Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Dev Cell ; 56(15): 2207-2222.e7, 2021 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-34256011

RESUMO

Cells counter DNA damage through repair or apoptosis, yet a direct mechanism for this choice has remained elusive. When facing interstrand crosslinks (ICLs), the ICL-repair protein FANCI heterodimerizes with FANCD2 to initiate ICL excision. We found that FANCI alternatively interacts with a pro-apoptotic factor, PIDD1, to enable PIDDosome (PIDD1-RAIDD-caspase-2) formation and apoptotic death. FANCI switches from FANCD2/repair to PIDD1/apoptosis signaling in the event of ICL-repair failure. Specifically, removing key endonucleases downstream of FANCI/FANCD2, increasing ICL levels, or allowing damaged cells into mitosis (when repair is suppressed) all suffice for switching. Reciprocally, apoptosis-committed FANCI reverts from PIDD1 to FANCD2 after a failed attempt to assemble the PIDDosome. Monoubiquitination and deubiquitination at FANCI K523 impact interactor selection. These data unveil a repair-or-apoptosis switch in eukaryotes. Beyond ensuring the removal of unrepaired genomes, the switch's bidirectionality reveals that damaged cells can offset apoptotic defects via de novo attempts at lesion repair.


Assuntos
Apoptose/fisiologia , Reparo do DNA/fisiologia , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Animais , Proteína Adaptadora de Sinalização CRADD/metabolismo , Linhagem Celular Tumoral , Cromatina/metabolismo , DNA/metabolismo , Dano ao DNA/fisiologia , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Células HeLa , Humanos , Ubiquitinação , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
2.
Nucleic Acids Res ; 48(10): 5467-5484, 2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32329774

RESUMO

Transcription-replication (T-R) conflicts are profound threats to genome integrity. However, whilst much is known about the existence of T-R conflicts, our understanding of the genetic and temporal nature of how cells respond to them is poorly established. Here, we address this by characterizing the early cellular response to transient T-R conflicts (TRe). This response specifically requires the DNA recombination repair proteins BLM and BRCA2 as well as a non-canonical monoubiquitylation-independent function of FANCD2. A hallmark of the TRe response is the rapid co-localization of these three DNA repair factors at sites of T-R collisions. We find that the TRe response relies on basal activity of the ATR kinase, yet it does not lead to hyperactivation of this key checkpoint protein. Furthermore, specific abrogation of the TRe response leads to DNA damage in mitosis, and promotes chromosome instability and cell death. Collectively our findings identify a new role for these well-established tumor suppressor proteins at an early stage of the cellular response to conflicts between DNA transcription and replication.


Assuntos
Replicação do DNA , Reparo de DNA por Recombinação , Transcrição Gênica , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteína BRCA2/fisiologia , Linhagem Celular , Sobrevivência Celular , Quinase 9 Dependente de Ciclina/metabolismo , DNA/metabolismo , Dano ao DNA , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Humanos , Mitose/genética , Regiões Promotoras Genéticas , RNA/metabolismo , RNA Polimerase II/metabolismo , Splicing de RNA , RecQ Helicases/fisiologia , Ubiquitinação
3.
Nat Chem Biol ; 16(3): 291-301, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31873223

RESUMO

DNA-damage repair is implemented by proteins that are coordinated by specialized molecular signals. One such signal in the Fanconi anemia (FA) pathway for the repair of DNA interstrand crosslinks is the site-specific monoubiquitination of FANCD2 and FANCI. The signal is mediated by a multiprotein FA core complex (FA-CC) however, the mechanics for precise ubiquitination remain elusive. We show that FANCL, the RING-bearing module in FA-CC, allosterically activates its cognate ubiqutin-conjugating enzyme E2 UBE2T to drive site-specific FANCD2 ubiquitination. Unlike typical RING E3 ligases, FANCL catalyzes ubiquitination by rewiring the intraresidue network of UBE2T to influence the active site. Consequently, a basic triad unique to UBE2T engages a structured acidic patch near the target lysine on FANCD2. This three-dimensional complementarity, between the E2 active site and substrate surface, induced by FANCL is central to site-specific monoubiquitination in the FA pathway. Furthermore, the allosteric network of UBE2T can be engineered to enhance FANCL-catalyzed FANCD2-FANCI di-monoubiquitination without compromising site specificity.


Assuntos
Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação L da Anemia de Fanconi/metabolismo , Regulação Alostérica/fisiologia , Sequência de Aminoácidos , Dano ao DNA , Reparo do DNA , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Proteína do Grupo de Complementação L da Anemia de Fanconi/fisiologia , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Humanos , Ligação Proteica , Especificidade por Substrato , Enzimas de Conjugação de Ubiquitina/metabolismo , Enzimas de Conjugação de Ubiquitina/fisiologia , Ubiquitinação
4.
Pediatr Blood Cancer ; 66(1): e27460, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30255556

RESUMO

BACKGROUND: Fanconi anemia is an inherited bone marrow failure disorder associated with a high incidence of leukemia and solid tumors. Currently, no interventions to prevent or delay the formation of solid tumors are available. PROCEDURE: Two of the most important hallmarks of Fanconi anemia are inflammation and oxidative stress. In this study, we administrated the antioxidant atorvastatin and the anti-inflammatory drug celecoxib to cohorts of Fancd2-/- /Trp53+/- mice, a model of Fanconi anemia. Treatment started at weaning and continued until the mice developed a palpable mass or suffered from >20% weight loss. Tumor samples and selected tissues were subjected to histopathological examination. χ2 test was performed to analyze tumor incidence, and Kaplan-Meier survival curves were evaluated with log-rank test. In addition, a small cohort of mice was monitored for the safety of the drugs. RESULTS: The combined oral administration of both drugs significantly delayed tumor onset in Fancd2-/- /Trp53+/- mice. Specifically, the treatment delayed the onset of ovarian tumors in Fancd2-/- /Trp53+/- mice and increased the mean ovarian tumor-free survival time by 17%, whereas this combinatorial drug regimen did not have a significant effect on other tumor types. In addition, no detrimental effects on hematopoiesis from the drug treatment were observed during a 12-month safety monitoring. CONCLUSIONS: The data presented here suggest that a combination of atorvastatin and celecoxib may be a good candidate for chemoprevention in Fanconi anemia.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Modelos Animais de Doenças , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Anemia de Fanconi/tratamento farmacológico , Proteína Supressora de Tumor p53/fisiologia , Animais , Atorvastatina/administração & dosagem , Celecoxib/administração & dosagem , Anemia de Fanconi/patologia , Feminino , Masculino , Camundongos , Camundongos Knockout , Taxa de Sobrevida
5.
PLoS Genet ; 14(10): e1007643, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30335751

RESUMO

The Fanconi Anemia (FA) pathway is important for repairing interstrand crosslinks (ICLs) between the Watson-Crick strands of the DNA double helix. An initial and essential stage in the repair process is the detection of the ICL. Here, we report the identification of UHRF2, a paralogue of UHRF1, as an ICL sensor protein. UHRF2 is recruited to ICLs in the genome within seconds of their appearance. We show that UHRF2 cooperates with UHRF1, to ensure recruitment of FANCD2 to ICLs. A direct protein-protein interaction is formed between UHRF1 and UHRF2, and between either UHRF1 and UHRF2, and FANCD2. Importantly, we demonstrate that the essential monoubiquitination of FANCD2 is stimulated by UHRF1/UHRF2. The stimulation is mediating by a retention of FANCD2 on chromatin, allowing for its monoubiquitination by the FA core complex. Taken together, we uncover a mechanism of ICL sensing by UHRF2, leading to FANCD2 recruitment and retention at ICLs, in turn facilitating activation of FANCD2 by monoubiquitination.


Assuntos
Reparo do DNA/fisiologia , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Sequência de Aminoácidos , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/fisiologia , Linhagem Celular , Núcleo Celular/metabolismo , Cromatina/metabolismo , DNA/metabolismo , Dano ao DNA/fisiologia , Anemia de Fanconi/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Células HEK293 , Células HeLa , Humanos , Domínios e Motivos de Interação entre Proteínas , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
6.
J Biol Chem ; 292(49): 20184-20195, 2017 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-29021208

RESUMO

In response to DNA cross-linking damage, the Fanconi anemia (FA) core complex activates the FA pathway by monoubiquitinating Fanconi anemia complementation group D2 (FANCD2) for the initiation of the nucleolytic processing of the DNA cross-links and stabilization of stalled replication forks. Given that all the classic FA proteins coordinately monoubiquitinate FANCD2, it is unclear why losses of individual classic FA genes yield varying cellular sensitivities to cross-linking damage. To address this question, we generated cellular knock-out models of FA core complex components and FANCD2 and found that FANCD2-null mutants display higher levels of spontaneous chromosomal damage and hypersensitivity to replication-blocking lesions than Fanconi anemia complementation group L (FANCL)-null mutants, suggesting that FANCD2 provides a basal level of DNA protection countering endogenous lesions in the absence of monoubiquitination. FANCD2's ubiquitination-independent function is likely involved in optimized recruitment of nucleolytic activities for the processing and protection of stressed replication forks. Our results reveal that FANCD2 has a ubiquitination-independent role in countering endogenous levels of replication stress, a function that is critical for the maintenance of genomic stability.


Assuntos
Dano ao DNA/genética , Replicação do DNA/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Estresse Fisiológico , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Técnicas de Introdução de Genes , Técnicas de Silenciamento de Genes , Instabilidade Genômica , Células HeLa , Humanos , Ubiquitinação
7.
Sci Rep ; 6: 36539, 2016 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-27827420

RESUMO

Proteins involved in genetic stability maintenance and safeguarding DNA replication act not only against cancer initiation but could also play a major role in sustaining cancer progression. Here, we report that the FANC pathway is highly expressed in metastatic melanoma harboring the oncogenic microphthalmia-associated transcription factor (MiTF). We show that MiTF downregulation in melanoma cells lowers the expression of several FANC genes and proteins. Moreover, we observe that, similarly to the consequence of MiTF downregulation, FANC pathway silencing alters proliferation, migration and senescence of human melanoma cells. We demonstrate that the FANC pathway acts downstream MiTF and establish the existence of an epistatic relationship between MiTF and the FANC pathway. Our findings point to a central role of the FANC pathway in cellular and chromosomal resistance to both DNA damage and targeted therapies in melanoma cells. Thus, the FANC pathway is a promising new therapeutic target in melanoma treatment.


Assuntos
Proliferação de Células , Sobrevivência Celular , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Melanoma/patologia , Fator de Transcrição Associado à Microftalmia/metabolismo , Animais , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Inativação Gênica , Humanos , Indóis/uso terapêutico , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Camundongos , Camundongos Nus , Fator de Transcrição Associado à Microftalmia/genética , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Sulfonamidas/uso terapêutico , Vemurafenib
8.
Nucleic Acids Res ; 44(22): 10758-10771, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27694619

RESUMO

The FANCI-FANCD2 (I-D) complex is considered to work with RAD51 to protect the damaged DNA in the stalled replication fork. However, the means by which this DNA protection is accomplished have remained elusive. In the present study, we found that the I-D complex directly binds to RAD51, and stabilizes the RAD51-DNA filament. Unexpectedly, the DNA binding activity of FANCI, but not FANCD2, is explicitly required for the I-D complex-mediated RAD51-DNA filament stabilization. The RAD51 filament stabilized by the I-D complex actually protects the DNA end from nucleolytic degradation by an FA-associated nuclease, FAN1. This DNA end protection is not observed with the RAD51 mutant from FANCR patient cells. These results clearly answer the currently enigmatic question of how RAD51 functions with the I-D complex to prevent genomic instability at the stalled replication fork.


Assuntos
Proteínas Aviárias/fisiologia , DNA/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Rad51 Recombinase/fisiologia , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Galinhas , Sequência Conservada , DNA/metabolismo , Dano ao DNA , Reparo do DNA , Replicação do DNA , Instabilidade Genômica , Humanos , Ligação Proteica , Estabilidade Proteica , Ubiquitinação
9.
Nat Struct Mol Biol ; 23(8): 755-757, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27322732

RESUMO

The tumor suppressor BRCA2 plays a key role in genome integrity by promoting replication-fork stability and homologous recombination (HR) DNA repair. Here we report that human cancer cells lacking BRCA2 rely on the Fanconi anemia protein FANCD2 to limit replication-fork progression and genomic instability. Our results identify a new role of FANCD2 in limiting constitutive replication stress in BRCA2-deficient cells, thereby affecting cell survival and treatment responses.


Assuntos
Proteína BRCA2/metabolismo , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular , Dano ao DNA , Replicação do DNA , Genoma Humano , Instabilidade Genômica , Células HEK293 , Humanos , Ftalazinas/farmacologia , Piperazinas/farmacologia
10.
Nat Commun ; 7: 11091, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-27033104

RESUMO

Germline mutations affecting telomere maintenance or DNA repair may, respectively, cause dyskeratosis congenita or Fanconi anaemia, two clinically related bone marrow failure syndromes. Mice expressing p53(Δ31), a mutant p53 lacking the C terminus, model dyskeratosis congenita. Accordingly, the increased p53 activity in p53(Δ31/Δ31) fibroblasts correlated with a decreased expression of 4 genes implicated in telomere syndromes. Here we show that these cells exhibit decreased mRNA levels for additional genes contributing to telomere metabolism, but also, surprisingly, for 12 genes mutated in Fanconi anaemia. Furthermore, p53(Δ31/Δ31) fibroblasts exhibit a reduced capacity to repair DNA interstrand crosslinks, a typical feature of Fanconi anaemia cells. Importantly, the p53-dependent downregulation of Fanc genes is largely conserved in human cells. Defective DNA repair is known to activate p53, but our results indicate that, conversely, an increased p53 activity may attenuate the Fanconi anaemia DNA repair pathway, defining a positive regulatory feedback loop.


Assuntos
Reparo do DNA , Regulação para Baixo , Anemia de Fanconi/genética , Proteína Supressora de Tumor p53/fisiologia , Animais , Células Cultivadas , Fator de Transcrição E2F4/genética , Fator de Transcrição E2F4/metabolismo , Fator de Transcrição E2F4/fisiologia , 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 D2 da Anemia de Fanconi/fisiologia , Humanos , Camundongos , Células NIH 3T3 , Transcriptoma
11.
Nucleic Acids Res ; 43(17): 8325-39, 2015 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-26187992

RESUMO

REV1 is a eukaryotic member of the Y-family of DNA polymerases involved in translesion DNA synthesis and genome mutagenesis. Recently, REV1 is also found to function in homologous recombination. However, it remains unclear how REV1 is recruited to the sites where homologous recombination is processed. Here, we report that loss of mammalian REV1 results in a specific defect in replication-associated gene conversion. We found that REV1 is targeted to laser-induced DNA damage stripes in a manner dependent on its ubiquitin-binding motifs, on RAD18, and on monoubiquitinated FANCD2 (FANCD2-mUb) that associates with REV1. Expression of a FANCD2-Ub chimeric protein in RAD18-depleted cells enhances REV1 assembly at laser-damaged sites, suggesting that FANCD2-mUb functions downstream of RAD18 to recruit REV1 to DNA breaks. Consistent with this suggestion we found that REV1 and FANCD2 are epistatic with respect to sensitivity to the double-strand break-inducer camptothecin. REV1 enrichment at DNA damage stripes also partially depends on BRCA1 and BRCA2, components of the FANCD2/BRCA supercomplex. Intriguingly, analogous to FANCD2-mUb and BRCA1/BRCA2, REV1 plays an unexpected role in protecting nascent replication tracts from degradation by stabilizing RAD51 filaments. Collectively these data suggest that REV1 plays multiple roles at stalled replication forks in response to replication stress.


Assuntos
Dano ao DNA , Replicação do DNA , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Proteínas Nucleares/fisiologia , Nucleotidiltransferases/fisiologia , Camptotecina/toxicidade , Linhagem Celular , DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , DNA Polimerase Dirigida por DNA , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Conversão Gênica , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Nucleotidiltransferases/química , Nucleotidiltransferases/metabolismo , Domínios e Motivos de Interação entre Proteínas , Estresse Fisiológico/genética , Inibidores da Topoisomerase I/toxicidade , Ubiquitina-Proteína Ligases
12.
Clin Cancer Res ; 20(14): 3884-95, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24787670

RESUMO

PURPOSE: Alveolar rhabdomyosarcoma that harbors the PAX3-FOXO1 fusion gene (t-ARMS) is a common and lethal subtype of this childhood malignancy. Improvement in clinical outcomes in this disease is predicated upon the identification of novel therapeutic targets. EXPERIMENTAL DESIGN: Robust mouse models were used for in vivo analysis, and molecular studies were performed on xenografts treated in parallel. Two independent patient sets (n = 101 and 124) of clinically annotated tumor specimens were used for analysis of FANCD2 levels and its association with clinical and molecular characteristics and outcomes. RESULTS: Our xenograft studies reveal a selective suppression of FANCD2 by m-TOR kinase inhibition and radiosensitization of the t-ARMS line only. In the initial patient set, we show that FANCD2 transcript levels are prognostic in univariate analysis, and are significantly associated with metastatic disease and that the copresence of the translocation and high expression of FANCD2 is independently prognostic. We also demonstrate a significant and nonrandom enrichment of mTOR-associated genes that correlate with FANCD2 gene expression within the t-ARMS samples, but not within other cases. In the second patient set, we show that on a protein level, FANCD2 expression correlates with PAX3-FOXO1 fusion gene and is strongly associated with phospho-P70S6K expression in cases with the fusion gene. CONCLUSIONS: Our data demonstrate that FANCD2 may have a significant role in the radiation resistance and virulence of t-ARMS. Indirectly targeting this DNA repair protein, through mTOR inhibition, may represent a novel and selective treatment strategy.


Assuntos
Biomarcadores Tumorais/metabolismo , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Proteínas de Fusão Oncogênica/metabolismo , Fatores de Transcrição Box Pareados/metabolismo , Rabdomiossarcoma Alveolar/metabolismo , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Quimiorradioterapia , Feminino , Humanos , Camundongos SCID , Morfolinas/farmacologia , Tolerância a Radiação , Rabdomiossarcoma Alveolar/tratamento farmacológico , Rabdomiossarcoma Alveolar/radioterapia , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Pediatr Blood Cancer ; 61(4): 740-2, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24115584

RESUMO

Fanconi anemia (FA) patients suffer from progressive bone marrow failure and often develop cancers. Previous studies showed that antioxidants tempol and resveratrol (RV) delayed tumor onset and reduced hematologic defects in FA murine models, respectively. Here we tested whether antioxidants N-acetylcysteine (NAC) or RV could delay cancer in tumor prone Fancd2(-/-) /Trp53(+/-) mice. Unlike tempol, neither compound had any significant chemopreventive effect in this model. We conclude that not all anti-oxidants are chemopreventive in FA. In addition, when given to Fancd2(-/-) mice, NAC helped maintain Fancd2(-/-) KSL cells in quiescence while tempol did not. The mechanisms behind the different actions of these antioxidants await further investigation.


Assuntos
Acetilcisteína/uso terapêutico , Antioxidantes/uso terapêutico , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Anemia de Fanconi/prevenção & controle , Sequestradores de Radicais Livres/uso terapêutico , Estilbenos/uso terapêutico , Proteína Supressora de Tumor p53/fisiologia , Animais , Anemia de Fanconi/genética , Anemia de Fanconi/patologia , Citometria de Fluxo , Camundongos , Camundongos Knockout , Resveratrol
14.
Immunity ; 39(5): 874-84, 2013 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-24184056

RESUMO

Severe sepsis remains a poorly understood systemic inflammatory condition with high mortality rates and limited therapeutic options in addition to organ support measures. Here we show that the clinically approved group of anthracyclines acts therapeutically at a low dose regimen to confer robust protection against severe sepsis in mice. This salutary effect is strictly dependent on the activation of DNA damage response and autophagy pathways in the lung, as demonstrated by deletion of the ataxia telangiectasia mutated (Atm) or the autophagy-related protein 7 (Atg7) specifically in this organ. The protective effect of anthracyclines occurs irrespectively of pathogen burden, conferring disease tolerance to severe sepsis. These findings demonstrate that DNA damage responses, including the ATM and Fanconi Anemia pathways, are important modulators of immune responses and might be exploited to confer protection to inflammation-driven conditions, including severe sepsis.


Assuntos
Antraciclinas/farmacologia , Antibacterianos/farmacologia , Reparo do DNA/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Peritonite/tratamento farmacológico , Sepse/prevenção & controle , Infecções por Adenoviridae/imunologia , Animais , Antraciclinas/uso terapêutico , Antibacterianos/uso terapêutico , Proteínas Mutadas de Ataxia Telangiectasia/deficiência , Proteínas Mutadas de Ataxia Telangiectasia/fisiologia , Proteína 7 Relacionada à Autofagia , Ceco/lesões , Dano ao DNA , Epirubicina/administração & dosagem , Epirubicina/farmacologia , Epirubicina/uso terapêutico , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Inflamação , Mediadores da Inflamação/análise , Injeções Intraperitoneais , Pulmão/metabolismo , Meropeném , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Associadas aos Microtúbulos/fisiologia , Especificidade de Órgãos , Peritonite/etiologia , Peritonite/genética , Peritonite/imunologia , Peritonite/fisiopatologia , Infecções Respiratórias/imunologia , Choque Séptico/prevenção & controle , Tienamicinas/uso terapêutico , Irradiação Corporal Total
15.
Mol Cell ; 50(6): 908-18, 2013 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-23806336

RESUMO

Fanconi anemia (FA) is a rare genetic disorder characterized by an increased susceptibility to squamous cell cancers. Fifteen FA genes are known, and the encoded proteins cooperate in a common DNA repair pathway. A critical step is the monoubiquitination of the FANCD2 protein, and cells from most FA patients are deficient in this step. How monoubiquitinated FANCD2 suppresses squamous cell cancers is unknown. Here we show that Fancd2-deficient mice are prone to Ras-oncogene-driven skin carcinogenesis, while Usp1-deficient mice, expressing elevated cellular levels of Fancd2-Ub, are resistant to skin tumors. Moreover, Fancd2-Ub activates the transcription of the tumor suppressor TAp63, thereby promoting cellular senescence and blocking skin tumorigenesis. For FA patients, the reduction of FANCD2-Ub and TAp63 protein levels may account for their susceptibility to squamous cell neoplasia. Taken together, Usp1 inhibition may be a useful strategy for upregulating TAp63 and preventing or treating squamous cell cancers in the general non-FA population.


Assuntos
Transformação Celular Neoplásica/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Genes Supressores de Tumor , Fosfoproteínas/genética , Transativadores/genética , Ativação Transcricional , Animais , Proteínas de Arabidopsis , Proliferação de Células , Células Cultivadas , Senescência Celular , Dano ao DNA , Resistência à Doença/genética , Endopeptidases/deficiência , Endopeptidases/genética , Anemia de Fanconi/genética , Feminino , Genes ras , Predisposição Genética para Doença , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias de Células Escamosas/induzido quimicamente , Neoplasias de Células Escamosas/genética , Neoplasias de Células Escamosas/patologia , Fosfoproteínas/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Transativadores/metabolismo , Proteases Específicas de Ubiquitina , Ubiquitinação
16.
Leukemia ; 27(10): 2040-2046, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23538752

RESUMO

Hematopoietic stem/progenitor cells (HSPCs) function to give rise to mature blood cells. Effective DNA damage response (DDR) and maintenance of genomic stability are crucial for normal functioning of HSPCs. Mammalian target of rapamycin (mTOR) integrates signals from nutrients and growth factors to control protein synthesis, cell growth, survival and metabolism, and has been shown to regulate DDR in yeast and human cancer cells through the p53/p21 signaling cascade. Here, we show that gene targeting of mTOR in HSPCs causes a defective DDR due to a variety of DNA damage agents, mimicking that caused by deficient FANCD2, a key component of the Fanconi anemia (FA) DDR machinery. Mechanistically, mTOR(-/-) HSPCs express drastically reduced FANCD2. Consistent with these genetic findings, inactivation of mTOR in human lymphoblast cells by pp242 or Torin 1, mTOR kinase inhibitors, suppresses FANCD2 expression and causes a defective DDR that can be rescued by reconstitution of exogenous FANCD2. Further mechanistic studies show that mTOR deficiency or inactivation increases phosphorylation and nuclear translocation of nuclear factor (NF)-κB, which results in an enhanced NF-κB binding to FANCD2 promoter to suppress FANCD2 expression. Thus, mTOR regulates DDR and genomic stability in hematopoietic cells through a noncanonical pathway involving NF-κB-mediated FANCD2 expression.


Assuntos
Dano ao DNA , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Células-Tronco Hematopoéticas/patologia , Linfócitos/patologia , NF-kappa B/metabolismo , Serina-Treonina Quinases TOR/fisiologia , Animais , Apoptose , Western Blotting , Células Cultivadas , Quebra Cromossômica , Ensaio Cometa , Ensaio de Desvio de Mobilidade Eletroforética , Embrião de Mamíferos/citologia , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Imunofluorescência , Instabilidade Genômica , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Camundongos , Camundongos Knockout , NF-kappa B/genética , Naftiridinas/farmacologia , Fragmentos de Peptídeos/farmacologia , Fosforilação , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
18.
Leukemia ; 25(8): 1259-67, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21519342

RESUMO

Fanconi D2 (FANCD2) is monoubiquitinated on K561 (FANCD2-Ub) in response to DNA double-strand breaks (DSBs) to stimulate repair of these potentially lethal DNA lesions. FANCD2-Ub was upregulated in CD34+ chronic myeloid leukemia (CML) cells and in BCR-ABL1 kinase-positive cell lines in response to elevated levels of reactive oxygen species (ROS) and DNA cross-linking agent mitomycin C. Downregulation of FANCD2 and inhibition of FANCD2-Ub reduced the clonogenic potential of CD34+ CML cells and delayed BCR-ABL1 leukemogenesis in mice. Retarded proliferation of BCR-ABL1 positive FANCD2-/- leukemia cells could be rescued by FANCD2 expression. BCR-ABL1 positive FANCD2-/- cells accumulated more ROS-induced DSBs in comparison with BCR-ABL1 positive FANCD2+/+ cells. Antioxidants diminished the number of DSBs and enhanced proliferation of BCR-ABL1 positive FANCD2-/- cells. Expression of wild-type FANCD2 and FANCD2(S222A) phosphorylation-defective mutant (deficient in stimulation of intra-S phase checkpoint, but proficient in DSB repair), but not FANCD2(K561R) monoubiquitination-defective mutant (proficient in stimulation of intra-S phase checkpoint, but deficient in DSB repair) reduced the number of DSBs and facilitated proliferation of BCR-ABL1 positive FANCD2-/- cells. We hypothesize that FANCD2-Ub has an important role in BCR-ABL1 leukemogenesis because of its ability to facilitate the repair of numerous ROS-induced DSBs.


Assuntos
Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Proteínas de Fusão bcr-abl/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/etiologia , Ubiquitinação , Animais , Linhagem Celular , Quebras de DNA de Cadeia Dupla , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Mitomicina/farmacologia , Espécies Reativas de Oxigênio/metabolismo
19.
Biochem Biophys Res Commun ; 404(1): 206-10, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21111709

RESUMO

The role of the Fanconi anemia (FA) repair pathway for DNA damage induced by formaldehyde was examined in the work described here. The following cell types were used: mouse embryonic fibroblast cell lines FANCA(-/-), FANCC(-/-), FANCA(-/-)C(-/-), FANCD2(-/-) and their parental cells, the Chinese hamster cell lines FANCD1 mutant (mt), FANCGmt, their revertant cells, and the corresponding wild-type (wt) cells. Cell survival rates were determined with colony formation assays after formaldehyde treatment. DNA double strand breaks (DSBs) were detected with an immunocytochemical γH2AX-staining assay. Although the sensitivity of FANCA(-/-), FANCC(-/-) and FANCA(-/-)C(-/-) cells to formaldehyde was comparable to that of proficient cells, FANCD1mt, FANCGmt and FANCD2(-/-) cells were more sensitive to formaldehyde than the corresponding proficient cells. It was found that homologous recombination (HR) repair was induced by formaldehyde. In addition, γH2AX foci in FANCD1mt cells persisted for longer times than in FANCD1wt cells. These findings suggest that formaldehyde-induced DSBs are repaired by HR through the FA repair pathway which is independent of the FA nuclear core complex.


Assuntos
Dano ao DNA , Reparo do DNA/genética , DNA Recombinante , Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Animais , Proteína BRCA2/fisiologia , Células CHO , Cricetinae , Cricetulus , Proteína do Grupo de Complementação A da Anemia de Fanconi/fisiologia , Proteína do Grupo de Complementação C da Anemia de Fanconi/fisiologia , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Formaldeído/toxicidade , Histonas/metabolismo , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...