, i.e. BRCA1, BRCA2, Rad51, Rad50, Mre11 and NBS1 resulting in apoptosis induction due to persistent DNA damage. This study presents an effective delivery of curcumin in TNBC cancer cells and it could open the new frontiers in clinical cancer chemotherapy.


Assuntos
Antineoplásicos/farmacologia , Compostos de Cetrimônio/farmacologia , Curcumina/farmacologia , Ácido Láctico/farmacologia , Nanopartículas/administração & dosagem , Ácido Poliglicólico/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cetrimônio , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Feminino , Humanos , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Espécies Reativas de Oxigênio/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
Mol Cancer Ther ; 16(8): 1456-1469, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28566437

RESUMO

High-grade gliomas, such as glioblastoma (GBM) and diffuse intrinsic pontine glioma (DIPG), are characterized by an aggressive phenotype with nearly universal local disease progression despite multimodal treatment, which typically includes chemotherapy, radiotherapy, and possibly surgery. Radiosensitizers that have improved the effects of radiotherapy for extracranial tumors have been ineffective for the treatment of GBM and DIPG, in part due to poor blood-brain barrier penetration and rapid intracranial clearance of small molecules. Here, we demonstrate that nanoparticles can provide sustained drug release and minimal toxicity. When administered locally, these nanoparticles conferred radiosensitization in vitro and improved survival in rats with intracranial gliomas when delivered concurrently with a 5-day course of fractionated radiotherapy. Compared with previous work using locally delivered radiosensitizers and cranial radiation, our approach, based on the rational selection of agents and a clinically relevant radiation dosing schedule, produces the strongest synergistic effects between chemo- and radiotherapy approaches to the treatment of high-grade gliomas. Mol Cancer Ther; 16(8); 1456-69. ©2017 AACR.


Assuntos
Neoplasias do Tronco Encefálico/tratamento farmacológico , Reparo do DNA , Glioma/tratamento farmacológico , Radiossensibilizantes/uso terapêutico , Animais , Neoplasias do Tronco Encefálico/patologia , Linhagem Celular Tumoral , Convecção , DNA/metabolismo , Reparo do DNA/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Endocitose/efeitos dos fármacos , Glioma/patologia , Humanos , Masculino , Nanopartículas/química , Nanopartículas/ultraestrutura , Poliésteres/química , Polietilenoglicóis/química , Radiossensibilizantes/farmacologia , Ratos Endogâmicos F344 , Distribuição Tecidual/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA