Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 14(9): 2263-2272, 2016 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-26923600

RESUMO

Genome editing has now been reported in many systems using TALEN and CRISPR-Cas9 nucleases. Precise mutations can be introduced during homology-directed repair with donor DNA carrying the wanted sequence edit, but efficiency is usually lower than for gene knockout and optimal strategies have not been extensively investigated. Here, we show that using phosphorothioate-modified oligonucleotides strongly enhances genome editing efficiency of single-stranded oligonucleotide donors in cultured cells. In addition, it provides better design flexibility, allowing insertions more than 100 bp long. Despite previous reports of phosphorothioate-modified oligonucleotide toxicity, clones of edited cells are readily isolated and targeted sequence insertions are achieved in rats and mice with very high frequency, allowing for homozygous loxP site insertion at the mouse ROSA locus in particular. Finally, when detected, imprecise knockin events exhibit indels that are asymmetrically positioned, consistent with genome editing taking place by two steps of single-strand annealing.


Assuntos
Sistemas CRISPR-Cas , Endonucleases/genética , Edição de Genes , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição/genética , Animais , Sequência de Bases , Linhagem Celular Tumoral , Marcação de Genes , Humanos , Mutação INDEL , Camundongos , Oligonucleotídeos/genética , Ratos , Peixe-Zebra
2.
Sci Rep ; 5: 14410, 2015 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-26442875

RESUMO

The generation of genetically-modified organisms has been revolutionized by the development of new genome editing technologies based on the use of gene-specific nucleases, such as meganucleases, ZFNs, TALENs and CRISPRs-Cas9 systems. The most rapid and cost-effective way to generate genetically-modified animals is by microinjection of the nucleic acids encoding gene-specific nucleases into zygotes. However, the efficiency of the procedure can still be improved. In this work we aim to increase the efficiency of CRISPRs-Cas9 and TALENs homology-directed repair by using TALENs and Cas9 proteins, instead of mRNA, microinjected into rat and mouse zygotes along with long or short donor DNAs. We observed that Cas9 protein was more efficient at homology-directed repair than mRNA, while TALEN protein was less efficient than mRNA at inducing homology-directed repair. Our results indicate that the use of Cas9 protein could represent a simple and practical methodological alternative to Cas9 mRNA in the generation of genetically-modified rats and mice as well as probably some other mammals.


Assuntos
Sistemas CRISPR-Cas/genética , Engenharia de Proteínas , Reparo de DNA por Recombinação/genética , Zigoto/fisiologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Microinjeções , Ratos , Ratos Sprague-Dawley
3.
FASEB J ; 27(2): 703-11, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23150522

RESUMO

Despite the recent availability of gene-specific nucleases, such as zinc-finger nucleases (ZFNs) and transcription activator-like nucleases (TALENs), there is still a need for new tools to modify the genome of different species in an efficient, rapid, and less costly manner. One aim of this study was to apply, for the first time, engineered meganucleases to mutate an endogenous gene in animal zygotes. The second aim was to target the mouse and rat recombination activating gene 1 (Rag1) to describe, for the first time, Rag1 knockout immunodeficient rats. We microinjected a plasmid encoding a meganuclease for Rag1 into the pronucleus of mouse and rat zygotes. Mutant animals were detected by PCR sequencing of the targeted sequence. A homozygous RAG1-deficient rat line was generated and immunophenotyped. Meganucleases were efficient, because 3.4 and 0.6% of mouse and rat microinjected zygotes, respectively, generated mutated animals. RAG1-deficient rats showed significantly decreased proportions and numbers of immature and mature T and B lymphocytes and normal NK cells vs. littermate wild-type controls. In summary, we describe the use of engineered meganucleases to inactivate an endogenous gene with efficiencies comparable to those of ZFNs and TALENs. Moreover, we generated an immunodeficient rat line useful for studies in which there is a need for biological parameters to be analyzed in the absence of immune responses.


Assuntos
Técnicas de Inativação de Genes/métodos , Genes RAG-1 , Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas de Homeodomínio/genética , Animais , Sequência de Bases , DNA/administração & dosagem , DNA/genética , Endonucleases/genética , Endonucleases/metabolismo , Marcação de Genes/métodos , Engenharia Genética/métodos , Transplante de Coração/imunologia , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/metabolismo , Imunofenotipagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microinjeções , Dados de Sequência Molecular , Ratos , Ratos Endogâmicos Lew , Transplante Homólogo
4.
BMC Cancer ; 10: 395, 2010 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-20663219

RESUMO

BACKGROUND: Mutations of the MEN1 gene predispose to multiple endocrine neoplasia type 1 (MEN1) syndrome. Our group and others have shown that Men1 disruption in mice recapitulates MEN1 pathology. Intriguingly, rare lesions in hormone-dependent tissues, such as prostate and mammary glands, were also observed in the Men1 mutant mice. METHODS: To study the occurrence of prostate lesions, we followed a male mouse cohort of 47 Men1+/- mice and 23 age-matched control littermates, starting at 18 months of age, and analysed the prostate glands from the cohort. RESULTS: Six Men1+/- mice (12.8%) developed prostate cancer, including two adenocarcinomas and four in situ carcinomas, while none of the control mice developed cancerous lesions. The expression of menin encoded by the Men1 gene was found to be drastically reduced in all carcinomas, and partial LOH of the wild-type Men1 allele was detected in three of the five analysed lesions. Using immunostaining for the androgen receptor and p63, a basal epithelial cell marker, we demonstrated that the menin-negative prostate cancer cells did not display p63 expression and that the androgen receptor was expressed but more heterogeneous in these lesions. Furthermore, our data showed that the expression of the cyclin-dependent kinase inhibitor CDKN1B (p27), a Men1 target gene known to be inactivated during prostate cell tumorigenesis, was notably decreased in the prostate cancers that developed in the mutant mice. CONCLUSION: Our work suggests the possible involvement of Men1 inactivation in the tumorigenesis of the prostate gland.


Assuntos
Adenocarcinoma/genética , Adenocarcinoma/patologia , Perda de Heterozigosidade , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas/fisiologia , Envelhecimento , Animais , Southern Blotting , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Heterozigoto , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfoproteínas/metabolismo , Receptores Androgênicos/metabolismo , Transativadores/metabolismo
5.
BMC Genet ; 9: 83, 2008 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-19061508

RESUMO

BACKGROUND: The Nonsense-Mediated mRNA Decay (NMD) pathway detects and degrades mRNAs containing premature termination codons, thereby preventing the accumulation of potentially detrimental truncated proteins. Intertissue variation in the efficiency of this mechanism has been suggested, which could have important implications for the understanding of genotype-phenotype correlations in various genetic disorders. However, compelling evidence in favour of this hypothesis is lacking. Here, we have explored this question by measuring the ratio of mutant versus wild-type Men1 transcripts in thirteen tissues from mice carrying a heterozygous truncating mutation in the ubiquitously expressed Men1 gene. RESULTS: Significant differences were found between two groups of tissues. The first group, which includes testis, ovary, brain and heart, displays a strong decrease of the nonsense transcript (average ratio of 18% of mutant versus wild-type Men1 transcripts, identical to the value measured in murine embryonic fibroblasts). The second group, comprising lung, intestine and thymus, shows much less pronounced NMD (average ratio of 35%). Importantly, the extent of degradation by NMD does not correlate with the expression level of eleven genes encoding proteins involved in NMD or with the expression level of the Men1 gene. CONCLUSION: Mouse models are an attractive option to evaluate the efficiency of NMD in multiple mammalian tissues and organs, given that it is much easier to obtain these from a mouse than from a single individual carrying a germline truncating mutation. In this study, we have uncovered in the thirteen different murine tissues that we examined up to a two-fold difference in NMD efficiency.


Assuntos
Códon sem Sentido/genética , Especificidade de Órgãos/genética , Estabilidade de RNA/genética , Animais , Regulação da Expressão Gênica , Camundongos , Modelos Animais , Modelos Genéticos , Mutação/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
6.
J Endocrinol ; 199(2): 287-98, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18772165

RESUMO

Mutations of the multiple endocrine neoplasia type 1 (MEN1) gene predispose patients to MEN1 that affects mainly endocrine tissues, suggesting important physiological functions of the gene in adult endocrine cells. Homozygous disruption of Men1 in mice causes embryonic lethality, whereas the eventual involvement of the gene in embryonic development of the endocrine cells remains unknown. Here, we show that homozygous Men1 knockout mice demonstrate a reduced number of glucagon-positive cells in the E12.5 pancreatic bud associated with apoptosis, whereas the exocrine pancreas development in these mice is not affected. Our data suggest that menin is involved in the survival of the early pancreatic endocrine cells during the first developmental transition. Furthermore, chimerism assay revealed that menin has an autonomous and specific effect on the development of islet cells. In addition, using pancreatic bud culture mimicking the differentiation of alpha- and beta-cells during the second transition, we show that loss of menin leads to the failure of endocrine cell development, altered pancreatic structure and a markedly decreased number of cells expressing neurogenin 3, indicating that menin is also required at this stage of the endocrine pancreas development. Taken together, our results suggest that menin plays an indispensable role in the development of the pancreatic endocrine cells.


Assuntos
Células Endócrinas/citologia , Células Endócrinas/metabolismo , Pâncreas/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Animais , Apoptose/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Imunofluorescência , Imuno-Histoquímica , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Técnicas In Vitro , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Pâncreas/citologia , Pâncreas/embriologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Endocr Relat Cancer ; 15(1): 217-27, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18310289

RESUMO

Multiple endocrine neoplasia type 1 (MEN1) results from the mutation of the predisposing gene, MEN1. Heterozygous Men1 mutant mice previously generated by several laboratories, including ours, mimic largely MEN1 pathology. Interestingly, our heterozygous Men1 mutant mice exhibit not only the endocrine tumours commonly seen in MEN1 patients, but also Leydig cell tumours (LCT) with high frequency, accompanied systematically by loss of the wild-type Men1 allele. As there exists a similarity of tumour phenotype between these mice and those mutated for the components of anti-Mullerian hormone (AMH)/bone morphogenic protein (BMP) pathway belonging to transforming growth factor-beta (TGF-beta) family, we investigated the expression and the activity of this pathway, known to have an important biological role in Leydig cells. Here, we report that the expression of AMH receptor type 2 is reduced in Men1 LCTs. Both immunostaining and western blot analyses also demonstrate a markedly decreased nuclear expression of Smad1, 3, 4 and 5 in the tumours. More interestingly, we show that the reconstituted menin expression in Men1-deficient Leydig cells derived from LCTs can significantly increase the transcriptional activity of a BMP pathway target promoter, XVent2. Furthermore, we found that the expression of p18, p27 and cyclin dependant kinase 4 (Cdk4), targets of TGF-beta pathways, is altered in the Leydig cell lesions. Our data provide the evidence of the deregulation of AMH/BMP and TGF-beta pathways in mouse Men1 LCTs, highlighting their involvement in tumorigenesis of Leydig cells due to Men1 inactivation.


Assuntos
Hormônio Antimülleriano/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Tumor de Células de Leydig/metabolismo , Neoplasia Endócrina Múltipla Tipo 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Northern Blotting , Western Blotting , Inibidor de Quinase Dependente de Ciclina p18/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Heterozigoto , Técnicas Imunoenzimáticas , Imunoprecipitação , Tumor de Células de Leydig/patologia , Luciferases/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neoplasia Endócrina Múltipla Tipo 1/patologia , Plasmídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Peptídeos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Smad/metabolismo
8.
Eur J Cancer ; 43(2): 402-14, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17184987

RESUMO

Multiple endocrine neoplasia type 1 (MEN1) is a hereditary syndrome caused by the inactivation of the responsible gene, MEN1. To date, the lack of MEN1-deficient cell lines derived directly from MEN1 tumours has hampered the detailed study of the MEN1 gene. We have established several stable Men1-deficient Leydig cell tumour (LCT) lines derived from a Leydig cell tumour developed in a male heterozygous Men1 mutant mouse. Our data show that these cell lines maintain the basic characteristics of Leydig cells in terms of both androgen synthesis and gene expression. Interestingly, reconstituted menin expression in one of Men1-deficient LCT cell lines resulted in cell growth inhibition, suggesting that the function of cell growth suppression of the menin pathway, apart from menin itself, is essentially preserved in these cells. Furthermore, we show that menin re-expression in these Men1-deficient cells leads to a block in the transition from G0/G1 to S phase of the cell cycle and an increase in apoptosis, accompanied by a marked increase of p18INK4C and p27Kip1 expression. The current study therefore highlights the importance of menin expression in cell cycle and cell survival control in endocrine cells, and may provide insights into the mechanisms of tumour suppression by menin in related endocrine tumours.


Assuntos
Apoptose/genética , Ciclo Celular/genética , Tumor de Células de Leydig/genética , Neoplasia Endócrina Múltipla Tipo 1/genética , Mutação/genética , Proteínas Proto-Oncogênicas/genética , Animais , Linhagem Celular Tumoral , Imuno-Histoquímica , Tumor de Células de Leydig/metabolismo , Perda de Heterozigosidade/genética , Masculino , Camundongos , Neoplasia Endócrina Múltipla Tipo 1/metabolismo , Progesterona/metabolismo , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/metabolismo
9.
Fam Cancer ; 5(1): 49-54, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16528608

RESUMO

Multiple Endocrine Neoplasia type 1 (MEN1) is a hereditary disease characterised by the occurrence of multiple endocrine tumours. The biological functions of the responsible gene, MEN1, and its encoded protein, menin, remain so far largely elusive. The recent generation of Men1 mutant mice by our group and other laboratories provides powerful tools allowing for the identification of cellular and molecular events that occur after gene disruption. Interestingly, it has been recently reported that p27(Kip1) expression is regulated by menin and that decreased p27(Kip1) expression can be found in MEN1 insulinomas and parathyroid adenomas. In order to address whether and when p27(Kip1) expression alters during insulinoma development in pancreatic beta-cell-specific Men1 mutant mice, we analysed p27(Kip1) expression in islet lesions from mutant mice at different ages. Our data revealed that p27(Kip1) protein expression was reduced in 40 out of 52 (77%) insulinomas analysed, whereas the remaining 12 insulinomas (23%) did not show altered p27(Kip1) expression. No difference between the insulinomas with and without decreased p27(Kip1) expression could be observed in terms of histological features or menin inactivation. Furthermore, our analysis on hyperplastic and dysplastic islets developed in young mutant mice showed the lack of detectable alteration in p27(Kip1) expression, despite evident loss of menin expression in a substantial proportion of islet cells. Our work confirms the altered p27(Kip1) expression reported in tumours from MEN1 patients, whereas it suggests that other molecular events may also participate in the tumorigenesis process initiated by the Men1 gene inactivation.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/genética , Regulação Neoplásica da Expressão Gênica , Insulinoma/genética , Neoplasia Endócrina Múltipla Tipo 1/genética , Neoplasias Pancreáticas/genética , Animais , Western Blotting , Sobrevivência Celular , Modelos Animais de Doenças , Regulação para Baixo , Deleção de Genes , Imuno-Histoquímica , Insulinoma/patologia , Ilhotas Pancreáticas/citologia , Camundongos , Camundongos Mutantes , Neoplasia Endócrina Múltipla Tipo 1/patologia , Neoplasias Pancreáticas/patologia , Probabilidade , Proteínas Proto-Oncogênicas/genética , Sensibilidade e Especificidade , Células Tumorais Cultivadas
10.
J Biol Chem ; 279(23): 24477-84, 2004 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-15054094

RESUMO

Menin, the product of the tumor suppressor gene MEN1, is widely expressed in mammalian endocrine and non-endocrine tissues, including intestine. Its known abundant expression in several types of cells with high proliferative capacity led us to investigate the physiological function of the protein menin in intestinal epithelium, one of the most rapidly growing epithelia. Here we showed that the Men1 gene is mainly expressed in the crypt compartment of the proximal small intestine and that its expression was increased during fasting in vivo, both suggesting a role of menin in the control of cell growth. Indeed, specific reduction of menin expression by transfected antisense cDNA in the rat duodenal crypt-like cell line, IEC-17, increased cell proliferation. The latter is correlated to a loss of cell-cycle arrest in G(1) phase by resting cells and an overexpression of cyclin D1 and cyclin-dependent kinase (Cdk)-4. Furthermore, these cells lost the inhibition of proliferation induced by transforming growth factor-beta1, associated with a decrease of transforming growth factor-beta type II receptor expression. As a result of deregulated proliferation, antisense menin transfected IEC-17 cells became tumorigenic as shown in vitro as well as in vivo in immunosuppressed animals. These results indicate that menin contributes to proliferation control in intestinal epithelial cells. The present study reveals an unknown physiological function for menin in intestine that may be important in the regulation of epithelial homeostasis.


Assuntos
Células Epiteliais/metabolismo , Intestinos/citologia , Proteínas Proto-Oncogênicas/biossíntese , Ágar/metabolismo , Animais , Western Blotting , Ciclo Celular , Divisão Celular , Linhagem Celular , Separação Celular , Ciclina D1/metabolismo , Quinase 4 Dependente de Ciclina , Quinases Ciclina-Dependentes/metabolismo , Proteínas do Citoesqueleto/metabolismo , DNA Complementar/metabolismo , Regulação para Baixo , Jejum , Citometria de Fluxo , Fase G1 , Heterozigoto , Imuno-Histoquímica , Terapia de Imunossupressão , Hibridização In Situ , Intestino Delgado/metabolismo , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Oligonucleotídeos Antissenso/química , Oligonucleotídeos Antissenso/farmacologia , Plasmídeos/metabolismo , Proteínas Serina-Treonina Quinases , RNA Mensageiro/metabolismo , Ratos , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fatores de Tempo , Transativadores/metabolismo , Transfecção , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1 , beta Catenina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA