Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
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.
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
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.
Proc Natl Acad Sci U S A ; 116(7): 2561-2570, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30692263

RESUMO

Fanconi anemia (FA) is a disease of DNA repair characterized by bone marrow failure and a reduced ability to remove DNA interstrand cross-links. Here, we provide evidence that the FA protein FANCI also functions in ribosome biogenesis, the process of making ribosomes that initiates in the nucleolus. We show that FANCI localizes to the nucleolus and is functionally and physically tied to the transcription of pre-ribosomal RNA (pre-rRNA) and to large ribosomal subunit (LSU) pre-rRNA processing independent of FANCD2. While FANCI is known to be monoubiquitinated when activated for DNA repair, we find that it is predominantly in the deubiquitinated state in the nucleolus, requiring the nucleoplasmic deubiquitinase (DUB) USP1 and the nucleolar DUB USP36. Our model suggests a possible dual pathophysiology for FA that includes defects in DNA repair and in ribosome biogenesis.


Assuntos
Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Ribossomos/metabolismo , Western Blotting , Nucléolo Celular/metabolismo , Reparo do DNA/fisiologia , Eletroforese em Gel de Poliacrilamida , Anemia de Fanconi/fisiopatologia , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Células HEK293 , Células HeLa , Humanos , Mutação , Biossíntese de Proteínas , Precursores de RNA/genética , RNA Ribossômico/genética , Transcrição Gênica , Ubiquitinação
5.
J Cell Sci ; 130(16): 2657-2662, 2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28811338

RESUMO

Fanconi anemia (FA) is a rare disease, in which homozygous or compound heterozygous inactivating mutations in any of 21 genes lead to genomic instability, early-onset bone marrow failure and increased cancer risk. The FA pathway is essential for DNA damage response (DDR) to DNA interstrand crosslinks. However, proteins of the FA pathway have additional cytoprotective functions that may be independent of DDR. We have shown that many FA proteins participate in the selective autophagy pathway that is required for the destruction of unwanted intracellular constituents. In this Cell Science at a Glance and the accompanying poster, we briefly review the role of the FA pathway in DDR and recent findings that link proteins of the FA pathway to selective autophagy of viruses and mitochondria. Finally, we discuss how perturbations in FA protein-mediated selective autophagy may contribute to inflammatory as well as genotoxic stress.


Assuntos
Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Animais , Autofagia/genética , Reparo do DNA/genética , Instabilidade Genômica/genética , Humanos , Mutação/fisiologia , Transdução de Sinais/fisiologia
6.
Biochim Biophys Acta Gen Subj ; 1861(7): 1813-1824, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28347842

RESUMO

BACKGROUND: The degradation of heme significantly contributes to cytoprotective effects against oxidative stress and inflammation. The enzyme heme oxygenase-1 (HO-1), involved in the degradation of heme, forms carbon monoxide (CO), ferrous iron, and bilirubin in conjunction with biliverdin reductase, and is induced by various stimuli including oxidative stress and heavy metals. We examined the involvement of heme metabolism in the induction of HO-1 by the inducers sulforaphane and sodium arsenite. METHODS: We examined the expression of HO-1 in sulforaphane-, sodium arsenite- and CORM3-treated HEK293T cells, by measuring the transcriptional activity and levels of mRNA and protein. RESULTS: The blockade of heme biosynthesis by succinylacetone and N-methyl protoporphyrin, which are inhibitors of heme biosynthesis, markedly decreased the induction of HO-1. The knockdown of the first enzyme in the biosynthesis of heme, 5-aminolevulinic acid synthase, also decreased the induction of HO-1. The cessation of HO-1 induction occurred at the transcriptional and translational levels, and was mediated by the activation of the heme-binding transcriptional repressor Bach1 and translational factor HRI. CO appeared to improve the expression of HO-1 at the transcriptional and translational levels. CONCLUSIONS: We demonstrated the importance of heme metabolism in the stress-inducible expression of HO-1, and also that heme and its degradation products are protective factors for self-defense responses. GENERAL SIGNIFICANCE: The key role of heme metabolism in the stress-inducible expression of HO-1 may promote further studies on heme and its degradation products as protective factors of cellular stresses and iron homeostasis in specialized cells, organs, and whole animal systems.


Assuntos
Heme Oxigenase-1/genética , Heme/metabolismo , Arsenitos/farmacologia , Fatores de Transcrição de Zíper de Leucina Básica/fisiologia , Monóxido de Carbono/fisiologia , Indução Enzimática , Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Células HEK293 , Células HeLa , Heme Oxigenase-1/biossíntese , Heptanoatos/farmacologia , Humanos , Isotiocianatos/farmacologia , Protoporfirinas/farmacologia , Compostos de Sódio/farmacologia , Sulfóxidos
7.
Toxicology ; 363-364: 48-57, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27393035

RESUMO

Our previous studies demonstrated that tetrachlorobenzoquinone (TCBQ), an active metabolite of pentachlorophenol, has effects on the generation of reactive oxygen species (ROS) and oxidative stress in vitro and in vivo. Nuclear factor erythroid-derived 2-like 2 (Nrf2) is a cellular sensor of electrophilic or oxidative stress that regulates the expression of antioxidant enzymes and defensive proteins. We have illustrated that TCBQ activates Nrf2 signaling by promoting the formation of the Kelch-like ECH-associated protein 1 (Keap1) cross-linking dimer and the formation of an ubiquitination switch from Nrf2 to Keap1. The activation of Nrf2 by TCBQ may serve as an adaptive response to a TCBQ-induced oxidative insult. BTB and CNC homolog 1 (Bach1) compete with Nrf2, leading to the negative regulation of the antioxidant response element (ARE). In this report, we propose that TCBQ induces the dynamic inactivation of Bach1. We observed a rapid nuclear efflux of Bach1 and an accumulation of Nrf2 in nuclei upon TCBQ treatment that precedes the binding of Nrf2 with ARE. We found that the nuclear export of Bach1 is dependent on its chromosomal region maintenance 1 (Crm1) interaction and tyrosine phosphorylation. Although TCBQ induces the ubiquitination of Bach1, TCBQ also increases the mRNA and protein levels of Bach1, returning Bach1 to normal levels. Moreover, we found that TCBQ-induced activation of Nrf2 involves c-Jun N-terminal kinase (JNK)-P62 signaling.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/fisiologia , Benzoquinonas/farmacologia , Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Hidrocarbonetos Clorados/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/fisiologia , Fatores de Transcrição de Zíper de Leucina Básica/efeitos dos fármacos , Western Blotting , Cicloeximida/farmacologia , Proteínas de Grupos de Complementação da Anemia de Fanconi/efeitos dos fármacos , Células Hep G2 , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Fator 2 Relacionado a NF-E2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/fisiologia , Reação em Cadeia da Polimerase em Tempo Real , Ubiquitinação
8.
Nat Rev Mol Cell Biol ; 17(6): 337-49, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27145721

RESUMO

The Fanconi anaemia pathway repairs DNA interstrand crosslinks (ICLs) in the genome. Our understanding of this complex pathway is still evolving, as new components continue to be identified and new biochemical systems are used to elucidate the molecular steps of repair. The Fanconi anaemia pathway uses components of other known DNA repair processes to achieve proper repair of ICLs. Moreover, Fanconi anaemia proteins have functions in genome maintenance beyond their canonical roles of repairing ICLs. Such functions include the stabilization of replication forks and the regulation of cytokinesis. Thus, Fanconi anaemia proteins are emerging as master regulators of genomic integrity that coordinate several repair processes. Here, we summarize our current understanding of the functions of the Fanconi anaemia pathway in ICL repair, together with an overview of its connections with other repair pathways and its emerging roles in genome maintenance.


Assuntos
Reparo do DNA , Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Anemia de Fanconi/genética , Animais , Dano ao DNA , Replicação do DNA , Humanos
9.
J Clin Invest ; 125(4): 1523-32, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25751062

RESUMO

The Fanconi anemia/BRCA (FA/BRCA) pathway is a DNA repair pathway that is required for excision of DNA interstrand cross-links. The 17 known FA proteins, along with several FA-associated proteins (FAAPs), cooperate in this pathway to detect, unhook, and excise DNA cross-links and to subsequently repair the double-strand breaks generated in the process. In the current study, we identified a patient with FA with a point mutation in FANCA, which encodes a mutant FANCA protein (FANCAI939S). FANCAI939S failed to bind to the FAAP20 subunit of the FA core complex, leading to decreased stability. Loss of FAAP20 binding exposed a SUMOylation site on FANCA at amino acid residue K921, resulting in E2 SUMO-conjugating enzyme UBC9-mediated SUMOylation, RING finger protein 4-mediated (RNF4-mediated) polyubiquitination, and proteasome-mediated degradation of FANCA. Mutation of the SUMOylation site of FANCA rescued the expression of the mutant protein. Wild-type FANCA was also subject to SUMOylation, RNF4-mediated polyubiquitination, and degradation, suggesting that regulated release of FAAP20 from FANCA is a critical step in the normal FA pathway. Consistent with this model, cells lacking RNF4 exhibited interstrand cross-linker hypersensitivity, and the gene encoding RNF4 was epistatic with the other genes encoding members of the FA/BRCA pathway. Together, the results from our study underscore the importance of analyzing unique patient-derived mutations for dissecting complex DNA repair processes.


Assuntos
Proteína BRCA1/fisiologia , Reparo do DNA/fisiologia , Proteína do Grupo de Complementação A da Anemia de Fanconi/fisiologia , Anemia de Fanconi/genética , Proteínas Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Adulto , Linhagem Celular Tumoral , Reparo do DNA/genética , Proteína do Grupo de Complementação A da Anemia de Fanconi/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/antagonistas & inibidores , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Feminino , Genes BRCA1 , Humanos , Mutação de Sentido Incorreto , Linhagem , Fenótipo , Mutação Puntual , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Processamento de Proteína Pós-Traducional/fisiologia , Proteólise , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/fisiologia , Sumoilação , Neoplasias de Mama Triplo Negativas/genética , Ubiquitinação/fisiologia
10.
Nucleic Acids Res ; 43(2): 893-903, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25520194

RESUMO

Fanconi anemia (FA) patients exhibit bone marrow failure, developmental defects and cancer. The FA pathway maintains chromosomal stability in concert with replication fork maintenance and DNA double strand break (DSB) repair pathways including RAD51-mediated homologous recombination (HR). RAD51 is a recombinase that maintains replication forks and repairs DSBs, but also rearranges chromosomes. Two RecQ helicases, RECQL5 and Bloom syndrome mutated (BLM) suppress HR through nonredundant mechanisms. Here we test the impact deletion of RECQL5 and BLM has on mouse embryonic stem (ES) cells deleted for FANCB, a member of the FA core complex. We show that RECQL5, but not BLM, conferred resistance to mitomycin C (MMC, an interstrand crosslinker) and camptothecin (CPT, a type 1 topoisomerase inhibitor) in FANCB-defective cells. RECQL5 suppressed, while BLM caused, breaks and radials in FANCB-deleted cells exposed to CPT or MMC, respectively. RECQL5 protected the nascent replication strand from MRE11-mediated degradation and restarted stressed replication forks in a manner additive to FANCB. By contrast BLM restarted, but did not protect, replication forks in a manner epistatic to FANCB. RECQL5 also lowered RAD51 levels in FANCB-deleted cells at stressed replication sites implicating a rearrangement avoidance mechanism. Thus, RECQL5 and BLM impact FANCB-defective cells differently in response to replication stress with relevance to chemotherapeutic regimes.


Assuntos
Reparo do DNA , Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , RecQ Helicases/fisiologia , Animais , Células Cultivadas , Quebras de DNA de Cadeia Dupla , Replicação do DNA , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Deleção de Genes , Camundongos , RecQ Helicases/genética
11.
Nucleic Acids Res ; 42(14): 9087-95, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25038251

RESUMO

Genetic recombination is important for generating diversity and to ensure faithful segregation of chromosomes at meiosis. However, few crossovers (COs) are formed per meiosis despite an excess of DNA double-strand break precursors. This reflects the existence of active mechanisms that limit CO formation. We previously showed that AtFANCM is a meiotic anti-CO factor. The same genetic screen now identified AtMHF2 as another player of the same anti-CO pathway. FANCM and MHF2 are both Fanconi Anemia (FA) associated proteins, prompting us to test the other FA genes conserved in Arabidopsis for a role in CO control at meiosis. This revealed that among the FA proteins tested, only FANCM and its two DNA-binding co-factors MHF1 and MHF2 limit CO formation at meiosis.


Assuntos
Proteínas de Arabidopsis/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Meiose/genética , Arabidopsis/genética , Proteínas de Arabidopsis/genética , DNA Helicases/genética , Proteínas de Ligação a DNA/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Mutação , Recombinação Genética
12.
Blood ; 123(1): 26-34, 2014 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-24200684

RESUMO

The inherited bone marrow failure (BMF) syndromes are a rare and diverse group of genetic disorders that ultimately result in the loss of blood production. The molecular defects underlying many of these conditions have been elucidated, and great progress has been made toward understanding the normal function of these gene products. This review will focus on perhaps the most well-known and genetically heterogeneous BMF syndrome: Fanconi anemia. More specifically, this account will review the current state of our knowledge on why the bone marrow fails in this illness and what this might tell us about the maintenance of bone marrow function and hematopoiesis.


Assuntos
Doenças da Medula Óssea/fisiopatologia , Medula Óssea/fisiopatologia , Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Anemia de Fanconi/fisiopatologia , Animais , Doenças da Medula Óssea/genética , Transplante de Medula Óssea/métodos , Reagentes de Ligações Cruzadas/química , Dano ao DNA , Reparo do DNA , Anemia de Fanconi/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Humanos , Camundongos , Células-Tronco/citologia
13.
Cold Spring Harb Perspect Biol ; 5(10): a012732, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24086043

RESUMO

DNA interstrand cross-links (ICLs) are lesions caused by a variety of endogenous metabolites, environmental exposures, and cancer chemotherapeutic agents that have two reactive groups. The common feature of these diverse lesions is that two nucleotides on opposite strands are covalently joined. ICLs prevent the separation of two DNA strands and therefore essential cellular processes including DNA replication and transcription. ICLs are mainly detected in S phase when a replication fork stalls at an ICL. Damage signaling and repair of ICLs are promoted by the Fanconi anemia pathway and numerous posttranslational modifications of DNA repair and chromatin structural proteins. ICLs are also detected and repaired in nonreplicating cells, although the mechanism is less clear. A unique feature of ICL repair is that both strands of DNA must be incised to completely remove the lesion. This is accomplished in sequential steps to prevent creating multiple double-strand breaks. Unhooking of an ICL from one strand is followed by translesion synthesis to fill the gap and create an intact duplex DNA, harboring a remnant of the ICL. Removal of the lesion from the second strand is likely accomplished by nucleotide excision repair. Inadequate repair of ICLs is particularly detrimental to rapidly dividing cells, explaining the bone marrow failure characteristic of Fanconi anemia and why cross-linking agents are efficacious in cancer therapy. Herein, recent advances in our understanding of ICLs and the biological responses they trigger are discussed.


Assuntos
Reparo do DNA/fisiologia , DNA/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Modelos Genéticos , Reagentes de Ligações Cruzadas/toxicidade , Dano ao DNA , Replicação do DNA , Proteínas de Ligação a DNA , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Recombinação Homóloga , Humanos , Recombinação Genética , Transdução de Sinais
14.
J Biol Chem ; 287(40): 33533-44, 2012 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-22875853

RESUMO

The application of functional genomic analysis of breast cancer metastasis has led to the identification of a growing number of organ-specific metastasis genes, which often function in concert to facilitate different steps of the metastatic cascade. However, the gene regulatory network that controls the expression of these metastasis genes remains largely unknown. Here, we demonstrate a computational approach for the deconvolution of transcriptional networks to discover master regulators of breast cancer bone metastasis. Several known regulators of breast cancer bone metastasis such as Smad4 and HIF1 were identified in our analysis. Experimental validation of the networks revealed BACH1, a basic leucine zipper transcription factor, as the common regulator of several functional metastasis genes, including MMP1 and CXCR4. Ectopic expression of BACH1 enhanced the malignance of breast cancer cells, and conversely, BACH1 knockdown significantly reduced bone metastasis. The expression of BACH1 and its target genes was linked to the higher risk of breast cancer recurrence in patients. This study established BACH1 as the master regulator of breast cancer bone metastasis and provided a paradigm to identify molecular determinants in complex pathological processes.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/fisiologia , Neoplasias Ósseas/secundário , Neoplasias da Mama/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Regulação Neoplásica da Expressão Gênica , Animais , Fatores de Transcrição de Zíper de Leucina Básica/genética , Osso e Ossos/patologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Feminino , Redes Reguladoras de Genes , Humanos , Hipóxia , Neoplasias Mamárias Animais/metabolismo , Metaloproteinase 1 da Matriz/metabolismo , Camundongos , Camundongos Nus , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias/patologia , Regiões Promotoras Genéticas , Proteínas Smad/metabolismo , Transcrição Gênica
15.
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
16.
Cancer Sci ; 103(5): 897-903, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22348305

RESUMO

Cellular senescence prevents the aberrant proliferation of damaged cells. The transcription factor Bach1 binds to p53 to repress cellular senescence, but it is still unclear how the Bach1-p53 interaction is regulated. We found that the Bach1-p53 interaction was inhibited by oncogenic Ras, bleomycin, and hydrogen peroxide. Proteomics analysis of Bach1 complex revealed its interaction with p19(ARF), a tumor suppressor that competitively inhibited the Bach1-p53 interaction when overexpressed within cells. Reduction of MDM2 expression in wild-type murine embryonic fibroblasts (MEFs) did not result in slower proliferation, showing that Bach1 plays a role in keeping the proliferation of MEFs independent of MDM2. Consistent with this interpretation, expression of p21 was highly induced in MEFs when both Bach1 and MDM2 were abrogated. The level of Bach1 protein was reduced on knockdown of p53. These results suggest that p53 activation involves its dissociation from Bach1, achieved in part by the competitive binding of p19(ARF) to Bach1. The p19(ARF)-Bach1 interaction constitutes a regulatory pathway of p53 in parallel with the p19(ARF)-MDM2 pathway.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/fisiologia , Senescência Celular , Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Proteína Supressora de Tumor p14ARF/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Ligação Competitiva , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Fibroblastos/citologia , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Genes Supressores de Tumor , Humanos , Camundongos , Proteína Supressora de Tumor p53/genética
17.
Biochem J ; 441(3): 919-26, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22032289

RESUMO

BACH1 (BRCA1-associated C-terminal helicase 1), the product of the BRIP1 {BRCA1 [breast cancer 1, early onset]-interacting protein C-terminal helicase 1; also known as FANCJ [FA-J (Fanconi anaemia group J) protein]} gene mutated in Fanconi anaemia patients from complementation group J, has been implicated in DNA repair and damage signalling. BACH1 exerts DNA helicase activities and physically interacts with BRCA1 and MLH1 (mutL homologue 1), which differentially control DNA DSB (double-strand break) repair processes. The present study shows that BACH1 plays a role in both HR (homologous recombination) and MMEJ (microhomology-mediated non-homologous end-joining) and reveals discrete mechanisms underlying modulation of these pathways. Our results indicate that BACH1 stimulates HR, which depends on the integrity of the helicase domain. Disruption of the BRCA1-BACH1 complex through mutation of BACH1 compromised errorfree NHEJ (non-homologous end-joining) and accelerated error-prone MMEJ. Conversely, molecular changes in BACH1 abrogating MLH1 binding interfered neither with HR nor with MMEJ. Importantly, MMEJ is a mutagenic DSB repair pathway, which is derepressed in hereditary breast and ovarian carcinomas. Since BRCA1 and BACH1 mutations targeting the BRCA1-BACH1 interaction have been associated with breast cancer susceptibility, the results of the present study thus provide evidence for a novel role of BACH1 in tumour suppression.


Assuntos
Proteína BRCA1/fisiologia , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Sequência de Aminoácidos , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Sequência de Bases , Fatores de Transcrição de Zíper de Leucina Básica/fisiologia , Células Cultivadas , Reparo do DNA por Junção de Extremidades/genética , Regulação para Baixo/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Feminino , Predisposição Genética para Doença , Células HeLa , Humanos , Células K562 , Modelos Biológicos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Mutação/fisiologia , Ligação Proteica/genética , Ligação Proteica/fisiologia
18.
Nucleic Acids Res ; 40(4): 1485-98, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22021381

RESUMO

We have previously reported that DT40 cells deficient in the Y-family polymerase REV1 are defective in replicating G-quadruplex DNA. In vivo this leads to uncoupling of DNA synthesis from redeposition of histones displaced ahead of the replication fork, which in turn leads to loss of transcriptional repression due to failure to recycle pre-existing repressive histone post-translational modifications. Here we report that a similar process can also affect transcriptionally active genes, leading to their deactivation. We use this finding to develop an assay based on loss of expression of a cell surface marker to monitor epigenetic instability at the level of single cells. This assay allows us to demonstrate G4 DNA motif-associated epigenetic instability in mutants of three helicases previously implicated in the unwinding of G-quadruplex structures, FANCJ, WRN and BLM. Transcriptional profiling of DT40 mutants reveals that FANCJ coordinates two independent mechanisms to maintain epigenetic stability near G4 DNA motifs that are dependent on either REV1 or on the WRN and BLM helicases, suggesting a model in which efficient in vivo replication of G-quadruplexes often requires the established 5'-3'-helicase activity of FANCJ acting in concert with either a specialized polymerase or helicase operating in the opposite polarity.


Assuntos
DNA Helicases/metabolismo , DNA/química , Epigênese Genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Quadruplex G , Animais , Antígenos CD/genética , Linhagem Celular , Cromatina/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Humanos , Mutação , Proteínas Nucleares/genética , Motivos de Nucleotídeos , Nucleotidiltransferases/genética , RecQ Helicases/genética , Receptores de Superfície Celular/análise , Transcrição Gênica
19.
Chromosoma ; 121(1): 21-36, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22057367

RESUMO

Chromosomal aberrations are often associated with incomplete genome duplication, for instance at common fragile sites, or as a consequence of chemical alterations in the DNA template that block replication forks. Studies of the cancer-prone disease Fanconi anaemia (FA) have provided important insights into the resolution of replication problems. The repair of interstrand DNA crosslinks induced by chemotherapy drugs is coupled with DNA replication and controlled by FA proteins. We discuss here the recent discovery of new FA-associated proteins and the development of new tractable repair systems that have dramatically improved our understanding of crosslink repair. We focus also on how FA proteins protect against replication failure in the context of fragile sites and on the identification of reactive metabolites that account for the development of Fanconi anaemia symptoms.


Assuntos
Replicação do DNA/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/fisiologia , Animais , Dano ao DNA/genética , Dano ao DNA/fisiologia , Reparo do DNA/genética , Reparo do DNA/fisiologia , Replicação do DNA/fisiologia , Desoxirribonucleases/metabolismo , Desoxirribonucleases/fisiologia , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Humanos , Modelos Biológicos , Transdução de Sinais/genética
20.
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