Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Mol Psychiatry ; 21(10): 1417-33, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26830142

RESUMEN

Social interaction is a fundamental behavior in all animal species, but the developmental timing of the social neural circuit formation and the cellular and molecular mechanisms governing its formation are poorly understood. We generated a mouse model with mutations in two Disheveled genes, Dvl1 and Dvl3, that displays adult social and repetitive behavioral abnormalities associated with transient embryonic brain enlargement during deep layer cortical neuron formation. These phenotypes were mediated by the embryonic expansion of basal neural progenitor cells (NPCs) via deregulation of a ß-catenin/Brn2/Tbr2 transcriptional cascade. Transient pharmacological activation of the canonical Wnt pathway during this period of early corticogenesis rescued the ß-catenin/Brn2/Tbr2 transcriptional cascade and the embryonic brain phenotypes. Remarkably, this embryonic treatment prevented adult behavioral deficits and partially rescued abnormal brain structure in Dvl mutant mice. Our findings define a mechanism that links fetal brain development and adult behavior, demonstrating a fetal origin for social and repetitive behavior deficits seen in disorders such as autism.


Asunto(s)
Trastorno de Movimiento Estereotipado/genética , Trastorno de Movimiento Estereotipado/fisiopatología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Conducta Animal , Encéfalo/embriología , Encéfalo/metabolismo , Encéfalo/fisiología , Proteínas Dishevelled/genética , Proteínas Dishevelled/metabolismo , Humanos , Ratones , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/fisiología , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Factores del Dominio POU/metabolismo , Factores del Dominio POU/fisiología , Fosfoproteínas/genética , Transducción de Señal/fisiología , Conducta Estereotipada/fisiología , Proteínas de Dominio T Box/metabolismo , Proteínas de Dominio T Box/fisiología , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/genética , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo , beta Catenina/fisiología
2.
Mol Psychiatry ; 17(4): 451-66, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22124272

RESUMEN

Complex neuropsychiatric disorders are believed to arise from multiple synergistic deficiencies within connected biological networks controlling neuronal migration, axonal pathfinding and synapse formation. Here, we show that deletion of 14-3-3ζ causes neurodevelopmental anomalies similar to those seen in neuropsychiatric disorders such as schizophrenia, autism spectrum disorder and bipolar disorder. 14-3-3ζ-deficient mice displayed striking behavioural and cognitive deficiencies including a reduced capacity to learn and remember, hyperactivity and disrupted sensorimotor gating. These deficits are accompanied by subtle developmental abnormalities of the hippocampus that are underpinned by aberrant neuronal migration. Significantly, 14-3-3ζ-deficient mice exhibited abnormal mossy fibre navigation and glutamatergic synapse formation. The molecular basis of these defects involves the schizophrenia risk factor, DISC1, which interacts isoform specifically with 14-3-3ζ. Our data provide the first evidence of a direct role for 14-3-3ζ deficiency in the aetiology of neurodevelopmental disorders and identifies 14-3-3ζ as a central risk factor in the schizophrenia protein interaction network.


Asunto(s)
Proteínas 14-3-3/fisiología , Encéfalo/crecimiento & desarrollo , Predisposición Genética a la Enfermedad/genética , Hipocampo/crecimiento & desarrollo , Neurogénesis/fisiología , Esquizofrenia/genética , Proteínas 14-3-3/genética , Animales , Encéfalo/metabolismo , Movimiento Celular/genética , Movimiento Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Hipocampo/metabolismo , Hipocampo/patología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/fisiología , Neurogénesis/genética , Neuronas/metabolismo , Neuronas/fisiología , Reconocimiento en Psicología/fisiología , Filtrado Sensorial/fisiología
3.
Nat Genet ; 17(4): 453-6, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9398849

RESUMEN

Atm is part of a pathway that responds to DNA damage from ionizing radiation (IR). This pathway involves p53, as Atm-deficient cell lines and mice are defective in p53 induction after IR. p53 is a multi-functional protein that simultaneously regulates distinct downstream pathways controlling cell-cycle progression and apoptosis. However, the mechanisms by which p53 differentially activates downstream pathways are unknown. To determine the relationship between Atm and p53, we examined cell-cycle and apoptotic responses in Atm-, p53-(ref. 8) and p21-deficient mice after IR in the whole animal. As expected, p53 protein levels were not induced by IR in thymus of Atm-deficient mice. IR-induced cell-cycle checkpoint function was also defective, and induction of p21 was attenuated in thymus from Atm-deficient mice. However, IR-induced apoptosis and Bax induction were completely normal; both of which are mediated by p53. IR-induced thymic apoptosis was suppressed in Atm/p53 double-mutant mice but not in Atm/p21 double mutants, demonstrating p53 dependence and Atm independence. Thus, Atm deficiency results in lack of p53 induction by IR, but only selective disruption of p53-dependent functions. Our results support a model in which upstream effectors such as Atm selectively activate p53 to regulate specific downstream pathways, providing a mechanism for controlling distinct cell-cycle and apoptotic responses.


Asunto(s)
Apoptosis/genética , Ataxia Telangiectasia/genética , Ciclo Celular/genética , Proteínas Serina-Treonina Quinasas , Proteínas/genética , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis/efectos de la radiación , Ataxia Telangiectasia/patología , Proteínas de la Ataxia Telangiectasia Mutada , Ciclo Celular/efectos de la radiación , Proteínas de Ciclo Celular , Fragmentación del ADN/efectos de la radiación , ADN Nucleotidilexotransferasa , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Humanos , Hibridación in Situ , Pulmón/metabolismo , Pulmón/efectos de la radiación , Ratones , Ratones Noqueados , Proteínas/fisiología , Timo/metabolismo , Timo/efectos de la radiación , Proteínas Supresoras de Tumor , Irradiación Corporal Total
4.
Nat Genet ; 17(4): 462-6, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9398851

RESUMEN

Patients with the human disorder ataxia-telangiectasia (A-T; refs 1,2) and Atm-deficient mice have a pleiotropic phenotype that includes infertility. Here we demonstrate that male gametogenesis is severely disrupted in Atm-deficient mice in the earliest stages of meiotic prophase I, resulting in apoptotic degeneration. Atm is required for proper assembly of Rad51 onto the chromosomal axial elements during meiosis. In addition, p53, p21 and Bax are elevated in testes from Atm-deficient mice. To determine whether these elevated protein levels are important factors in the meiotic disruption of Atm-deficient mice, we analysed the meiotic phenotype of Atm/p53 or Atm/p21 double mutants. In these double mutants, meiosis progressed to later stages but was only partly rescued. Assembly of Rad51 foci on axial elements remained defective, and gametogenesis proceeded only to pachytene of prophase I. Previous results demonstrated that mice homozygous for a null mutation in Rad51 (ref. 6) display an early embryonic lethal phenotype that can be partly rescued by removing p53 and/or p21. Because Atm-deficient mice are viable but completely infertile, our studies suggest that the Rad51 assembly defects and elevated levels of p53, p21 and Bax represent tissue-specific responses to the absence of Atm.


Asunto(s)
Ataxia Telangiectasia/genética , Ciclinas/genética , Genes p53 , Profase/genética , Proteínas Serina-Treonina Quinasas , Proteínas/genética , Animales , Apoptosis/genética , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Proteínas de Unión al ADN/genética , Humanos , Masculino , Ratones , Ratones Noqueados , Recombinasa Rad51 , Espermatocitos/metabolismo , Espermatocitos/ultraestructura , Complejo Sinaptonémico/genética , Proteínas Supresoras de Tumor
5.
Nat Genet ; 11(1): 17-26, 1995 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-7550308

RESUMEN

We have examined the developmental expression of the murine breast and ovarian cancer susceptibility gene, Brca1, to investigate its role in the control of cell growth and differentiation. Specifically, we have analysed Brca1 expression during embryonic development, in adult tissues, and during postnatal mammary gland development, particularly in response to ovarian hormones. Our results suggest that Brca1 is expressed in rapidly proliferating cell types undergoing differentiation. In the mammary gland, Brca1 expression is induced during puberty, pregnancy, and following treatment of ovariectomized animals with 17 beta-estradiol and progesterone. These observations imply that Brca1 is involved in the processes of proliferation and differentiation in multiple tissues, notably in the mammary gland in response to ovarian hormones.


Asunto(s)
Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Glándulas Mamarias Animales/metabolismo , Proteínas de Neoplasias/biosíntesis , Factores de Transcripción/biosíntesis , Animales , Proteína BRCA1 , Secuencia de Bases , División Celular/genética , Células Cultivadas , Desarrollo Embrionario y Fetal , Epitelio/metabolismo , Estradiol/farmacología , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Hibridación in Situ , Lactancia , Masculino , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/embriología , Glándulas Mamarias Animales/crecimiento & desarrollo , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Oligonucleótidos Antisentido , Especificidad de Órganos , Ovariectomía , Paridad , Embarazo , Progesterona/farmacología , Maduración Sexual , Factores de Transcripción/genética
6.
Nat Genet ; 22(1): 37-43, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10319859

RESUMEN

Cre-mediated excision of exon 11 of the breast-tumour suppressor gene Brca1 in mouse mammary epithelial cells causes increased apoptosis and abnormal ductal development. Mammary tumour formation occurs after long latency and is associated with genetic instability characterized by aneuploidy, chromosomal rearrangements or alteration of Trp53 (encoding p53) transcription. To directly test the role of p53 in Brca1-associated tumorigenesis, we introduced a Trp53-null allele into mice with mammary epithelium-specific inactivation of Brca1. The loss of p53 accelerated the formation of mammary tumours in these females. Our results demonstrate that disruption of Brca1 causes genetic instability and triggers further alterations, including the inactivation of p53, that lead to tumour formation.


Asunto(s)
Proteína BRCA1/genética , Células Epiteliales/metabolismo , Glándulas Mamarias Animales/crecimiento & desarrollo , Neoplasias Mamarias Animales/genética , Alelos , Animales , Apoptosis/genética , Línea Celular , Células Epiteliales/patología , Femenino , Genes p53/genética , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Animales/patología , Ratones , Ratones Endogámicos , Mutación , Translocación Genética
7.
Nat Genet ; 19(4): 333-9, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9697693

RESUMEN

Heterozygous mutation or deletion of the beta subunit of platelet-activating factor acetylhydrolase (PAFAH1B1, also known as LIS1) in humans is associated with type I lissencephaly, a severe developmental brain disorder thought to result from abnormal neuronal migration. To further understand the function of PAFAH1B1, we produced three different mutant alleles in mouse Pafah1b1. Homozygous null mice die early in embryogenesis soon after implantation. Mice with one inactive allele display cortical, hippocampal and olfactory bulb disorganization resulting from delayed neuronal migration by a cell-autonomous neuronal pathway. Mice with further reduction of Pafah1b1 activity display more severe brain disorganization as well as cerebellar defects. Our results demonstrate an essential, dosage-sensitive neuronal-specific role for Pafah1b1 in neuronal migration throughout the brain, and an essential role in early embryonic development. The phenotypes observed are distinct from those of other mouse mutants with neuronal migration defects, suggesting that Pafah1b1 participates in a novel pathway for neuronal migration.


Asunto(s)
Anomalías Múltiples/patología , Movimiento Celular/fisiología , Corteza Cerebral/patología , Proteínas Asociadas a Microtúbulos , Neuronas/citología , Proteínas/fisiología , 1-Alquil-2-acetilglicerofosfocolina Esterasa , Anomalías Múltiples/genética , Animales , Células Cultivadas , Cerebelo/anomalías , Corteza Cerebral/anomalías , Corteza Cerebral/embriología , Desarrollo Embrionario y Fetal , Genotipo , Hipocampo/patología , Ratones , Ratones Noqueados , Bulbo Olfatorio/patología , Proteínas/genética
8.
Nat Genet ; 14(3): 312-5, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8896561

RESUMEN

Murine models of human carcinogenesis are exceedingly valuable tools to understand genetic mechanisms of neoplastic growth. The identification of recurrent chromosomal rearrangements by cytogenetic techniques serves as an initial screening test for tumour specific aberrations. In murine models of human carcinogenesis, however, karyotype analysis is technically demanding because mouse chromosomes are acrocentric and of similar size. Fluorescence in situ hybridization (FISH) with mouse chromosome specific painting probes can complement conventional banding analysis. Although sensitive and specific, FISH analyses are restricted to the visualization of only a few mouse chromosomes at a time. Here we apply a novel imaging technique that we developed recently for the visualization of human chromosomes to the simultaneous discernment of all mouse chromosomes. The approach is based on spectral imaging to measure chromosome-specific spectra after FISH with differentially labelled mouse chromosome painting probes. Utilizing a combination of Fourier spectroscopy, CCD-imaging and conventional optical microscopy, spectral imaging allows simultaneous measurement of the fluorescence emission spectrum at all sample points. A spectrum-based classification algorithm has been adapted to karyotype mouse chromosomes. We have applied spectral karyotyping (SKY) to chemically induced plasmocytomas, mammary gland tumours from transgenic mice overexpressing the c-myc oncogene and thymomas from mice deficient for the ataxia telangiectasia (Atm) gene. Results from these analyses demonstrate the potential of SKY to identify complex chromosomal aberrations in mouse models of human carcinogenesis.


Asunto(s)
Aberraciones Cromosómicas , Cromosomas , Cariotipificación/métodos , Proteínas Serina-Treonina Quinasas , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Genes myc , Humanos , Hibridación Fluorescente in Situ/métodos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos , Ratones Transgénicos , Neoplasias/genética , Plasmacitoma/genética , Proteínas/genética , Proteínas Supresoras de Tumor
9.
Nat Cell Biol ; 2(11): 767-75, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11056530

RESUMEN

Whereas total loss of Lis1 is lethal, disruption of one allele of the Lis1 gene results in brain abnormalities, indicating that developing neurons are particularly sensitive to a reduction in Lis1 dosage. Here we show that Lis1 is enriched in neurons relative to levels in other cell types, and that Lis1 interacts with the microtubule motor cytoplasmic dynein. Production of more Lis1 in non-neuronal cells increases retrograde movement of cytoplasmic dynein and leads to peripheral accumulation of microtubules. These changes may reflect neuron-like dynein behaviours induced by abundant Lis1. Lis1 deficiency produces the opposite phenotype. Our results indicate that abundance of Lis1 in neurons may stimulate specific dynein functions that function in neuronal migration and axon growth.


Asunto(s)
Dineínas/metabolismo , Proteínas Asociadas a Microtúbulos/biosíntesis , Centro Organizador de los Microtúbulos/fisiología , Microtúbulos/fisiología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Células COS , Centrómero/fisiología , Chlorocebus aethiops , Citoplasma/metabolismo , Complejo Dinactina , Fibroblastos/citología , Aparato de Golgi/metabolismo , Aparato de Golgi/fisiología , Interfase/fisiología , Membranas Intracelulares/metabolismo , Membranas Intracelulares/fisiología , Mamíferos , Proteínas Asociadas a Microtúbulos/metabolismo , Centro Organizador de los Microtúbulos/metabolismo , Microtúbulos/metabolismo , Neuronas/metabolismo , Ratas
10.
Neuron ; 28(3): 681-96, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11163259

RESUMEN

Mutations in mammalian Lis1 (Pafah1b1) result in neuronal migration defects. Several lines of evidence suggest that LIS1 participates in pathways regulating microtubule function, but the molecular mechanisms are unknown. Here, we demonstrate that LIS1 directly interacts with the cytoplasmic dynein heavy chain (CDHC) and NUDEL, a murine homolog of the Aspergillus nidulans nuclear migration mutant NudE. LIS1 and NUDEL colocalize predominantly at the centrosome in early neuroblasts but redistribute to axons in association with retrograde dynein motor proteins. NUDEL is phosphorylated by Cdk5/p35, a complex essential for neuronal migration. NUDEL and LIS1 regulate the distribution of CDHC along microtubules, and establish a direct functional link between LIS1, NUDEL, and microtubule motors. These results suggest that LIS1 and NUDEL regulate CDHC activity during neuronal migration and axonal retrograde transport in a Cdk5/p35-dependent fashion.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Dineínas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Sistema Nervioso/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterasa , Animales , Aspergillus nidulans , Transporte Axonal/fisiología , Movimiento Celular , Células Cultivadas , Centrosoma/metabolismo , Quinasa 5 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Citoplasma/metabolismo , Proteínas Fúngicas/genética , Humanos , Sustancias Macromoleculares , Ratones , Ratones Mutantes , Proteínas Asociadas a Microtúbulos/genética , Microtúbulos/metabolismo , Datos de Secuencia Molecular , Sistema Nervioso/citología , Sistema Nervioso/embriología , Neuronas/citología , Neuronas/metabolismo , Fosforilación , Subunidades de Proteína , Homología de Secuencia de Aminoácido , Células Madre/citología , Células Madre/metabolismo , Técnicas del Sistema de Dos Híbridos
13.
J Clin Invest ; 101(10): 2042-53, 1998 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-9593760

RESUMEN

The oculocerebrorenal syndrome of Lowe (OCRL) is an X-linked human genetic disorder characterized by mental retardation, congenital cataracts, and renal tubular dysfunction. The Lowe syndrome gene, OCRL1, encodes a phosphatidylinositol 4,5-bisphosphate 5-phosphatase in the Golgi complex. The pathogenesis of Lowe syndrome due to deficiency of a phosphatidylinositol 4,5-bisphosphate 5-phosphatase in the Golgi complex is unknown. We have used targeted disruption in embryonic stem cells to make mice deficient in Ocrl1, the mouse homologue for OCRL1, as an animal model for the disease. Surprisingly, mice deficient in Ocrl1 do not develop the congenital cataracts, renal Fanconi syndrome, or neurological abnormalities seen in the human disorder. We hypothesized that Ocrl1 deficiency is complemented in mice by inositol polyphosphate 5-phosphatase (Inpp5b), an autosomal gene that encodes a phosphatidylinositol bisphosphate 5-phosphatase highly homologous to Ocrl1. We created mice deficient in Inpp5b; the mice were viable and fertile without phenotype except for testicular degeneration in males beginning after sexual maturation. We crossed mice deficient in Ocrl1 to mice deficient in Inpp5b. No liveborn mice or embryos lacking both enzymes were found, demonstrating that Ocrl1 and Inpp5b have overlapping functions in mice and suggesting that the lack of phenotype in Ocrl1-deficient mice may be due to compensating Inpp5b function.


Asunto(s)
Síndrome Oculocerebrorrenal/genética , Monoéster Fosfórico Hidrolasas/genética , Proteínas/genética , Secuencia de Aminoácidos , Aminoácidos/orina , Animales , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Expresión Génica/genética , Marcación de Gen , Humanos , Inositol Polifosfato 5-Fosfatasas , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Actividad Motora/fisiología , Síndrome Oculocerebrorrenal/fisiopatología , Fenotipo , Monoéster Fosfórico Hidrolasas/fisiología , Proteínas/fisiología , Empalme del ARN , ARN Mensajero/metabolismo , Células Madre
14.
Mol Cell Biol ; 19(4): 3095-102, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10082576

RESUMEN

Both p53 and ATM are checkpoint regulators with roles in genetic stabilization and cancer susceptibility. ATM appears to function in the same DNA damage checkpoint pathway as p53. However, ATM's role in p53-dependent apoptosis and tumor suppression in response to cell cycle dysregulation is unknown. In this study, we tested the role of murine ataxia telangiectasia protein (Atm) in a transgenic mouse brain tumor model in which p53-mediated apoptosis results in tumor suppression. These p53-mediated activities are induced by tissue-specific inactivation of pRb family proteins by a truncated simian virus 40 large T antigen in brain epithelium. We show that p53-dependent apoptosis, transactivation, and tumor suppression are unaffected by Atm deficiency, suggesting that signaling in the DNA damage pathway is distinct from that in the oncogene-induced pathway. In addition, we show that Atm deficiency has no overall effect on tumor growth and progression in this model.


Asunto(s)
Apoptosis/genética , Transformación Celular Neoplásica/genética , Proteínas Serina-Treonina Quinasas , Proteínas/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Antígenos Virales de Tumores , Proteínas de la Ataxia Telangiectasia Mutada , Neoplasias Encefálicas/etiología , Neoplasias Encefálicas/genética , Proteínas de Ciclo Celular , Plexo Coroideo/patología , Proteínas de Unión al ADN , Epitelio/patología , Ratones , Ratones Transgénicos , Proteína de Retinoblastoma , Virus 40 de los Simios , Activación Transcripcional , Proteínas Supresoras de Tumor
15.
Mol Cell Biol ; 21(5): 1828-32, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11238919

RESUMEN

PARP-1 and ATM are both involved in the response to DNA strand breaks, resulting in induction of a signaling network responsible for DNA surveillance, cellular recovery, and cell survival. ATM interacts with double-strand break repair pathways and induces signals resulting in the control of the cell cycle-coupled checkpoints. PARP-1 acts as a DNA break sensor in the base excision repair pathway of DNA. Mice with mutations inactivating either protein show radiosensitivity and high radiation-induced chromosomal aberration frequencies. Embryos carrying double mutations of both PARP-1 and Atm genes were generated. These mutant embryos show apoptosis in the embryo but not in extraembryonic tissues and die at embryonic day 8.0, although extraembryonic tissues appear normal for up to 10.5 days of gestation. These results reveal a functional synergy between PARP-1 and ATM during a period of embryogenesis when cell cycle checkpoints are not active and the embryo is particularly sensitive to DNA damage. These results suggest that ATM and PARP-1 have synergistic phenotypes due to the effects of these proteins on signaling DNA damage and/or on distinct pathways of DNA repair.


Asunto(s)
Mutación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas/genética , Proteínas/fisiología , Animales , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada , Ciclo Celular , Proteínas de Ciclo Celular , Muerte Celular , División Celular , Supervivencia Celular , Aberraciones Cromosómicas , Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN , Embrión de Mamíferos/metabolismo , Genotipo , Heterocigoto , Ratones , Microscopía Electrónica , Modelos Biológicos , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas , Reacción en Cadena de la Polimerasa , Tolerancia a Radiación , Transducción de Señal , Factores de Tiempo , Proteínas Supresoras de Tumor
16.
Cancer Res ; 60(2): 395-9, 2000 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-10667593

RESUMEN

Ataxia telangiectasia (AT) patients have inactivating mutations in both copies of the ATM gene. The ATM protein that the gene encodes is involved in DNA double-strand break (DSB) recognition; in its absence, p53 response to DSBs is delayed and reduced. In addition, AT patients have a high propensity for cancer, and cells from these patients show chromosomal instability. Here, using an in vivo mouse model system with the pink-eyed unstable mutation, we demonstrate that the absence of functional Atm results in a significantly elevated frequency of intrachromosomal recombination resulting in deletion events (wild-type 17.73%, heterozygous Atm 15.72%, and mutant Atm 30.33%). No such increase was observed in mice heterozygous for Atm. These results further advocate the role of ATM in maintaining genomic integrity after the onset of endogenous damage. This system relies on the initiation of events during a relatively short time frame to produce an observable deletion product. AT patients have a lifelong exposure to endogenous damage and perhaps similarly acting external agents. Because 25% of our genome consists of repeated elements, genomic instability due to an increased level of homologous recombination between such repeats, as observed here, may contribute to carcinogenesis in AT patients.


Asunto(s)
Ataxia Telangiectasia/genética , Proteínas Serina-Treonina Quinasas/fisiología , Recombinación Genética , Eliminación de Secuencia , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular , Cruzamientos Genéticos , Proteínas de Unión al ADN , Femenino , Genotipo , Heterocigoto , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Supresoras de Tumor
17.
Cancer Res ; 59(19): 4781-3, 1999 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-10519383

RESUMEN

The presence of increased frequencies of blood-derived and solid tumors in ataxia-telangiectasia (A-T) patients, coupled with a role for the ATM (A-T mutation) protein in detecting specific forms of DNA damage, has led to the assumption of a mutator phenotype in A TM-deficient cells. Supporting this assumption are observations of increased rates of chromosomal aberrations and intrachromosomal homologous recombinational events in the cells of A-T patients. We have bred mice with knockout mutations for the selectable Aprt (adenine phosphoribosyltransferase) locus and the Atm locus to examine the frequency of second-step autosomal mutations in Atm-deficient cells. Two solid tissues were examined: (a) the ear, which yields predominately mesenchymal cells; and (b) the kidney, which yields predominately epithelial cells. We report here the lack of a mutator phenotype for inactivating autosomal mutations in solid tissues of the Atm-deficient mice.


Asunto(s)
Adenina Fosforribosiltransferasa/genética , Mutación , Proteínas Serina-Treonina Quinasas , Proteínas/genética , Animales , Ataxia Telangiectasia/genética , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular , Mapeo Cromosómico , Proteínas de Unión al ADN , Oído , Femenino , Genotipo , Heterocigoto , Homocigoto , Riñón/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Especificidad de Órganos , Proteínas/metabolismo , Proteínas Supresoras de Tumor
18.
J Neurosci ; 20(7): 2439-50, 2000 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-10729324

RESUMEN

Human cortical heterotopia and neuronal migration disorders result in epilepsy; however, the precise mechanisms remain elusive. Here we demonstrate severe neuronal dysplasia and heterotopia throughout the granule cell and pyramidal cell layers of mice containing a heterozygous deletion of Lis1, a mouse model of human 17p13.3-linked lissencephaly. Birth-dating analysis using bromodeoxyuridine revealed that neurons in Lis1+/- murine hippocampus are born at the appropriate time but fail in migration to form a defined cell layer. Heterotopic pyramidal neurons in Lis1+/- mice were stunted and possessed fewer dendritic branches, whereas dentate granule cells were hypertrophic and formed spiny basilar dendrites from which the principal axon emerged. Both somatostatin- and parvalbumin-containing inhibitory neurons were heterotopic and displaced into both stratum radiatum and stratum lacunosum-moleculare. Mechanisms of synaptic transmission were severely disrupted, revealing hyperexcitability at Schaffer collateral-CA1 synapses and depression of mossy fiber-CA3 transmission. In addition, the dynamic range of frequency-dependent facilitation of Lis1+/- mossy fiber transmission was less than that of wild type. Consequently, Lis1+/- hippocampi are prone to interictal electrographic seizure activity in an elevated [K(+)](o) model of epilepsy. In Lis1+/- hippocampus, intense interictal bursting was observed on elevation of extracellular potassium to 6.5 mM, a condition that resulted in only minimal bursting in wild type. These anatomical and physiological hippocampal defects may provide a neuronal basis for seizures associated with lissencephaly.


Asunto(s)
Coristoma/patología , Modelos Animales de Enfermedad , Epilepsia/etiología , Hipocampo/anomalías , Malformaciones del Sistema Nervioso/patología , 1-Alquil-2-acetilglicerofosfocolina Esterasa , Animales , Bromodesoxiuridina/farmacología , Movimiento Celular , Aparato de Golgi/ultraestructura , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/análisis , Neuronas/fisiología , Parvalbúminas/análisis , Somatostatina/análisis
19.
Mol Endocrinol ; 14(7): 1038-52, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10894153

RESUMEN

Restricted germ cell loss through apoptosis is initiated in the fetal gonad around embryonic day 13.5 (E13.5) as part of normal germ cell development. The mechanism of this germ cell attrition is unknown. We show that Bcl-x plays a crucial role in maintaining the survival of mouse germ cells during gonadogenesis. A bcl-x hypomorphic mouse was generated through the introduction of a neomycin (neo) gene into the promoter of the bcl-x gene by homologous recombination. Mice that contained two copies of the hypomorphic allele had severe reproductive defects attributed to compromised germ cell development. Males with two mutant alleles lacked spermatogonia and were sterile; females showed a severely reduced population of primordial and primary follicles and exhibited greatly impaired fertility. Primordial germ cells (PGCs) in bcl-x hypomorph mice migrated to the genital ridge by E12.5 but were depleted by E15.5, a time when Bcl-x and Bax were present. Two additional bcl-x transcripts were identified in fetal germ cells more than 300 bp upstream of previously reported start sites. Insertion of a neo cassette led to a down-regulation of the bcl-x gene at E12.5 in the hypomorph. Bax was detected by immunohistochemistry in germ cells from bcl-x hypomorph and control testes at E12.5 and E13.5. Bcl-x function was restored, and animals of both genders were fertile after removal of the neo selection cassette using Cre-mediated recombination. Alternatively, the loss of Bcl-x function in the hypomorph was corrected by the deletion of both copies of the bax gene, resulting in a restoration of germ cell survival. These findings demonstrate that the balance of Bcl-x and Bax control PGC survival and apoptosis.


Asunto(s)
Embrión de Mamíferos/citología , Células Germinativas/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas/genética , Animales , Secuencia de Bases , Supervivencia Celular/genética , Desarrollo Embrionario y Fetal/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos , Ratones Mutantes , Datos de Secuencia Molecular , Oocitos/patología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Espermatogonias/patología , Testículo/embriología , Transcripción Genética , Proteína X Asociada a bcl-2 , Proteína bcl-X
20.
Genes Brain Behav ; 3(1): 51-62, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14960015

RESUMEN

Dvl1 is one of three murine Dishevelled genes widely expressed in embryonic development and in the adult central nervous system. Dishevelled proteins are a necessary component of the Wnt and planar cell polarity developmental signaling pathways. We reported previously that mice deficient in Dvl1 exhibited abnormal social interaction and sensorimotor gating. To assess the validity of our earlier findings, we replicated the previous behavioral tests and included several new assays. The behaviors assessed included: social interaction, sensorimotor reflexes, motor activity, nociception, prepulse inhibition of acoustic startle (PPI) and learning and memory. Assessments with an explicit social component included: social dominance test, whisker trimming, nest building, home-cage huddling and ultrasonic vocalization rate analysis in pups. In addition, separate cohorts of wildtype and Dvl1-null mice were assessed for social recognition of a conspecific. Replicating the original report, Dvl1-null mice were impaired in several tasks containing an explicit social component. However, no impairment was observed in the social memory task. A previously observed deficit in PPI did not replicate in two institutions. In conclusion, we provide evidence that the social interaction phenotype of Dvl1-deficient mice has a strong genetic influence, but the sensorimotor gating deficit was subject to environmental influences. The specificity of observed social interaction deficits also suggests that lack of Dvl1 is associated with deficits in the recognition of social hierarchy and dominance.


Asunto(s)
Inhibición Neural/genética , Fosfoproteínas/genética , Reflejo de Sobresalto/genética , Conducta Social , Proteínas Adaptadoras Transductoras de Señales , Animales , Conducta Animal/fisiología , Proteínas Dishevelled , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Mutantes , Comportamiento de Nidificación/fisiología , Fosfoproteínas/fisiología , Desempeño Psicomotor/fisiología , Vibrisas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA