Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Nat Struct Mol Biol ; 28(6): 487-500, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34117478

RESUMO

Fanconi anemia (FA) is a devastating hereditary disease characterized by bone marrow failure (BMF) and acute myeloid leukemia (AML). As FA-deficient cells are hypersensitive to DNA interstrand crosslinks (ICLs), ICLs are widely assumed to be the lesions responsible for FA symptoms. Here, we show that FA-mutated cells are hypersensitive to persistent replication stress and that FA proteins play a role in the break-induced-replication (BIR)-like pathway for fork restart. Both the BIR-like pathway and ICL repair share almost identical molecular mechanisms of 53BP1-BRCA1-controlled signaling response, SLX4- and FAN1-mediated fork cleavage and POLD3-dependent DNA synthesis, suggesting that the FA pathway is intrinsically one of the BIR-like pathways. Replication stress not only triggers BMF in FA-deficient mice, but also specifically induces monosomy 7, which is associated with progression to AML in patients with FA, in FA-deficient cells.


Assuntos
Replicação do DNA , Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Anemia de Fanconi/genética , Aneuploidia , Animais , Transtornos da Insuficiência da Medula Óssea/etiologia , Linhagem Celular Transformada , Galinhas , Quebra Cromossômica , Deleção Cromossômica , Cromossomos Humanos Par 7/genética , DNA Polimerase III/fisiologia , Replicação do DNA/genética , Progressão da Doença , Anemia de Fanconi/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Feminino , Células HCT116 , Células HEK293 , Humanos , Hidroxiureia/farmacologia , Leucemia Mieloide Aguda/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Genéticos , Especificidade da Espécie , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/fisiologia , Ubiquitina-Proteína Ligases/fisiologia
2.
Gynecol Oncol ; 159(3): 869-876, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33032822

RESUMO

OBJECTIVE: Pathogenic variations in the homologous recombination (HR) gene, BRCA1 interacting protein C-terminal helicase 1 (BRIP1) increase the risk for ovarian cancer. PARP inhibitors (PARPi) exert a synthetic lethal effect in BRCA-mutated ovarian cancers. Effective HR requires cooperation between BRCA1 and BRIP1; therefore, BRIP1-incompetancy may predict vulnerability to synthetic lethality. Here we investigated the response of ovarian epithelial cells with defective BRIP1 function to PARPi, and compared these cells to those lacking BRCA1 activity. METHODS: We engineered Chinese Hamster ovarian (CHO) epithelial cells to express deficient BRIP1 or BRCA1, and exposed them to olaparib with or without carboplatin or cisplatin. We assessed cellular proliferation and survival; we calculated inhibitory concentrations and combination and reduction drug indices. RESULTS: BRIP1 and BRCA1 inactivation impedes HR activity, decreases cellular proliferation and compromises DNA damage recovery. Platinum agent exposure impairs cellular survival. Olaparib exposure alone decreases cell viability in BRCA1-deficient cells, although has no effect on BRIP1-deficient cells. Combining carboplatin or cisplatin with olaparib synergistically attenuates cellular survival, consistent with synthetic lethality. CONCLUSIONS: BRIP1-deficient ovarian epithelial cells exhibit defective HR, resulting in synthetic lethality when exposed to a platinum agent/PARPi combination. PARPi alone had no effect; this lack of effect may result from distinguishing molecular properties of BRIP1and/or consequences of genomic background. Our study identifies altered BRIP1 as a target for precision medicine-based therapies for ovarian cancers. This investigation supports consideration of the use of a platinum agent/PARPi combination in ovarian cancers depending upon genetic profile and genomic background.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Neoplasias Ovarianas/tratamento farmacológico , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , RNA Helicases/genética , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Proteína BRCA1/genética , Células CHO , Carboplatina/farmacologia , Carboplatina/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Cricetulus , Sinergismo Farmacológico , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Feminino , Humanos , Terapia de Alvo Molecular/métodos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Ftalazinas/farmacologia , Ftalazinas/uso terapêutico , Piperazinas/farmacologia , Piperazinas/uso terapêutico , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Medicina de Precisão/métodos , RNA Helicases/deficiência , Reparo de DNA por Recombinação/efeitos dos fármacos , Mutações Sintéticas Letais/efeitos dos fármacos
3.
Chem Res Toxicol ; 33(10): 2659-2667, 2020 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-32876438

RESUMO

Doxorubicin (DOX) is a widely used classical broad-spectrum anticancer drug. The major mechanism of DOX-mediated anticancer activity at clinically relevant concentrations is believed to be via DNA double-strand breaks due to topoisomerase IIα. However, other mechanisms by which DOX causes cytotoxicity have been proposed, including formaldehyde-dependent virtual interstrand cross-linking (ICL) formation. In this study, a method was established whereby cytotoxicity caused by virtual ICL derived from DOX is turned on and off using a cell culture system. Using this strategy, DOX-mediated cytotoxicity in Fanconi anemia group gene (FANC)/breast cancer susceptibility gene (BRCA)-deficient cells increased up to 70-fold compared to that in cells proficient in DNA repair pathways by increasing intracellular formaldehyde (FA) concentration. This approach also demonstrated that cytotoxicity introduced by DOX-mediated FA-dependent virtual ICL is completely independent of the toxicity induced by topoisomerase II inhibition at the cellular level. The potential of dual-targeting by DOX treatment was verified using an acid-specific FA donor. Overall, anticancer therapy targeting tumors deficient in the FANC/BRCA pathway may be possible by minimizing DOX-induced toxicity in normal cells.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Proteína BRCA1/antagonistas & inibidores , Doxorrubicina/farmacologia , Proteínas de Grupos de Complementação da Anemia de Fanconi/antagonistas & inibidores , Formaldeído/análise , Animais , Antibióticos Antineoplásicos/química , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Células Cultivadas , Galinhas , Dano ao DNA , Doxorrubicina/química , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Formaldeído/metabolismo , Estrutura Molecular
4.
Nat Commun ; 9(1): 2280, 2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29891926

RESUMO

Defects in DNA repair can cause various genetic diseases with severe pathological phenotypes. Fanconi anemia (FA) is a rare disease characterized by bone marrow failure, developmental abnormalities, and increased cancer risk that is caused by defective repair of DNA interstrand crosslinks (ICLs). Here, we identify the deubiquitylating enzyme USP48 as synthetic viable for FA-gene deficiencies by performing genome-wide loss-of-function screens across a panel of human haploid isogenic FA-defective cells (FANCA, FANCC, FANCG, FANCI, FANCD2). Thus, as compared to FA-defective cells alone, FA-deficient cells additionally lacking USP48 are less sensitive to genotoxic stress induced by ICL agents and display enhanced, BRCA1-dependent, clearance of DNA damage. Consequently, USP48 inactivation reduces chromosomal instability of FA-defective cells. Our results highlight a role for USP48 in controlling DNA repair and suggest it as a potential target that could be therapeutically exploited for FA.


Assuntos
Reparo do DNA/genética , Reparo do DNA/fisiologia , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteases Específicas de Ubiquitina/genética , Proteases Específicas de Ubiquitina/metabolismo , Proteína BRCA1/metabolismo , Sistemas CRISPR-Cas , Linhagem Celular , Instabilidade Cromossômica , Dano ao DNA , Anemia de Fanconi/terapia , Proteína do Grupo de Complementação A da Anemia de Fanconi/deficiência , Proteína do Grupo de Complementação A da Anemia de Fanconi/genética , Proteína do Grupo de Complementação A da Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação C da Anemia de Fanconi/deficiência , Proteína do Grupo de Complementação C da Anemia de Fanconi/genética , Proteína do Grupo de Complementação C da Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/deficiência , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação G da Anemia de Fanconi/deficiência , Proteína do Grupo de Complementação G da Anemia de Fanconi/genética , Proteína do Grupo de Complementação G da Anemia de Fanconi/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Técnicas de Inativação de Genes , Terapia Genética , Histonas/metabolismo , Humanos , Mutação , Rad51 Recombinase/metabolismo , Proteases Específicas de Ubiquitina/deficiência , Ubiquitinação
5.
Clin Cancer Res ; 24(11): 2700-2709, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29530934

RESUMO

Purpose: Mutations in Fanconi anemia (FA) genes are common in sporadic squamous cell carcinoma of the head and neck (HNSCC), and we have previously demonstrated that FA pathway depletion in HNSCC cell lines stimulates invasion. The goal of our studies was to use a systems approach in order to define FA pathway-dependent lipid metabolism and to extract lipid-based signatures and effectors of invasion in FA-deficient cells.Experimental Design: We subjected FA-isogenic HNSCC keratinocyte cell lines to untargeted and targeted lipidomics analyses to discover novel biomarkers and candidate therapeutic targets in FA-deficient cells. Cellular invasion assays were carried out in the presence and absence of N-butyldeoxynojirimycin (NB-DNJ), a biosynthetic inhibitor of the newly identified class of gangliosides, to investigate the requirement of ganglioside upregulation in FA-deficient HNSCC cells.Results: The most notable element of the lipid profiling results was a consistent elevation of glycosphingolipids, and particularly the accumulation of gangliosides. Conversely, repression of this same class of lipids was observed upon genetic correction of FA patient-derived HNSCC cells. Functional studies demonstrate that ganglioside upregulation is required for HNSCC cell invasion driven by FA pathway loss. The motility of nontransformed keratinocytes in response to FA loss displayed a similar dependence, thus supporting early and late roles for the FA pathway in controlling keratinocyte invasion through lipid regulation.Conclusions: Elevation of glycosphingolipids including the ganglioside GM3 in response to FA loss stimulates invasive characteristics of immortalized and transformed keratinocytes. An inhibitor of glycosphingolipid biosynthesis NB-DNJ attenuates invasive characteristics of FA-deficient HNSCC cells. Clin Cancer Res; 24(11); 2700-9. ©2018 AACR.


Assuntos
Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Glicoesfingolipídeos/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Metabolismo dos Lipídeos , Lipidômica , Redes e Vias Metabólicas , Animais , Linhagem Celular Transformada , Linhagem Celular Tumoral , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Gangliosídeo G(M3)/metabolismo , Glicoesfingolipídeos/química , Neoplasias de Cabeça e Pescoço/etiologia , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Queratinócitos/metabolismo , Lipidômica/métodos , Metabolômica/métodos , Camundongos
6.
Cell Death Differ ; 25(11): 1967-1979, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29511342

RESUMO

Metabolically reactive formaldehyde is a genotoxin and a carcinogen. Mice lacking the main formaldehyde-detoxifying gene Adh5 combined with the loss of the Fanconi anemia (FA) DNA repair pathway rapidly succumbed to bone marrow failure (BMF) primarily due to the extensive ablation of the hematopoietic stem cell (HSC) pool. However, the mechanism by which formaldehyde mediates these toxic effects is still unknown. We uncover a detrimental role of tetrahydrofolic acid (THF) in cells lacking Adh5 or the FA repair pathway. We show that Adh5- or FA-deficient cells are hypersensitive to formaldehyde and to THF, presenting DNA damage and genome instability. THF cytotoxicity involved imbalance of the nucleotide pool by deregulation of the thymidylate synthase (TYMS) enzyme, which stalled replication forks. In mice, THF exposure had widespread effects on hematopoiesis, affecting the frequency and the viability of myeloid- and lymphoid-committed precursor cells. Moreover, the hematopoietic stem and progenitor cells (HSPC) showed genomic instability, reduced colony-forming capacity and increased frequency of cycling and apoptotic HSCs upon THF exposure. Overall, our data reveal that the physiological pool of THF and formaldehyde challenge the stability of the genome of HSPCs that might lead to blood disorders.


Assuntos
Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Tetra-Hidrofolatos/toxicidade , Álcool Desidrogenase/deficiência , Álcool Desidrogenase/genética , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Galinhas , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Instabilidade Genômica/efeitos dos fármacos , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Histonas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Timidilato Sintase/metabolismo
7.
Oncotarget ; 7(47): 76590-76603, 2016 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-27740936

RESUMO

Cells that are deficient in homologous recombination, such as those that have mutations in any of the Fanconi Anemia (FA)/BRCA genes, are hypersensitive to inhibition of poly(ADP-ribose) polymerase (PARP). However, FA/BRCA-deficient tumors represent a small fraction of breast cancers, which might restrict the therapeutic utility of PARP inhibitor monotherapy. The gene encoding the serine-threonine protein kinase p21-activated kinase 1 (PAK1) is amplified and/or overexpressed in several human cancer types including 25-30% of breast tumors. This enzyme controls many cellular processes by phosphorylating both cytoplasmic and nuclear substrates. Here, we show that depletion or pharmacological inhibition of PAK1 down-regulated the expression of genes involved in the FA/BRCA pathway and compromised the ability of cells to repair DNA by Homologous Recombination (HR), promoting apoptosis and reducing colony formation. Combined inhibition of PAK1 and PARP in PAK1 overexpressing breast cancer cells had a synergistic effect, enhancing apoptosis, suppressing colony formation, and delaying tumor growth in a xenograft setting. Because reduced PAK1 activity impaired FA/BRCA function, inhibition of this kinase in PAK1 amplified and/or overexpressing breast cancer cells represents a plausible strategy for expanding the utility of PARP inhibitors to FA/BRCA-proficient cancers.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Quinases Ativadas por p21/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Cromossomos Humanos Par 11/genética , Dano ao DNA/efeitos dos fármacos , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/genética , Sinergismo Farmacológico , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Feminino , Amplificação de Genes , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Recombinação Homóloga , Humanos , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto , Quinases Ativadas por p21/genética
8.
Sci Rep ; 5: 18127, 2015 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-26658157

RESUMO

Fanconi anemia (FA) is a genetic disorder characterized by bone marrow failure, variable congenital malformations and a predisposition to malignancies. FANCB (also known as FAAP95), is the only X-linked FA gene discovered thus far. In the present study, we investigated hematopoiesis in adult Fancb deficient (Fancb(-/y)) mice and found that Fancb(-/y) mice have decreased hematopoietic stem cell (HSC) quiescence accompanied by reduced progenitor activity in vitro and reduced repopulating capacity in vivo. Like other FA mouse models previously reported, the hematopoietic system of Fancb(-/y) mice is hypersensitive to DNA cross-linking agent mitomycin C (MMC), which induces bone marrow failure in Fancb(-/y) mice. Furthermore, Fancb(-/y) BM exhibits slower recovery kinetics and less tolerance to myelotoxic stress induced by 5-fluorouracil than wild-type littermates. RNA-seq analysis reveals altered expression of genes involved in HSC function and cell cycle regulation in Fancb(-/y) HSC and progenitor cells. Thus, this Fancb(-/y) mouse model provides a novel approach for studying the critical role of the FA pathway not only in germ cell development but also in the maintenance of HSC function.


Assuntos
Modelos Animais de Doenças , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Animais , Antineoplásicos/farmacologia , Contagem de Células Sanguíneas , Medula Óssea/metabolismo , Medula Óssea/patologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Anemia de Fanconi/sangue , Anemia de Fanconi/genética , Anemia de Fanconi/patologia , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Feminino , Fluoruracila/farmacologia , Perfilação da Expressão Gênica/métodos , Hematopoese/genética , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitomicina/farmacologia
9.
Genes Dev ; 29(24): 2532-46, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26637282

RESUMO

Microsatellites are short tandem repeat sequences that are highly prone to expansion/contraction due to their propensity to form non-B-form DNA structures, which hinder DNA polymerases and provoke template slippage. Although error correction by mismatch repair plays a key role in preventing microsatellite instability (MSI), which is a hallmark of Lynch syndrome, activities must also exist that unwind secondary structures to facilitate replication fidelity. Here, we report that Fancj helicase-deficient mice, while phenotypically resembling Fanconi anemia (FA), are also hypersensitive to replication inhibitors and predisposed to lymphoma. Whereas metabolism of G4-DNA structures is largely unaffected in Fancj(-/-) mice, high levels of spontaneous MSI occur, which is exacerbated by replication inhibition. In contrast, MSI is not observed in Fancd2(-/-) mice but is prevalent in human FA-J patients. Together, these data implicate FANCJ as a key factor required to counteract MSI, which is functionally distinct from its role in the FA pathway.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Anemia de Fanconi/fisiopatologia , Linfoma/genética , Instabilidade de Microssatélites , Animais , Antineoplásicos/farmacologia , Fatores de Transcrição de Zíper de Leucina Básica/deficiência , Camptotecina/farmacologia , Linhagem Celular , Células Cultivadas , Dano ao DNA/genética , Anemia de Fanconi/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/efeitos da radiação , Predisposição Genética para Doença , Humanos , Masculino , Camundongos Knockout , Mitomicina/farmacologia , Neoplasias Epiteliais e Glandulares/genética , RNA Helicases , Raios Ultravioleta
10.
Nucleic Acids Res ; 42(2): 860-9, 2014 01.
Artigo em Inglês | MEDLINE | ID: mdl-24163102

RESUMO

It has been proposed that guanine-rich DNA forms four-stranded structures in vivo called G-quadruplexes or G4 DNA. G4 DNA has been implicated in several biological processes, but tools to study G4 DNA structures in cells are limited. Here we report the development of novel murine monoclonal antibodies specific for different G4 DNA structures. We show that one of these antibodies designated 1H6 exhibits strong nuclear staining in most human and murine cells. Staining intensity increased on treatment of cells with agents that stabilize G4 DNA and, strikingly, cells deficient in FANCJ, a G4 DNA-specific helicase, showed stronger nuclear staining than controls. Our data strongly support the existence of G4 DNA structures in mammalian cells and indicate that the abundance of such structures is increased in the absence of FANCJ. We conclude that monoclonal antibody 1H6 is a valuable tool for further studies on the role of G4 DNA in cell and molecular biology.


Assuntos
DNA/química , Quadruplex G , Animais , Anticorpos Monoclonais/imunologia , Núcleo Celular/genética , Cromossomos/imunologia , DNA/análise , DNA/imunologia , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Humanos , Camundongos
11.
Cell Biochem Biophys ; 66(3): 431-41, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23238643

RESUMO

Intermittent hypoxia (IH) is a hallmark feature in obstructive sleep apnea (OSA) which is increasingly recognized as an independent risk factor for atherosclerosis. Oxidative stress, inflammation, and cell apoptosis are major pathological events initiating or accelerating atherogenesis. This study addressed whether IH would affect these proatherogenic factors in endothelial cells and the mechanistic pathways involved. EA.hy926 cells were exposed to intermittent normoxia or IH for different numbers of cycles (32, 64, or 96). IH exposure time-dependently raised cellular GSSG/GSH ratio, increased production of IL-6 and IL-8, and accelerated cell apoptosis and death, concurrent with activation of NF-κB and inhibition of Nrf2/HO-1 pathways. At 64 cycles, inhibition of NF-κB attenuated IH-induced cellular oxidative stress and accumulation of inflammatory cytokines in cell culture medium but aggravated IH-induced cell apoptosis, while stimulation of HO-1 suppressed IH-induced cellular oxidative stress and cell apoptosis without affecting accumulation of inflammatory cytokines in cell culture medium. We demonstrated that early stage of exposure to IH-induced oxidative and inflammatory stresses leading to acceleration of cell apoptosis via NF-κB and Nrf2/HO-1 pathways in endothelial cells, suggesting the potential mechanisms for IH-induced vascular pathogenesis, in resemblance to OSA.


Assuntos
Células Endoteliais/citologia , Heme Oxigenase-1/metabolismo , NF-kappa B/metabolismo , Apoptose/efeitos dos fármacos , Fatores de Transcrição de Zíper de Leucina Básica/deficiência , Fatores de Transcrição de Zíper de Leucina Básica/genética , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular , Meios de Cultura/química , Citocinas/metabolismo , Células Endoteliais/efeitos dos fármacos , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Técnicas de Silenciamento de Genes , Hemina/farmacologia , Humanos , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/deficiência , NF-kappa B/genética , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Tiofenos/farmacologia
12.
Blood ; 119(18): 4142-51, 2012 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-22408259

RESUMO

Oxidative stress has been implicated in the pathogenesis of many human diseases including Fanconi anemia (FA), a genetic disorder associated with BM failure and cancer. Here we show that major antioxidant defense genes are down-regulated in FA patients, and that gene down-regulation is selectively associated with increased oxidative DNA damage in the promoters of the antioxidant defense genes. Assessment of promoter activity and DNA damage repair kinetics shows that increased initial damage, rather than a reduced repair rate, contributes to the augmented oxidative DNA damage. Mechanistically, FA proteins act in concert with the chromatin-remodeling factor BRG1 to protect the promoters of antioxidant defense genes from oxidative damage. Specifically, BRG1 binds to the promoters of the antioxidant defense genes at steady state. On challenge with oxidative stress, FA proteins are recruited to promoter DNA, which correlates with significant increase in the binding of BRG1 within promoter regions. In addition, oxidative stress-induced FANCD2 ubiquitination is required for the formation of a FA-BRG1-promoter complex. Taken together, these data identify a role for the FA pathway in cellular antioxidant defense.


Assuntos
Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Anemia de Fanconi/metabolismo , Regulação da Expressão Gênica , Estresse Oxidativo/genética , Regiões Promotoras Genéticas/genética , 8-Hidroxi-2'-Desoxiguanosina , Antioxidantes , Células da Medula Óssea/metabolismo , Células Cultivadas/efeitos dos fármacos , Dano ao DNA , DNA Helicases/metabolismo , Reparo do DNA , Desoxiguanosina/análogos & derivados , Desoxiguanosina/análise , Regulação para Baixo , Anemia de Fanconi/genética , Anemia de Fanconi/patologia , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Humanos , Peróxido de Hidrogênio/toxicidade , Complexos Multiproteicos , Proteínas Nucleares/metabolismo , Oxirredução , Ligação Proteica , Processamento de Proteína Pós-Traducional , Fatores de Transcrição/metabolismo , Ubiquitinação
13.
Hum Mol Genet ; 18(18): 3484-95, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19561169

RESUMO

The Fanconi anemia (FA) core complex member FANCM remodels synthetic replication forks and recombination intermediates. Thus far, only one FA patient with FANCM mutations has been described, but the relevance of these mutations for the FA phenotype is uncertain. To provide further experimental access to the FA-M complementation group we have generated Fancm-deficient mice by deleting exon 2. FANCM deficiency caused hypogonadism in mice and hypersensitivity to cross-linking agents in mouse embryonic fibroblasts (MEFs), thus phenocopying other FA mouse models. However, Fancm(Delta2/Delta2) mice also showed unique features atypical for FA mice, including underrepresentation of female Fancm(Delta2/Delta2) mice and decreased overall and tumor-free survival. This increased cancer incidence may be correlated to the role of FANCM in the suppression of spontaneous sister chromatid exchanges as observed in MEFs. In addition, FANCM appeared to have a stimulatory rather than essential role in FANCD2 monoubiquitination. The FA-M mouse model presented here suggests that FANCM functions both inside and outside the FA core complex to maintain genome stability and to prevent tumorigenesis.


Assuntos
Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Alelos , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Células Cultivadas , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patologia , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovário/anormalidades , Ovário/metabolismo , Fenótipo , Troca de Cromátide Irmã , Taxa de Sobrevida , Testículo/anormalidades , Testículo/metabolismo
14.
Blood ; 112(5): 2062-70, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18550849

RESUMO

Fanconi anemia (FA) proteins are thought to play a role in chromosome stability and repair of DNA cross-links; however, these functions may not fully explain the developmental abnormalities and bone marrow failure that are characteristic of FA individuals. Here we associate the FA proteins with the Notch1 developmental pathway through a direct protein-protein interaction between the FA core complex and the hairy enhancer of split 1 (HES1). HES1 interaction with FA core complex members is dependent on a functional FA pathway. Cells depleted of HES1 exhibit an FA-like phenotype that includes cellular hypersensitivity to mitomycin C (MMC) and lack of FANCD2 monoubiquitination and foci formation. HES1 is also required for proper nuclear localization or stability of some members of the core complex. Our results suggest that HES1 is a novel interacting protein of the FA core complex.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Proteínas de Homeodomínio/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem Celular , Linhagem Celular Transformada , Resistência a Medicamentos/genética , Resistência a Medicamentos/fisiologia , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação C da Anemia de Fanconi/deficiência , Proteína do Grupo de Complementação C da Anemia de Fanconi/genética , Proteína do Grupo de Complementação C da Anemia de Fanconi/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/química , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Células HeLa , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Camundongos Knockout , Mitomicina/farmacologia , Complexos Multiproteicos , Ligação Proteica , RNA Interferente Pequeno/genética , Receptor Notch1/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Fatores de Transcrição HES-1 , Técnicas do Sistema de Duplo-Híbrido , Ubiquitinação
15.
J Biol Chem ; 283(33): 22363-70, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18550526

RESUMO

Intracellular heme is a redox active molecule that can be detrimental to cells at high concentrations or under oxidizing conditions. To prevent accumulation, the inducible enzyme heme oxygenase-1 (HMOX1) catalyzes degradation of heme. In the absence of elevated intracellular heme or oxidative stress, the basic region leucine zipper transcriptional regulator BACH1 binds HMOX1 antioxidant response elements and represses transcription. Conversely, increased intracellular heme or sulfhydryl oxidation inactivate BACH1, permitting transcriptional induction of HMOX1. Here, we investigate the effect of BACH1 inactivation on the induction of HMOX1 and as a mechanism for broader gene induction. We show that BACH1 is inactivated at low micromolar arsenite concentrations and that BACH1 inactivation is necessary and sufficient for transcriptional induction of HMOX1. Because BACH1 is thought to interact with antioxidant response element motifs, we further examined the role of BACH1 as a regulator of inducible antioxidant gene expression by assessing the global profile of gene expression following BACH1 knockdown using small interfering RNA. The loss of BACH1 function in human keratinocytes results almost exclusively in HMOX1 induction, suggesting that BACH1 may function as a rheostat regulating levels of intracellular free heme.


Assuntos
Arsenitos/farmacologia , Fatores de Transcrição de Zíper de Leucina Básica/antagonistas & inibidores , Fatores de Transcrição de Zíper de Leucina Básica/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/antagonistas & inibidores , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Heme Oxigenase-1/biossíntese , Queratinócitos/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/deficiência , Primers do DNA , Indução Enzimática , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Heme/metabolismo , Humanos , Zíper de Leucina/genética , Análise de Sequência com Séries de Oligonucleotídeos , Oxirredução , RNA/genética , RNA/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Pathol Biol (Paris) ; 55(1): 19-28, 2007 Feb.
Artigo em Francês | MEDLINE | ID: mdl-16904272

RESUMO

Fanconi anemia (FA) is a recessive human cancer prone syndrome featuring bone marrow failure, developmental abnormalities and hypersensitivity to DNA crosslinking agents exposure. 11 among 12 FA gene have been isolated. The biochemical functions of the FANC proteins remain poorly understood. Anyhow, to cope with DNA crosslinks a cell needs a functional FANC pathway. Moreover, the FANC proteins appear to be involved in cell protection against oxidative damage and in the control of TNF-alpha activity. In this review, we describe the current understanding of the FANC pathway and we present how it may be integrated in the complex networks of proteins involved in maintaining the cellular homeostasis.


Assuntos
Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Anemia de Fanconi/patologia , Transplante de Medula Óssea , Cromossomos Humanos/efeitos dos fármacos , Reagentes de Ligações Cruzadas/efeitos adversos , DNA/efeitos dos fármacos , Dano ao DNA , Reparo do DNA/genética , Anemia de Fanconi/diagnóstico , Anemia de Fanconi/genética , Anemia de Fanconi/terapia , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Genes Recessivos , Teste de Complementação Genética , Heterogeneidade Genética , Terapia Genética , Hematopoese , Homeostase , Humanos , Mutagênicos/efeitos adversos , Estresse Oxidativo , Fenótipo , Fator de Necrose Tumoral alfa/fisiologia
17.
J Med Genet ; 44(4): 241-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17105750

RESUMO

BACKGROUND: Fanconi anaemia is a heterogeneous genetic disease, where 12 complementation groups have been already described. Identifying the complementation group in patients with Fanconi anaemia constitutes a direct procedure to confirm the diagnosis of the disease and is required for the recruitment of these patients in gene therapy trials. OBJECTIVE: To determine the subtype of Fanconi anaemia patients in Spain, a Mediterranean country with a relatively high population (23%) of Fanconi anaemia patients belonging to the gypsy race. METHODS: Most patients could be subtyped by retroviral complementation approaches in peripheral blood T cells, although some mosaic patients were subtyped in cultured skin fibroblasts. Other approaches, mainly based on western blot analysis and generation of nuclear RAD51 and FANCJ foci, were required for the subtyping of a minor number of patients. RESULTS AND CONCLUSIONS: From a total of 125 patients included in the Registry of Fanconi Anaemia, samples from 102 patients were available for subtyping analyses. In 89 cases the subtype could be determined and in 8 cases exclusions of common complementation groups were made. Compared with other international studies, a skewed distribution of complementation groups was observed in Spain, where 80% of the families belonged to the Fanconi anaemia group A (FA-A) complementation group. The high proportion of gypsy patients, all of them FA-A, and the absence of patients with FA-C account for this characteristic distribution of complementation groups.


Assuntos
Algoritmos , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Anemia de Fanconi/classificação , Heterogeneidade Genética , Roma (Grupo Étnico)/genética , Células Cultivadas/química , Células Cultivadas/efeitos dos fármacos , Consanguinidade , Resistência a Medicamentos/genética , Compostos de Epóxi/farmacologia , Anemia de Fanconi/epidemiologia , Anemia de Fanconi/genética , Proteína do Grupo de Complementação A da Anemia de Fanconi/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/análise , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Fibroblastos/química , Fibroblastos/patologia , Teste de Complementação Genética , Genótipo , Humanos , Incidência , Mitomicina/farmacologia , Mosaicismo , Sistema de Registros , Retroviridae/genética , Espanha/epidemiologia , Linfócitos T/química , Linfócitos T/efeitos dos fármacos , Linfócitos T/patologia , Transdução Genética
18.
Clin Cancer Res ; 11(20): 7508-15, 2005 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16243825

RESUMO

PURPOSE: BRCA2, FANCC, and FANCG gene mutations are present in a subset of pancreatic cancer. Defects in these genes could lead to hypersensitivity to interstrand cross-linkers in vivo and a more optimal treatment of pancreatic cancer patients based on the genetic profile of the tumor. EXPERIMENTAL DESIGN: Two retrovirally complemented pancreatic cancer cell lines having defects in the Fanconi anemia pathway, PL11 (FANCC-mutated) and Hs766T (FANCG-mutated), as well as several parental pancreatic cancer cell lines with or without mutations in the Fanconi anemia/BRCA2 pathway, were assayed for in vitro and in vivo sensitivities to various chemotherapeutic agents. RESULTS: A distinct dichotomy of drug responses was observed. Fanconi anemia-defective cancer cells were hypersensitive to the cross-linking agents mitomycin C (MMC), cisplatin, chlorambucil, and melphalan but not to 5-fluorouracil, gemcitabine, doxorubicin, etoposide, vinblastine, or paclitaxel. Hypersensitivity to cross-linking agents was confirmed in vivo; FANCC-deficient xenografts of PL11 and BRCA2-deficient xenografts of CAPAN1 regressed on treatment with two different regimens of MMC whereas Fanconi anemia-proficient xenografts did not. The MMC response comprised cell cycle arrest, apoptosis, and necrosis. Xenografts of PL11 also regressed after a single dose of cyclophosphamide whereas xenografts of genetically complemented PL11(FANCC) did not. CONCLUSIONS: MMC or other cross-linking agents as a clinical therapy for pancreatic cancer patients with tumors harboring defects in the Fanconi anemia/BRCA2 pathway should be specifically investigated.


Assuntos
Reagentes de Ligações Cruzadas/farmacologia , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Mitomicina/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proteína BRCA2/deficiência , Proteína BRCA2/genética , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Clorambucila/farmacologia , Cisplatino/farmacologia , Reagentes de Ligações Cruzadas/uso terapêutico , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Etoposídeo/farmacologia , Proteína do Grupo de Complementação C da Anemia de Fanconi/deficiência , Proteína do Grupo de Complementação C da Anemia de Fanconi/genética , Proteína do Grupo de Complementação G da Anemia de Fanconi/deficiência , Proteína do Grupo de Complementação G da Anemia de Fanconi/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/deficiência , Feminino , Fluoruracila/farmacologia , Humanos , Concentração Inibidora 50 , Melfalan/farmacologia , Camundongos , Camundongos Nus , Mitomicina/uso terapêutico , Mutação , Paclitaxel/farmacologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Fatores de Tempo , Vimblastina/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Gencitabina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA