Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Semin Cell Dev Biol ; 24(10-12): 724-35, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23994285

RESUMEN

The regulation of blastocyst implantation in the uterus is orchestrated by the ovarian hormones estrogen and progesterone. These hormones act via their nuclear receptors to direct the transcriptional activity of the endometrial compartments and create a defined period in which the uterus is permissive to embryo implantation termed the "window of receptivity". Additional members of the nuclear receptor family have also been described to have a potential role in endometrial function. Much of what we know about the function of these nuclear receptors during implantation we have learned from the use of mouse models. Transgenic murine models with targeted gene ablation have allowed us to identify a complex network of paracrine signaling between the endometrial epithelium and stroma. While some of the critical molecules have been identified, the mechanism underlying the intricate communication between endometrial compartments during the implantation window has not been fully elucidated. Defining this mechanism will help identify markers of a receptive uterine environment, ultimately providing a useful tool to help improve the fertility outlook for reproductively challenged couples. The aim of this review is to outline our current understanding of how nuclear receptors and their effector molecules regulate blastocyst implantation in the endometrium.


Asunto(s)
Implantación del Embrión , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Ritmo Circadiano , Humanos , Transducción de Señal
2.
Nat Med ; 5(9): 1018-25, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10470078

RESUMEN

The molecular mechanism(s) of immunoglobulin A (IgA) nephropathy, the most common primary renal glomerular disease worldwide, is unknown. Its pathologic features include hematuria, high levels of circulating IgA-fibronectin (Fn) complexes, and glomerular deposition of IgA, complement C3, Fn and collagen. We report here that two independent mouse models (gene knockout and antisense transgenic), both manifesting deficiency of an anti-inflammatory protein, uteroglobin (UG), develop almost all of the pathologic features of human IgA nephropathy. We further demonstrate that Fn-UG heteromerization, reported to prevent abnormal glomerular deposition of Fn and collagen, also abrogates both the formation of IgA-Fn complexes and their binding to glomerular cells. Moreover, UG prevents glomerular accumulation of exogenous IgA in UG-null mice. These results define an essential role for UG in preventing mouse IgA nephropathy and warrant further studies to determine if a similar mechanism(s) underlies the human disease.


Asunto(s)
Glomerulonefritis por IGA/patología , Inmunoglobulina A/inmunología , Glomérulos Renales/patología , Uteroglobina/fisiología , Animales , Complejo Antígeno-Anticuerpo/análisis , Complejo Antígeno-Anticuerpo/sangre , Complejo Antígeno-Anticuerpo/efectos de los fármacos , Complejo Antígeno-Anticuerpo/inmunología , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Complemento C3/análisis , Complemento C3/inmunología , Modelos Animales de Enfermedad , Fibronectinas/análisis , Fibronectinas/sangre , Fibronectinas/genética , Fibronectinas/inmunología , Eliminación de Gen , Mesangio Glomerular/citología , Mesangio Glomerular/efectos de los fármacos , Mesangio Glomerular/inmunología , Mesangio Glomerular/patología , Glomerulonefritis por IGA/genética , Glomerulonefritis por IGA/inmunología , Glomerulonefritis por IGA/fisiopatología , Hematuria/patología , Hematuria/orina , Humanos , Inmunoglobulina A/análisis , Inmunoglobulina A/sangre , Inmunoglobulina A/efectos de los fármacos , Glomérulos Renales/inmunología , Glomérulos Renales/metabolismo , Glomérulos Renales/fisiopatología , Ratones , Ratones Noqueados , Ratones Transgénicos , Factor de Crecimiento Derivado de Plaquetas/análisis , Factor de Crecimiento Derivado de Plaquetas/genética , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-sis , ARN sin Sentido/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Uteroglobina/deficiencia , Uteroglobina/genética , Uteroglobina/farmacología
3.
Science ; 208(4451): 1468-9, 1980 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-6770463

RESUMEN

Homologous sperm and ova of either squirrel monkeys or hamsters were placed in the oviducts of pseudopregnant rabbits. Xenogenous fertilization rates of 36 and 60 percent were obtained for squirrel monkey and hamster gametes, respectively.


Asunto(s)
Fertilización , Oviductos/fisiología , Óvulo/trasplante , Animales , Cricetinae , Femenino , Haplorrinos , Saimiri , Trasplante Heterólogo
4.
Science ; 289(5485): 1751-4, 2000 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-10976068

RESUMEN

Progesterone regulates reproductive function through two intracellular receptors, progesterone receptor-A (PR-A) and progesterone receptor-B (PR-B), that arise from a single gene and function as transcriptional regulators of progesterone-responsive genes. Although in vitro studies show that PR isoforms can display different transcriptional regulatory activities, their physiological significance is unknown. By selective ablation of PR-A in mice, we show that the PR-B isoform modulates a subset of reproductive functions of progesterone by regulation of a subset of progesterone-responsive target genes. Thus, PR-A and PR-B are functionally distinct mediators of progesterone action in vivo and should provide suitable targets for generation of tissue-selective progestins.


Asunto(s)
Implantación del Embrión , Progesterona/fisiología , Receptores de Progesterona/fisiología , Reproducción , Útero/fisiología , Animales , División Celular/efectos de los fármacos , Cruzamientos Genéticos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Estradiol/farmacología , Femenino , Regulación de la Expresión Génica , Masculino , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/efectos de los fármacos , Ratones , Ratones Noqueados , Ovariectomía , Ovulación , Progesterona/farmacología , Isoformas de Proteínas , Receptores de Progesterona/genética , Útero/citología , Útero/efectos de los fármacos , Útero/metabolismo
5.
Science ; 279(5358): 1922-5, 1998 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-9506940

RESUMEN

The in vivo biological function of a steroid receptor coactivator was assessed in mice in which the SRC-1 gene was inactivated by gene targeting. Although in both sexes the homozygous mutants were viable and fertile, target organs such as uterus, prostate, testis, and mammary gland exhibited decreased growth and development in response to steroid hormones. Expression of RNA encoding TIF2, a member of the SRC-1 family, was increased in the SRC-1 null mutant, perhaps compensating partially for the loss of SRC-1 function in target tissues. The results indicate that SRC-1 mediates steroid hormone responses in vivo and that loss of its coactivator function results in partial resistance to hormone.


Asunto(s)
Genitales Masculinos/crecimiento & desarrollo , Hormonas Esteroides Gonadales/farmacología , Glándulas Mamarias Animales/crecimiento & desarrollo , Factores de Transcripción/fisiología , Útero/crecimiento & desarrollo , Animales , Línea Celular , Resistencia a Medicamentos , Estradiol/sangre , Estradiol/farmacología , Femenino , Marcación de Gen , Genitales Masculinos/efectos de los fármacos , Histona Acetiltransferasas , Masculino , Glándulas Mamarias Animales/efectos de los fármacos , Ratones , Coactivador 1 de Receptor Nuclear , Coactivador 2 del Receptor Nuclear , Tamaño de los Órganos/efectos de los fármacos , Embarazo , Progesterona/sangre , Progesterona/farmacología , Próstata/efectos de los fármacos , Próstata/crecimiento & desarrollo , Células Madre , Testículo/efectos de los fármacos , Testículo/crecimiento & desarrollo , Testosterona/sangre , Testosterona/farmacología , Factores de Transcripción/genética , Útero/efectos de los fármacos
6.
Science ; 276(5317): 1408-12, 1997 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-9162006

RESUMEN

Despite myriads of biological activities ascribed to uteroglobin (UG), a steroid-inducible secreted protein, its physiological functions are unknown. Mice in which the uteroglobin gene was disrupted had severe renal disease that was associated with massive glomerular deposition of predominantly multimeric fibronectin (Fn). The molecular mechanism that normally prevents Fn deposition appears to involve high-affinity binding of UG with Fn to form Fn-UG heteromers that counteract Fn self-aggregation, which is required for abnormal tissue deposition. Thus, UG is essential for maintaining normal renal function in mice, which raises the possibility that an analogous pathogenic mechanism may underlie genetic Fn-deposit human glomerular disease.


Asunto(s)
Fibronectinas/metabolismo , Glomérulos Renales , Uteroglobina/fisiología , Animales , Células Cultivadas , Cruzamientos Genéticos , Marcación de Gen , Humanos , Enfermedades Renales/embriología , Enfermedades Renales/genética , Enfermedades Renales/patología , Glomérulos Renales/embriología , Glomérulos Renales/metabolismo , Glomérulos Renales/ultraestructura , Ratones , Ratones Endogámicos C57BL , Uteroglobina/deficiencia , Uteroglobina/genética
7.
Sci Rep ; 9(1): 11966, 2019 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-31427604

RESUMEN

Progesterone receptor (PGR) co-ordinately regulates ovulation, fertilisation and embryo implantation through tissue-specific actions, but the mechanisms for divergent PGR action are poorly understood. Here we characterised PGR activity in mouse granulosa cells using combined ChIP-seq for PGR and H3K27ac and gene expression microarray. Comparison of granulosa, uterus and oviduct PGR-dependent genes showed almost complete tissue specificity in PGR target gene profiles. In granulosa cells 82% of identified PGR-regulated genes bound PGR within 3 kb of the gene and PGR binding sites were highly enriched in proximal promoter regions in close proximity to H3K27ac-modified active chromatin. Motif analysis showed highly enriched PGR binding to the PGR response element (GnACAnnnTGTnC), but PGR also interacted significantly with other transcription factor binding motifs. In uterus PGR showed far more tendency to bind intergenic chromatin regions and low evidence of interaction with other transcription factors. This is the first genome-wide description of PGR action in granulosa cells and systematic comparison of diverse PGR action in different reproductive tissues. It clarifies finely-tuned contextual PGR-chromatin interactions with implications for more targeted reproductive medicine.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Regulación de la Expresión Génica , Progesterona/metabolismo , Receptores de Progesterona/metabolismo , Secuencia de Bases , Sitios de Unión , Femenino , Células de la Granulosa/metabolismo , Histonas/metabolismo , Humanos , Motivos de Nucleótidos , Especificidad de Órganos , Ovario/metabolismo , Ovulación/genética , Posición Específica de Matrices de Puntuación , Unión Proteica , Elementos de Respuesta
8.
J Clin Invest ; 83(4): 1183-90, 1989 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-2784798

RESUMEN

Circulating alpha 1-antitrypsin is synthesized primarily in the liver and secreted into the bloodstream, where it serves as the major protease inhibitor. The PiZ variant of alpha 1-antitrypsin is associated with decreased levels of the protein in sera as a result of its retention within hepatocytes. Homozygosity for the variant allele predisposes individuals to the development of pulmonary emphysema and an increased risk for liver disease. We and others have previously demonstrated that the normal PiM human alpha 1-antitrypsin gene can be properly expressed in the livers of transgenic mice. The PiZ variant of the human alpha 1-antitrypsin gene was introduced into the germline of mice to determine whether the mutant protein would accumulate in mouse hepatocytes and if such accumulation would result in the development of liver damage in an animal model. As expected, the mutant human protein was abundantly synthesized in the livers of the transgenic animals and accumulated within the rough endoplasmic reticulum of hepatocytes as it does in human patients. PiZ mice developed significantly more liver necrosis and inflammation than PiM transgenic mice or control littermates. The degree of liver damage was correlated with the amount of PiZ alpha 1-antitrypsin accumulated in the liver of the different pedigrees of mice. Although 40% of PiZ mice tested were seropositive for mouse hepatitis virus (MHV), the degree of liver damage was not influenced by the MHV seropositivity; rather, it was related only to the presence of accumulated PiZ protein.


Asunto(s)
Hígado/patología , alfa 1-Antitripsina/metabolismo , Animales , Humanos , Hígado/efectos de los fármacos , Hígado/ultraestructura , Cirrosis Hepática Experimental/etiología , Cirrosis Hepática Experimental/metabolismo , Cirrosis Hepática Experimental/patología , Ratones , Ratones Transgénicos , Necrosis , Fenotipo , Especificidad de la Especie , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/fisiología
9.
J Clin Invest ; 104(12): 1683-92, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10606622

RESUMEN

Certain mutations in genes for sarcomeric proteins cause hypertrophic cardiomyopathy (HCM). We have developed a transgenic rabbit model for HCM caused by a common point mutation in the beta-myosin heavy chain (MyHC) gene, R400Q. Wild-type and mutant human beta-MyHC cDNAs were cloned 3' to a 7-kb murine beta-MyHC promoter. We injected purified transgenes into fertilized zygotes to generate two lines each of the wild-type and mutant transgenic rabbits. Expression of transgene mRNA and protein were confirmed by Northern blotting and 2-dimensional gel electrophoresis followed by immunoblotting, respectively. Animals carrying the mutant transgene showed substantial myocyte disarray and a 3-fold increase in interstitial collagen expression in their myocardia. Mean septal thicknesses were comparable between rabbits carrying the wild type transgene and their nontransgenic littermates (NLMs) but were significantly increased in the mutant transgenic animals. Posterior wall thickness and left ventricular mass were also increased, but dimensions and systolic function were normal. Premature death was more common in mutant than in wild-type transgenic rabbits or in NLMs. Thus, cardiac expression of beta-MyHC-Q(403) in transgenic rabbits induced hypertrophy, myocyte and myofibrillar disarray, interstitial fibrosis, and premature death, phenotypes observed in humans patients with HCM due to beta-MyHC-Q(403).


Asunto(s)
Cardiomiopatía Hipertrófica/etiología , Modelos Animales de Enfermedad , Cadenas Pesadas de Miosina/genética , Animales , Animales Modificados Genéticamente , Colágeno/análisis , Humanos , Cadenas Pesadas de Miosina/fisiología , ARN Mensajero/análisis , Conejos
10.
Mol Cell Biol ; 7(3): 1276-9, 1987 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-3031486

RESUMEN

We have used a bovine papillomavirus-based vector to generate transgenic mice. Transgenic mice result from either pronuclear or cytoplasmic injections of the vector into fertilized eggs. Of 30 mice generated by microinjection, 27 (90%) contained the vector in its episomal form, at less than one copy per cell. This represents a highly efficient means of gene transfer in which the transgene is in a controlled genetic environment.


Asunto(s)
Genes Virales , Vectores Genéticos , Plásmidos , Transformación Genética , Animales , Papillomavirus Bovino 1/genética , Transferencia de Embrión , Femenino , Regulación de la Expresión Génica , Masculino , Ratones , Microinyecciones , Embarazo
11.
Mol Cell Biol ; 16(5): 2056-64, 1996 May.
Artículo en Inglés | MEDLINE | ID: mdl-8628271

RESUMEN

This report defines the elements between bp -800 and -166 that regulate the quantitative level of mouse CC10 (mCC10) transcription in the lungs. The elements in this promoter domain are the response elements for the NKx2.1 homeobox protein, thyroid transcription factor 1 (TTF1). DNase I footprint analysis identified five binding sites for TTF1 between bp -800 and - 166. These sites are located at bp -344 to -335, - 282 to -273, -268 to -263, -258 to -249, and - 199 to - 190. In addition to these enhancer elements, two TTF1 binding sites were identified in the proximal promoter region (bp - 166 to + 1), at bp -74 to -69 and -49 to -39. An identical footprint of the mCC10 promoter region was also observed with another member of the NKx family, NKx 2.5, the cardiac muscle-specific homeobox protein (CSX). Deletion and linker-scanner mutational analyses of the TTF1 binding sites in the mCC10 distal promoter region with transient cotransfection into CV1 cells with either TTF1 or CSX identified the site located between bp -282 and -273 as the major regulator of CC10 expression, with minor regulation by sites at bp -344 to -335 and -258 to -249. The importance of the NKx binding site at bp -282 to -273 was verified in vivo. Transgenic mice generated with the human growth hormone gene fused to 800 bp of the mCC10 promoter containing a mutation in the TTF1 binding site at bp -282 to -273 showed a reduction in transgene expression equal to that of the mice generated with only 166 bp of 5'-flanking DNA. This report emphasizes the importance of TTF1 or related factors as major regulators of pulmonary gene expression and demonstrates the potential of NKx proteins to bind and activate heterologous target genes.


Asunto(s)
Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Proteínas Nucleares/metabolismo , Biosíntesis de Proteínas , Factores de Transcripción/metabolismo , Transcripción Genética , Uteroglobina , Animales , Secuencia de Bases , Línea Celular , Cloranfenicol O-Acetiltransferasa/biosíntesis , Cartilla de ADN , Inhibidores Enzimáticos , Proteína Homeótica Nkx-2.5 , Humanos , Pulmón , Ratones , Datos de Secuencia Molecular , Mutagénesis Insercional , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes/biosíntesis , Secuencias Reguladoras de Ácidos Nucleicos , Mapeo Restrictivo , Factor Nuclear Tiroideo 1 , Transfección , beta-Galactosidasa/biosíntesis
12.
Mol Cell Biol ; 20(12): 4462-73, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10825210

RESUMEN

CDC37 encodes a 50-kDa protein that targets intrinsically unstable oncoprotein kinases including Cdk4, Raf-1, and v-src to the molecular chaperone Hsp90, an interaction that is thought to be important for the establishment of signaling pathways. CDC37 is required for proliferation in budding yeast and is coexpressed with cyclin D1 in proliferative zones during mouse development, a finding consistent with a positive role in cell proliferation. CDC37 expression may not only be required to support proliferation in cells that are developmentally programmed to proliferate but may also be required in cells that are inappropriately induced to initiate proliferation by oncogenes. Here we report that mouse mammary tumor virus (MMTV)-CDC37 transgenic mice develop mammary gland tumors at a rate comparable to that observed previously in MMTV-cyclin D1 mice. Moreover, CDC37 was found to collaborate with MMTV-c-myc in the transformation of multiple tissues, including mammary and salivary glands in females and testis in males, and also collaborates with cyclin D1 to transform the female mammary gland. These data indicate that CDC37 can function as an oncogene in mice and suggests that the establishment of protein kinase pathways mediated by Cdc37-Hsp90 can be a rate-limiting event in epithelial cell transformation.


Asunto(s)
Proteínas de Ciclo Celular/genética , Transformación Celular Neoplásica , Ciclina D1/genética , Proteínas de Drosophila , Genes myc , Chaperonas Moleculares , Animales , Proteínas de Ciclo Celular/metabolismo , Ciclina D1/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Ratones , Ratones Transgénicos , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo
13.
Mol Cell Biol ; 21(2): 663-77, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11134352

RESUMEN

The dynamic embryonic expression of germ cell nuclear factor (GCNF), an orphan nuclear receptor, suggests that it may play an important role during early development. To determine the physiological role of GCNF, we have generated a targeted mutation of the GCNF gene in mice. Germ line mutation of the GCNF gene proves that the orphan nuclear receptor is essential for embryonic survival and normal development. GCNF(-/-) embryos cannot survive beyond 10.5 days postcoitum (dpc), probably due to cardiovascular failure. Prior to death, GCNF(-/-) embryos suffer significant defects in posterior development. Unlike GCNF(+/+) embryos, GCNF(-/-) embryos do not turn and remain in a lordotic position, the majority of the neural tube remains open, and the hindgut fails to close. GCNF(-/-) embryos also suffer serious defects in trunk development, specifically in somitogenesis, which terminates by 8.75 dpc. The maximum number of somites in GCNF(-/-) embryos is 13 instead of 25 as in the GCNF(+/+) embryos. Interestingly, the tailbud of GCNF(-/-) embryos develops ectopically outside the yolk sac. Indeed, alterations in expression of multiple marker genes were identified in the posterior of GCNF(-/-) embryos, including the primitive streak, the node, and the presomitic mesoderm. These results suggest that GCNF is required for maintenance of somitogenesis and posterior development and is essential for embryonic survival. These results suggest that GCNF regulates a novel and critical developmental pathway involved in normal anteroposterior development.


Asunto(s)
Coristoma/embriología , Proteínas de Unión al ADN/metabolismo , Eliminación de Gen , Esbozos de los Miembros/anomalías , Receptores Citoplasmáticos y Nucleares/metabolismo , Cola (estructura animal)/anomalías , Animales , Diferenciación Celular , Coristoma/metabolismo , Proteínas de Unión al ADN/genética , Desarrollo Embrionario y Fetal , Muerte Fetal , Regulación del Desarrollo de la Expresión Génica , Marcación de Gen , Marcadores Genéticos , Histocitoquímica , Hibridación in Situ , Esbozos de los Miembros/citología , Esbozos de los Miembros/embriología , Esbozos de los Miembros/metabolismo , Ratones , Ratones Noqueados , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares , ARN Mensajero/análisis , ARN Mensajero/genética , Receptores Citoplasmáticos y Nucleares/genética , Recombinación Genética/genética , Somitos/citología , Somitos/metabolismo , Cola (estructura animal)/citología , Cola (estructura animal)/embriología , Cola (estructura animal)/metabolismo
14.
Nat Biotechnol ; 15(3): 239-43, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9062922

RESUMEN

Transgenic mice have been used as models for tissue-specific gene regulation and to examine the molecular and cellular effects of altered expression of specific gene in disease processes such as tumorigenesis. Because of the deleterious effects of constitutive expression of transgenes, which frequently result in prenatal or postnatal death, only a limited number of disease models have been established in transgenic mice. We report an inducible binary transactivation system that permits the control of transgene expression in a tissue-specific and inducible fashion in mice. In this system, transcription of the target transgene is kept silent until turned on by the administration of an exogenous compound. We also demonstrate that expression level of the target gene can be induced three to four orders of magnitude and can be controlled by the administrated compound in a dose-dependent manner.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Hígado/metabolismo , Animales , Células Cultivadas , Hormona de Crecimiento Humana/genética , Ligandos , Hígado/citología , Hígado/efectos de los fármacos , Ratones , Ratones Transgénicos , Mifepristona/farmacocinética , Mifepristona/farmacología , Fenotipo , Transgenes
15.
J Med Genet ; 43(8): 653-9, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16571646

RESUMEN

BACKGROUND: Andersen-Tawil syndrome (ATS) is a rare inherited disorder, characterised by periodic paralysis, cardiac dysarrhythmias, and dysmorphic features, and is caused by mutations in the gene KCNJ2, which encodes the inward rectifier potassium channel, Kir2.1. This study sought to analyse KCNJ2 in patients with familial ATS and to determine the functional characteristics of the mutated gene. METHODS AND RESULTS: We screened a family with inherited ATS for the mutation in KCNJ2, using direct DNA sequencing. A missense mutation (T75R) of Kir2.1, located in the highly conserved cytoplasmic N-terminal domain, was identified in three affected members of this family. Using the Xenopus oocyte expression system and whole cell voltage clamp analyses, we found that the T75R mutant was non-functional and possessed a strong dominant negative effect when co-expressed with the same amount of wild type Kir2.1. Transgenic (Tg) mice expressing the mutated form of Kir2.1 in the heart had prolonged QTc intervals compared with mice expressing the wild type protein. Ventricular tachyarrhythmias were observed in 5 of 14 T75R-Tg mice compared with 1 of 7 Wt-Tg and none of 6 non-transgenic littermates. In three of five T75R-Tg mice with ventricular tachycardia, their ECG disclosed bidirectional tachycardia as in our proband. CONCLUSIONS: The in vitro studies revealed that the T75R mutant of Kir2.1 had a strong dominant negative effect in the Xenopus oocyte expression system. It still preserved the ability to co-assemble and traffic to the cell membrane in mammalian cells. For in vivo studies, the T75R-Tg mice had bidirectional ventricular tachycardia after induction and longer QT intervals.


Asunto(s)
Síndrome de Andersen/genética , Predisposición Genética a la Enfermedad , Mutación/genética , Canales de Potasio de Rectificación Interna/genética , Adolescente , Animales , Análisis Mutacional de ADN , Electrocardiografía , Electrofisiología , Femenino , Humanos , Ratones , Ratones Transgénicos , Miocardio/citología , Miocardio/patología , Miocitos Cardíacos/citología , Xenopus
16.
J Endocrinol ; 189(3): 473-84, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16731779

RESUMEN

Progesterone (P4) and its cognate receptor, the progesterone receptor (PGR), have important roles in the establishment and maintenance of pregnancy in the murine uterus. In previous studies, using high-density DNA microarray analysis, we identified a subset of genes whose expression is repressed by chronic P4-PGR activation in the uterus. The Clca3 gene is one of the genes whose expression is the most significantly downregulated by P4 and PGR. In the present study, we performed real-time RT-PCR and in situ hybridization to investigate the regulation of Clca3 by P4 and determine the pattern of expression of Clca3 in the uterus during early pregnancy. This analysis shows that Clca3 mRNA transcripts were detected in the luminal and glandular epithelium of the pseudopregnant uterus at day 0.5 and that the expression of Clca3 was not detected after day 3.5. P4 represses Clca3 mRNA synthesis in the luminal epithelial and glandular epithelial cells of the uterus in ovariectomized wild-type mice, but not in Pgr knockout (PRKO) mice. Conversely, estrogen (E2) induces Clca3 expression in the luminal epithelium and glandular epithelium, and this induction was repressed by P4 in the murine uterus. Analysis of the promoter region of Clca3 by in silico and transient transfection analysis in HEC-1A cells identified the regulation of Clca3 by estrogen receptor-alpha (ESR1) within the first 528 bp of 5'-flanking region of the Clca3 gene. Our studies identified Clca3 as a novel downregulated gene of PGR that is a direct target of E2 regulation.


Asunto(s)
Canales de Cloruro/genética , Regulación de la Expresión Génica , Progesterona/metabolismo , Regiones Promotoras Genéticas , Receptores de Progesterona/metabolismo , Útero/metabolismo , Animales , Línea Celular , Estradiol/metabolismo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Hibridación in Situ/métodos , Ratones , Ratones Noqueados , Ovariectomía , Embarazo , Progesterona/genética , Seudoembarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección/métodos
17.
Mol Cell Endocrinol ; 247(1-2): 82-90, 2006 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-16406265

RESUMEN

SSTR1 is found on the majority of human pancreatic beta cells, however, its role in insulin secretion has yet to be elucidated. In this study, we used the SSTR1 knockout mouse model to examine the role of SSTR1 in insulin secretion and glucose homeostasis in mice. Despite the reported effect of SSTR1 in inhibiting growth hormone secretion, SSTR1-/- mice had significantly reduced body weight with growth retardation. Perfusion of isolated mouse pancreata at 3 months of age demonstrated a significant increase in insulin secretion in SSTR1-/- mice compared with that of WT controls. We also found that at 3 months of age, SSTR1-/- mice had significantly decreased levels of systemic insulin secretion and were glucose intolerant. However, SSTR1 gene-ablated mice had a much higher rate of insulin clearance compared to WT mice at the same age. When challenged at 12 months of age, we found SSTR1-/- mice had increased glucose tolerance with exaggerated increase of insulin levels at the end of the experiment. Immunochemical analysis showed that the pancreatic islets of SSTR1-/- mice had significantly decreased levels of somatostatin staining and a significant decrease of SSTR5 expression. These results demonstrate that SSTR1 plays an important role in the regulation of insulin secretion in the endocrine pancreas in mice.


Asunto(s)
Glucosa/metabolismo , Islotes Pancreáticos/metabolismo , Receptores de Somatostatina/metabolismo , Factores de Edad , Animales , Crecimiento , Homeostasis , Insulina/metabolismo , Resistencia a la Insulina , Secreción de Insulina , Islotes Pancreáticos/patología , Ratones , Ratones Noqueados , Receptores de Somatostatina/genética
18.
J Steroid Biochem Mol Biol ; 102(1-5): 41-50, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17067792

RESUMEN

The ovarian steroid hormone progesterone is a major regulator of uterine function. The actions of this hormone is mediated through its cognate receptor, the progesterone receptor, Pgr. Ablation of the Pgr has shown that this receptor is critical for all female reproductive functions including the ability of the uterus to support and maintain the development of the implanting mouse embryo. High density DNA microarray analysis has identified direct and indirect targets of Pgr action. One of the targets of Pgr action is a member of the Hedgehog morphogen Indian Hedgehog, Ihh. Ihh and members of the Hh signaling cascade show a coordinate expression pattern in the mouse uterus during the preimplantation period of pregnancy. The expression of Ihh and its receptor Patched-1, Ptc1, as well as, down stream targets of Ihh-Ptch1 signaling, such as the orphan nuclear receptor COUP-TF II show that this morphogen pathway mediates communication between the uterine epithelial and stromal compartments. The members of the Ihh signaling axis may function to coordinate the proliferation, vascularization and differentiation of the uterine stroma during pregnancy. This analysis demonstrates that progesterone regulates uterine function in the mouse by coordinating the signals from the uterine epithelium to stroma in the preimplantation mouse uterus.


Asunto(s)
Receptores de Progesterona/metabolismo , Útero/fisiología , Animales , Femenino , Proteínas Hedgehog/metabolismo , Humanos , Transducción de Señal , Útero/citología
19.
Circ Res ; 88(6): 587-92, 2001 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-11282892

RESUMEN

Cardiac-restricted expression of Cre recombinase can provoke lineage-specific gene excision in the myocardium. However, confounding early lethality may still preclude using loss-of-function models to study the postnatal heart. Here, we have tested whether inducible, heart-specific recombination can be triggered after birth by transgenic expression of a Cre fusion protein that incorporates a mutated progesterone receptor ligand binding domain (PR1) that is activated by the synthetic antiprogestin, RU486, but not by endogenous steroid hormones. CrePR1 driven by the alpha-myosin heavy chain (alphaMHC) promoter was expressed specifically in heart. Translocation of CrePR1 from cytoplasm to nuclei in ventricular myocytes was induced by RU486. To establish whether this approach can mediate cardiac-specific, drug-dependent excision between loxP sites in vivo, we mated alphaMHC-CrePR1 mice with a ubiquitously expressed (ROSA26) Cre reporter line. Offspring harboring alphaMHC-CrePR1 and/or the floxed allele were injected with RU486 versus vehicle, and the prevalence of beta-galactosidase (beta-gal)-positive cells was determined, indicative of Cre-mediated excision. Little or no baseline recombination was seen 1 week after birth. Cardiac-restricted, RU486-inducible recombination was demonstrated in bigenic mice at age 3 and 6 weeks, using each of 3 independent CrePR1 lines. Recombination in the absence of ligand paralleled the levels of CrePR1 protein expression and was more evident at 6 weeks. Thus, conditional, posttranslational activation of a Cre fusion protein can bypass potential embryonic and perinatal effects on the heart and permits inducible recombination in cardiac muscle. High levels of the chimeric Cre protein, in particular, were associated with progressive recombination in the absence of drug.


Asunto(s)
Hormonas/farmacología , Integrasas/genética , Miocardio/metabolismo , Proteínas Virales , Animales , Animales Recién Nacidos , Transporte Biológico/efectos de los fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Marcación de Gen , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Integrasas/metabolismo , Ratones , Ratones Transgénicos , Mifepristona/farmacología , Miocardio/citología , Cadenas Pesadas de Miosina/genética , Regiones Promotoras Genéticas/genética , Ratas , Ratas Sprague-Dawley , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Proteínas Recombinantes de Fusión/efectos de los fármacos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Recombinación Genética/efectos de los fármacos
20.
Cancer Res ; 56(18): 4096-102, 1996 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-8797572

RESUMEN

We have previously reported the development of a transgenic mouse model for prostate cancer derived from PB-Tag transgenic line 8247, henceforth designated the TRAMP (transgenic adenocarcinoma mouse prostate) model. We now describe the temporal and spatial consequences of transgene expression and report the identification and characterization of metastatic disease in the TRAMP model. TRAMP mice characteristically express the T antigen oncoprotein by 8 weeks of age and develop distinct pathology in the epithelium of the dorsolateral prostate by 10 weeks of age. Distant site metastases can be detected as early as 12 weeks of age. The common sites of metastases are the periaortic lymph nodes and lungs, with occasional metastases to the kidney, adrenal gland, and bone. By 28 weeks of age, 100% harbor metastatic prostate cancer in the lymph nodes or lungs. We have also demonstrated the loss of normal E-cadherin expression, as observed in human prostate cancer, as primary tumors become less differentiated and metastasize. The TRAMP model provides a consistent source of primary and metastatic tumors for histopathobiological and molecular analysis to further define the earliest molecular events involved in the genesis, progression, and metastasis of prostate cancer.


Asunto(s)
Adenocarcinoma/patología , Antígenos Virales de Tumores/biosíntesis , Metástasis de la Neoplasia , Neoplasias de la Próstata/patología , Envejecimiento , Animales , Animales Modificados Genéticamente , Antígenos Virales de Tumores/análisis , Antígenos Virales de Tumores/genética , Línea Celular , Humanos , Inmunohistoquímica , Metástasis Linfática , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Especificidad de Órganos , Recombinación Genética , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA