Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nature ; 600(7890): 748-753, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34853474

RESUMO

Centromeric integrity is key for proper chromosome segregation during cell division1. Centromeres have unique chromatin features that are essential for centromere maintenance2. Although they are intrinsically fragile and represent hotspots for chromosomal rearrangements3, little is known about how centromere integrity in response to DNA damage is preserved. DNA repair by homologous recombination requires the presence of the sister chromatid and is suppressed in the G1 phase of the cell cycle4. Here we demonstrate that DNA breaks that occur at centromeres in G1 recruit the homologous recombination machinery, despite the absence of a sister chromatid. Mechanistically, we show that the centromere-specific histone H3 variant CENP-A and its chaperone HJURP, together with dimethylation of lysine 4 in histone 3 (H3K4me2), enable a succession of events leading to the licensing of homologous recombination in G1. H3K4me2 promotes DNA-end resection by allowing DNA damage-induced centromeric transcription and increased formation of DNA-RNA hybrids. CENP-A and HJURP interact with the deubiquitinase USP11, enabling formation of the RAD51-BRCA1-BRCA2 complex5 and rendering the centromeres accessible to RAD51 recruitment and homologous recombination in G1. Finally, we show that inhibition of homologous recombination in G1 leads to centromeric instability and chromosomal translocations. Our results support a model in which licensing of homologous recombination at centromeric breaks occurs throughout the cell cycle to prevent the activation of mutagenic DNA repair pathways and preserve centromeric integrity.


Assuntos
Proteínas Cromossômicas não Histona , Reparo do DNA , Proteínas de Ligação a DNA , Centrômero/genética , Centrômero/metabolismo , Proteína Centromérica A , Proteínas Cromossômicas não Histona/metabolismo , Segregação de Cromossomos , DNA , Proteínas de Ligação a DNA/metabolismo , Histonas/metabolismo , Recombinação Homóloga
2.
Nature ; 600(7888): 329-333, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34819671

RESUMO

Efficient humoral responses rely on DNA damage, mutagenesis and error-prone DNA repair. Diversification of B cell receptors through somatic hypermutation and class-switch recombination are initiated by cytidine deamination in DNA mediated by activation-induced cytidine deaminase (AID)1 and by the subsequent excision of the resulting uracils by uracil DNA glycosylase (UNG) and by mismatch repair proteins1-3. Although uracils arising in DNA are accurately repaired1-4, how these pathways are co-opted to generate mutations and double-strand DNA breaks in the context of somatic hypermutation and class-switch recombination is unknown1-3. Here we performed a genome-wide CRISPR-Cas9 knockout screen for genes involved in class-switch recombination and identified FAM72A, a protein that interacts with the nuclear isoform of UNG (UNG2)5 and is overexpressed in several cancers5. We show that the FAM72A-UNG2 interaction controls the levels of UNG2 and that class-switch recombination is defective in Fam72a-/- B cells due to the upregulation of UNG2. Moreover, we show that somatic hypermutation is reduced in Fam72a-/- B cells and that its pattern is skewed upon upregulation of UNG2. Our results are consistent with a model in which FAM72A interacts with UNG2 to control its physiological level by triggering its degradation, regulating the level of uracil excision and thus the balance between error-prone and error-free DNA repair. Our findings have potential implications for tumorigenesis, as reduced levels of UNG2 mediated by overexpression of Fam72a would shift the balance towards mutagenic DNA repair, rendering cells more prone to acquire mutations.


Assuntos
Linfócitos B , Reparo de Erro de Pareamento de DNA , Switching de Imunoglobulina , Região de Troca de Imunoglobulinas , Mutação , Hipermutação Somática de Imunoglobulina , Animais , Feminino , Masculino , Camundongos , Linfócitos B/metabolismo , Sistemas CRISPR-Cas/genética , Genoma/genética , Switching de Imunoglobulina/genética , Região de Troca de Imunoglobulinas/genética , Hipermutação Somática de Imunoglobulina/genética , Regulação para Cima , Uracila/metabolismo
3.
Mol Cell ; 81(12): 2596-2610.e7, 2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-33961796

RESUMO

p53-binding protein 1 (53BP1) regulates both the DNA damage response and p53 signaling. Although 53BP1's function is well established in DNA double-strand break repair, how its role in p53 signaling is modulated remains poorly understood. Here, we identify the scaffolding protein AHNAK as a G1 phase-enriched interactor of 53BP1. We demonstrate that AHNAK binds to the 53BP1 oligomerization domain and controls its multimerization potential. Loss of AHNAK results in hyper-accumulation of 53BP1 on chromatin and enhanced phase separation, culminating in an elevated p53 response, compromising cell survival in cancer cells but leading to senescence in non-transformed cells. Cancer transcriptome analyses indicate that AHNAK-53BP1 cooperation contributes to the suppression of p53 target gene networks in tumors and that loss of AHNAK sensitizes cells to combinatorial cancer treatments. These findings highlight AHNAK as a rheostat of 53BP1 function, which surveys cell proliferation by preventing an excessive p53 response.


Assuntos
Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Cromatina/metabolismo , DNA/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Fase G1/fisiologia , Histonas/metabolismo , Humanos , Células MCF-7 , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Transdução de Sinais/fisiologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/fisiologia
4.
Cell Death Differ ; 26(9): 1615-1630, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30442946

RESUMO

PARP3 has been shown to be a key driver of TGFß-induced epithelial-to-mesenchymal transition (EMT) and stemness in breast cancer cells, emerging as an attractive therapeutic target. Nevertheless, the therapeutic value of PARP3 inhibition has not yet been assessed. Here we investigated the impact of the absence of PARP3 or its inhibition on the tumorigenicity of BRCA1-proficient versus BRCA1-deficient breast cancer cell lines, focusing on the triple-negative breast cancer subtype (TNBC). We show that PARP3 knockdown exacerbates centrosome amplification and genome instability and reduces survival of BRCA1-deficient TNBC cells. Furthermore, we engineered PARP3-/- BRCA1-deficient or BRCA1-proficient TNBC cell lines using the CRISPR/nCas9D10A gene editing technology and demonstrate that the absence of PARP3 selectively suppresses the growth, survival and in vivo tumorigenicity of BRCA1-deficient TNBC cells, mechanistically via effects associated with an altered Rictor/mTORC2 signaling complex resulting from enhanced ubiquitination of Rictor. Accordingly, PARP3 interacts with and ADP-ribosylates GSK3ß, a positive regulator of Rictor ubiquitination and degradation. Importantly, these phenotypes were rescued by re-expression of a wild-type PARP3 but not by a catalytic mutant, demonstrating the importance of PARP3's catalytic activity. Accordingly, reduced survival and compromised Rictor/mTORC2 signaling were also observed using a cell-permeable PARP3-specific inhibitor. We conclude that PARP3 and BRCA1 are synthetic lethal and that targeting PARP3's catalytic activity is a promising therapeutic strategy for BRCA1-associated cancers via the Rictor/mTORC2 signaling pathway.


Assuntos
Proteína BRCA1/genética , Proteínas de Ciclo Celular/genética , Poli(ADP-Ribose) Polimerases/genética , Proteína Companheira de mTOR Insensível à Rapamicina/genética , Neoplasias de Mama Triplo Negativas/genética , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Transição Epitelial-Mesenquimal/genética , Feminino , Xenoenxertos , Humanos , Alvo Mecanístico do Complexo 2 de Rapamicina/genética , Camundongos , Transdução de Sinais , Fator de Crescimento Transformador beta/genética , Neoplasias de Mama Triplo Negativas/patologia
5.
Cell Rep ; 23(8): 2443-2454, 2018 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-29791854

RESUMO

Nup133 belongs to the Y-complex, a key component of the nuclear pore complex (NPC) scaffold. Studies on a null mutation in mice previously revealed that Nup133 is essential for embryonic development but not for mouse embryonic stem cell (mESC) proliferation. Using single-pore detection and average NE-fluorescence intensity, we find that Nup133 is dispensable for interphase and postmitotic NPC scaffold assembly in pluripotent mESCs. However, loss of Nup133 specifically perturbs the formation of the nuclear basket as manifested by the absence of Tpr in about half of the NPCs combined with altered dynamics of Nup153. We further demonstrate that its central domain mediates Nup133's role in assembling Tpr and Nup153 into a properly configured nuclear basket. Our findings thus revisit the role of the Y-complex in pore biogenesis and provide insights into the interplay between NPC scaffold architecture, nuclear basket assembly, and the generation of heterogeneity among NPCs.


Assuntos
Antígenos de Histocompatibilidade Menor/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Poro Nuclear/metabolismo , Animais , Feminino , Imageamento Tridimensional , Interfase , Masculino , Camundongos , Antígenos de Histocompatibilidade Menor/química , Complexo de Proteínas Formadoras de Poros Nucleares/química , Domínios Proteicos , Proteínas Proto-Oncogênicas/metabolismo
6.
Front Immunol ; 9: 373, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29599769

RESUMO

Systemic lupus erythematosus (SLE) is a severe and heterogeneous autoimmune disease with a complex genetic etiology, characterized by the production of various pathogenic autoantibodies, which participate in end-organ damages. The majority of human SLE occurs in adults as a polygenic disease, and clinical flares interspersed with silent phases of various lengths characterize the usual evolution of the disease in time. Trying to understand the mechanism of the different phenotypic traits of the disease, and considering the central role of B cells in SLE, we previously performed a detailed wide analysis of gene expression variation in B cells from quiescent SLE patients. This analysis pointed out an overexpression of TRIB1. TRIB1 is a pseudokinase that has been implicated in the development of leukemia and also metabolic disorders. It is hypothesized that Trib1 plays an adapter or scaffold function in signaling pathways, notably in MAPK pathways. Therefore, we planned to understand the functional significance of TRIB1 overexpression in B cells in SLE. We produced a new knock-in model with B-cell-specific overexpression of Trib1. We showed that overexpression of Trib1 specifically in B cells does not impact B cell development nor induce any development of SLE symptoms in the mice. By contrast, Trib1 has a negative regulatory function on the production of immunoglobulins, notably IgG1, but also on the production of autoantibodies in an induced model. We observed a decrease of Erk activation in BCR-stimulated Trib1 overexpressing B cells. Finally, we searched for Trib1 partners in B cells by proteomic analysis in order to explore the regulatory function of Trib1 in B cells. Interestingly, we find an interaction between Trib1 and CD72, a negative regulator of B cells whose deficiency in mice leads to the development of autoimmunity. In conclusion, the overexpression of Trib1 could be one of the molecular pathways implicated in the negative regulation of B cells during SLE.


Assuntos
Linfócitos B/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lúpus Eritematoso Sistêmico/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Adulto , Animais , Formação de Anticorpos/genética , Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos B/metabolismo , Autoanticorpos/metabolismo , Autoimunidade/genética , Células Cultivadas , Feminino , Humanos , Imunoglobulina G/biossíntese , Imunomodulação , Peptídeos e Proteínas de Sinalização Intracelular/genética , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transgenes/genética
7.
J Cell Biol ; 217(4): 1537-1552, 2018 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-29440513

RESUMO

Fluorescent labeling of endogenous proteins for live-cell imaging without exogenous expression of tagged proteins or genetic manipulations has not been routinely possible. We describe a simple versatile antibody-based imaging approach (VANIMA) for the precise localization and tracking of endogenous nuclear factors. Our protocol can be implemented in every laboratory allowing the efficient and nonharmful delivery of organic dye-conjugated antibodies, or antibody fragments, into different metazoan cell types. Live-cell imaging permits following the labeled probes bound to their endogenous targets. By using conventional and super-resolution imaging we show dynamic changes in the distribution of several nuclear transcription factors (i.e., RNA polymerase II or TAF10), and specific phosphorylated histones (γH2AX), upon distinct biological stimuli at the nanometer scale. Hence, considering the large panel of available antibodies and the simplicity of their implementation, VANIMA can be used to uncover novel biological information based on the dynamic behavior of transcription factors or posttranslational modifications in the nucleus of single live cells.


Assuntos
Núcleo Celular/metabolismo , Técnica Direta de Fluorescência para Anticorpo , Histonas/metabolismo , Microscopia Confocal , Análise de Célula Única/métodos , Fatores de Transcrição/metabolismo , Animais , Apoptose , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Núcleo Celular/patologia , Proliferação de Células , Fibroblastos/metabolismo , Humanos , Cinética , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Osteossarcoma/genética , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Fosforilação , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Fatores Associados à Proteína de Ligação a TATA/genética , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Proteína de Ligação a TATA-Box/genética , Proteína de Ligação a TATA-Box/metabolismo , Fator de Transcrição TFIID/genética , Fator de Transcrição TFIID/metabolismo , Fatores de Transcrição/genética
8.
Nat Commun ; 8(1): 113, 2017 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-28740167

RESUMO

DNA breaks are complex DNA lesions that can be repaired by two alternative mechanisms: non-homologous end-joining and homologous recombination. The decision between them depends on the activation of the DNA resection machinery, which blocks non-homologous end-joining and stimulates recombination. On the other hand, post-translational modifications play a critical role in DNA repair. We have found that the SUMO E3 ligase CBX4 controls resection through the key factor CtIP. Indeed, CBX4 depletion impairs CtIP constitutive sumoylation and DNA end processing. Importantly, mutating lysine 896 in CtIP recapitulates the CBX4-depletion phenotype, blocks homologous recombination and increases genomic instability. Artificial fusion of CtIP and SUMO suppresses the effects of both the non-sumoylatable CtIP mutant and CBX4 depletion. Mechanistically, CtIP sumoylation is essential for its recruitment to damaged DNA. In summary, sumoylation of CtIP at lysine 896 defines a subpopulation of the protein that is involved in DNA resection and recombination.The choice between non-homologous end-joining and homologous recombination to repair a DNA double-strand break depends on activation of the end resection machinery. Here the authors show that SUMO E3 ligase CBX4 sumoylates subpopulation of CtIP to regulate recruitment to breaks and resection.


Assuntos
Proteínas de Transporte/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Ligases/metabolismo , Proteínas Nucleares/metabolismo , Proteínas do Grupo Polycomb/metabolismo , Western Blotting , Proteínas de Transporte/genética , Linhagem Celular Tumoral , DNA/genética , DNA/metabolismo , Endodesoxirribonucleases , Células HEK293 , Recombinação Homóloga , Humanos , Ligases/genética , Microscopia Confocal , Proteínas Nucleares/genética , Proteínas do Grupo Polycomb/genética , Interferência de RNA , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação
9.
EMBO Mol Med ; 9(10): 1398-1414, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28729482

RESUMO

Maintenance of genome integrity requires the functional interplay between Fanconi anemia (FA) and homologous recombination (HR) repair pathways. Endogenous acetaldehyde, a product of cellular metabolism, is a potent source of DNA damage, particularly toxic to cells and mice lacking the FA protein FANCD2. Here, we investigate whether HR-compromised cells are sensitive to acetaldehyde, similarly to FANCD2-deficient cells. We demonstrate that inactivation of HR factors BRCA1, BRCA2, or RAD51 hypersensitizes cells to acetaldehyde treatment, in spite of the FA pathway being functional. Aldehyde dehydrogenases (ALDHs) play key roles in endogenous acetaldehyde detoxification, and their chemical inhibition leads to cellular acetaldehyde accumulation. We find that disulfiram (Antabuse), an ALDH2 inhibitor in widespread clinical use for the treatment of alcoholism, selectively eliminates BRCA1/2-deficient cells. Consistently, Aldh2 gene inactivation suppresses proliferation of HR-deficient mouse embryonic fibroblasts (MEFs) and human fibroblasts. Hypersensitivity of cells lacking BRCA2 to acetaldehyde stems from accumulation of toxic replication-associated DNA damage, leading to checkpoint activation, G2/M arrest, and cell death. Acetaldehyde-arrested replication forks require BRCA2 and FANCD2 for protection against MRE11-dependent degradation. Importantly, acetaldehyde specifically inhibits in vivo the growth of BRCA1/2-deficient tumors and ex vivo in patient-derived tumor xenograft cells (PDTCs), including those that are resistant to poly (ADP-ribose) polymerase (PARP) inhibitors. The work presented here therefore identifies acetaldehyde metabolism as a potential therapeutic target for the selective elimination of BRCA1/2-deficient cells and tumors.


Assuntos
Acetaldeído/metabolismo , Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Rad51 Recombinase/metabolismo , Aldeído-Desidrogenase Mitocondrial/genética , Aldeído-Desidrogenase Mitocondrial/metabolismo , Animais , Proteína BRCA1/genética , Proteína BRCA2/genética , Linhagem Celular Tumoral , Dano ao DNA , Anemia de Fanconi/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Fibroblastos , Recombinação Homóloga , Humanos , Camundongos , Camundongos Nus , Rad51 Recombinase/genética , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Eur J Immunol ; 47(4): 665-676, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28105679

RESUMO

To mount highly specific and adapted immune responses, B lymphocytes assemble and diversify their antibody repertoire through mechanisms involving the formation of programmed DNA damage. Immunoglobulin class switch recombination (CSR) is triggered by DNA lesions induced by activation-induced cytidine deaminase, which are processed to double-stranded DNA break (DSB) intermediates. These DSBs activate the cellular DNA damage response and enroll numerous DNA repair factors, involving poly(ADP-ribose) polymerases Parp1, Parp2, and Parp3 to promote appropriate DNA repair and efficient long-range recombination. The macroParp Parp9, which is overexpressed in certain lymphomas, has been recently implicated in DSB repair, acting together with Parp1. Here, we examine the contribution of Parp9 to the resolution of physiological DSBs incurred during V(D)J recombination and CSR by generating Parp9-/- mice. We find that Parp9-deficient mice are viable, fertile, and do not show any overt phenotype. Moreover, we find that Parp9 is dispensable for B-cell development. Finally, we show that CSR and DNA end-joining are robust in the absence of Parp9, indicating that Parp9 is not essential in vivo to achieve physiological DSB repair, or that strong compensatory mechanisms exist.


Assuntos
Linfócitos B/fisiologia , Reparo do DNA por Junção de Extremidades , Switching de Imunoglobulina , Poli(ADP-Ribose) Polimerase-1/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Imunidade Adaptativa , Animais , Células Cultivadas , Quebras de DNA de Cadeia Dupla , Dano ao DNA , Reparo do DNA , Imunoglobulinas/genética , Imunoglobulinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Poli(ADP-Ribose) Polimerases/genética
11.
Cell Rep ; 16(11): 2967-2979, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27601299

RESUMO

Paralog of XRCC4 and XLF (PAXX) is a member of the XRCC4 superfamily and plays a role in nonhomologous end-joining (NHEJ), a DNA repair pathway critical for lymphocyte antigen receptor gene assembly. Here, we find that the functions of PAXX and XLF in V(D)J recombination are masked by redundant joining activities. Thus, combined PAXX and XLF deficiency leads to an inability to join RAG-cleaved DNA ends. Additionally, we demonstrate that PAXX function in V(D)J recombination depends on its interaction with Ku. Importantly, we show that, unlike XLF, the role of PAXX during the repair of DNA breaks does not overlap with ATM and the RAG complex. Our findings illuminate the role of PAXX in V(D)J recombination and support a model in which PAXX and XLF function during NHEJ repair of DNA breaks, whereas XLF, the RAG complex, and the ATM-dependent DNA damage response promote end joining by stabilizing DNA ends.


Assuntos
Linfócitos B/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Proteínas de Ligação a DNA/química , Homologia de Sequência de Aminoácidos , Recombinação V(D)J/genética , Animais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Sistemas CRISPR-Cas/genética , Dano ao DNA , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Deleção de Genes , Edição de Genes , Rearranjo Gênico do Linfócito B , Imunoglobulinas/genética , Autoantígeno Ku/metabolismo , Modelos Biológicos , Proteínas Oncogênicas v-abl/metabolismo
12.
Genes Dev ; 28(22): 2450-63, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25366693

RESUMO

Faithful DNA repair is essential to avoid chromosomal rearrangements and promote genome integrity. Nuclear organization has emerged as a key parameter in the formation of chromosomal translocations, yet little is known as to whether DNA repair can efficiently occur throughout the nucleus and whether it is affected by the location of the lesion. Here, we induce DNA double-strand breaks (DSBs) at different nuclear compartments and follow their fate. We demonstrate that DSBs induced at the nuclear membrane (but not at nuclear pores or nuclear interior) fail to rapidly activate the DNA damage response (DDR) and repair by homologous recombination (HR). Real-time and superresolution imaging reveal that DNA DSBs within lamina-associated domains do not migrate to more permissive environments for HR, like the nuclear pores or the nuclear interior, but instead are repaired in situ by alternative end-joining. Our results are consistent with a model in which nuclear position dictates the choice of DNA repair pathway, thus revealing a new level of regulation in DSB repair controlled by spatial organization of DNA within the nucleus.


Assuntos
Núcleo Celular/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Linhagem Celular Tumoral , Cromatina/genética , Células HeLa , Recombinação Homóloga/genética , Humanos , Membrana Nuclear/metabolismo , Lâmina Nuclear/metabolismo
13.
Mol Aspects Med ; 34(6): 1138-52, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23454615

RESUMO

To cope with the devastating insults constantly inflicted to their genome by intrinsic and extrinsic DNA damaging sources, cells have evolved a sophisticated network of interconnected DNA caretaking mechanisms that will detect, signal and repair the lesions. Among the underlying molecular mechanisms that regulate these events, PARylation catalyzed by Poly(ADP-ribose) polymerases (PARPs), appears as one of the earliest post-translational modification at the site of the lesion that is known to elicit recruitment and regulation of many DNA damage response proteins. In this review we discuss how the complex PAR molecule operates in stress-induced DNA damage signaling and genome maintenance but also in various physiological settings initiated by developmentally programmed DNA breakage. To illustrate the latter, particular emphasis will be placed on the emerging contribution of PARPs to B cell receptor assembly and diversification.


Assuntos
Dano ao DNA , Reparo do DNA , Genoma/fisiologia , Poli Adenosina Difosfato Ribose/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Animais , Linfócitos B/metabolismo , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Processamento de Proteína Pós-Traducional
14.
J Exp Med ; 209(11): 2099-111, 2012 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-23008333

RESUMO

Antibody diversification requires the DNA deaminase AID to induce DNA instability at immunoglobulin (Ig) loci upon B cell stimulation. For efficient cytosine deamination, AID requires single-stranded DNA and needs to gain access to Ig loci, with RNA pol II transcription possibly providing both aspects. To understand these mechanisms, we isolated and characterized endogenous AID-containing protein complexes from the chromatin of diversifying B cells. The majority of proteins associated with AID belonged to RNA polymerase II elongation and chromatin modification complexes. Besides the two core polymerase subunits, members of the PAF complex, SUPT5H, SUPT6H, and FACT complex associated with AID. We show that AID associates with RNA polymerase-associated factor 1 (PAF1) through its N-terminal domain, that depletion of PAF complex members inhibits AID-induced immune diversification, and that the PAF complex can serve as a binding platform for AID on chromatin. A model is emerging of how RNA polymerase II elongation and pausing induce and resolve AID lesions.


Assuntos
Diversidade de Anticorpos , Linfócitos B/metabolismo , Citidina Desaminase/metabolismo , Proteínas Nucleares/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Citidina Desaminase/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células HEK293 , Células HeLa , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Humanos , Switching de Imunoglobulina , Imunoglobulinas/genética , Imunoprecipitação , Proteínas Nucleares/genética , Ligação Proteica , Interferência de RNA , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fatores de Elongação da Transcrição/genética , Fatores de Elongação da Transcrição/metabolismo
15.
Mol Cell Biol ; 31(3): 442-9, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21135131

RESUMO

Activation-induced cytidine deaminase (AID) is a mutator enzyme that initiates class switch recombination and somatic hypermutation of immunoglobulin genes (Ig) in B lymphocytes. However, AID also produces off-target DNA damage, including mutations in oncogenes and double-stranded breaks that can serve as substrates for oncogenic chromosomal translocations. AID is strictly regulated by a number of mechanisms, including phosphorylation at serine 38 and threonine 140, which increase activity. Here we show that phosphorylation can also suppress AID activity in vivo. Serine 3 is a novel phospho-acceptor which, when mutated to alanine, leads to increased class switching and c-myc/IgH translocations without affecting AID levels or catalytic activity. Conversely, increasing AID phosphorylation specifically on serine 3 by interfering with serine/threonine protein phosphatase 2A (PP2A) leads to decreased class switching. We conclude that AID activity and its oncogenic potential can be downregulated by phosphorylation of serine 3 and that this process is controlled by PP2A.


Assuntos
Citidina Desaminase/química , Citidina Desaminase/metabolismo , Cadeias Pesadas de Imunoglobulinas/genética , Proteínas Proto-Oncogênicas c-myc/genética , Translocação Genética , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Animais , Inibidores Enzimáticos/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Switching de Imunoglobulina/efeitos dos fármacos , Camundongos , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Proteína Fosfatase 2/antagonistas & inibidores , Proteína Fosfatase 2/metabolismo , Recombinação Genética/efeitos dos fármacos , Recombinação Genética/genética , Hipermutação Somática de Imunoglobulina/efeitos dos fármacos , Hipermutação Somática de Imunoglobulina/genética , Translocação Genética/efeitos dos fármacos
16.
Cell ; 143(1): 122-33, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20887897

RESUMO

Activation-induced cytidine deaminase (AID) initiates antibody gene diversification by creating U:G mismatches. However, AID is not specific for antibody genes; Off-target lesions can activate oncogenes or cause chromosome translocations. Despite its importance in these transactions little is known about how AID finds its targets. We performed an shRNA screen to identify factors required for class switch recombination (CSR) of antibody loci. We found that Spt5, a factor associated with stalled RNA polymerase II (Pol II) and single stranded DNA (ssDNA), is required for CSR. Spt5 interacts with AID, it facilitates association between AID and Pol II, and AID recruitment to its Ig and non-Ig targets. ChIP-seq experiments reveal that Spt5 colocalizes with AID and stalled Pol II. Further, Spt5 accumulation at sites of Pol II stalling is predictive of AID-induced mutation. We propose that AID is targeted to sites of Pol II stalling in part via its association with Spt5.


Assuntos
Linfócitos B/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Citidina Desaminase/metabolismo , Switching de Imunoglobulina , RNA Polimerase II/metabolismo , Fatores de Elongação da Transcrição/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Fibroblastos/metabolismo , Humanos , Imunoglobulinas/genética , Camundongos
17.
J Exp Med ; 206(5): 1047-56, 2009 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-19364882

RESUMO

Immunoglobulin class switch recombination (CSR) is initiated by DNA breaks triggered by activation-induced cytidine deaminase (AID). These breaks activate DNA damage response proteins to promote appropriate repair and long-range recombination. Aberrant processing of these breaks, however, results in decreased CSR and/or increased frequency of illegitimate recombination between the immunoglobulin heavy chain locus and oncogenes like c-myc. Here, we have examined the contribution of the DNA damage sensors Parp1 and Parp2 in the resolution of AID-induced DNA breaks during CSR. We find that although Parp enzymatic activity is induced in an AID-dependent manner during CSR, neither Parp1 nor Parp2 are required for CSR. We find however, that Parp1 favors repair of switch regions through a microhomology-mediated pathway and that Parp2 actively suppresses IgH/c-myc translocations. Thus, we define Parp1 as facilitating alternative end-joining and Parp2 as a novel translocation suppressor during CSR.


Assuntos
Dano ao DNA , Genes myc , Cadeias Pesadas de Imunoglobulinas/genética , Região de Troca de Imunoglobulinas/genética , Poli(ADP-Ribose) Polimerases/fisiologia , Recombinação Genética/imunologia , Translocação Genética/imunologia , Linfócitos B/imunologia , Citidina Desaminase/genética , Reparo do DNA , Humanos , Imunoglobulinas/genética , Ativação Linfocitária , Poli(ADP-Ribose) Polimerase-1 , Poli Adenosina Difosfato Ribose/genética
18.
Cell ; 135(6): 1028-38, 2008 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-19070574

RESUMO

Chromosomal translocation requires formation of paired double-strand DNA breaks (DSBs) on heterologous chromosomes. One of the most well characterized oncogenic translocations juxtaposes c-myc and the immunoglobulin heavy-chain locus (IgH) and is found in Burkitt's lymphomas in humans and plasmacytomas in mice. DNA breaks in IgH leading to c-myc/IgH translocations are created by activation-induced cytidine deaminase (AID) during antibody class switch recombination or somatic hypermutation. However, the source of DNA breaks at c-myc is not known. Here, we provide evidence for the c-myc promoter region being required in targeting AID-mediated DNA damage to produce DSBs in c-myc that lead to c-myc/IgH translocations in primary B lymphocytes. Thus, in addition to producing somatic mutations and DNA breaks in antibody genes, AID is also responsible for the DNA lesions in oncogenes that are required for their translocation.


Assuntos
Citidina Desaminase/metabolismo , Genes de Cadeia Pesada de Imunoglobulina , Genes myc , Translocação Genética , Animais , Linfócitos B/metabolismo , Linfoma de Burkitt/genética , Linfoma de Burkitt/metabolismo , Quebras de DNA de Cadeia Dupla , Células-Tronco Embrionárias , Humanos , Integrases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Plasmocitoma/genética , Plasmocitoma/metabolismo
19.
Immunity ; 28(5): 630-8, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18455451

RESUMO

MicroRNAs (miRNAs) are small noncoding RNAs that regulate vast networks of genes that share miRNA target sequences. To examine the physiologic effects of an individual miRNA-mRNA interaction in vivo, we generated mice that carry a mutation in the putative microRNA-155 (miR-155) binding site in the 3'-untranslated region of activation-induced cytidine deaminase (AID), designated Aicda(155) mice. AID is required for immunoglobulin gene diversification in B lymphocytes, but it also promotes chromosomal translocations. Aicda(155) caused an increase in steady-state Aicda mRNA and protein amounts by increasing the half-life of the mRNA, resulting in a high degree of Myc-Igh translocations. A similar but more pronounced translocation phenotype was also found in miR-155-deficient mice. Our experiments indicate that miR-155 can act as a tumor suppressor by reducing potentially oncogenic translocations generated by AID.


Assuntos
Linfócitos B/enzimologia , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Genes myc , Cadeias Pesadas de Imunoglobulinas/genética , MicroRNAs/metabolismo , Translocação Genética , Regiões 3' não Traduzidas , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Genes de Imunoglobulinas , Switching de Imunoglobulina , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Mutantes , MicroRNAs/genética , Mutação , Estabilidade de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Hipermutação Somática de Imunoglobulina
20.
Chem Biol ; 15(4): 383-92, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18420145

RESUMO

Vitamin D nuclear receptor (VDR), a ligand-dependent transcriptional regulator, is an important target for multiple clinical applications, such as osteoporosis and cancer. Since exacerbated increase of calcium serum level is currently associated with VDR ligands action, superagonists with low calcium serum levels have been developed. Based on the crystal structures of human VDR (hVDR) bound to 1alpha,25-dihydroxyvitamin D(3) and superagonists-notably, KH1060-we designed a superagonist ligand. In order to optimize the aliphatic side chain conformation with a subsequent entropy benefit, we incorporated an oxolane ring and generated two stereo diasteromers, AMCR277A and AMCR277B. Only AMCR277A exhibits superagonist activity in vitro, but is as calcemic in vivo as the natural ligand. The crystal structures of the complexes between the ligand binding domain of hVDR and these ligands provide a rational approach to the design of more potent superagonist ligands for potential clinical application.


Assuntos
Desenho de Fármacos , Receptores de Calcitriol/agonistas , Receptores de Calcitriol/metabolismo , Vitamina D/química , Vitamina D/farmacologia , Animais , Calcitriol/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Cristalografia por Raios X , Humanos , Ligantes , Masculino , Camundongos , Receptores de Calcitriol/genética , Estereoisomerismo , Transcrição Gênica/efeitos dos fármacos , Vitamina D/análogos & derivados , Vitamina D/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA