Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 14(11): 753, 2023 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-37980415

RESUMO

Pathogenic variants in BRCA2 are known to significantly increase the lifetime risk of developing breast and ovarian cancers. Sequencing-based genetic testing has resulted in the identification of thousands of BRCA2 variants that are considered to be variants of uncertain significance (VUS) because the disease risk associated with them is unknown. One such variant is p.Arg3052Gln, which has conflicting interpretations of pathogenicity in the ClinVar variant database. Arginine at position 3052 in BRCA2 plays an important role in stabilizing its C-terminal DNA binding domain. We have generated a knock-in mouse model expressing this variant to examine its role on growth and survival in vivo. Homozygous as well as hemizygous mutant mice are viable, fertile and exhibit no overt phenotype. While we did not observe any hematopoietic defects in adults, we did observe a marked reduction in the in vitro proliferative ability of fetal liver cells that were also hypersensitive to PARP inhibitor, olaparib. In vitro studies performed on embryonic and adult fibroblasts derived from the mutant mice showed significant reduction in radiation induced RAD51 foci formation as well as increased genomic instability after mitomycin C treatment. We observed mis-localization of a fraction of R3052Q BRCA2 protein to the cytoplasm which may explain the observed in vitro phenotypes. Our findings suggest that BRCA2 R3052Q should be considered as a hypomorphic variant.


Assuntos
Antineoplásicos , Neoplasias da Mama , Neoplasias Ovarianas , Humanos , Feminino , Camundongos , Animais , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Testes Genéticos , Neoplasias Ovarianas/genética , Homozigoto , Neoplasias da Mama/genética , Proteína BRCA1/genética , Predisposição Genética para Doença
2.
Sci Rep ; 12(1): 7200, 2022 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-35504930

RESUMO

Signaling pathways play an important role in cell fate determination in stem cells and regulate a plethora of developmental programs, the dysregulation of which can lead to human diseases. Growth factors (GFs) regulating these signaling pathways therefore play a major role in the plasticity of adult stem cells and modulate cellular differentiation and tissue repair outcomes. We consider murine mammary organoid generation from self-organizing adult stem cells as a tool to understand the role of GFs in organ development and tissue regeneration. The astounding capacity of mammary organoids to regenerate a gland in vivo after transplantation makes it a convenient model to study organ regeneration. We show organoids grown in suspension with minimal concentration of Matrigel and in the presence of a cocktail of GFs regulating EGF and FGF signaling can recapitulate key epithelial layers of adult mammary gland. We establish a toolkit utilizing in vivo whole animal imaging and ultrasound imaging combined with ex vivo approaches including tissue clearing and confocal imaging to study organ regeneration and ductal morphogenesis. Although the organoid structures were severely impaired in vitro when cultured in the presence of individual GFs, ex vivo imaging revealed ductal branching after transplantation albeit with significantly reduced number of terminal end buds. We anticipate these imaging modalities will open novel avenues to study mammary gland morphogenesis in vivo and can be beneficial for monitoring mammary tumor progression in pre-clinical and clinical settings.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular , Organoides , Animais , Fatores Imunológicos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Glândulas Mamárias Animais/metabolismo , Camundongos , Morfogênese , Organoides/crescimento & desenvolvimento , Organoides/metabolismo , Regeneração
3.
Nat Commun ; 13(1): 1751, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35365640

RESUMO

The interaction between tumor suppressor BRCA2 and DSS1 is essential for RAD51 recruitment and repair of DNA double stand breaks (DSBs) by homologous recombination (HR). We have generated mice with a leucine to proline substitution at position 2431 of BRCA2, which disrupts this interaction. Although a significant number of mutant mice die during embryogenesis, some homozygous and hemizygous mutant mice undergo normal postnatal development. Despite lack of radiation induced RAD51 foci formation and a severe HR defect in somatic cells, mutant mice are fertile and exhibit normal RAD51 recruitment during meiosis. We hypothesize that the presence of homologous chromosomes in close proximity during early prophase I may compensate for the defect in BRCA2-DSS1 interaction. We show the restoration of RAD51 foci in mutant cells when Topoisomerase I inhibitor-induced single strand breaks are converted into DSBs during DNA replication. We also partially rescue the HR defect by tethering the donor DNA to the site of DSBs using streptavidin-fused Cas9. Our findings demonstrate that the BRCA2-DSS1 complex is dispensable for RAD51 loading when the homologous DNA is close to the DSB.


Assuntos
Quebras de DNA de Cadeia Dupla , Rad51 Recombinase , Animais , DNA , Reparo do DNA/genética , Recombinação Homóloga , Camundongos , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo
4.
Cell Death Dis ; 12(9): 838, 2021 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-34489406

RESUMO

Hereditary non-polyposis colorectal cancer, now known as Lynch syndrome (LS) is one of the most common cancer predisposition syndromes and is caused by germline pathogenic variants (GPVs) in DNA mismatch repair (MMR) genes. A common founder GPV in PMS2 in the Canadian Inuit population, NM_000535.5: c.2002A>G, leads to a benign missense (p.I668V) but also acts as a de novo splice site that creates a 5 bp deletion resulting in a truncated protein (p.I668*). Individuals homozygous for this GPV are predisposed to atypical constitutional MMR deficiency with a delayed onset of first primary malignancy. We have generated mice with an equivalent germline mutation (Pms2c.1993A>G) and demonstrate that it results in a splicing defect similar to those observed in humans. Homozygous mutant mice are viable like the Pms2 null mice. However, unlike the Pms2 null mice, these mutant mice are fertile, like humans homozygous for this variant. Furthermore, these mice exhibit a significant increase in microsatellite instability and intestinal adenomas on an Apc mutant background. Rectification of the splicing defect in human and murine fibroblasts using antisense morpholinos suggests that this novel mouse model can be valuable in evaluating the efficacy aimed at targeting the splicing defect in PMS2 that is highly prevalent among the Canadian Inuits.


Assuntos
Reparo de Erro de Pareamento de DNA/genética , Efeito Fundador , Endonuclease PMS2 de Reparo de Erro de Pareamento/genética , Mutação/genética , Splicing de RNA/genética , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Sequência de Bases , Modelos Animais de Doenças , Éxons/genética , Fertilidade/genética , Fibroblastos/metabolismo , Masculino , Meiose , Camundongos Endogâmicos C57BL , Instabilidade de Microssatélites , Endonuclease PMS2 de Reparo de Erro de Pareamento/metabolismo , Morfolinos/farmacologia , Pólipos/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espermatozoides/patologia , Testículo/patologia
5.
Nat Commun ; 9(1): 537, 2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29416040

RESUMO

BRCA2 is essential for maintaining genomic integrity. BRCA2-deficient primary cells are either not viable or exhibit severe proliferation defects. Yet, BRCA2 deficiency contributes to tumorigenesis. It is believed that mutations in genes such as TRP53 allow BRCA2 heterozygous cells to overcome growth arrest when they undergo loss of heterozygosity. Here, we report the use of an insertional mutagenesis screen to identify a role for BRE (Brain and Reproductive organ Expressed, also known as BRCC45), known to be a part of the BRCA1-DNA damage sensing complex, in the survival of BRCA2-deficient mouse ES cells. Cell viability by BRE overexpression is mediated by deregulation of CDC25A phosphatase, a key cell cycle regulator and an oncogene. We show that BRE facilitates deubiquitylation of CDC25A by recruiting ubiquitin-specific-processing protease 7 (USP7) in the presence of DNA damage. Additionally, we uncovered the role of CDC25A in BRCA-mediated tumorigenesis, which can have implications in cancer treatment.


Assuntos
Dano ao DNA , Proteínas do Tecido Nervoso/fisiologia , Peptidase 7 Específica de Ubiquitina/metabolismo , Fosfatases cdc25/metabolismo , Animais , Proteína BRCA1/genética , Proteína BRCA1/fisiologia , Proteína BRCA2/genética , Proteína BRCA2/fisiologia , Carcinogênese , Sobrevivência Celular/fisiologia , Células Cultivadas , Estabilidade Enzimática , Heterozigoto , Humanos , Perda de Heterozigosidade , Células MCF-7 , Camundongos , Camundongos Knockout , Mutagênese Insercional , Processamento de Proteína Pós-Traducional , Proteína Supressora de Tumor p53/genética , Ubiquitinação
6.
Genetics ; 207(4): 1335-1345, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29021281

RESUMO

BRCA2 loss-of-heterozygosity (LOH) is frequently observed in BRCA2-mutated tumors, but its biallelic loss causes embryonic lethality in mice and inhibits proliferation of normal somatic cells. Therefore, it remains unclear how loss of BRCA2 contributes to tumorigenesis. One possibility is that mutation in potential genetic interactors of BRCA2, such as TRP53, is required for cell survival/proliferation in the absence of BRCA2. In this study, using an insertional mutagenesis screen in mouse embryonic stem cells (mESC), we have identified GIPC3 (GAIP-interacting protein C-terminus 3) as a BRCA2 genetic interactor that contributes to survival of Brca2-null mESC. GIPC3 does not compensate for BRCA2 loss in the repair of double-strand breaks. Mass-spectrometric analysis resulted in the identification of G-protein signaling transducers, APPL1 and APPL2, as potential GIPC3-binding proteins. A mutant GIPC3 (His155Ala) that does not bind to APPL1/2 failed to rescue the lethality of Brca2-null mESC, suggesting that the cell viability by GIPC3 is mediated via APPL1/2. Finally, the physiological significance of GIPC3 as a genetic interactor of BRCA2 is supported by the observation that Brca2-null embryos with Gipc3 overexpression are developmentally more advanced than their control littermates. Taken together, we have uncovered a novel role for GIPC3 as a BRCA2 genetic interactor.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteína BRCA2/genética , Neoplasias da Mama/genética , Carcinogênese/genética , Animais , Proteína BRCA2/deficiência , Neoplasias da Mama/patologia , Proteínas de Transporte/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Perda de Heterozigosidade/genética , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Mutagênese Insercional , Mutação
7.
Nat Commun ; 7: 12425, 2016 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-27498558

RESUMO

Poly (ADP-ribose) polymerase (PARP) inhibitor (PARPi) olaparib has been approved for treatment of advanced ovarian cancer associated with BRCA1 and BRCA2 mutations. BRCA1- and BRCA2-mutated cells, which are homologous recombination (HR) deficient, are hypersensitive to PARPi through the mechanism of synthetic lethality. Here we examine the effect of PARPi on HR-proficient cells. Olaparib pretreatment, PARP1 knockdown or Parp1 heterozygosity of Brca2(cko/ko) mouse embryonic stem cells (mESCs), carrying a null (ko) and a conditional (cko) allele of Brca2, results in viable Brca2(ko/ko) cells. PARP1 deficiency does not restore HR in Brca2(ko/ko) cells, but protects stalled replication forks from MRE11-mediated degradation through its impaired recruitment. The functional consequence of Parp1 heterozygosity on BRCA2 loss is demonstrated by a significant increase in tumorigenesis in Brca2(cko/cko) mice. Thus, while olaparib efficiently kills BRCA2-deficient cells, we demonstrate that it can also contribute to the synthetic viability if PARP is inhibited before BRCA2 loss.


Assuntos
Proteína BRCA2/deficiência , Poli(ADP-Ribose) Polimerase-1/deficiência , Animais , Proteína BRCA2/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Recombinação Homóloga/efeitos dos fármacos , Humanos , Integrases/metabolismo , Proteína Homóloga a MRE11/metabolismo , Camundongos , Modelos Biológicos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/metabolismo , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Poli(ADP-Ribose) Polimerase-1/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia
8.
Hum Mol Genet ; 25(10): 1934-1945, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26920070

RESUMO

The breast cancer gene, BRCA2, is essential for viability, yet patients with Fanconi anemia-D1 subtype are born alive with biallelic mutations in this gene. The hypomorphic nature of the mutations is believed to support viability, but this is not always apparent. One such mutation is IVS7+2T>G, which causes premature protein truncation due to skipping of exon 7. We previously identified a transcript lacking exons 4-7, which restores the open-reading frame, encodes a DNA repair proficient protein and is expressed in IVS7+2T>G carriers. However, because the exons 4-7 encoded region contains several residues required for normal cell-cycle regulation and cytokinesis, this transcript's ability to support viability can be argued. To address this, we generated a Brca2 knock-in mouse model lacking exons 4-7 and demonstrated that these exons are dispensable for viability as well as tumor-free survival. This study provides the first in vivo evidence of the functional significance of a minor transcript of BRCA2 that can play a major role in the survival of humans who are homozygous for a clearly pathogenic mutation. Our results highlight the importance of assessing protein function restoration by premature truncating codon bypass by alternative splicing when evaluating the functional significance of variants such as nonsense and frame-shift mutations that are assumed to be clearly pathogenic. Our findings will impact not only the assessment of variants that map to this region, but also influence counseling paradigms and treatment options for such mutation carriers.


Assuntos
Proteína BRCA2/genética , Neoplasias da Mama/genética , Anemia de Fanconi/genética , Predisposição Genética para Doença , Processamento Alternativo/genética , Animais , Neoplasias da Mama/patologia , Éxons/genética , Anemia de Fanconi/patologia , Técnicas de Introdução de Genes , Mutação em Linhagem Germinativa , Humanos , Camundongos , Mutação , Linhagem , Sítios de Splice de RNA
9.
Oncotarget ; 7(10): 11094-112, 2016 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-26848978

RESUMO

The oncogenic role of microRNA-155 (miR-155) in leukemia is well established but its role in other cancers, especially breast cancer, is gradually emerging. In this study we examined the effect of mir-155 loss in a well-characterized spontaneous breast cancer mouse model where Brca1 and Trp53 are deleted by K14-Cre. miR-155 is known to be up-regulated in BRCA1-deficient tumors. Surprisingly, complete loss of miR-155 (miR-155ko/ko) did not alter the tumor free survival of the mutant mice. However, we found increased infiltration of myeloid derived suppressor cells (MDSCs) in miR-155 deficient tumors. In addition, cytokine/chemokine array analysis revealed altered level of cytokines that are implicated in the recruitment of MDSCs. Mechanistically, we identified C/EBP-ß, a known miR-155 target, to regulate the expression of these cytokines in the miR-155-deficient cells. Furthermore, using an allograft model, we showed that inhibition of miR-155 in cancer cells suppressed in vivo growth, which was restored by the loss of miR-155 in the microenvironment. Taken together, we have uncovered a novel tumor suppressive function of miR-155 in the tumor microenvironment, which is also dependent on miR-155 expression in the tumor cells. Because of the oncogenic as well as tumor suppressive roles of miR-155, our findings warrant caution against a systemic inhibition of miR-155 for anticancer therapy.


Assuntos
Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , MicroRNAs , Células Supressoras Mieloides/imunologia , Microambiente Tumoral/imunologia , Animais , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Camundongos , Camundongos Mutantes , Oncogenes
10.
Hum Mol Genet ; 21(18): 3993-4006, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22678057

RESUMO

Single-nucleotide substitutions and small in-frame insertions or deletions identified in human breast cancer susceptibility genes BRCA1 and BRCA2 are frequently classified as variants of unknown clinical significance (VUS) due to the availability of very limited information about their functional consequences. Such variants can most reliably be classified as pathogenic or non-pathogenic based on the data of their co-segregation with breast cancer in affected families and/or their co-occurrence with a pathogenic mutation. Biological assays that examine the effect of variants on protein function can provide important information that can be used in conjunction with available familial data to determine the pathogenicity of VUS. In this report, we have used a previously described mouse embryonic stem (mES) cell-based functional assay to characterize eight BRCA2 VUS that affect highly conserved amino acid residues and map to the N-terminal PALB2-binding or the C-terminal DNA-binding domains. For several of these variants, very limited co-segregation information is available, making it difficult to determine their pathogenicity. Based on their ability to rescue the lethality of Brca2-deficient mES cells and their effect on sensitivity to DNA-damaging agents, homologous recombination and genomic integrity, we have classified these variants as pathogenic or non-pathogenic. In addition, we have used homology-based modeling as a predictive tool to assess the effect of some of these variants on the structural integrity of the C-terminal DNA-binding domain and also generated a knock-in mouse model to analyze the physiological significance of a residue reported to be essential for the interaction of BRCA2 with meiosis-specific recombinase, DMC1.


Assuntos
Proteína BRCA2/genética , Neoplasias da Mama/genética , Células-Tronco Embrionárias/metabolismo , Mutação , Proteínas Nucleares/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Sequência de Aminoácidos , Animais , Proteína BRCA2/química , Proteínas de Ciclo Celular , Sobrevivência Celular , Células Cultivadas , Mapeamento Cromossômico , Sequência Conservada , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Proteínas de Ligação a DNA , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/fisiologia , Proteína do Grupo de Complementação N da Anemia de Fanconi , Feminino , Estudos de Associação Genética , Humanos , Funções Verossimilhança , Masculino , Camundongos , Camundongos Transgênicos , Mitomicina/farmacologia , Modelos Moleculares , Mutagênicos/farmacologia , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas/genética , Estrutura Quaternária de Proteína , Homologia Estrutural de Proteína
11.
Nat Med ; 17(10): 1275-82, 2011 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-21946536

RESUMO

BRCA1, a well-known tumor suppressor with multiple interacting partners, is predicted to have diverse biological functions. However, so far its only well-established role is in the repair of damaged DNA and cell cycle regulation. In this regard, the etiopathological study of low-penetrant variants of BRCA1 provides an opportunity to uncover its other physiologically important functions. Using this rationale, we studied the R1699Q variant of BRCA1, a potentially moderate-risk variant, and found that it does not impair DNA damage repair but abrogates the repression of microRNA-155 (miR-155), a bona fide oncomir. Mechanistically, we found that BRCA1 epigenetically represses miR-155 expression via its association with HDAC2, which deacetylates histones H2A and H3 on the miR-155 promoter. We show that overexpression of miR-155 accelerates but the knockdown of miR-155 attenuates the growth of tumor cell lines in vivo. Our findings demonstrate a new mode of tumor suppression by BRCA1 and suggest that miR-155 is a potential therapeutic target for BRCA1-deficient tumors.


Assuntos
Proteína BRCA1/metabolismo , Reparo do DNA/fisiologia , Epigênese Genética/fisiologia , Regulação Neoplásica da Expressão Gênica/genética , MicroRNAs/metabolismo , Análise de Variância , Proteína BRCA1/genética , Southern Blotting , Imunoprecipitação da Cromatina , Reparo do DNA/genética , Epigênese Genética/genética , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Histona Desacetilase 2/metabolismo , Humanos , Imuno-Histoquímica , Imunoprecipitação , Hibridização In Situ , Análise em Microsséries , Mutação de Sentido Incorreto/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
J Clin Invest ; 119(10): 3160-71, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19770520

RESUMO

To date, inheritance of a mutant BRCA1 or BRCA2 gene is the best-established indicator of an increased risk of developing breast cancer. Sequence analysis of these genes is being used to identify BRCA1/2 mutation carriers, though these efforts are hampered by the high frequency of variants of unknown clinical significance (VUSs). Functional evaluation of such variants has been restricted due to lack of a physiologically relevant assay. In this study we developed a functional assay using mouse ES cells to study variants of BRCA1. We introduced BAC clones with human wild-type BRCA1 or variants into Brca1-null ES cells and confirmed that only wild-type and a known neutral variant rescued cell lethality. The same neutral variant was also able to rescue embryogenesis in Brca1-null mice. A test of several BRCT domain mutants revealed all to be deleterious, including a VUS. Furthermore, we used this assay to determine the effects of BRCA1 variants on cell cycle regulation, differentiation, and genomic stability. Importantly, we discovered that ES cells rescued by S1497A BRCA1 exhibited significant hypersensitivity after gamma-irradiation. Our results demonstrate that this ES cell-based assay is a powerful and reliable method for analyzing the functional impact of BRCA1 variants, which we believe could be used to determine which patients may require preventative treatments.


Assuntos
Proteína BRCA1 , Células-Tronco Embrionárias/fisiologia , Mutação , Animais , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Ciclo Celular/fisiologia , Diferenciação Celular/fisiologia , Dano ao DNA , Reparo do DNA , Células-Tronco Embrionárias/citologia , Feminino , Predisposição Genética para Doença , Humanos , Camundongos , Camundongos Nus , Camundongos Transgênicos , Polimorfismo de Nucleotídeo Único
13.
Cancer Res ; 69(3): 863-72, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19155299

RESUMO

RecA/Rad51 protein family members (Rad51, Rad51b, Rad51c, Rad51d, Xrcc2, and Xrcc3) are essential for DNA repair by homologous recombination, and their role in cancers has been anticipated. Here we provide the first direct evidence for a tumor suppressor function for a member of the Rad51 family. We show that Rad51c deficiency leads to early embryonic lethality, which can be delayed on a Trp53-null background. To uncover the role of Rad51c in tumorigenesis, we have exploited the fact that Rad51c and Trp53 are both closely located on the mouse chromosome 11. We have generated double heterozygous (DH) mice carrying mutant alleles of both genes either on different (DH-trans) or on the same chromosome (DH-cis), the latter allowing for a deletion of wild-type alleles of both genes by loss of heterozygosity. DH-trans mice, in contrast to DH-cis, developed tumors with latency and spectrum similar to Trp53 heterozygous mice. Strikingly, Rad51c mutation in DH-cis mice promoted the development of tumors of specialized sebaceous glands and suppressed tumors characteristic of Trp53 mutation. In addition, DH-cis females developed tumors significantly earlier than any other group.


Assuntos
Transformação Celular Neoplásica/genética , Genes Supressores de Tumor , Rad51 Recombinase/genética , Proteína Supressora de Tumor p53/genética , Adenocarcinoma Sebáceo/genética , Alelos , Animais , Proteínas de Ligação a DNA , Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Feminino , Perda de Heterozigosidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Rad51 Recombinase/deficiência , Neoplasias das Glândulas Sebáceas/genética , Fatores Sexuais
14.
J Cell Biol ; 176(5): 581-92, 2007 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-17312021

RESUMO

RAD51C is a member of the RecA/RAD51 protein family, which is known to play an important role in DNA repair by homologous recombination. In mice, it is essential for viability. Therefore, we have generated a hypomorphic allele of Rad51c in addition to a null allele. A subset of mice expressing the hypomorphic allele is infertile. This infertility is caused by sexually dimorphic defects in meiotic recombination, revealing its two distinct functions. Spermatocytes undergo a developmental arrest during the early stages of meiotic prophase I, providing evidence for the role of RAD51C in early stages of RAD51-mediated recombination. In contrast, oocytes can progress normally to metaphase I after superovulation but display precocious separation of sister chromatids, aneuploidy, and broken chromosomes at metaphase II. These defects suggest a possible late role of RAD51C in meiotic recombination. Based on the marked reduction in Holliday junction (HJ) resolution activity in Rad51c-null mouse embryonic fibroblasts, we propose that this late function may be associated with HJ resolution.


Assuntos
Cromátides/genética , Prófase Meiótica I/genética , Metáfase/genética , Rad51 Recombinase/fisiologia , Recombinação Genética , Alelos , Animais , Aberrações Cromossômicas , DNA Cruciforme/metabolismo , Proteínas de Ligação a DNA , Feminino , Infertilidade/genética , Masculino , Prófase Meiótica I/fisiologia , Metáfase/fisiologia , Camundongos , Modelos Genéticos , Oócitos/citologia , Oócitos/metabolismo , Oócitos/ultraestrutura , Ovário/citologia , Ovário/metabolismo , Ovário/ultraestrutura , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Fatores Sexuais , Espermatócitos/citologia , Espermatócitos/metabolismo , Espermatócitos/ultraestrutura , Testículo/citologia , Testículo/metabolismo , Testículo/ultraestrutura
15.
Development ; 131(1): 131-42, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14660434

RESUMO

The role of Brca2 in gametogenesis has been obscure because of embryonic lethality of the knockout mice. We generated Brca2-null mice carrying a human BAC with the BRCA2 gene. This construct rescues embryonic lethality and the mice develop normally. However, there is poor expression of the transgene in the gonads and the mice are infertile, allowing examination of the function of BRCA2 in gametogenesis. BRCA2-deficient spermatocytes fail to progress beyond the early prophase I stage of meiosis. Observations on localization of recombination-related and spermatogenic-related proteins suggest that the spermatocytes undergo early steps of recombination (DNA double strand break formation), but fail to complete recombination or initiate spermiogenic development. In contrast to the early meiotic prophase arrest of spermatocytes, some mutant oocytes can progress through meiotic prophase I, albeit with a high frequency of nuclear abnormalities, and can be fertilized and produce embryos. Nonetheless, there is marked depletion of germ cells in adult females. These studies provide evidence for key roles of the BRCA2 protein in mammalian gametogenesis and meiotic success.


Assuntos
Proteína BRCA2/genética , Desenvolvimento Embrionário e Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Genes BRCA2 , Meiose/genética , Animais , Proteína BRCA2/deficiência , Feminino , Morte Fetal/genética , Humanos , Hibridização In Situ , Infertilidade Masculina/genética , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Oócitos/citologia , Oócitos/fisiologia , Reação em Cadeia da Polimerase , Espermatócitos/citologia , Espermatócitos/fisiologia
16.
Hum Mol Genet ; 12(17): 2121-31, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12915465

RESUMO

Numerous missense mutations in human BRCA1 gene have been linked to predisposition to breast cancer. However, the functional significance of the majority of these mutations remains unknown. We have examined the molecular basis for three such cancer-causing mutations. The first mutation, a T-->G transversion in codon 64, is predicted to change a conserved cysteine residue to glycine in the RING finger domain of the 1863 amino acid BRCA1 protein. Using a humanized mouse model we demonstrate that this missense mutation actually results in a functionally null protein. This striking result occurs because the single base alteration generates a new 5' splice site in exon 5 and also disrupts a putative exonic splicing enhancer motif. Consequently, the normal splice donor site is disrupted and an internal cryptic splice site is activated. This results in a 22-nucleotide deletion and the aberrant transcript is predicted to encode a severely truncated protein consisting of only 63 amino acids. To identify other missense mutations in BRCA1 that may result in aberrant splicing, we screened various mutations using the Genscan program. We demonstrate that at least two other missense mutations in codons 1495 and 1823 result in aberrant splicing due to the possible disruption of cis-acting splicing regulatory elements. In conclusion, our study demonstrates for the first time the application of a humanized mouse model for functional analysis of human mutations in mice and also shows the need for a careful examination of the functional consequences of single base alterations and single nucleotide polymorphisms identified in human disease-causing genes.


Assuntos
Processamento Alternativo/genética , Proteína BRCA1/genética , Éxons/genética , Modelos Animais , Mutação de Sentido Incorreto , Animais , Proteína BRCA1/metabolismo , Células Cultivadas , Cromossomos Artificiais Bacterianos , Feminino , Humanos , Linfócitos/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Sítios de Splice de RNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...