Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
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
2.
Eur J Immunol ; 53(7): e2350373, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37143384

RESUMO

During immune responses, B cells engaging a cognate antigen are recruited to GCs in secondary lymphoid organs where they will diversify their BCR to generate highly specific and adapted humoral responses. They do so, by inducing the expression of activation-induced cytidine deaminase (AID), which initiates somatic hypermutation (SHM) and class switch recombination (CSR). AID deaminates cytosines in ss DNA, generating U:G mismatches that are processed to induce ds DNA break intermediates during CSR that result in the expression of a different antibody isotype. Interestingly, hypoxia regions have been reported in GCs and suggesting that hypoxia could modulate the humoral response. Furthermore, hypoxia inducible transcription factor (HIF) can bind to the AID promoter and induce AID expression in a non-B-cell setting, suggesting that it might be involved in the transcriptional induction of AID in B cells, hence, regulating SHM and CSR. We, thus, hypothesized that HIF could regulate the efficiency of CSR. Here, we show that the inactivation of both the HIF-1α and HIF-1ß subunits of the HIF transcription factor in murine CH12 B cells results in defective CSR and that this is due to the suboptimal induction of AID expression.


Assuntos
Citidina Desaminase , Regulação da Expressão Gênica , Animais , Camundongos , Linfócitos B , Citidina Desaminase/metabolismo , Switching de Imunoglobulina , Isotipos de Imunoglobulinas/metabolismo , Hipermutação Somática de Imunoglobulina , Fatores de Transcrição/genética
3.
Mol Cell ; 63(2): 293-305, 2016 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-27397684

RESUMO

Repetitive DNA is packaged into heterochromatin to maintain its integrity. We use CRISPR/Cas9 to induce DSBs in different mammalian heterochromatin structures. We demonstrate that in pericentric heterochromatin, DSBs are positionally stable in G1 and recruit NHEJ factors. In S/G2, DSBs are resected and relocate to the periphery of heterochromatin, where they are retained by RAD51. This is independent of chromatin relaxation but requires end resection and RAD51 exclusion from the core. DSBs that fail to relocate are engaged by NHEJ or SSA proteins. We propose that the spatial disconnection between end resection and RAD51 binding prevents the activation of mutagenic pathways and illegitimate recombination. Interestingly, in centromeric heterochromatin, DSBs recruit both NHEJ and HR proteins throughout the cell cycle. Our results highlight striking differences in the recruitment of DNA repair factors between pericentric and centromeric heterochromatin and suggest a model in which the commitment to specific DNA repair pathways regulates DSB position.


Assuntos
Centrômero/metabolismo , Montagem e Desmontagem da Cromatina , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Heterocromatina/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Sistemas CRISPR-Cas , Centrômero/química , Centrômero/genética , Reparo do DNA por Junção de Extremidades , Fase G2 , Heterocromatina/química , Heterocromatina/genética , Histonas/genética , Histonas/metabolismo , Autoantígeno Ku/genética , Autoantígeno Ku/metabolismo , Camundongos , Células NIH 3T3 , Interferência de RNA , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Proteína Rad52 de Recombinação e Reparo de DNA/genética , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Reparo de DNA por Recombinação , Fase S , Fatores de Tempo , Transfecção
4.
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
5.
Biol Cell ; 112(1): 22-37, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31789463

RESUMO

BACKGROUND: The centrosome regulates cell spatial organisation by controlling the architecture of the microtubule (MT) cytoskeleton. Conversely, the position of the centrosome within the cell depends on cytoskeletal networks it helps organizing. In mammalian cells, centrosome positioning involves a population of MT stably anchored at centrioles, the core components of the centrosome. An MT-anchoring complex containing the proteins ninein and Cep170 is enriched at subdistal appendages (SAP) that decorate the older centriole (called mother centriole) and at centriole proximal ends. Here, we studied the role played at the centrosome by hVFL3/CCDC61, the human ortholog of proteins required for anchoring distinct sets of cytoskeletal fibres to centrioles in unicellular eukaryotes. RESULTS: We show that hVFL3 co-localises at SAP and at centriole proximal ends with components of the MT-anchoring complex, and physically interacts with Cep170. Depletion of hVFL3 increased the distance between mother and daughter centrioles without affecting the assembly of a filamentous linker that tethers the centrioles and contains the proteins rootletin and C-Nap1. When the linker was disrupted by inactivating C-Nap1, hVFL3-depletion exacerbated centriole splitting, a phenotype also observed following depletion of other SAP components. This supported that hVFL3 is required for SAP function, which we further established by showing that centrosome positioning is perturbed in hVFL3-depleted interphase cells. Finally, we found that hVFL3 is an MT-binding protein. CONCLUSIONS AND SIGNIFICANCE: Together, our results support that hVFL3 is required for anchoring MT at SAP during interphase and ensuring proper centrosome cohesion and positioning. The role of the VFL3 family of proteins thus appears to have been conserved in evolution despite the great variation in the shape of centriole appendages in different eukaryotic species.


Assuntos
Proteínas de Transporte/metabolismo , Centríolos , Centrossomo , Tubulina (Proteína)/metabolismo , Animais , Sistemas CRISPR-Cas , Proteínas de Transporte/genética , Linhagem Celular , Centríolos/metabolismo , Centríolos/ultraestrutura , Centrossomo/metabolismo , Centrossomo/ultraestrutura , Cílios/ultraestrutura , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Humanos , Microscopia Eletrônica , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , RNA Interferente Pequeno
6.
Eur J Immunol ; 48(4): 720-723, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29293266

RESUMO

The Mediator complex is known to orchestrate transcription. Here we show that B cell conditional deficient mice for the Med1 subunit display robust somatic hypermutation. Nevertheless, the mutation frequency at A residues is decreased and the expected A/T ratio is abolished, implicating Mediator in the second phase of somatic hypermutation.


Assuntos
Linfócitos B/citologia , Subunidade 1 do Complexo Mediador/deficiência , Subunidade 1 do Complexo Mediador/genética , Hipermutação Somática de Imunoglobulina/genética , Animais , Linfócitos B/imunologia , Centro Germinativo/citologia , Centro Germinativo/imunologia , Camundongos , Camundongos Transgênicos
7.
PLoS Genet ; 11(5): e1005240, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-26000965

RESUMO

To generate highly specific and adapted immune responses, B cells diversify their antibody repertoire through mechanisms involving the generation of programmed DNA damage. Somatic hypermutation (SHM) and class switch recombination (CSR) are initiated by the recruitment of activation-induced cytidine deaminase (AID) to immunoglobulin loci and by the subsequent generation of DNA lesions, which are differentially processed to mutations during SHM or to double-stranded DNA break intermediates during CSR. The latter activate the DNA damage response and mobilize multiple DNA repair factors, including Parp1 and Parp2, to promote DNA repair and long-range recombination. We examined the contribution of Parp3 in CSR and SHM. We find that deficiency in Parp3 results in enhanced CSR, while SHM remains unaffected. Mechanistically, this is due to increased occupancy of AID at the donor (Sµ) switch region. We also find evidence of increased levels of DNA damage at switch region junctions and a bias towards alternative end joining in the absence of Parp3. We propose that Parp3 plays a CSR-specific role by controlling AID levels at switch regions during CSR.


Assuntos
Regulação da Expressão Gênica , Switching de Imunoglobulina/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Animais , Linfócitos B/metabolismo , Sequência de Bases , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Loci Gênicos , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Região de Troca de Imunoglobulinas/genética , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Poli(ADP-Ribose) Polimerases/genética , Recombinação Genética , Hipermutação Somática de Imunoglobulina/genética
8.
Elife ; 112022 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-35319462

RESUMO

Centrioles are formed by microtubule triplets in a ninefold symmetric arrangement. In flagellated protists and animal multiciliated cells, accessory structures tethered to specific triplets render the centrioles rotationally asymmetric, a property that is key to cytoskeletal and cellular organization in these contexts. In contrast, centrioles within the centrosome of animal cells display no conspicuous rotational asymmetry. Here, we uncover rotationally asymmetric molecular features in human centrioles. Using ultrastructure expansion microscopy, we show that LRRCC1, the ortholog of a protein originally characterized in flagellate green algae, associates preferentially to two consecutive triplets in the distal lumen of human centrioles. LRRCC1 partially co-localizes and affects the recruitment of another distal component, C2CD3, which also has an asymmetric localization pattern in the centriole lumen. Together, LRRCC1 and C2CD3 delineate a structure reminiscent of a filamentous density observed by electron microscopy in flagellates, termed the 'acorn.' Functionally, the depletion of LRRCC1 in human cells induced defects in centriole structure, ciliary assembly, and ciliary signaling, supporting that LRRCC1 cooperates with C2CD3 to organizing the distal region of centrioles. Since a mutation in the LRRCC1 gene has been identified in Joubert syndrome patients, this finding is relevant in the context of human ciliopathies. Taken together, our results demonstrate that rotational asymmetry is an ancient property of centrioles that is broadly conserved in human cells. Our work also reveals that asymmetrically localized proteins are key for primary ciliogenesis and ciliary signaling in human cells.


Assuntos
Proteínas de Ciclo Celular , Centríolos , Ciliopatias , Proteínas Associadas aos Microtúbulos , Animais , Proteínas de Ciclo Celular/genética , Centríolos/metabolismo , Centrossomo/metabolismo , Cílios/metabolismo , Humanos , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/metabolismo
9.
J Exp Med ; 217(11)2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-32865561

RESUMO

The autosomal recessive immunodeficiency, centromeric instability, and facial anomalies (ICF) syndrome is a genetically heterogeneous disorder. Despite the identification of the underlying gene defects, it is unclear how mutations in any of the four known ICF genes cause a primary immunodeficiency. Here we demonstrate that loss of ZBTB24 in B cells from mice and ICF2 patients affects nonhomologous end-joining (NHEJ) during immunoglobulin class-switch recombination and consequently impairs immunoglobulin production and isotype balance. Mechanistically, we found that ZBTB24 associates with poly(ADP-ribose) polymerase 1 (PARP1) and stimulates its auto-poly(ADP-ribosyl)ation. The zinc-finger in ZBTB24 binds PARP1-associated poly(ADP-ribose) chains and mediates the PARP1-dependent recruitment of ZBTB24 to DNA breaks. Moreover, through its association with poly(ADP-ribose) chains, ZBTB24 protects them from degradation by poly(ADP-ribose) glycohydrolase (PARG). This facilitates the poly(ADP-ribose)-dependent assembly of the LIG4/XRCC4 complex at DNA breaks, thereby promoting error-free NHEJ. Thus, we uncover ZBTB24 as a regulator of PARP1-dependent NHEJ and class-switch recombination, providing a molecular basis for the immunodeficiency in ICF2 syndrome.


Assuntos
Reparo do DNA por Junção de Extremidades/genética , Face/anormalidades , Switching de Imunoglobulina/genética , Mutação , Doenças da Imunodeficiência Primária/genética , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Animais , Linfócitos B/imunologia , Quebras de DNA , Face/patologia , Células HEK293 , Humanos , Região de Troca de Imunoglobulinas , Camundongos , Poli(ADP-Ribose) Polimerase-1/metabolismo , Doenças da Imunodeficiência Primária/sangue , Doenças da Imunodeficiência Primária/patologia , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Transfecção
10.
PLoS One ; 14(8): e0220694, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31461461

RESUMO

A large number of genetic studies in yeast rely on the use of expression vectors. To facilitate the experimental approach of these studies, several collections of expression vectors have been generated (YXplac, pRS series, etc.). Subsequently, these collections have been expanded by adding more diversity to many of the plasmid features, including new selection markers and new promoter sequences. However, the ever growing number of plasmid features makes it unrealistic for research labs to maintain an up-to-date collection of plasmids. Here, we developed the COSPLAY toolbox: a Golden Gate approach based on the scheme of a simple modular plasmid that recapitulates and completes all the properties of the pRS plasmids. The COSPLAY toolbox contains a basal collection of individual functional modules. Moreover, we standardized a simple and rapid, software-assisted protocol which facilitates the addition of new personalized modules. Finally, our toolbox includes the possibility to select a genomic target location and to perform a single copy integration of the expression vector.


Assuntos
Clonagem Molecular/métodos , Vetores Genéticos/genética , Plasmídeos/genética , Saccharomyces cerevisiae/genética , Biblioteca Gênica , Genes Reporter , Engenharia Genética/métodos , Software , Transformação Genética
11.
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
12.
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
13.
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
14.
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
15.
J Exp Med ; 213(3): 303-12, 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-26903242

RESUMO

Immunoglobulin (Ig) class switch recombination (CSR) is initiated by the transcription-coupled recruitment of activation-induced cytidine deaminase (AID) to Ig switch regions (S regions). During CSR, the IgH locus undergoes dynamic three-dimensional structural changes in which promoters, enhancers, and S regions are brought to close proximity. Nevertheless, little is known about the underlying mechanisms. In this study, we show that Med1 and Med12, two subunits of the mediator complex implicated in transcription initiation and long-range enhancer/promoter loop formation, are dynamically recruited to the IgH locus enhancers and the acceptor regions during CSR and that their knockdown in CH12 cells results in impaired CSR. Furthermore, we show that conditional inactivation of Med1 in B cells results in defective CSR and reduced acceptor S region transcription. Finally, we show that in B cells undergoing CSR, the dynamic long-range contacts between the IgH enhancers and the acceptor regions correlate with Med1 and Med12 binding and that they happen at a reduced frequency in Med1-deficient B cells. Our results implicate the mediator complex in the mechanism of CSR and are consistent with a model in which mediator facilitates the long-range contacts between S regions and the IgH locus enhancers during CSR and their transcriptional activation.


Assuntos
Switching de Imunoglobulina/genética , Cadeias Pesadas de Imunoglobulinas/genética , Subunidade 1 do Complexo Mediador/metabolismo , Complexo Mediador/metabolismo , Recombinação Genética/genética , Ativação Transcricional/genética , Animais , Linfócitos B/metabolismo , Células Cultivadas , Técnicas de Silenciamento de Genes , Loci Gênicos , Camundongos , Ligação Proteica , Transcrição Gênica
17.
J Exp Med ; 210(12): 2495-502, 2013 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-24145512

RESUMO

Immunoglobulin (Ig) class switch recombination (CSR) is initiated by the transcription-coupled recruitment of activation-induced cytidine deaminase (AID) to switch regions and by the subsequent generation of double-stranded DNA breaks (DSBs). These DNA breaks are ultimately resolved through the nonhomologous end joining (NHEJ) pathway. We show that during CSR, AID associates with subunits of cohesin, a complex previously implicated in sister chromatid cohesion, DNA repair, and the formation of DNA loops between enhancers and promoters. Furthermore, we implicate the cohesin complex in the mechanism of CSR by showing that cohesin is dynamically recruited to the Sµ-Cµ region of the IgH locus during CSR and that knockdown of cohesin or its regulatory subunits results in impaired CSR and increased usage of microhomology-based end joining.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Switching de Imunoglobulina , Animais , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Linhagem Celular , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Proteínas Cromossômicas não Histona/antagonistas & inibidores , Proteínas Cromossômicas não Histona/genética , Citidina Desaminase/metabolismo , Reparo do DNA por Junção de Extremidades , Técnicas de Silenciamento de Genes , Camundongos , Recombinação Genética , Coesinas
18.
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
19.
J Exp Med ; 208(8): 1649-60, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21746811

RESUMO

Immunoglobulin class switch recombination (CSR) is initiated by double-stranded DNA breaks (DSBs) in switch regions triggered by activation-induced cytidine deaminase (AID). Although CSR correlates with epigenetic modifications at the IgH locus, the relationship between these modifications and AID remains unknown. In this study, we show that during CSR, AID forms a complex with KAP1 (KRAB domain-associated protein 1) and HP1 (heterochromatin protein 1) that is tethered to the donor switch region (Sµ) bearing H3K9me3 (trimethylated histone H3 at lysine 9) in vivo. Furthermore, in vivo disruption of this complex results in impaired AID recruitment to Sµ, inefficient DSB formation, and a concomitant defect in CSR but not in somatic hypermutation. We propose that KAP1 and HP1 tether AID to H3K9me3 residues at the donor switch region, thus providing a mechanism linking AID to epigenetic modifications during CSR.


Assuntos
Linfócitos B/citologia , Citidina Desaminase/imunologia , Epigênese Genética/imunologia , Switching de Imunoglobulina/imunologia , Região de Troca de Imunoglobulinas/imunologia , Proteínas Nucleares/imunologia , Proteínas Repressoras/imunologia , Animais , Linfócitos B/imunologia , Western Blotting , Imunoprecipitação da Cromatina , Cromatografia em Gel , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/metabolismo , Análise Mutacional de DNA , Primers do DNA/genética , Citometria de Fluxo , Histonas/metabolismo , Hibridização in Situ Fluorescente , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteína 28 com Motivo Tripartido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA