Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Gene ; 715: 144028, 2019 Oct 05.
Article in English | MEDLINE | ID: mdl-31374326

ABSTRACT

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.


Subject(s)
Chromogranins/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/biosynthesis , Gene Expression Regulation , Insulin Secretion , Insulin-Secreting Cells/metabolism , Aged , Animals , Cell Line , Chromogranins/genetics , Cyclic AMP/genetics , Cyclic AMP/metabolism , Female , GTP-Binding Protein alpha Subunits, Gs/genetics , Humans , Insulin-Secreting Cells/cytology , Male , Middle Aged , Organ Specificity , Peptide Elongation Factor 1/genetics , Peptide Elongation Factor 1/metabolism , Rats
2.
Nat Commun ; 8: 15161, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28443644

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Chromogranins/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Myelin Sheath/metabolism , Phosphoproteins/metabolism , SOXE Transcription Factors/metabolism , Schwann Cells/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins , Cell Differentiation , Cell Line , Cell Proliferation , Chromogranins/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/biosynthesis , Gene Expression Regulation/genetics , HEK293 Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/genetics , Rats , Repressor Proteins/metabolism , Trans-Activators , Transcription Factor HES-1/metabolism , YAP-Signaling Proteins
3.
PLoS One ; 10(2): e0117378, 2015.
Article in English | MEDLINE | ID: mdl-25659103

ABSTRACT

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.


Subject(s)
DNA Methylation/physiology , DNA, Intergenic/metabolism , GTP-Binding Protein alpha Subunits, Gs/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/metabolism , Multigene Family/physiology , Transcription, Genetic/physiology , Alleles , Animals , Chromogranins , CpG Islands/physiology , DNA, Intergenic/genetics , Female , GTP-Binding Protein alpha Subunits, Gs/genetics , Male , Mice , Mice, Mutant Strains
4.
J Bone Miner Res ; 29(11): 2357-68, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24764158

ABSTRACT

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.


Subject(s)
Disease Models, Animal , Fibrous Dysplasia of Bone , GTP-Binding Protein alpha Subunits, Gs , Gene Expression , Mutation, Missense , Age Factors , Amino Acid Substitution , Animals , Bone Remodeling/genetics , Embryo, Mammalian/enzymology , Embryo, Mammalian/pathology , Fibrous Dysplasia of Bone/enzymology , Fibrous Dysplasia of Bone/genetics , Fibrous Dysplasia of Bone/pathology , GTP-Binding Protein alpha Subunits, Gs/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/genetics , Humans , Mice , Mice, Transgenic , Osteogenesis/genetics
5.
Horm Res Paediatr ; 80(4): 229-41, 2013.
Article in English | MEDLINE | ID: mdl-24107509

ABSTRACT

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.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs , Gene Expression Regulation , Genetic Diseases, Inborn , Genetic Loci , Genomic Imprinting , Mutation , Chromogranins , GTP-Binding Protein alpha Subunits, Gs/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/genetics , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Humans , Organ Specificity/genetics
6.
Genet Mol Res ; 12(3): 3479-87, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-24065686

ABSTRACT

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.


Subject(s)
Esthesioneuroblastoma, Olfactory/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Nose Neoplasms/genetics , Receptors, Odorant/genetics , Esthesioneuroblastoma, Olfactory/pathology , GTP-Binding Protein alpha Subunits, Gs/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , Nasal Cavity/pathology , Nose Neoplasms/pathology , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/metabolism , Receptors, Odorant/biosynthesis , Smell/genetics
7.
Pharmacol Rep ; 64(2): 256-65, 2012.
Article in English | MEDLINE | ID: mdl-22661174

ABSTRACT

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.


Subject(s)
Hippocampus/drug effects , Phenols/pharmacology , Receptors, Serotonin/metabolism , Serotonin Antagonists/pharmacology , Sulfonamides/pharmacology , Animals , Antidepressive Agents, Tricyclic/pharmacology , Binding Sites , Cell Membrane/drug effects , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Electrophysiological Phenomena , GTP-Binding Protein alpha Subunits, G12-G13/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/biosynthesis , Hippocampus/metabolism , Imipramine/pharmacology , Male , Phenols/administration & dosage , Protein Binding , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Serotonin/analogs & derivatives , Serotonin/pharmacology , Serotonin Antagonists/administration & dosage , Sulfonamides/administration & dosage
8.
Am J Physiol Heart Circ Physiol ; 302(8): H1591-602, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22268112

ABSTRACT

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.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/biosynthesis , Hydrogen Peroxide/pharmacology , Myocytes, Smooth Muscle/physiology , Oxidants/pharmacology , Receptors, Platelet-Derived Growth Factor/physiology , Adenylyl Cyclases/metabolism , Animals , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Cyclic AMP/biosynthesis , Cyclic AMP/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gs/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/genetics , Mitogen-Activated Protein Kinases/physiology , Oncogene Protein v-akt/biosynthesis , Oncogene Protein v-akt/genetics , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , RNA, Small Interfering/pharmacology , Rats , Receptors, Growth Factor/antagonists & inhibitors , Signal Transduction/drug effects , Transcriptional Activation/physiology
9.
Pflugers Arch ; 462(4): 529-43, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21811789

ABSTRACT

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.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Myocardium/metabolism , Receptors, Adrenergic, beta/drug effects , Signal Transduction/drug effects , Systole/drug effects , Animals , Calcium Channels, L-Type/metabolism , Colforsin/pharmacology , Cyclic AMP/metabolism , Down-Regulation/drug effects , G-Protein-Coupled Receptor Kinase 2/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/biosynthesis , Guinea Pigs , Isoproterenol/pharmacology , KCNQ1 Potassium Channel/metabolism , Male , Myocardial Contraction/drug effects , Receptors, Adrenergic, beta/physiology , Refractory Period, Electrophysiological , Ventricular Remodeling/drug effects
10.
J Biol Chem ; 286(4): 2625-35, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21115479

ABSTRACT

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).


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/isolation & purification , GTP-Binding Protein alpha Subunits/isolation & purification , Glutathione Transferase/isolation & purification , Recombinant Fusion Proteins/isolation & purification , Adenylyl Cyclases/chemistry , Adenylyl Cyclases/metabolism , Animals , Baculoviridae/genetics , Enzyme Activation , GTP-Binding Protein alpha Subunits/biosynthesis , GTP-Binding Protein alpha Subunits/chemistry , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits, Gs/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/chemistry , GTP-Binding Protein alpha Subunits, Gs/genetics , Gene Expression , Glutathione Transferase/biosynthesis , Glutathione Transferase/chemistry , Glutathione Transferase/genetics , HEK293 Cells , Humans , Isoenzymes/chemistry , Isoenzymes/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Spodoptera
11.
Gynecol Oncol ; 118(2): 160-6, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20537689

ABSTRACT

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.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/genetics , Ovarian Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Carboplatin/administration & dosage , Chemotherapy, Adjuvant , Chromogranins , Disease-Free Survival , Epithelial Cells/pathology , Female , GTP-Binding Protein alpha Subunits, Gs/biosynthesis , Gene Amplification , Humans , Middle Aged , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/surgery , Paclitaxel/administration & dosage , Predictive Value of Tests , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction
12.
Proc Natl Acad Sci U S A ; 105(4): 1209-14, 2008 Jan 29.
Article in English | MEDLINE | ID: mdl-18212126

ABSTRACT

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.


Subject(s)
Bone Density/physiology , GTP-Binding Protein alpha Subunits, Gs/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/genetics , Osteoblasts/chemistry , Osteoblasts/metabolism , Protein Engineering , Receptors, Serotonin, 5-HT4/biosynthesis , Receptors, Serotonin, 5-HT4/genetics , Amino Acid Sequence , Animals , Cell Line , Female , GTP-Binding Protein alpha Subunits, Gs/metabolism , GTP-Binding Protein alpha Subunits, Gs/physiology , Humans , Ligands , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Protein Engineering/methods , Serotonin 5-HT4 Receptor Agonists , Signal Transduction/genetics , Signal Transduction/physiology
13.
Am J Physiol Heart Circ Physiol ; 294(2): H775-84, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18055527

ABSTRACT

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.


Subject(s)
Adenylyl Cyclase Inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/biosynthesis , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Peroxynitrous Acid/pharmacology , Signal Transduction/physiology , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Blotting, Western , Carbazoles/pharmacology , Cells, Cultured , GTP-Binding Protein alpha Subunits, Gs/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/physiology , Indoles/pharmacology , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/physiology , Muscle Relaxation/drug effects , Muscle Relaxation/physiology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Oxadiazoles/pharmacology , Peroxynitrous Acid/antagonists & inhibitors , Quinoxalines/pharmacology , Rats , Receptor, Angiotensin, Type 1/biosynthesis , Receptors, Atrial Natriuretic Factor/biosynthesis , Signal Transduction/drug effects
14.
Auton Neurosci ; 132(1-2): 52-62, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17185044

ABSTRACT

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.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/biosynthesis , Ganglia, Sympathetic/metabolism , Superior Cervical Ganglion/metabolism , Animals , Chromogranins , Female , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Neurons/metabolism , Rats , Sex Factors , Species Specificity
15.
Anticancer Res ; 25(3B): 2075-83, 2005.
Article in English | MEDLINE | ID: mdl-16158948

ABSTRACT

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.


Subject(s)
Calcitonin/pharmacology , Hyaluronan Receptors/biosynthesis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Adenylyl Cyclases/metabolism , Animals , Calcitonin/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunits, Gs/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/metabolism , Guanylyl Imidodiphosphate/pharmacology , Humans , Hyaluronan Receptors/genetics , In Situ Hybridization , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Prostatic Neoplasms/genetics , Protein Isoforms , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
16.
Horm Metab Res ; 37(6): 343-6, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16001325

ABSTRACT

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.


Subject(s)
Multiple Endocrine Neoplasia Type 1/metabolism , Protein Biosynthesis , Signal Transduction , Animals , Chromogranins , Chromosomes, Human, Pair 11/genetics , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit , Cyclic AMP-Dependent Protein Kinases , Education , GTP-Binding Protein alpha Subunits, Gs/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Multiple Endocrine Neoplasia Type 1/genetics , Protein Biosynthesis/genetics , Proteins/genetics , Proteins/metabolism , Signal Transduction/genetics
17.
Protein Expr Purif ; 41(1): 27-37, 2005 May.
Article in English | MEDLINE | ID: mdl-15802218

ABSTRACT

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.


Subject(s)
Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Animals , Cell-Free System , DNA/genetics , Detergents , Digitonin , Escherichia coli/genetics , Escherichia coli/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/biosynthesis , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/isolation & purification , GTP-Binding Protein alpha Subunits, Gs/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/genetics , GTP-Binding Protein alpha Subunits, Gs/isolation & purification , Genetic Vectors , Humans , In Vitro Techniques , Kinetics , Membrane Fusion , Polidocanol , Polyethylene Glycols , Protein Biosynthesis , Rats , Receptor, Muscarinic M2/biosynthesis , Receptor, Muscarinic M2/genetics , Receptor, Muscarinic M2/isolation & purification , Receptors, Adrenergic, beta-2/biosynthesis , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/isolation & purification , Receptors, G-Protein-Coupled/isolation & purification , Receptors, Neurotensin/biosynthesis , Receptors, Neurotensin/genetics , Receptors, Neurotensin/isolation & purification , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Solubility , Thioredoxins/biosynthesis , Thioredoxins/genetics , Thioredoxins/isolation & purification
18.
Biol Pharm Bull ; 27(7): 1130-2, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15256754

ABSTRACT

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.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/genetics , Isoproterenol/pharmacology , Myocardium/metabolism , Receptors, Adrenergic, beta-1/biosynthesis , Receptors, Adrenergic, beta-1/genetics , Adrenergic beta-1 Receptor Agonists , Animals , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Male , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Rats , Rats, Wistar
19.
Neuropsychopharmacology ; 29(3): 494-501, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14694347

ABSTRACT

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.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/biosynthesis , Psychomotor Performance/physiology , Reflex, Startle/physiology , Acoustic Stimulation/methods , Animals , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Cyclic AMP-Dependent Protein Kinases/genetics , Female , GTP-Binding Protein alpha Subunits, Gs/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Schizophrenia/genetics , Schizophrenia/metabolism
20.
Methods Find Exp Clin Pharmacol ; 25(9): 723-6, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14685300

ABSTRACT

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.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/genetics , Isoproterenol/administration & dosage , Myocardium/metabolism , RNA, Messenger/biosynthesis , Animals , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Heart/drug effects , Male , Organ Size/drug effects , RNA, Messenger/genetics , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...