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











Intervalo de año de publicación
1.
Am J Reprod Immunol ; 89(3): e13662, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36458539

RESUMEN

PROBLEM: Protective effects for adult neurological disorders have been attributed to sex hormones. Using a murine model of prematurity, we evaluated the role of estrogen signaling in the process of perinatal brain injury following exposure to intrauterine inflammation. METHOD OF STUDY: Intrauterine lipopolysaccharide (LPS) was used to invoke preterm labor and fetal neuroinflammation. Fetal brains were analyzed for changes in Esr1, Esr2 and Cyp19. Dams heterozygous for the Esr1 knockout allele were also given intrauterine LPS to compare delivery and offspring viability to wild type controls. RESULTS: The upregulation in inflammatory cytokines was accompanied by an increase in Esr1 and Esr2 transcripts, though protein levels declined. Cyp19 did not differ by mRNA or protein abundance. Offspring from Esr1 mutants were larger, had a longer gestation and significantly greater mortality. CONCLUSIONS: Estrogen signaling is altered in the fetal brains of preterm offspring exposed to neuroinflammatory injury. The reduction of Esr1 and Esr2 proteins with LPS suggests that these proteins are degraded. It is possible that transcriptional upregulation of Esr1 and Esr2 occurs to compensate for the loss of these proteins. Alternatively, the translation of Esr1 and Esr2 mRNAs may be disrupted with LPS while a feedback mechanism upregulates transcription. Intact Esr1 signaling is also associated with early preterm delivery following exposure to intrauterine LPS. A loss of one Esr1 allele delays this process, but appears to do so at the cost of fetal viability. These results suggest estrogen signaling plays opposing roles between maternal and fetal responses to preterm birth.


Asunto(s)
Receptor alfa de Estrógeno , Viabilidad Fetal , Nacimiento Prematuro , Animales , Femenino , Ratones , Embarazo , Aromatasa , Modelos Animales de Enfermedad , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Viabilidad Fetal/genética , Lipopolisacáridos , Nacimiento Prematuro/genética , Nacimiento Prematuro/metabolismo
2.
J Reprod Immunol ; 123: 48-57, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28915450

RESUMEN

In addition to natural killer cells, other innate lymphoid cells have recently been identified in the mouse and human uterus, but their roles in successful pregnancy remain poorly defined. In this study, we examined the dynamic changes of uterine innate lymphoid cells throughout pregnancy in mice. We found that the total number of uterine innate lymphoid cells markedly increased at early-gestation. Among the three groups of uterine innate lymphoid cells, the number of the group 2 uterine innate lymphoid cells increased the most during pregnancy. We also determined that the depletion of uterine innate lymphoid cells in Rag1-/- mice resulted in impaired uterine spiral artery remodeling. These results suggest that uterine innate lymphoid cells may play an important role in mouse reproduction.


Asunto(s)
Endometrio/irrigación sanguínea , Proteínas de Homeodominio/genética , Linfocitos/inmunología , Embarazo/inmunología , Útero/patología , Animales , Células Cultivadas , Citocinas/metabolismo , Femenino , Viabilidad Fetal/genética , Proteínas de Homeodominio/metabolismo , Humanos , Inmunidad Innata , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células TH1/inmunología , Remodelación Vascular/genética
3.
Eur J Hum Genet ; 25(8): 924-929, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28561018

RESUMEN

It has been shown previously that homozygous and compound-heterozygous variants affecting protein function in the human NLRP genes impact reproduction and/or fetal imprinting patterns. These variants represent so-called 'maternal effect mutations', that is, although female variant carriers are healthy, they are at risk of reproductive failure, and their offspring may develop aberrant methylation and imprinting disorders. In contrast, the relevance to reproductive failure of maternal heterozygous NLRP7 variants remains unclear. The present report describes the identification of a heterozygous NLRP7 variant in a healthy 28-year-old woman with a history of recurrent reproductive failure, and the molecular findings in two of the deceased offspring. Next-generation sequencing (NGS) for NLRP variants was performed. In the tissues of two offspring (one fetus; one deceased premature neonate) methylation of imprinted loci was tested using methylation-specific assays. Both pregnancies had been characterized by the presence of elevated human chorionic gonadotropin (hCG) levels and ovarian cysts. In the mother, a heterozygous nonsense 2-bp deletion in exon 5 of the NLRP7 gene was identified (NM_001127255.1:c.2010_2011del, p.(Phe671Glnfs*18)). In the two investigated offspring, heterogeneous aberrant methylation patterns were detected at imprinted loci. The present data support the hypothesis that heterozygous NLRP7 variants contribute to reproductive wastage, and that these variants represent autosomal dominant maternal effect variants which lead to aberrant imprinting marks in the offspring. Specific screening and close prenatal monitoring of NLRP7 variant carriers is proposed. Egg donation might facilitate successful pregnancy in heterozygous NLRP7 variant carriers.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Viabilidad Fetal/genética , Impresión Genómica , Adulto , Alelos , Codón sin Sentido , Metilación de ADN , Femenino , Muerte Fetal/etiología , Heterocigoto , Humanos , Recién Nacido , Recien Nacido Prematuro , Muerte Perinatal/etiología , Embarazo
4.
Development ; 139(19): 3531-42, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22899851

RESUMEN

The molecular mechanisms that regulate and coordinate signaling between the extracellular matrix (ECM) and cells contributing to the developing vasculature are complex and poorly understood. Myocardin-like protein 2 (MKL2) is a transcriptional co-activator that in response to RhoA and cytoskeletal actin signals physically associates with serum response factor (SRF), activating a subset of SRF-regulated genes. We now report the discovery of a previously undescribed MKL2/TGFß signaling pathway in embryonic stem (ES) cells that is required for maturation and stabilization of the embryonic vasculature. Mkl2(-/-) null embryos exhibit profound derangements in the tunica media of select arteries and arterial beds, which leads to aneurysmal dilation, dissection and hemorrhage. Remarkably, TGFß expression, TGFß signaling and TGFß-regulated genes encoding ECM are downregulated in Mkl2(-/-) ES cells and the vasculature of Mkl2(-/-) embryos. The gene encoding TGFß2, the predominant TGFß isoform expressed in vascular smooth muscle cells and embryonic vasculature, is activated directly via binding of an MKL2/SRF protein complex to a conserved CArG box in the TGFß2 promoter. Moreover, Mkl2(-/-) ES cells exhibit derangements in cytoskeletal organization, cell adhesion and expression of ECM that are rescued by forced expression of TGFß2. Taken together, these data demonstrate that MKL2 regulates a conserved TGF-ß signaling pathway that is required for angiogenesis and ultimately embryonic survival.


Asunto(s)
Vasos Sanguíneos/embriología , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción/fisiología , Factor de Crecimiento Transformador beta2/genética , Animales , Fístula Arteriovenosa/embriología , Fístula Arteriovenosa/genética , Vasos Sanguíneos/metabolismo , Células Cultivadas , Embrión de Mamíferos , Células Madre Embrionarias/fisiología , Viabilidad Fetal/genética , Hemorragia/embriología , Hemorragia/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/fisiología , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo
5.
Int J Dev Biol ; 56(4): 255-61, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22562201

RESUMEN

Phenotypic characterization of Akt1 and Igf2 null mice has revealed roles for each in the regulation of placentation, and fetal and postnatal growth. Insulin-like growth factor 2 (IGF2) is encoded by the Igf2 gene and influences cellular function, at least in part, through activation of an intracellular serine/threonine kinase called AKT1. Akt1 and Igf2 null mice were originally characterized on inbred and mixed genetic backgrounds, prohibiting direct comparisons of their phenotypes. The impact of loss of AKT1 or IGF2 on placental, fetal, and postnatal function were examined following transfer of Akt1 and Igf2 null mutations to an outbred CD1 genetic background. Disruption of IGF2 did not affect AKT expression or activation. Both Akt1-/- and Igf2-/- mice exhibited decreased placental weight, fetal weight and viability. Deregulation of placental growth was similar in Akt1 and Igf2 nulls; however, disruption of Igf2 had a more severe impact on prenatal survival and postnatal growth. Placental structure, including organization of junctional and labyrinth zones and development of the interstitial, invasive, trophoblast lineage, were similar in mutant and wild-type mice. Akt1 and Igf2 null mutations affected postnatal growth. The relative impact of each gene differed during pre-weaning versus post-weaning growth phases. AKT1 had a more significant role during pre-weaning growth, whereas IGF2 was a bigger contributor to post-weaning growth. Akt1 and Igf2 null mutations impact placental, fetal and postnatal growth. Placental phenotypes are similar; however, fetal and postnatal growth patterns are unique to each mutation.


Asunto(s)
Desarrollo Fetal/fisiología , Factor II del Crecimiento Similar a la Insulina/metabolismo , Placentación/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Animales Recién Nacidos , Western Blotting , Femenino , Desarrollo Fetal/genética , Viabilidad Fetal/genética , Viabilidad Fetal/fisiología , Inmunohistoquímica , Factor II del Crecimiento Similar a la Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Placenta/metabolismo , Placentación/genética , Embarazo , Proteínas Proto-Oncogénicas c-akt/genética , Destete
6.
Mamm Genome ; 22(9-10): 506-17, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21611832

RESUMEN

Replication origin licensing builds a fundamental basis for DNA replication in all eukaryotes. This occurs during the late M to early G1 phases in which chromatin is licensed by loading of the MCM2-7 complex, an essential component of the replicative helicase. In the following S phase, only a minor fraction of chromatin-bound MCM2-7 complexes are activated to unwind the DNA. Therefore, it is proposed that the vast majority of MCM2-7 complexes license dormant origins that can be used as backups. Consistent with this idea, it has been repeatedly demonstrated that a reduction (~60%) in chromatin-bound MCM2-7 complexes has little effect on the density of active origins. In this study, however, we describe the first exception to this observation. A reduction of licensed origins due to Mcm4 ( chaos3 ) homozygosity reduces active origin density in primary embryonic fibroblasts (MEFs) in a C57BL/6J (B6) background. We found that this is associated with an intrinsically lower level of active origins in this background compared to others. B6 Mcm4 ( chaos3/chaos3 ) cells proliferate slowly due to p53-dependent upregulation of p21. In fact, the development of B6 Mcm4 ( chaos3/chaos3 ) mice is impaired and a significant fraction of them die at birth. While inactivation of p53 restores proliferation in B6 Mcm4 ( chaos3/chaos3 ) MEFs, it paradoxically does not rescue animal lethality. These findings indicate that a reduction of licensed origins may cause a more profound effect on cell types with lower densities of active origins. Moreover, p53 is required for the development of mice that suffer from intrinsic replication stress.


Asunto(s)
ADN Helicasas/genética , Replicación del ADN , Origen de Réplica , Animales , Proliferación Celular , Transformación Celular Neoplásica/genética , Cromatina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Daño del ADN , Viabilidad Fetal/genética , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Componente 4 del Complejo de Mantenimiento de Minicromosoma , Antígeno Nuclear de Célula en Proliferación/metabolismo , Recombinasa Rad51/genética , Especificidad de la Especie , Transcripción Genética , Proteína p53 Supresora de Tumor/genética , Regulación hacia Arriba/genética
7.
Biol Reprod ; 84(3): 537-45, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20980686

RESUMEN

The importance of placental circulation is exemplified by the correlation of placental size and blood flow with fetal weight and survival during normal and compromised human pregnancies in such conditions as preeclampsia and intrauterine growth restriction (IUGR). Using noninvasive magnetic resonance imaging, we evaluated the role of PKBalpha/AKT1, a major mediator of angiogenesis, on placental vascular function. PKBalpha/AKT1 deficiency reduced maternal blood volume fraction without affecting the integrity of the fetomaternal blood barrier. In addition to angiogenesis, PKBalpha/AKT1 regulates additional processes related to survival and growth. In accordance with reports in adult mice, we demonstrated a role for PKBalpha/AKT1 in regulating chondrocyte organization in fetal long bones. Using tetraploid complementation experiments with PKBalpha/AKT1-expressing placentas, we found that although placental PKBalpha/AKT1 restored fetal survival, fetal PKBalpha/AKT1 regulated fetal size, because tetraploid complementation did not prevent intrauterine growth retardation. Histological examination of rescued fetuses showed reduced liver blood vessel and renal glomeruli capillary density in PKBalpha/Akt1 null fetuses, both of which were restored by tetraploid complementation. However, bone development was still impaired in tetraploid-rescued PKBalpha/Akt1 null fetuses. Although PKBalpha/AKT1-expressing placentas restored chondrocyte cell number in the hypertrophic layer of humeri, fetal PKBalpha/AKT1 was found to be necessary for chondrocyte columnar organization. Remarkably, a dose-dependent phenotype was exhibited for PKBalpha/AKT1 when examining PKBalpha/Akt1 heterozygous fetuses as well as those complemented by tetraploid placentas. The differential role of PKBalpha/AKT1 on mouse fetal survival and growth may shed light on its roles in human IUGR.


Asunto(s)
Tamaño Corporal/genética , Viabilidad Fetal/genética , Feto/fisiología , Placenta/metabolismo , Proteínas Proto-Oncogénicas c-akt/fisiología , Animales , Embrión de Mamíferos , Femenino , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/metabolismo , Retardo del Crecimiento Fetal/patología , Viabilidad Fetal/fisiología , Feto/metabolismo , Edad Gestacional , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Embarazo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo
8.
Nat Genet ; 39(1): 93-8, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17143284

RESUMEN

Mcm4 (minichromosome maintenance-deficient 4 homolog) encodes a subunit of the MCM2-7 complex (also known as MCM2-MCM7), the replication licensing factor and presumptive replicative helicase. Here, we report that the mouse chromosome instability mutation Chaos3 (chromosome aberrations occurring spontaneously 3), isolated in a forward genetic screen, is a viable allele of Mcm4. Mcm4(Chaos3) encodes a change in an evolutionarily invariant amino acid (F345I), producing an apparently destabilized MCM4. Saccharomyces cerevisiae strains that we engineered to contain a corresponding allele (resulting in an F391I change) showed a classical minichromosome loss phenotype. Whereas homozygosity for a disrupted Mcm4 allele (Mcm4(-)) caused preimplantation lethality, Mcm(Chaos3/-) embryos died late in gestation, indicating that Mcm4(Chaos3) is hypomorphic. Mutant embryonic fibroblasts were highly susceptible to chromosome breaks induced by the DNA replication inhibitor aphidicolin. Most notably, >80% of Mcm4(Chaos3/Chaos3) females succumbed to mammary adenocarcinomas with a mean latency of 12 months. These findings suggest that hypomorphic alleles of the genes encoding the subunits of the MCM2-7 complex may increase breast cancer risk.


Asunto(s)
Adenocarcinoma/genética , Inestabilidad Cromosómica/genética , ADN Helicasas/genética , Neoplasias Mamarias Animales/genética , Secuencia de Aminoácidos , Animales , Células Cultivadas , Mapeo Cromosómico , Análisis Mutacional de ADN , Femenino , Viabilidad Fetal/genética , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Componente 4 del Complejo de Mantenimiento de Minicromosoma , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
9.
Dev Cell ; 11(6): 859-71, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17141160

RESUMEN

The mTOR kinase controls cell growth, proliferation, and survival through two distinct multiprotein complexes, mTORC1 and mTORC2. mTOR and mLST8 are in both complexes, while raptor and rictor are part of only mTORC1 and mTORC2, respectively. To investigate mTORC1 and mTORC2 function in vivo, we generated mice deficient for raptor, rictor, or mLST8. Like mice null for mTOR, those lacking raptor die early in development. However, mLST8 null embryos survive until e10.5 and resemble embryos missing rictor. mLST8 is necessary to maintain the rictor-mTOR, but not the raptor-mTOR, interaction, and both mLST8 and rictor are required for the hydrophobic motif phosphorylation of Akt/PKB and PKCalpha, but not S6K1. Furthermore, insulin signaling to FOXO3, but not to TSC2 or GSK3beta, requires mLST8 and rictor. Thus, mTORC1 function is essential in early development, mLST8 is required only for mTORC2 signaling, and mTORC2 is a necessary component of the Akt-FOXO and PKCalpha pathways.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Proteína Quinasa C-alfa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal , Transactivadores/fisiología , Animales , Citoesqueleto/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Desarrollo Fetal/genética , Viabilidad Fetal/genética , Proteína Forkhead Box O3 , Marcación de Gen , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Insulina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones/embriología , Ratones Noqueados , Complejos Multiproteicos , Fosforilación , Unión Proteica , Proteínas , Serina-Treonina Quinasas TOR , Transactivadores/genética , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/metabolismo
10.
Dev Cell ; 11(4): 583-9, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16962829

RESUMEN

The rapamycin-insensitive mTOR complex 2 (mTORC2) has been suggested to play an important role in growth factor-dependent signaling. To explore this possibility further in a mammalian model system, we disrupted the expression of rictor, a specific component of mTORC2, in mice by using a multiallelic gene targeting strategy. Embryos that lack rictor develop normally until E9.5, and then exhibit growth arrest and die by E11.5. Although placental defects occur in null embryos, an epiblast-specific knockout of rictor only delayed lethality by a few days, thereby suggesting other important roles for this complex in the embryo proper. Analyses of rictor null embryos and fibroblasts indicate that mTORC2 is a primary kinase for Ser473 of Akt/PKB. Rictor null fibroblasts exhibit low proliferation rates, impaired Akt/PKB activity, and diminished metabolic activity. Taken together, these findings indicate that both rictor and mTORC2 are essential for the development of both embryonic and extraembryonic tissues.


Asunto(s)
Alelos , Proteínas Portadoras/metabolismo , Desarrollo Fetal/genética , Viabilidad Fetal/genética , Proteínas Quinasas/metabolismo , Actinas/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Fibroblastos/fisiología , Marcación de Gen , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Proteínas Quinasas/análisis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina , Serina-Treonina Quinasas TOR
11.
Mol Cell Biol ; 23(18): 6542-52, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12944480

RESUMEN

The E2F transcription factors are key downstream targets of the retinoblastoma protein (pRB) tumor suppressor. We have previously shown that E2F3 plays a critical role in mediating the mitogen-induced activation of E2F-responsive genes and contributes to both the inappropriate proliferation and the p53-dependent apoptosis that arise in pRB-deficient embryos. Here we show that E2F3 also has a significant effect on the phenotype of tumor-prone Rb(+/-) mice. The absence of E2F3 results in a significant expansion in the life spans of these animals that correlates with a dramatic alteration in the tumor spectrum. E2F3 loss suppresses the development of the pituitary tumors that normally account for the death of Rb(+/-) mice. However, it also promotes the development of medullary thyroid carcinomas yielding metastases at a high frequency. This increased aggressiveness does not seem to result from any change in p53 levels or activity in these tumors. We show that, instead, E2F3 loss leads to an increase in the rate of tumor initiation. Finally, analysis of Rb(+/-); E2f3(+/-) mice shows that this tumor-suppressive function of E2F3 is dose dependent.


Asunto(s)
Carcinoma Medular/etiología , Carcinoma Medular/patología , Neoplasias Hipofisarias/genética , Proteína de Retinoblastoma/deficiencia , Neoplasias de la Tiroides/etiología , Neoplasias de la Tiroides/patología , Factores de Transcripción/genética , Animales , Animales Recién Nacidos , Factor de Transcripción E2F3 , Femenino , Viabilidad Fetal/genética , Dosificación de Gen , Heterocigoto , Longevidad , Masculino , Ratones , Ratones Mutantes , Mutación , Metástasis de la Neoplasia/genética , Fenotipo , Neoplasias Hipofisarias/prevención & control , Proteína de Retinoblastoma/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor
12.
J Neurosci ; 23(17): 6956-64, 2003 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-12890790

RESUMEN

Huntington's disease (HD) is caused by a polyglutamine expansion in the disease protein huntingtin. The polyglutamine expansion causes huntingtin to interact abnormally with a number of proteins. However, it is unclear whether, and how, huntingtin-associated proteins are involved in the neurodegeneration in HD. Here, we show that huntingtin-associated protein-1 (HAP1), which is involved in intracellular trafficking of epidermal growth factor receptor (EGFR), is highly expressed in the hypothalamus. Mice lacking HAP1 die after birth because of depressed feeding activity. Terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling staining and electron microscopic examination revealed the degeneration in hypothalamic regions that control feeding behavior. Hypothalamic degeneration was also observed in HD transgenic mice that have a significant loss of body weight. Inhibition of HAP1 expression decreases EGFR signaling and cell viability, whereas overexpression of HAP1 enhances this signaling activity and inhibits mutant huntingtin-mediated cytotoxicity. These results suggest that the effect of mutant huntingtin on HAP1 and EGFR signaling may contribute to the hypothalamic neurodegeneration and loss of body weight in HD.


Asunto(s)
Enfermedad de Huntington/genética , Hipotálamo/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Neuronas/metabolismo , Animales , Peso Corporal/genética , Muerte Celular/genética , Supervivencia Celular/genética , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Receptores ErbB/metabolismo , Conducta Alimentaria , Viabilidad Fetal/genética , Marcación de Gen , Homocigoto , Proteína Huntingtina , Enfermedad de Huntington/patología , Hipotálamo/patología , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/patología , Neuronas/ultraestructura , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenotipo , Unión Proteica/genética , Ratas , Transducción de Señal
13.
J Clin Invest ; 111(10): 1519-27, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12750401

RESUMEN

Chronic hypoxia induces pulmonary vascular remodeling, leading to pulmonary hypertension, right ventricular hypertrophy, and heart failure. Heterozygous deficiency of hypoxia-inducible factor-1alpha (HIF-1alpha), which mediates the cellular response to hypoxia by increasing expression of genes involved in erythropoiesis and angiogenesis, has been previously shown to delay hypoxia-induced pulmonary hypertension. HIF-2alpha is a homologue of HIF-1alpha and is abundantly expressed in the lung, but its role in pulmonary hypertension remains unknown. Therefore, we analyzed the pulmonary response of WT and viable heterozygous HIF-2alpha-deficient (Hif2alpha(+/-)) mice after exposure to 10% O(2) for 4 weeks. In contrast to WT mice, Hif2alpha(+/-) mice were fully protected against pulmonary hypertension and right ventricular hypertrophy, unveiling a critical role of HIF-2alpha in hypoxia-induced pulmonary vascular remodeling. Pulmonary expression levels of endothelin-1 and plasma catecholamine levels were increased threefold and 12-fold respectively in WT but not in Hif2alpha(+/-) mice after hypoxia, suggesting that HIF-2alpha-mediated upregulation of these vasoconstrictors contributes to the development of hypoxic pulmonary vascular remodeling.


Asunto(s)
Hipertensión Pulmonar/prevención & control , Hipoxia/complicaciones , Transactivadores/deficiencia , Disfunción Ventricular Derecha/prevención & control , Animales , Cámaras de Exposición Atmosférica , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Catecolaminas/sangre , Endotelina-1/genética , Endotelina-1/metabolismo , Viabilidad Fetal/genética , Hematócrito , Heterocigoto , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Pulmón/irrigación sanguínea , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Transgénicos , Fenotipo , Arteria Pulmonar/patología , ARN Mensajero/metabolismo , Tiempo , Transactivadores/genética , Regulación hacia Arriba , Disfunción Ventricular Derecha/etiología
14.
J Neurosci ; 23(3): 748-57, 2003 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-12574403

RESUMEN

RAS proteins are critical regulators of mitosis and are mutationally activated in many human tumors. RAS signaling is also known to mediate long-term potentiation (LTP) and long-term memory formation in postmitotic neurons, in part through activation of the RAF-MEK-ERK pathway. The RAS effector RIN1 appears to function through competitive inhibition of RAS-RAF binding and also through diversion of RAS signaling to alternate pathways. We show that RIN1 is preferentially expressed in postnatal forebrain neurons in which it is localized in dendrites and physically associated with RAS, suggesting a role in RAS-mediated postsynaptic neuronal plasticity. Mice with an Rin1 gene disruption showed a striking enhancement in amygdala LTP. In addition, two independent behavioral tests demonstrated elevated amygdala-dependent aversive memory in Rin1(-/-) mice. These results indicate that RIN1 serves as an inhibitory modulator of neuronal plasticity in aversive memory formation.


Asunto(s)
Proteínas Portadoras/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Potenciación a Largo Plazo/fisiología , Memoria/fisiología , Plasticidad Neuronal/fisiología , Proteínas de Unión al GTP rab , Proteínas ras/metabolismo , Amígdala del Cerebelo/fisiología , Animales , Proteínas Portadoras/genética , Recuento de Células , Condicionamiento Clásico , Dendritas/metabolismo , Miedo/fisiología , Viabilidad Fetal/genética , Hipocampo/fisiología , Humanos , Aprendizaje/fisiología , Potenciación a Largo Plazo/genética , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuronas/ultraestructura , Prosencéfalo/química , Prosencéfalo/citología , Prosencéfalo/metabolismo , Unión Proteica/fisiología
15.
J Neurosci ; 22(18): 7931-40, 2002 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12223546

RESUMEN

Dopamine receptor subtypes D1 and D2, and many other seven-transmembrane receptors including adenosine receptor A2A, are colocalized in striatum of brain. These receptors stimulate or inhibit adenylyl cyclases (ACs) to produce distinct physiological and pharmacological responses and interact with each other synergistically or antagonistically at various levels. The identity of the AC isoform that is coupled to each of these receptors, however, remains unknown. To investigate the in vivo role of the type 5 adenylyl cyclase (AC5), which is preferentially expressed in striatum, mice deficient for the AC5 gene were generated. The genetic ablation of the AC5 gene eliminated >80% of forskolin-induced AC activity and 85-90% of AC activity stimulated by either D1 or A2A receptor agonists in striatum. However, D1- or A2A-specific pharmaco-behaviors were basically preserved, whereas the signal cascade from D2 to AC was completely abolished in AC5(-/-), and motor activity of AC5(-/-) was not suppressed by treatment of cataleptic doses of the antipsychotic drugs haloperidol and sulpiride. Interestingly, both haloperidol and clozapine at low doses remarkably increased the locomotion of AC5(-/-) in the open field test that was produced in part by a common mechanism that involved the increased activation of D1 dopamine receptors. Together, these results suggest that AC5 is the principal AC integrating signals from multiple receptors including D1, D2, and A2A in striatum and the cascade involving AC5 among diverse D2 signaling pathways is essential for neuroleptic effects of antipsychotic drugs.


Asunto(s)
Adenilil Ciclasas/deficiencia , Encéfalo/metabolismo , Isoenzimas/deficiencia , Receptores de Dopamina D2/metabolismo , Adenilil Ciclasas/genética , Animales , Antipsicóticos/farmacología , Encéfalo/efectos de los fármacos , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Colforsina/farmacología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , AMP Cíclico , Agonistas de Dopamina/farmacología , Activación Enzimática/efectos de los fármacos , Activadores de Enzimas/farmacología , Viabilidad Fetal/genética , Marcación de Gen , Heterocigoto , Homocigoto , Isoenzimas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Receptor de Adenosina A2A , Receptores de Dopamina D1/metabolismo , Receptores Purinérgicos P1/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
16.
J Cell Biol ; 154(2): 293-307, 2001 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-11470819

RESUMEN

Cajal bodies (CBs) are nuclear suborganelles involved in the biogenesis of small nuclear ribonucleoproteins (snRNPs). In addition to snRNPs, they are highly enriched in basal transcription and cell cycle factors, the nucleolar proteins fibrillarin (Fb) and Nopp140 (Nopp), the survival motor neuron (SMN) protein complex, and the CB marker protein, p80 coilin. We report the generation of knockout mice lacking the COOH-terminal 487 amino acids of coilin. Northern and Western blot analyses demonstrate that we have successfully removed the full-length coilin protein from the knockout animals. Some homozygous mutant animals are viable, but their numbers are reduced significantly when crossed to inbred backgrounds. Analysis of tissues and cell lines from mutant animals reveals the presence of extranucleolar foci that contain Fb and Nopp but not other typical nucleolar markers. These so-called "residual" CBs neither condense Sm proteins nor recruit members of the SMN protein complex. Transient expression of wild-type mouse coilin in knockout cells results in formation of CBs and restores these missing epitopes. Our data demonstrate that full-length coilin is essential for proper formation and/or maintenance of CBs and that recruitment of snRNP and SMN complex proteins to these nuclear subdomains requires sequences within the coilin COOH terminus.


Asunto(s)
Cuerpos Enrollados/metabolismo , Ratones Noqueados/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/deficiencia , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Animales , Autoantígenos/metabolismo , Northern Blotting , Línea Celular , Nucléolo Celular/metabolismo , Nucléolo Celular/ultraestructura , Proteínas Cromosómicas no Histona/metabolismo , Cuerpos Enrollados/genética , Cuerpos Enrollados/ultraestructura , Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Viabilidad Fetal/genética , Expresión Génica/efectos de los fármacos , Marcación de Gen , Proteínas Fluorescentes Verdes , Homocigoto , Proteínas Luminiscentes/genética , Ratones , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Especificidad de Órganos , Fosfoproteínas/metabolismo , Empalme del ARN , ARN Mensajero , Proteínas de Unión al ARN , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Proteínas del Complejo SMN , Tasa de Supervivencia , Proteínas Nucleares snRNP
17.
Circulation ; 104(3): 358-64, 2001 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-11457758

RESUMEN

BACKGROUND: Vascular endothelial growth factors (VEGFs) and their receptors are essential regulators of vasculogenesis and angiogenesis in both embryos and adults. One of the factors with a still unknown physiological function is VEGF-B, which is expressed in many tissues, including the heart. METHODS AND RESULTS: Mice carrying a targeted deletion in the VEGF-B gene were developed. In VEGF-B(-/-) animals, no gross abnormalities were observed in organs that normally show high expression of VEGF-B, such as the heart, muscle, and kidney. Analysis of heart function by ECG showed that adult VEGF-B(-/-) mice have an atrial conduction abnormality characterized by a prolonged PQ interval. VEGF- or basic fibroblast growth factor-induced corneal angiogenesis was similar in normal and VEGF-B(-/-) mice. CONCLUSIONS: VEGF-B seems to be required for normal heart function in adult animals but is not required for proper development of the cardiovascular system either during development or for angiogenesis in adults.


Asunto(s)
Factores de Crecimiento Endotelial/deficiencia , Atrios Cardíacos/fisiopatología , Sistema de Conducción Cardíaco/fisiopatología , Animales , Recuento de Células Sanguíneas , Electrocardiografía , Técnicas Electrofisiológicas Cardíacas , Factores de Crecimiento Endotelial/genética , Factores de Crecimiento Endotelial/metabolismo , Factores de Crecimiento Endotelial/farmacología , Ojo/irrigación sanguínea , Ojo/efectos de los fármacos , Femenino , Fertilidad/genética , Viabilidad Fetal/genética , Factor 2 de Crecimiento de Fibroblastos/farmacología , Expresión Génica/fisiología , Marcación de Gen , Atrios Cardíacos/crecimiento & desarrollo , Homocigoto , Linfocinas/farmacología , Masculino , Ratones , Ratones Noqueados , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Tamaño de los Órganos , Fenotipo , ARN Mensajero/metabolismo , Factor A de Crecimiento Endotelial Vascular , Factor B de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
18.
Cell ; 98(6): 779-90, 1999 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-10499795

RESUMEN

Transcription factors of the Myc proto-oncogene family promote cell division, but how they do this is poorly understood. Here we address the functions of Drosophila Myc (dMyc) during development. Using mosaic analysis in the fly wing, we show that loss of dMyc retards cellular growth (accumulation of cell mass) and reduces cell size, whereas dMyc overproduction increases growth rates and cell size. dMyc-induced growth promotes G1/S progression but fails to accelerate cell division because G2/M progression is independently controlled by Cdc25/String. We also show that the secreted signal Wingless patterns growth in the wing primordium by modulating dMyc expression. Our results indicate that dMyc links patterning signals to cell division by regulating primary targets involved in cellular growth and metabolism.


Asunto(s)
Proteínas de Unión al ADN , Proteínas de Drosophila , Drosophila/crecimiento & desarrollo , Proteínas Proto-Oncogénicas c-myc/genética , Factores de Transcripción/genética , Alelos , Animales , Constitución Corporal/genética , Tipificación del Cuerpo/genética , Ciclo Celular/genética , Tamaño de la Célula/genética , Drosophila/genética , Femenino , Viabilidad Fetal/genética , Infertilidad , Mosaicismo , Mutación , Alas de Animales/crecimiento & desarrollo
19.
Autoimmunity ; 22(4): 199-207, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-8781712

RESUMEN

The viable motheaten (mev) mice are characterized by a moth-eaten appearance of the coat, immunodeficiency, autoimmunity, generalized inflammatory disease, paws necroses, and early death. The target of the single point mev mutation is PTP1C, a protein tyrosine phosphatase whose deficient expression in hematopoietic cells should explain all phenotypic features of mev mice, particularly their autoimmune and inflammatory pathologies. In order to evaluate their role in the development of the mev mouse disease, we constructed mevscid congenics to probe the impact of autoimmunity and mevbeige congenics to probe the impact of elastase and cathepsine G neutrophil activities. Both mevscid and mevbeige mice were nearly equivalent to mev mice with regards to moth-eaten appearance, paw necroses and early death. Thus, autoimmunity does neither initiate nor substantially enhance the mev mouse syndrome. Moreover, the beige mutation-linked deficiency of protease activity of neutrophils is unable to significantly reduce the mev mutation-dependent inflammatory pathology.


Asunto(s)
Cruzamientos Genéticos , Viabilidad Fetal/genética , Viabilidad Fetal/fisiología , Enfermedades de la Piel/genética , Enfermedades de la Piel/patología , Animales , Femenino , Crecimiento/fisiología , Inmunoglobulinas/sangre , Esperanza de Vida/tendencias , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones SCID , Mortalidad/tendencias , Enfermedades de la Piel/mortalidad
20.
Genet Res ; 56(2-3): 223-36, 1990.
Artículo en Inglés | MEDLINE | ID: mdl-2272513

RESUMEN

A null allele of the Gpi-1s structural gene, that encodes glucose phosphate isomerase (GPI-1; E.C. 5.3.1.9), arose in a mutation experiment and was designated Gpi-1sa-m1H. The viability of homozygotes has been investigated. No offspring homozygous for the null allele were produced by intercrossing two heterozygotes, so the homozygous condition was presumed to be embryonic lethal. Embryos were produced by crossing Gpi-1sa/null heterozygous females and Gpi-1sb/null heterozygous males. Homozygous null embryos were identified at different stages of development by electrophoresis and staining either for GPI-1 alone or GPI-1 plus phosphoglycerate kinase (PGK) activity. At 6 1/2 and 7 1/2 days post coitum homozygous null embryos were present at approximately the expected 25% frequency (37/165; 22.4% overall) although at 7 1/2 days the homozygous null embryos tended to be small. By 8 1/2 days most homozygous null embryos were developmentally retarded and had not developed significantly further than at 7 1/2 days; some were dead or dying. By 9 1/2 days the homozygous null conceptus was characterised by a small implantation site that contained trophoblast and often a small amount of extraembryonic membrane. Surviving trophoblast tissue was also detectable at 10 1/2 days. Previous studies have shown that oocyte-coded GPI-1 persists only until 5 1/2 or 6 1/2 days. Survival of homozygous null embryos to 7 1/2 or 8 1/2 days and survival of certain extraembryonic tissue to 10 1/2 days suggests that the homozygous null condition may not be cell-lethal although it is certainly embryo-lethal. Mutant cells that are deficient in glycolysis may use the pentose phosphate shunt to bypass the block in glycolysis created by the deficiency of glucose phosphate isomerase, and/or might be rescued by the transport, from the maternal blood, of energy sources other than glucose (such as glutamine). Either strategy may only permit slow cell growth that would not be adequate to support normal embryogenesis. Transport of maternal nutrients would be more efficient to the trophoblast and extraembryonic membranes and this may help to explain why these tissues survive for longer than the embryo itself. The morphological similarity between homozygous nulls and androgenetic conceptuses, where the trophoblast also survives better than the embryo, is discussed.


Asunto(s)
Viabilidad Fetal/genética , Glucosa-6-Fosfato Isomerasa/genética , Animales , Cruzamientos Genéticos , Femenino , Genes , Glucosa-6-Fosfato Isomerasa/metabolismo , Masculino , Ratones , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA