Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Gene ; 715: 144028, 2019 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-31374326

RESUMEN

BACKGROUND: Type 2 diabetes (T2D) is a complex polygenic disease with unclear mechanism. In an attempt to identify novel genes involved in ß-cell function, we harness a bioinformatics method called Loss-of-function tool (LoFtool) gene score. METHODS: RNA-sequencing data from human islets were used to cross-reference genes within the 1st quartile of most intolerant LoFtool score with the 100th most expressed genes in human islets. Out of these genes, GNAS and EEF1A1 genes were selected for further investigation in diabetic islets, metabolic tissues along with their correlation with diabetic phenotypes. The influence of GNAS and EEF1A1 on insulin secretion and ß-cell function were validated in INS-1 cells. RESULTS: A comparatively higher expression level of GNAS and EEF1A1 was observed in human islets than fat, liver and muscle tissues. Furthermore, diabetic islets displayed a reduced expression of GNAS, but not of EEF1A, compared to non-diabetic islets. The expression of GNAS was positively correlated with insulin secretory index, GLP1R, GIPR and inversely correlated with HbA1c. Diabetic human islets displayed a reduced cAMP generation and insulin secretory capacity in response to glucose. Moreover, siRNA silencing of GNAS in INS-1 cells reduced insulin secretion, insulin content, and cAMP production. In addition, the expression of Insulin, PDX1, and MAFA was significantly down-regulated in GNAS-silenced cells. However, cell viability and apoptosis rate were unaffected. CONCLUSION: LoFtool is a powerful tool to identify genes associated with pancreatic islets dysfunction. GNAS is a crucial gene for the ß-cell insulin secretory capacity.


Asunto(s)
Cromograninas/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Regulación de la Expresión Génica , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Anciano , Animales , Línea Celular , Cromograninas/genética , AMP Cíclico/genética , AMP Cíclico/metabolismo , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Humanos , Células Secretoras de Insulina/citología , Masculino , Persona de Mediana Edad , Especificidad de Órganos , Factor 1 de Elongación Peptídica/genética , Factor 1 de Elongación Peptídica/metabolismo , Ratas
2.
Nat Commun ; 8: 15161, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28443644

RESUMEN

Schwann cell (SC) myelination in the peripheral nervous system is essential for motor function, and uncontrolled SC proliferation occurs in cancer. Here, we show that a dual role for Hippo effectors TAZ and YAP in SC proliferation and myelination through modulating G-protein expression and interacting with SOX10, respectively. Developmentally regulated mutagenesis indicates that TAZ/YAP are critical for SC proliferation and differentiation in a stage-dependent manner. Genome-wide occupancy mapping and transcriptome profiling reveal that nuclear TAZ/YAP promote SC proliferation by activating cell cycle regulators, while targeting critical differentiation regulators in cooperation with SOX10 for myelination. We further identify that TAZ targets and represses Gnas, encoding Gαs-protein, which opposes TAZ/YAP activities to decelerate proliferation. Gnas deletion expands SC precursor pools and blocks peripheral myelination. Thus, the Hippo/TAZ/YAP and Gαs-protein feedback circuit functions as a fulcrum balancing SC proliferation and differentiation, providing insights into molecular programming of SC lineage progression and homeostasis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cromograninas/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Vaina de Mielina/metabolismo , Fosfoproteínas/metabolismo , Factores de Transcripción SOXE/metabolismo , Células de Schwann/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Ciclo Celular , Diferenciación Celular , Línea Celular , Proliferación Celular , Cromograninas/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfoproteínas/genética , Ratas , Proteínas Represoras/metabolismo , Transactivadores , Factor de Transcripción HES-1/metabolismo , Proteínas Señalizadoras YAP
3.
PLoS One ; 10(2): e0117378, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25659103

RESUMEN

Differential marking of genes in female and male gametes by DNA methylation is essential to genomic imprinting. In female gametes transcription traversing differentially methylated regions (DMRs) is a common requirement for de novo methylation at DMRs. At the imprinted Gnas cluster oocyte specific transcription of a protein-coding transcript, Nesp, is needed for methylation of two DMRs intragenic to Nesp, namely the Nespas-Gnasxl DMR and the Exon1A DMR, thereby enabling expression of the Gnas transcript and repression of the Gnasxl transcript. On the paternal allele, Nesp is repressed, the germline DMRs are unmethylated, Gnas is repressed and Gnasxl is expressed. Using mutant mouse models, we show that on the paternal allele, ectopic transcription of Nesp traversing the intragenic Exon1A DMR (which regulates Gnas expression) results in de novo methylation of the Exon1A DMR and de-repression of Gnas just as on the maternal allele. However, unlike the maternal allele, methylation on the mutant paternal allele occurs post-fertilisation, i.e. in somatic cells. This, to our knowledge is the first example of transcript/transcription driven DNA methylation of an intragenic CpG island, in somatic tissues, suggesting that transcription driven de novo methylation is not restricted to the germline in the mouse. Additionally, Gnasxl is repressed on a paternal chromosome on which Nesp is ectopically expressed. Thus, a paternally inherited Gnas cluster showing ectopic expression of Nesp is "maternalised" in terms of Gnasxl and Gnas expression. We show that these mice have a phenotype similar to mutants with two expressed doses of Gnas and none of Gnasxl.


Asunto(s)
Metilación de ADN/fisiología , ADN Intergénico/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Familia de Multigenes/fisiología , Transcripción Genética/fisiología , Alelos , Animales , Cromograninas , Islas de CpG/fisiología , ADN Intergénico/genética , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Masculino , Ratones , Ratones Mutantes
4.
J Bone Miner Res ; 29(11): 2357-68, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24764158

RESUMEN

Fibrous dysplasia of bone (FD) is a crippling skeletal disease associated with postzygotic mutations (R201C, R201H) of the gene encoding the α subunit of the stimulatory G protein, Gs. By causing a characteristic structural subversion of bone and bone marrow, the disease results in deformity, hypomineralization, and fracture of the affected bones, with severe morbidity arising in childhood or adolescence. Lack of inheritance of the disease in humans is thought to reflect embryonic lethality of germline-transmitted activating Gsα mutations, which would only survive through somatic mosaicism. We have generated multiple lines of mice that express Gsα(R201C) constitutively and develop an inherited, histopathologically exact replica of human FD. Robust transgene expression in neonatal and embryonic tissues and embryonic stem (ES) cells were associated with normal development of skeletal tissues and differentiation of skeletal cells. As in humans, FD lesions in mice developed only in the postnatal life; a defined spatial and temporal pattern characterized the onset and progression of lesions across the skeleton. In individual bones, lesions developed through a sequence of three distinct histopathological stages: a primary modeling phase defined by endosteal/medullary excess bone formation and normal resorption; a secondary phase, with excess, inappropriate remodeling; and a tertiary fibrous dysplastic phase, which reproduced a full-blown replica of the human bone pathology in mice of age ≥1 year. Gsα mutations are sufficient to cause FD, and are per se compatible with germline transmission and normal embryonic development in mice. Our novel murine lines constitute the first model of FD.


Asunto(s)
Modelos Animales de Enfermedad , Displasia Fibrosa Ósea , Subunidades alfa de la Proteína de Unión al GTP Gs , Expresión Génica , Mutación Missense , Factores de Edad , Sustitución de Aminoácidos , Animales , Remodelación Ósea/genética , Embrión de Mamíferos/enzimología , Embrión de Mamíferos/patología , Displasia Fibrosa Ósea/enzimología , Displasia Fibrosa Ósea/genética , Displasia Fibrosa Ósea/patología , Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Humanos , Ratones , Ratones Transgénicos , Osteogénesis/genética
5.
Horm Res Paediatr ; 80(4): 229-41, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24107509

RESUMEN

GNAS is a complex imprinted locus leading to several different gene products that show exclusive monoallelic expression. GNAS also encodes the α-subunit of the stimulatory G protein (Gsα), a ubiquitously expressed signaling protein that is essential for the actions of many hormones and other endogenous molecules. Gsα is expressed biallelically in most tissues but its expression is silenced from the paternal allele in a small number of tissues. The tissue-specific paternal silencing of Gsα results in different parent-of-origin-specific phenotypes in patients who carry inactivating GNAS mutations. In this paper, we review the GNAS complex locus and discuss how disruption of Gsα expression and the expression of other GNAS products shape the phenotypes of human disorders caused by mutations in this gene.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs , Regulación de la Expresión Génica , Enfermedades Genéticas Congénitas , Sitios Genéticos , Impresión Genómica , Mutación , Cromograninas , Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/metabolismo , Humanos , Especificidad de Órganos/genética
6.
Genet Mol Res ; 12(3): 3479-87, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-24065686

RESUMEN

Olfactory neuroblastoma (ONB) is a malignant tumor found in the human nasal cavity. These tumors are rare and poorly characterized at the molecular level. In this study, we asked whether olfactory-specific genes are expressed in ONBs by using reverse-transcriptase-polymerase chain reaction. We found that the olfactory marker protein and the RIC-8B genes, which are specifically expressed in mature olfactory neurons, are expressed in ONBs. Importantly, we also found that ONBs express a large variety of odorant receptor genes, representative of different odorant receptor gene subfamilies. Our results show that the ONBs express genes that are normally expressed in mature olfactory neurons and indicate that they are derived from progenitor or immature cells in the olfactory epithelium and not from a clonal expansion of a single or few mature olfactory neurons.


Asunto(s)
Estesioneuroblastoma Olfatorio/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Neoplasias Nasales/genética , Receptores Odorantes/genética , Estesioneuroblastoma Olfatorio/patología , Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Regulación Neoplásica de la Expresión Génica , Humanos , Cavidad Nasal/patología , Neoplasias Nasales/patología , Neuronas Receptoras Olfatorias/citología , Neuronas Receptoras Olfatorias/metabolismo , Receptores Odorantes/biosíntesis , Olfato/genética
7.
Pharmacol Rep ; 64(2): 256-65, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22661174

RESUMEN

BACKGROUND: SB 269970, a 5-HT(7) receptor antagonist may produce a faster antidepressant-like effect in animal models, than do antidepressant drugs, e.g., imipramine. The present work was aimed at examining the effect of single and repeated (14 days) administration of SB 269970 on the 5-HT(7) receptor in the hippocampus. METHODS: The reactivity of 5-HT(7) receptors was determined using 5-carboxamidotryptamine (5-CT), which increased the bursting frequency of spontaneous epileptiform activity in hippocampal slices. Additionally, the effects of SB 269970 administration on the affinity and density of 5-HT(7) receptors were investigated using [(3)H]-SB 269970 and the influence of SB 269970 and imipramine on mRNA expression levels of Gα(s) and Gα(12) mRNA were studied using RT-qPCR. RESULTS: Acute and repeated treatment with SB 269970 led to attenuation of the excitatory effects of activation of 5-HT(7) receptors. Neither single nor repeated administration of SB 269970 changed the mean affinity of 5-HT(7) receptors for [(3)H]-SB 269970. Repeated, but not single, administration of SB 269970 decreased the maximum density of [(3)H]-SB 269970 binding sites. While administration of imipramine did not change the expression of mRNAs for Gα(s) and Gα(12) proteins after both single and repeated administration of SB 269970, a reduction in Gα(s) and Gα(12) mRNA expression levels was evident. CONCLUSIONS: These findings indicate that even single administration of SB269970 induces functional desensitization of the 5-HT(7) receptor system, which precedes changes in the receptor density. This mechanism may be responsible for the rapid antidepressant-like effect of the 5-HT(7) antagonist in animal models.


Asunto(s)
Hipocampo/efectos de los fármacos , Fenoles/farmacología , Receptores de Serotonina/metabolismo , Antagonistas de la Serotonina/farmacología , Sulfonamidas/farmacología , Animales , Antidepresivos Tricíclicos/farmacología , Sitios de Unión , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Fenómenos Electrofisiológicos , Subunidades alfa de la Proteína de Unión al GTP G12-G13/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Hipocampo/metabolismo , Imipramina/farmacología , Masculino , Fenoles/administración & dosificación , Unión Proteica , ARN Mensajero/biosíntesis , Ratas , Ratas Wistar , Serotonina/análogos & derivados , Serotonina/farmacología , Antagonistas de la Serotonina/administración & dosificación , Sulfonamidas/administración & dosificación
8.
Am J Physiol Heart Circ Physiol ; 302(8): H1591-602, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22268112

RESUMEN

Oxidative stress has been shown to increase the expression of G(i)α proteins in vascular smooth muscle cells (VSMC) from spontaneously hypertensive rats. The present study was undertaken to examine if H(2)O(2), which induces oxidative stress, could also enhance the expression of G(i)α proteins in VSMC and to further explore the underlying signaling pathways responsible for this response. Treatment of VSMC with H(2)O(2) increased the expression of G(i)α proteins and not of G(s)α protein in a concentration- and time-dependent manner. A maximal increase of ∼40-50% was observed at 100 µM and 1 h and was restored to control levels by AG1295 and AG1478, inhibitors of epidermal growth factor receptor (EGF-R) and platelet-derived growth factor receptor (PDGF-R), respectively, and PD98059 and U126, inhibitors of extracellular signal-regulated kinase (ERK1/2), and wortmannin and AKT inhibitor VIII, inhibitors of PKB/AKT, respectively. In addition, H(2)O(2) also increased the phosphorylation of EGF-R, PDGF-R, ERK1/2, and AKT, which was attenuated by the respective inhibitors, whereas the inhibitors of EGF-R and PDGE-R also inhibited the enhanced phosphorylation of ERK1/2 and AKT. Furthermore, transfection of cells with short interfering RNA of EGF-R and PDGF-R restored the H(2)O(2)-induced enhanced expression of G(i)α proteins to control levels. The increased expression of G(i)α proteins was reflected in enhanced G(i) functions as demonstrated by enhanced inhibition of adenylyl cyclase by inhibitory hormones and forskolin-stimulated adenylyl cyclase activity by a low concentration of GTPγS, whereas G(s)α-mediated stimulations of AC were significantly decreased. Furthermore, H(2)O(2)-induced enhanced proliferation of VSMC was attenuated by dibutyryl-cAMP. These results suggest that H(2)O(2) increases the expression of G(i)α proteins in VSMC through the transactivation of EGF-R/PDGF-R and ERK1/2 and phosphatidylinositol-3 kinase signaling pathways.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/biosíntesis , Peróxido de Hidrógeno/farmacología , Miocitos del Músculo Liso/fisiología , Oxidantes/farmacología , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología , Adenilil Ciclasas/metabolismo , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Células Cultivadas , AMP Cíclico/biosíntesis , AMP Cíclico/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Proteínas Quinasas Activadas por Mitógenos/fisiología , Proteína Oncogénica v-akt/biosíntesis , Proteína Oncogénica v-akt/genética , Fosfatidilinositol 3-Quinasas/fisiología , Fosforilación , ARN Interferente Pequeño/farmacología , Ratas , Receptores de Factores de Crecimiento/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Activación Transcripcional/fisiología
9.
Pflugers Arch ; 462(4): 529-43, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21811789

RESUMEN

It is uncertain if downregulation of ß-adrenoceptor signaling pathway is promoted by an enhanced adrenergic tone at an early stage of cardiac disease, or it develops secondary to detrimental local myocardial changes in advanced heart failure. We examined the integrity of ß-adrenoceptor signaling pathway upon chronic infusion of isoproterenol, a ß-adrenoceptor agonist, at a dose producing no structural left ventricular (LV) remodeling and systolic dysfunction. Subcutaneous isoproterenol infusion (400 µg kg(-1) h(-1) over 16 days) to guinea pigs using osmotic minipumps produced no change in cardiac weights, LV internal dimensions, myocyte cross-sectional area, extent of interstitial fibrosis, and basal contractile function. Isolated, perfused heart preparations from isoproterenol-treated guinea pigs exhibited attenuated responsiveness to acute ß-adrenoceptor stimulation, as evidenced by reduced LV developed pressure increase, less shortening of LV epicardial monophasic action potential and effective refractory period, and less myocardial cyclic adenosine monophosphate elevation, in response to isoproterenol exposure, when compared to saline-treated controls. Pharmacological responses to forskolin, an activator of the adenylate cyclase catalytic subunit, were well preserved in isoproterenol-treated hearts. Downregulation of ß-adrenoceptor-mediated effects upon chronic isoproterenol infusion was associated with markedly reduced stimulatory G-protein α-subunit (G(sα)) myocardial expression levels. No change in expression levels of ß-adrenoceptors, G-protein-coupled receptor kinase 2, inhibitory G-protein α-subunit (G(iα2)), and Ca(v)1.2 and K(v)7.1 ion channels was determined in isoproterenol-treated hearts. We therefore conclude that sustained adrenergic overstimulation may promote downregulation of myocardial ß-adrenoceptor-mediated effects independently of structural LV remodeling and systolic failure, an effect attributed to ß-adrenoceptor uncoupling from adenylate cyclase due to reduced G(sα)-protein expression.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Miocardio/metabolismo , Receptores Adrenérgicos beta/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sístole/efectos de los fármacos , Animales , Canales de Calcio Tipo L/metabolismo , Colforsina/farmacología , AMP Cíclico/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Quinasa 2 del Receptor Acoplado a Proteína-G/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Cobayas , Isoproterenol/farmacología , Canal de Potasio KCNQ1/metabolismo , Masculino , Contracción Miocárdica/efectos de los fármacos , Receptores Adrenérgicos beta/fisiología , Periodo Refractario Electrofisiológico , Remodelación Ventricular/efectos de los fármacos
10.
J Biol Chem ; 286(4): 2625-35, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21115479

RESUMEN

Ric-8A and Ric-8B are nonreceptor G protein guanine nucleotide exchange factors that collectively bind the four subfamilies of G protein α subunits. Co-expression of Gα subunits with Ric-8A or Ric-8B in HEK293 cells or insect cells greatly promoted Gα protein expression. We exploited these characteristics of Ric-8 proteins to develop a simplified method for recombinant G protein α subunit purification that was applicable to all Gα subunit classes. The method allowed production of the olfactory adenylyl cyclase stimulatory protein Gα(olf) for the first time and unprecedented yield of Gα(q) and Gα(13). Gα subunits were co-expressed with GST-tagged Ric-8A or Ric-8B in insect cells. GST-Ric-8·Gα complexes were isolated from whole cell detergent lysates with glutathione-Sepharose. Gα subunits were dissociated from GST-Ric-8 with GDP-AlF(4)(-) (GTP mimicry) and found to be >80% pure, bind guanosine 5'-[γ-thio]triphosphate (GTPγS), and stimulate appropriate G protein effector enzymes. A primary characterization of Gα(olf) showed that it binds GTPγS at a rate marginally slower than Gα(s short) and directly activates adenylyl cyclase isoforms 3, 5, and 6 with less efficacy than Gα(s short).


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs/aislamiento & purificación , Subunidades alfa de la Proteína de Unión al GTP/aislamiento & purificación , Glutatión Transferasa/aislamiento & purificación , Proteínas Recombinantes de Fusión/aislamiento & purificación , Adenilil Ciclasas/química , Adenilil Ciclasas/metabolismo , Animales , Baculoviridae/genética , Activación Enzimática , Subunidades alfa de la Proteína de Unión al GTP/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP/química , Subunidades alfa de la Proteína de Unión al GTP/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gs/química , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Expresión Génica , Glutatión Transferasa/biosíntesis , Glutatión Transferasa/química , Glutatión Transferasa/genética , Células HEK293 , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Spodoptera
11.
Gynecol Oncol ; 118(2): 160-6, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20537689

RESUMEN

OBJECTIVES: The purpose of this study was to identify genes that predict progression-free survival (PFS) in advanced epithelial ovarian cancer (aEOC) receiving standard therapy. METHODS: We performed microarray analysis on laser microdissected aEOC cells. All cases received staging laparotomy and adjuvant chemotherapy (carboplatin+paclitaxel) as primary therapy. RESULTS: Microarray analysis identified 50 genes differentially expressed between tumors of patients with no evidence of disease (NED) or evidence of disease (ED) (p<0.001). Six genes (13%) were located at 8q24, and 9 genes (19.6%), at 20q11-13. The ratio of selected gene set/analyzed gene set in chromosomes 8 and 20 are significantly higher than that in other chromosome regions (6/606 vs. 32/13656, p=0.01) and (12/383 vs. 32/13656, p=1.3 x 10(-)(16)). We speculate that the abnormal chromosomal distribution is due to genomic alteration and that these genes may play an important role in aEOC and choose GNAS (GNAS complex locus, NM_000516) on 20q13 based on the p value and fold change. Genomic PCR of aEOC cells also showed that amplification of GNAS was significantly correlated with unfavorable PFS (p=0.011). Real-time quantitative RT-PCR analysis of independent samples revealed that high mRNA expression levels of the GNAS genes, located at chromosome 20q13, was significantly unfavorable indicators of progression-free survival (PFS). Finally, GNAS amplification was an independent prognostic factor for PFS. CONCLUSIONS: Our results suggest that GNAS gene amplification may be an independent, qualitative, and reproducible biomarker of PFS in aEOC.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Neoplasias Ováricas/genética , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/genética , Carboplatino/administración & dosificación , Quimioterapia Adyuvante , Cromograninas , Supervivencia sin Enfermedad , Células Epiteliales/patología , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Amplificación de Genes , Humanos , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Neoplasias Ováricas/cirugía , Paclitaxel/administración & dosificación , Valor Predictivo de las Pruebas , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/genética , ARN Neoplásico/aislamiento & purificación , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Proc Natl Acad Sci U S A ; 105(4): 1209-14, 2008 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-18212126

RESUMEN

Osteoblasts are essential for maintaining bone mass, avoiding osteoporosis, and repairing injured bone. Activation of osteoblast G protein-coupled receptors (GPCRs), such as the parathyroid hormone receptor, can increase bone mass; however, the anabolic mechanisms are poorly understood. Here we use "Rs1," an engineered GPCR with constitutive G(s) signaling, to evaluate the temporal and skeletal effects of G(s) signaling in murine osteoblasts. In vivo, Rs1 expression induces a dramatic anabolic skeletal response, with midfemur girth increasing 1,200% and femur mass increasing 380% in 9-week-old mice. Bone volume, cellularity, areal bone mineral density, osteoblast gene markers, and serum bone turnover markers were also elevated. No such phenotype developed when Rs1 was expressed after the first 4 weeks of postnatal life, indicating an exquisite temporal sensitivity of osteoblasts to Rs1 expression. This pathway may represent an important determinant of bone mass and may open future avenues for enhancing bone repair and treating metabolic bone diseases.


Asunto(s)
Densidad Ósea/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Osteoblastos/química , Osteoblastos/metabolismo , Ingeniería de Proteínas , Receptores de Serotonina 5-HT4/biosíntesis , Receptores de Serotonina 5-HT4/genética , Secuencia de Aminoácidos , Animales , Línea Celular , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/fisiología , Humanos , Ligandos , Masculino , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Ingeniería de Proteínas/métodos , Agonistas del Receptor de Serotonina 5-HT4 , Transducción de Señal/genética , Transducción de Señal/fisiología
13.
Am J Physiol Heart Circ Physiol ; 294(2): H775-84, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18055527

RESUMEN

We previously showed that S-nitroso-N-acetylpenicillamine, a nitric oxide donor, decreased the levels and functions of G(i)alpha proteins by formation of peroxynitrite (ONOO(-)) in vascular smooth muscle cells (VSMC). The present studies were undertaken to investigate whether ONOO(-) can modulate the expression of G(i)alpha protein and associated adenylyl cyclase signaling in VSMC. Treatment of A-10 and aortic VSMC with ONOO(-) for 24 h decreased the expression of G(i)alpha-2 and G(i)alpha-3, but not G(s)alpha, protein in a concentration-dependent manner; expression was restored toward control levels by (111)Mn-tetralis(benzoic acid porphyrin) and uric acid, but not by 1H[1,2,4]oxadiazole[4,3-a]quinoxaline-1-one (ODQ) and KT-5823. cGMP levels were increased by approximately 50% and 150% by 0.1 and 0.5 mM ONOO(-), respectively, and attenuated toward control levels by ODQ. In addition, 0.5 mM ONOO(-) attenuated the inhibition of adenylyl cyclase by ANG II and C-type atrial natriuretic peptide (C-ANP(4-23)), as well as the inhibition of forskolin-stimulated adenylyl cyclase activity by GTPgammaS, whereas, the G(s)-mediated stimulations were augmented. In addition, 0.5 mM ONOO(-) decreased phosphorylation of ERK1/2 and p38 MAP kinase and enhanced JNK phosphorylation but did not affect AKT1/3 phosphorylation. These results suggest that ONOO(-) decreased the expression of G(i) proteins and associated functions in VSMC through a cGMP-independent mechanism and may involve the MAP kinase signaling pathway.


Asunto(s)
Inhibidores de Adenilato Ciclasa , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/biosíntesis , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/fisiología , Ácido Peroxinitroso/farmacología , Transducción de Señal/fisiología , Animales , Aorta Torácica/citología , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Western Blotting , Carbazoles/farmacología , Células Cultivadas , Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gs/fisiología , Indoles/farmacología , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/fisiología , Relajación Muscular/efectos de los fármacos , Relajación Muscular/fisiología , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Oxadiazoles/farmacología , Ácido Peroxinitroso/antagonistas & inhibidores , Quinoxalinas/farmacología , Ratas , Receptor de Angiotensina Tipo 1/biosíntesis , Receptores del Factor Natriurético Atrial/biosíntesis , Transducción de Señal/efectos de los fármacos
14.
Auton Neurosci ; 132(1-2): 52-62, 2007 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-17185044

RESUMEN

Neuroendocrine secretory protein 55 (NESP55) is a soluble, acidic and heat-stable protein, belonging to the class of chromogranins. It is expressed specifically in endocrine cells and the nervous system, and is probably involved in both constitutive and regulated secretion. In the present study, we investigated the distribution of NESP55 in various rat sympathetic ganglia by immunohistochemistry. The expression of NESP55-IR was detected in a subpopulation of principal neurons in the rat SCG, which was also TH positive, and, thus, adrenergic. In the rat stellate ganglion, more than two thirds of NESP55 positive neurons were adrenergic. Colocalization of NESP55 and calcitonin gene-related peptide (CGRP) in cholinergic neurons was also observed. In the rat thoracic chain, however, the majority of NESP55 positive neurons appeared to lack TH. No detectable NESP55-IR was found in the mouse SCG. Furthermore, in the sexually dimorphic SCG, it was demonstrated that, 80% of the NESP55 positive principal neurons were also NPY positive in the male rat, while a slightly higher, but statistically significant proportion, 87%, was found in the female. Whether or not this small difference is physiologically significant is unknown. The present data provide basic knowledge about the expression of NESP55 in the sympathetic autonomic nervous system of rat, which may further our understanding of the functional significance of NESP55.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Ganglios Simpáticos/metabolismo , Ganglio Cervical Superior/metabolismo , Animales , Cromograninas , Femenino , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos BALB C , Neuronas/metabolismo , Ratas , Factores Sexuales , Especificidad de la Especie
15.
Anticancer Res ; 25(3B): 2075-83, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16158948

RESUMEN

BACKGROUND: Calcitonin (CT) exerts an autocrine/paracrine influence on prostatic tumor invasion through coupling to transduction protein Gsalpha. Cell adhesion glycoprotein CD44 variant v7-v10 also faciliates invasion, but its modulation by the CT-Gsalpha system was unexplored. MATERIALS AND METHODS: LnCaP, PC-3 and metastasis-derived PC-3M cell lines were studied, including cells modified therefrom: Gsalpha-QL, expressing mutant constitutively active Gsalpha protein, and CT+, overexpressing CT. CD44 variant expression was evaluated in vivo after orthotopic implantion into nude mice, and in vitro by real-time RT-PCR and Western blotting. RESULTS: Both mRNA and protein levels of the CD44 variant were minimal in PC-3M tumor implants, but elevated in Gsalpha-QL. Exogenous CT stimulated invasion into Matrigel strongly in LnCaP and CT+, and less in PC-3 and Gsalpha-QL. By Western blot analysis, untreated Gsalpha-QL and CT+ cells overexpressed CD44 variant compared with LnCaP or PC-3. By quantitative RT-PCR, exogenous CT dose-dependently increased CD44 variant mRNA to seven-fold. Pharmacologic agents that stimulated or inhibited Gsalpha activity or stimulated adenylyl cyclase produced proportionate dose-dependent effects on both CD44 variant expression and Matrigel invasion. CONCLUSION: This paracrine factor, acting though cyclic AMP, regulates the expression of CD44v7-10, which modulates the tumor phenotype.


Asunto(s)
Calcitonina/farmacología , Receptores de Hialuranos/biosíntesis , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Adenilil Ciclasas/metabolismo , Animales , Calcitonina/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Guanilil Imidodifosfato/farmacología , Humanos , Receptores de Hialuranos/genética , Hibridación in Situ , Masculino , Ratones , Ratones Desnudos , Invasividad Neoplásica , Neoplasias de la Próstata/genética , Isoformas de Proteínas , ARN Mensajero/biosíntesis , ARN Mensajero/genética
16.
Horm Metab Res ; 37(6): 343-6, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16001325

RESUMEN

Medicine is an ever changing art, continuously adjusting to the shifting principles of philosophy and constant discoveries of science; it was beautifully said by Hippocrates: "... medicine does not do the same thing at this moment and the next..." Unabated dissemination of information is the only way that patients are assured that physicians will continue to practice medicine that reflects contemporary science. In the field of Multiple Endocrine Neoplasias (MENs), this is at least in part accomplished by biennially held International Workshops. The articles led by this editorial are the second and last installment of a collection of state-of-the-art presentations given in the course of the ninth such workshop that was held in Bethesda, MD, June 19-22, 2004. In addition to serving as an introduction to the articles that were written by some of the leaders in the field, the text of the editorial review that follows also supports the notion that MENs are poised to lead translational research in endocrinology: these disorders have benefited remarkably from the discoveries of the human genome project and are at a unique position to take advantage of new modalities in basic and clinical science.


Asunto(s)
Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Biosíntesis de Proteínas , Transducción de Señal , Animales , Cromograninas , Cromosomas Humanos Par 11/genética , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico , Educación , Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasia Endocrina Múltiple Tipo 1/genética , Biosíntesis de Proteínas/genética , Proteínas/genética , Proteínas/metabolismo , Transducción de Señal/genética
17.
Protein Expr Purif ; 41(1): 27-37, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15802218

RESUMEN

In Escherichia coli and other cell-based expression systems, there are critical difficulties in synthesizing membrane proteins, such as the low protein expression levels and the formation of insoluble aggregates. However, structure determinations by X-ray crystallography require the purification of milligram quantities of membrane proteins. In this study, we tried to solve these problems by using cell-free protein expression with an E. coli S30 extract, with G protein coupled receptors (GPCRs) as the target integral membrane proteins. In this system, the thioredoxin-fusion vector induced high protein expression levels as compared with the non-fusion and hexa-histidine-tagged proteins. Two detergents, Brij35 and digitonin, effectively solubilized the produced GPCRs, with little or no effect on the protein yields. The synthesized proteins were detected by Coomassie brilliant blue staining within 1h of reaction initiation, and were easily reconstituted within phospholipid vesicles. Surprisingly, the unpurified, reconstituted thioredoxin-fused receptor proteins had functional activity, in that a specific affinity binding value of an antagonist was obtained for the receptor. This cell-free translation system (about 1mg/ml of reaction volume for 6-8 h) has biophysical and biochemical advantages for the synthesis of integral membrane proteins.


Asunto(s)
Receptores Acoplados a Proteínas G/biosíntesis , Receptores Acoplados a Proteínas G/genética , Animales , Sistema Libre de Células , ADN/genética , Detergentes , Digitonina , Escherichia coli/genética , Escherichia coli/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/aislamiento & purificación , Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/aislamiento & purificación , Vectores Genéticos , Humanos , Técnicas In Vitro , Cinética , Fusión de Membrana , Polidocanol , Polietilenglicoles , Biosíntesis de Proteínas , Ratas , Receptor Muscarínico M2/biosíntesis , Receptor Muscarínico M2/genética , Receptor Muscarínico M2/aislamiento & purificación , Receptores Adrenérgicos beta 2/biosíntesis , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/aislamiento & purificación , Receptores Acoplados a Proteínas G/aislamiento & purificación , Receptores de Neurotensina/biosíntesis , Receptores de Neurotensina/genética , Receptores de Neurotensina/aislamiento & purificación , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Solubilidad , Tiorredoxinas/biosíntesis , Tiorredoxinas/genética , Tiorredoxinas/aislamiento & purificación
18.
Biol Pharm Bull ; 27(7): 1130-2, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15256754

RESUMEN

We investigated the gene expression of beta(1)-adrenergic receptor (beta(1)AR) and stimulatory G-protein Gsalpha, important signal transduction elements for regulating heart rate and contractility, in ventricle after chronic treatment with isoproterenol (ISO) in rat. Rats were treated with ISO (4 mg/kg, intraperitoneal) twice a day for 4 d. Ventricle weight of the heart and ventricle weight/body weight ratio were increased by 23% and 25% compared with control, respectively. Positive inotropic responses to ISO in left atrial muscle preparations isolated from ISO-treated rats were markedly decreased. Northern blot hybridization showed that the mRNA transcript of beta(1)AR was significantly decreased in ventricle of ISO-treated rats, whereas Gsalpha mRNA level was unchanged. Present results demonstrate that the gene expression of myocardial beta(1)AR, but not Gsalpha, was decreased in rat myocardium of ISO-induced cardiac hypertrophy, and suggesting that decrease in the gene expression of beta(1)AR may be one of the mechanisms responsible for the diminished cardiac function.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Isoproterenol/farmacología , Miocardio/metabolismo , Receptores Adrenérgicos beta 1/biosíntesis , Receptores Adrenérgicos beta 1/genética , Agonistas de Receptores Adrenérgicos beta 1 , Animales , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Masculino , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/fisiología , Ratas , Ratas Wistar
19.
Neuropsychopharmacology ; 29(3): 494-501, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14694347

RESUMEN

Schizophrenia is a complex disorder characterized by wide-ranging cognitive impairments, including deficits in learning as well as sensory gating. The causes of schizophrenia are unknown, but alterations in intracellular G-protein signaling pathways are among the molecular changes documented in patients with schizophrenia. Using the CaMKIIalpha promoter to drive expression in neurons within the forebrain, we have developed transgenic mice that express a constitutively active form of G(s)alpha (G(s)alpha(*)), the G protein that couples receptors such as the D(1) and D(5) dopamine receptors to adenylyl cyclase. We have also generated mice in which the CaMKIIalpha promoter drives expression of a dominant-negative form of protein kinase A, R(AB). Here, we examine startle responses and prepulse inhibition of the startle reflex (PPI) in these G(s)alpha(*) and R(AB) transgenic mice. G(s)alpha(*) transgenic mice exhibited selective deficits in PPI, without exhibiting alterations in the startle response, whereas no deficit in startle or PPI was found in the R(AB) transgenic mice. Thus, overstimulation of the cAMP/PKA pathway disrupts PPI, but the cAMP/PKA pathway may not be essential for sensorimotor gating. G(s)alpha(*) transgenic mice may provide an animal model of certain endophenotypes of schizophrenia, because of the similarities between them and patients with schizophrenia in G-protein function, hippocampus-dependent learning, and sensorimotor gating.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Desempeño Psicomotor/fisiología , Reflejo de Sobresalto/fisiología , Estimulación Acústica/métodos , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/biosíntesis , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Esquizofrenia/genética , Esquizofrenia/metabolismo
20.
Methods Find Exp Clin Pharmacol ; 25(9): 723-6, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14685300

RESUMEN

We investigated the influence of chronic beta-adrenergic stimulation on Gsalpha protein gene expression and beta-adrenoceptor responsiveness in rat ventricular myocardium. The rats received twice-daily injections of 4 mg/kg isoproterenol (ISO) alone or with 8 mg/kg propranolol (PROP) for 4 days. In ventricular myocardium, Gsalpha mRNA expression decreased by 27% after ISO treatment. Dose-dependent (10 nM to 100 micro M) positive inotropic responses by ISO in the left papillary muscles were lower after ISO treatment than in saline-treated myocardium with decreases in ED50 values. PROP itself had no effect, although it antagonized both ISO-induced effects. These results suggest that impaired Galpha mRNA expression may explain the loss of cardiac Gsalpha subunit levels after chronic beta-adrenergic stimulation, and that these changes can provide one mechanism for the progress of long-term desensitization.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Isoproterenol/administración & dosificación , Miocardio/metabolismo , ARN Mensajero/biosíntesis , Animales , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Corazón/efectos de los fármacos , Masculino , Tamaño de los Órganos/efectos de los fármacos , ARN Mensajero/genética , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...