Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Virology ; 536: 101-109, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31415943

RESUMO

Bovine viral diarrhea viruses (BVDV), segregated in BVDV-1 and BVDV-2 species, lead to substantial economic losses to the cattle industry worldwide. It has been hypothesized that there could be differences in level of replication, pathogenesis and tissue tropism between BVDV-1 and BVDV-2 strains. Thus, this study developed an in vitro method to evaluate virus competition between BVDV-1 and BVDV-2 strains. To this end the competitive dynamics of BVDV-1a, BVDV-1b, and BVDV-2a strains in cell cultures was evaluated by a PrimeFlow RNA assay. Similar results were observed in this study, as was observed in an earlier in vivo transmission study. Competitive exclusion was observed as the BVDV-2a strains dominated and excluded the BVDV-1a and BVDV-1b strains. The in vitro model developed can be used to identify viral variations that result in differences in frequency of subgenotypes detected in the field, vaccine failure, pathogenesis, and strain dependent variation in immune responses.


Assuntos
Bioensaio , Vírus da Diarreia Viral Bovina Tipo 1/genética , Vírus da Diarreia Viral Bovina Tipo 2/genética , Células Epiteliais/virologia , RNA Viral/genética , Animais , Doença das Mucosas por Vírus da Diarreia Viral Bovina/diagnóstico , Doença das Mucosas por Vírus da Diarreia Viral Bovina/virologia , Bovinos , Linhagem Celular , Coinfecção , Vírus da Diarreia Viral Bovina Tipo 1/classificação , Vírus da Diarreia Viral Bovina Tipo 1/isolamento & purificação , Vírus da Diarreia Viral Bovina Tipo 1/metabolismo , Vírus da Diarreia Viral Bovina Tipo 2/classificação , Vírus da Diarreia Viral Bovina Tipo 2/isolamento & purificação , Vírus da Diarreia Viral Bovina Tipo 2/metabolismo , Cães , Células Epiteliais/patologia , Feminino , Células Madin Darby de Rim Canino , Gravidez , RNA/genética , RNA/metabolismo , Sondas RNA/genética , Sondas RNA/metabolismo , RNA Viral/metabolismo , Tropismo Viral , Replicação Viral
2.
Virology ; 492: 225-31, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26971244

RESUMO

Like other members from the Pestivirus genus, 'HoBi'-like pestiviruses cause economic losses for cattle producers due to both acute and persistent infections. The present study analyzed for the first time PI animals derived from a controlled infection with two different 'HoBi'-like strains where the animals were maintained under conditions where superinfection by other pestiviruses could be excluded. The sequence of the region coding for viral glycoproteins E1/E2 of variants within the swarms of viruses present in the PI calves and two viral inoculums used to generate them were compared. Differences in genetic composition of the viral swarms were observed suggesting that host factors can play a role in genetic variations among PIs. Moreover, PIs generated with the same inoculum showed amino acid substitutions in similar sites of the polyprotein, even in serum from PIs with different quasispecies composition, reinforcing that some specific sites in E2 are important for host adaptation.


Assuntos
Infecções por Pestivirus/virologia , Pestivirus/genética , Filogenia , Proteínas do Envelope Viral/genética , Adaptação Fisiológica , Animais , Bovinos , Clonagem Molecular , Células Epiteliais/patologia , Células Epiteliais/virologia , Expressão Gênica , Pestivirus/classificação , Pestivirus/isolamento & purificação , Cultura Primária de Células , Proteínas Recombinantes/genética , Análise de Sequência de DNA , Conchas Nasais/patologia , Conchas Nasais/virologia
3.
Virus Res ; 126(1-2): 96-105, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17376555

RESUMO

Bovine viral diarrhea virus (BVDV), a pestivirus of the Flaviviridae family, is an economically important cattle pathogen with a worldwide distribution. Besides the segregation into two distinct species (BVDV1/BVDV2) two different biotypes, a cytopathic (cp) and a noncytopathic (ncp) biotype, are distinguished based on their behavior in epithelial cell cultures. One of the most serious forms of BVDV infection affecting immunocompetent animals of all ages is severe acute BVD (sa BVD) which is caused by highly virulent ncp BVDV2 strains. Previous studies revealed that these highly virulent ncp viruses cause cell death in a lymphoid cell line (BL3) which is not clearly associated with typical apoptotic changes (e.g. PARP cleavage) observed after infection with cp BVDV. To further characterize the underlying molecular mechanisms, we first analyzed the role of the mitochondria and caspases as key mediators of apoptosis. Compared to infection with cp BVDV2, infection with highly virulent ncp BVDV2 resulted in a delayed and less pronounced disruption of the mitochondrial transmembrane potential (DeltaPsi(m)) and a weaker activation of the caspase cascade. In contrast, infection with low virulence ncp BVDV2 showed no significant differences from the uninfected control cells. Since different pro- and anti-apoptotic cellular signaling pathways may become activated upon virus infection, we compared the effect of different BVDV2 strains on cellular signaling pathways in BL3 cells. Stress-mediated p38 MAPK phosphorylation was detected only in cells infected with cp BVDV2. Interestingly, infection with highly virulent ncp BVDV2 was found to influence the phosphoinositide 3-kinase (PI3K)-Akt pathway. This indicates that BL3 cells respond differently to infection with BVDV depending on virulence and biotype.


Assuntos
Vírus da Diarreia Viral Bovina Tipo 2/patogenicidade , Animais , Apoptose , Doença das Mucosas por Vírus da Diarreia Viral Bovina/patologia , Doença das Mucosas por Vírus da Diarreia Viral Bovina/fisiopatologia , Doença das Mucosas por Vírus da Diarreia Viral Bovina/virologia , Caspases/metabolismo , Bovinos , Linhagem Celular , Efeito Citopatogênico Viral , Vírus da Diarreia Viral Bovina Tipo 2/classificação , Ativação Enzimática , Potencial da Membrana Mitocondrial , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Virulência
4.
Arch Physiol Biochem ; 110(1-2): 129-36, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11935410

RESUMO

Mammalian gonadotropin-releasing hormone (GnRH I) is a hypothalamic decapeptide that governs gonadotropin secretion through interaction with its seven transmembrane (7TM), G protein-coupled receptor (GPCR) expressed by anterior pituitary cells. A second decapeptide, GnRH II, originally discovered in the chicken hypothalamus was recently reported to be expressed in the mammalian hypothalamus as well. A search of the recently-sequenced human genome identified a 7TM/GPCR on chromosome 1 that exhibited a higher identity with non-mammalian vertebrate GnRH II receptors (55%) than with the human GnRH I receptor (39%). Molecular cloning and nucleotide sequencing of this putative GnRH II receptor cDNA from monkey pituitary gland revealed a 379 amino acid receptor that, unlike the GnRH I receptor, possessed a C-terminal tail. Heterologous expression and functional testing of the receptor in COS-1 cells confirmed its identity as a GnRH II receptor: measurement of 3H-inositol phosphate accumulation revealed EC(50)s for GnRH II of 0.86 nM and for GnRH I of 337 nM. Ubiquitous tissue expression of GnRH II receptor mRNA was observed using a human tissue RNA expression array and a 32P-labeled antisense riboprobe representing the 7TM region of human GnRH II receptor cDNA. As predicted by the presence of its C-terminal tail, the GnRH II receptor was desensitized by GnRH II treatment whereas the naturally tail-less GnRH I receptor was not desensitized by GnRH I. Pharmacological analysis of the GnRH II receptor revealed that GnRH I 'superagonists' were more potent than GnRH I but less potent than GnRH II. Numerous GnRH I antagonists showed neither antagonistic nor agonistic activity with the GnRH II receptor. The functions of the GnRH II receptor are unknown but may include regulation of gonadotropin secretion, female sexual behavior, or tumor cell growth.


Assuntos
Receptores LHRH/classificação , Animais , Haplorrinos , Humanos , Especificidade de Órgãos , RNA Mensageiro/metabolismo , Receptores LHRH/antagonistas & inibidores , Receptores LHRH/química , Receptores LHRH/genética , Análise de Sequência de DNA
6.
BMC Cell Biol ; 2: 21, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11716781

RESUMO

BACKGROUND: Luteinizing hormone secreted by the anterior pituitary gland regulates gonadal function. Luteinizing hormone secretion is regulated both by alterations in gonadotrope responsiveness to hypothalamic gonadotropin releasing hormone and by alterations in gonadotropin releasing hormone secretion. The mechanisms that determine gonadotrope responsiveness are unknown but may involve regulators of G protein signaling (RGSs). These proteins act by antagonizing or abbreviating interaction of Galpha proteins with effectors such as phospholipase Cbeta. Previously, we reported that gonadotropin releasing hormone-stimulated second messenger inositol trisphosphate production was inhibited when RGS3 and gonadotropin releasing hormone receptor cDNAs were co-transfected into the COS cell line. Here, we present evidence for RGS3 inhibition of gonadotropin releasing hormone-induced luteinizing hormone secretion from cultured rat pituitary cells. RESULTS: A truncated version of RGS3 (RGS3T = RGS3 314-519) inhibited gonadotropin releasing hormone-stimulated inositol trisphosphate production more potently than did RSG3 in gonadotropin releasing hormone receptor-bearing COS cells. An RSG3/glutathione-S-transferase fusion protein bound more 35S-Gqalpha than any other member of the G protein family tested. Adenoviral-mediated RGS3 gene transfer in pituitary gonadotropes inhibited gonadotropin releasing hormone-stimulated luteinizing hormone secretion in a dose-related fashion. Adeno-RGS3 also inhibited gonadotropin releasing hormone stimulated 3H-inositol phosphate accumulation, consistent with a molecular site of action at the Gqalpha protein. CONCLUSIONS: RGS3 inhibits gonadotropin releasing hormone-stimulated second messenger production (inositol trisphosphate) as well as luteinizing hormone secretion from rat pituitary gonadotropes apparently by binding and suppressing the transduction properties of Gqalpha protein function. A version of RGS3 that is amino-terminally truncated is even more potent than intact RGS3 at inhibiting gonadotropin releasing hormone-stimulated inositol trisphosphate production.


Assuntos
Proteínas de Ligação ao GTP , Proteínas Ativadoras de GTPase , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Hormônio Luteinizante/metabolismo , Adeno-Hipófise/metabolismo , Proteínas RGS/fisiologia , Proteínas Repressoras , Animais , Células COS , Sinalização do Cálcio , Células Cultivadas , Feminino , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Inositol 1,4,5-Trifosfato/metabolismo , Proteínas RGS/genética , Ratos , Receptores LHRH/metabolismo , Deleção de Sequência
7.
Biochem Biophys Res Commun ; 282(4): 1012-8, 2001 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-11352653

RESUMO

Mammalian gonadotropin-releasing hormone (GnRH I) is a hypothalamic decapeptide that stimulates gonadotropic hormone secretion upon interaction with its membrane receptors (type I) on pituitary cells, thereby governing reproductive processes. A second releasing hormone (GnRH II) expressed in mammals was shown earlier to be expressed in nonmammals and to have its own receptor. Here we demonstrate that a second receptor (type II) gene is present in the human genome, and report the cloning and characterization of its cDNA from monkeys. The cDNA encodes a G-protein-coupled/7 transmembrane receptor having a C-terminal cytoplasmic tail; it resembles more closely the type II receptors of amphibians and fish (approximately 55% identity) than it does the type I receptor of humans (approximately 39%). The GnRH type II receptor proved to be experimentally functional with, and specific for, GnRH II. GnRH receptor type II RNA is expressed ubiquitously in human tissues. This is the first report of a GnRH type II receptor in mammals. Its identification will permit exploration of its role in regulating gonadotropin secretion, female sexual behavior, and tumor cell growth.


Assuntos
Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/metabolismo , Receptores LHRH/genética , Sequência de Aminoácidos , Animais , Células COS , Clonagem Molecular , Humanos , Macaca mulatta , Dados de Sequência Molecular , RNA Mensageiro/biossíntese , Receptores LHRH/biossíntese , Receptores LHRH/fisiologia , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , Transfecção
8.
Life Sci ; 64(24): 2215-23, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10374911

RESUMO

The mechanisms of GnRH-induced desensitization of LH secretion are poorly understood. Protein kinase C (PKC) and protein kinase A (PKA) desensitize some receptors of the 7-membrane type, and the GnRH receptor has consensus phosphorylation sites for PKC in the first and third intracellular loops, and a site for PKA in the first intracellular loop. In the first set of experiments we determined whether synthetic peptides representing the three intracellular loops of the receptor could be phosphorylated in vitro by purified PKC and PKA. As compared with a model substrate peptide for PKC, the third intracellular loop was phosphorylated 74% and the first intracellular loop 21%; PKA-phosphorylated the first intracellular loop peptide 17% as well as a model peptide substrate. In the second set of experiments, we used phorbol 12-myristate 13 acetate (PMA), an established PKC stimulator, and cholera toxin (CTX), established to activate the Gs protein and presumed to activate PKA, to treat cultured rat pituitary cells followed by LH measurements. Treatment with both drugs severely impaired GnRH-stimulated LH secretion whereas neither drug alone reduced LH secretion. Dibutyryl cAMP did not duplicate the effects of cholera toxin suggesting that the CTX action could not be explained by an increase in cAMP. These results suggest that more than one intracellular signaling pathway requires activation in order to induce desensitization; one pathway involves PKC and the other involves a pathway stimulated by cholera toxin, presumably Gs protein, which does not involve PKA.


Assuntos
Hormônio Liberador de Gonadotropina/farmacologia , Hormônio Luteinizante/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Bucladesina/farmacologia , Células Cultivadas , Toxina da Cólera/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ativação Enzimática/efeitos dos fármacos , Proteínas de Ligação ao GTP/metabolismo , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fosforilação , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/metabolismo , Proteína Quinase C/metabolismo , Ratos , Receptores LHRH/química , Receptores LHRH/genética , Receptores LHRH/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
9.
Cell Mol Life Sci ; 55(4): 653-62, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10357233

RESUMO

The molecular recognition hypothesis for peptides is that binding sites of ligands and their receptors are encoded by short, complementary segments of DNA. A corollary hypothesis for nonpeptide ligands posited here is that peptide replicas may be encoded by the DNA segment complementary to the receptor binding sites for nonpeptides. This corollary was tested for digitalis. a family of cardiotonic and natriuretic steroids including ouabain. A hexapeptide (ouabain-like peptide, OLP) complementary to a ouabain binding site on sodium potassium dependent adenosine triphosphatase (Na+ K+ ATPase) exhibited activity in a digitalis bioassay. Antisera to the complementary peptide (OLP) stained the neurohypophysis in an immunocytochemical procedure. The complementary peptide was found to share an identical 4-amino acid region with the 39-amino acid glycopeptide moiety of the vasopressin-neurophysin precursor. This glycopeptide was isolated from pituitary extracts; it exhibited digitalis-like activity in the submicromolar range and cross-reacted with complementary peptide antibodies. Another digitalis-like substance with high activity also was detected in the extracts. These results demonstrate that the vasopressin-neurophysin glycopeptide has digitalis-like activity. Moreover, the findings are consistent with the hypothesis that peptide mimetics of nonpeptides are encoded in the genome.


Assuntos
Inibidores Enzimáticos/metabolismo , Ouabaína/metabolismo , Peptídeos/metabolismo , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Sítios de Ligação , Glicosídeos Digitálicos/isolamento & purificação , Dados de Sequência Molecular , Neurofisinas/metabolismo , Neuro-Hipófise/metabolismo , Neuro-Hipófise/patologia , Precursores de Proteínas/metabolismo , Coloração e Rotulagem/métodos , Suínos , Vasopressinas/metabolismo
10.
Endocrinology ; 140(6): 2562-9, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10342843

RESUMO

The level of LH secretion is determined by both alterations in gonadotrope responsiveness and alterations in GnRH secretion. The molecular mechanisms underlying gonadotrope responsiveness are unknown, but may include G protein-coupled receptor kinases (GRKs). Typically, GRKs phosphorylate the intracellular regions of seven-transmembrane receptors permitting beta-arrestin to bind, which prevents receptor activation of its G protein. Previously, we reported that heterologous expression of GRK2, -3, and -6 in GnRH receptor-expressing COS cells by complementary DNA transfection suppressed GnRH-stimulated inositol trisphosphate production, and that coexpression of GRK2 and beta-arrestin-2 was more inhibitory than either expressed alone. Here, we have investigated the effect of GRK2 on GnRH-stimulated LH secretion using adenovirus-mediated gene transfer in normal pituitary gonadotropes. Pituitary cells were infected with adeno-GRK2 or adeno-beta-galactosidase constructs at a multiplicity of infection of 60 (number of viral particles per cell). Seventy-two hours later, GRK2 expression was measured by enzyme-linked immunosorbent assay, and GnRH-stimulated LH secretion (10(-7) M GnRH-A for 90 min) was assayed by RIA. Adeno-beta-galactosidase infected 96-99% of the cells based on X-Gal staining. Uninfected and adeno-beta-galactosidase-infected cells exhibited endogenous GRK immunoreactivity of about 0.5 (OD405), and LH secretion of 14.8-17.7 ng/ml. Adeno-GRK2-infected cells showed a GRK2 immunoreactivity of about 2.5 (OD405) and LH secretion of 2.5 ng/ml. Therefore, adeno-GRK2 infection resulted in a 5-fold increase in the GRK2 OD405 value, which was accompanied by an 80-85% decrease in GnRH-stimulated LH secretion. GnRH-stimulated inositol trisphosphate production by gonadotropes also was inhibited, suggesting a site of action for GRK2 at phospholipase Cbeta or earlier in the signal transduction pathway. The significance of these findings is 2-fold: 1) adenoviral-mediated gene transfer permits investigation of the regulatory role of gene products in the cell of interest, the gonadotrope, rather than in heterologous cell systems; and 2) additional, stronger evidence is provided that supports a role for GRKs in setting the responsiveness of GnRH receptor signaling.


Assuntos
Adenoviridae/genética , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Técnicas de Transferência de Genes , Hormônio Luteinizante/metabolismo , Hipófise/metabolismo , Animais , Quinase 2 de Receptor Acoplado a Proteína G , Hormônio Liberador de Gonadotropina/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores LHRH/metabolismo , Quinases de Receptores Adrenérgicos beta
11.
Neuroendocrinology ; 67(4): 244-59, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9588694

RESUMO

We have investigated exocytotic proteins in ovine pituitary cells and sought to identify changes in expression of these proteins related to the effects of estrogen on luteinising hormone (LH) secretion in the ovariectomised ewe. Sheep were treated with either oestradiol benzoate, or oil (i.m.) and blood samples collected for LH assay. Pituitaries were perfusion-fixed and dual-label immunohistochemistry was performed to identify hormone-secreting cells, and colocalise synaptic proteins within different cell types. Synaptophysin, SNAP-25, VAMP-2, rab3A, Munc-18-1, alpha/beta-SNAP, csp, and secretogranin II were detected in gonadotropes and somatotropes. Lactotropes were positive for SNAP-25 and synaptophysin (other synaptic proteins not investigated). Synaptotagmin I was detected in gonadotropes and lactotropes, but not somatotropes. Synaptophysin, SNAP-25, synaptotagmins I, II and III, VAMP-2, rab3A, Munc-18-1, alpha/beta-SNAP, csp, and secretogranin II were detected in nerve fibres of the posterior lobe. Membrane staining for SNAP-25 and weak cytoplasmic labelling for both synaptotagmin I and secretogranin II were detected in the intermediate lobe. Syntaxin and complexin II antibodies did not label any region of the ovine pituitary. Oestrogen treatment, to induce a pre-ovulatory-like LH surge, caused migration of LH-containing secretory granules toward the plasma membrane of gonadotropes, but did not alter the percentage of gonadotropes expressing each exocytotic protein. Oestrogen treatment caused a similar redistribution of csp and secretogranin II staining in gonadotropes. We conclude that synaptic protein expression is not altered in the anterior pituitary at the time when LH secretion is maximal. The ubiquitous distribution of many exocytotic proteins suggests that all hormone-secreting cells of the pituitary gland contain the same, or similar exocytotic machinery, but distinct 'activating factors' are required to selectively trigger the secretion of individual hormones.


Assuntos
Estradiol/farmacologia , Exocitose/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Ovário/fisiologia , Adeno-Hipófise/efeitos dos fármacos , Animais , Feminino , Hormônio Foliculoestimulante/metabolismo , Imuno-Histoquímica , Hormônio Luteinizante/metabolismo , Microscopia Confocal , Proteínas do Tecido Nervoso/análise , Ovariectomia , Adeno-Hipófise/citologia , Hormônios Hipofisários/análise , Taxa Secretória/efeitos dos fármacos , Ovinos
12.
Endocrinology ; 139(4): 1781-8, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9528962

RESUMO

GnRH stimulates gonadotropin secretion, which desensitizes unless the releasing hormone is secreted or administered in a pulsatile fashion. The mechanism of desensitization is unknown, but as the GnRH receptor is G protein coupled, it might involve G protein-coupled receptor kinases (GRKs). Such kinases phosphorylate the intracellular regions of seven-transmembrane receptors, permitting beta-arrestin to bind, which prevents the receptor from activating G proteins. Here, we tested the effect of GRKs and beta-arrestins on GnRH-induced inositol trisphosphate (IP3) production in COS cells transfected with the GnRH receptor complementary DNA. GRK2, -3, and -6 overexpression inhibited IP3 production by 50-75% during the 30 sec of GnRH treatment. Coexpression of GRK2 and beta-arrestin-2 suppressed GnRH-induced IP3 production more than that of either alone. Immunocytochemical staining of rat anterior pituitary revealed that all cells expressed GRK2, -3, and -6; all cells also expressed the beta-arrestins. Western blots on cytosolic extracts of rat pituitaries revealed the presence of GRK2/3 and beta-arrestin-1 and -2. The expression of GRKs and beta-arrestins by gonadotropes and their inhibition of GnRH-stimulated IP3 production in COS-1 cells expressing the GnRH receptor suggest a potential regulatory role for the GRK/beta arrestin paradigm in GnRH receptor signaling.


Assuntos
Arrestinas/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptores LHRH/fisiologia , Transdução de Sinais , Animais , Arrestinas/análise , Células COS , Bovinos , Hormônio Liberador de Gonadotropina/farmacologia , Imuno-Histoquímica , Fosfatos de Inositol/biossíntese , Cinética , Fosforilação , Adeno-Hipófise/química , Ratos , Receptores Proteína Tirosina Quinases/análise , Transfecção , beta-Arrestina 1 , beta-Arrestina 2 , beta-Arrestinas
13.
Mol Cell Endocrinol ; 132(1-2): 53-9, 1997 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-9324046

RESUMO

The classical mode of luteinizing hormone (LH) secretory desensitization in the rat appears after 3-6 h of continuous in vitro administration of gonadotropin (GnRH). A second mode has been reported to occur very rapidly (< 2 min) after the onset of GnRH administration, and to reverse within 3 min after its withdrawal. Here, the existence of a third mode of desensitization is reported. occurring at 40-50 min after initiation of continuous GnRH administration. Rat pituitary cells were perifused with 10(-8) M GnRH for 6 h: 10 min samples were collected for LH measurements by radioimmunoassay. As expected, the pattern of LH release was biphasic: LH levels peaked in the first phase at 30 min, decreased at 40-50 min, increased in the second phase to maximal levels at 90-110 min, and then decreased in the classical desensitization mode to near-baseline values by 300-360 min. Static incubations of pituitary cells in Petri dishes in the presence of high (10(-8) M) or submaximal (10(-9) M) GnRH concentrations confirmed the decrease in LH secretion at 40-50 min. Measurement of LH by reverse hemolytic plaque assay (RHPA) confirmed the existence of this new mode of desensitization; since 93% of all gonadotropes had become secretory at 40-50 min, the possibility of two subpopulations of gonadotropes accounting for the two phases of LH secretion appears to be ruled-out. GnRH receptor binding studies demonstrated a approximately 50% decrease in cell-surface binding in association with the desensitization at 40-50 min. These studies suggest the existence of a third mode of GnRH-induced LH secretory desensitization that is not due to gonadotrope subpopulations but may be causally associated with decreased GnRH receptor binding.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Luteinizante/metabolismo , Hipófise/metabolismo , Animais , Células Cultivadas , Feminino , Hormônio Liberador de Gonadotropina/farmacologia , Radioimunoensaio , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley
14.
Mol Cell Endocrinol ; 127(2): 143-54, 1997 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-9099910

RESUMO

The molecular cloning and nucleotide sequencing of the gonadotropin-releasing hormone (GnRH) receptor represented an enhanced step in the experimental effort to understand this key molecule in the reproductive process at a cell and molecular level. A subsequent step in this broad effort is heterologous expression of the receptor in model cell systems for studies of signal transduction and desensitization, processes that may require immunologic detection of the receptor. Therefore, the GnRH receptor was tagged at its N-terminus using recombinant DNA procedures with the HA-1 epitope that is bound by a monoclonal antibody (12CA5). COS-1 cells expressing this receptor bound [(125)I]D-Ala6-desGly10-GnRH ethylamide (GnRH-A) with the expected high affinity (IC(50) = 0.47 nM), and were immunocytochemically stained by the 12CA5 antibody. Signal transduction was demonstrated by GnRH-induced [(3)H]inositol phosphate accumulation in receptor-expressing COS-1 cells. Western blotting of COS-1 cell membranes expressing the receptor revealed protein bands at 67, 57, and 32 kDa. Immunoprecipitation occurred when the solubilized receptor from COS-1 cell membranes was reacted with 12CA5 antibody and anti-mouse IgG Sepharose, and the presence of the receptor demonstrated either by its binding of [(125)I]GnRH-A or by its detection on Western blots. Desensitization of inositol 1,4,5-trisphosphate (IP(3)) production by N-epitope-tagged GnRH receptor expressing COS-1 cells was evoked by a five min GnRH pretreatment; [(32)P]i labeling of such cells during desensitization followed by immunoprecipitation of the N-epitope-tagged receptor was not associated with receptor phosphorylation. Finally, the epitope tagged receptor was expressed in the high-yield baculovirus/insect Sf9 cell system: the membrane receptor bound [(125)I]GnRH-A with slightly lowered affinity (IC(50) = 1.4 nM), and in Western blots yielded protein bands of 32, 56/57, 69, and 120/140 kDa. The development and validation of these heterologous systems will permit the study of several GnRH receptor-mediated processes that are poorly understood.


Assuntos
Células COS , Epitopos , Receptores LHRH/biossíntese , Transdução de Sinais/fisiologia , Spodoptera , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Epitopos/análise , Expressão Gênica , Hormônio Liberador de Gonadotropina/metabolismo , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Inositol 1,4,5-Trifosfato/análise , Fosfatos de Inositol/metabolismo , Dados de Sequência Molecular , Peso Molecular , Fosforilação , RNA Mensageiro/análise , Ensaio Radioligante , Receptores LHRH/química , Receptores LHRH/genética , Receptores LHRH/metabolismo , Proteínas Recombinantes de Fusão , Especificidade da Espécie , Transfecção
15.
Endocrinology ; 138(2): 843-6, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9003025

RESUMO

The cellular and molecular mechanisms of gonadotrope desensitization are unknown but transduction of the GnRH signal is known to involve sequentially the GnRH receptor, Gq alpha protein, phospholipase C beta-1, inositol-1,4,5-trisphosphate (IP3), and intracellular Ca+2 release. Here, we report the results of studies of a new family of proteins known as regulators of G protein signaling (RGS) that recently have been implicated in desensitization of several ligand induced processes. Using DNA-mediated transfection, we co-expressed the GnRH receptor and RGS1,2,3, or 4 in COS-1 cells. Control cells and those expressing RGS1,2, and 4 produced five fold increases in IP3 levels during the 30 sec after treatment with GnRH. In contrast, RGS3 expression suppressed by 75% the GnRH-induced IP3 responses. RGS3 was shown to bind Gq alpha protein in a model in vitro system: recombinant RGS3-glutathione-S-transferase (GST) fusion protein bound five-fold more 35S-met labeled Gq alpha protein than did with GST alone, suggesting that the mechanism of RGS3 action is attenuation of Gq alpha protein activation of phospholipase C. RGS3 mRNA and protein were observed to be expressed endogenously in the gonadotropic alpha T3-1 cell line. These results suggest a potential role for RGS3 in modulating the LH secretory responsiveness of the pituitary gonadotrope to GnRH.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Proteínas Ativadoras de GTPase , Hormônio Liberador de Gonadotropina/farmacologia , Proteínas/fisiologia , Proteínas RGS , Proteínas Repressoras , Transdução de Sinais , Animais , Células COS , Linhagem Celular , Glutationa Transferase/metabolismo , Inositol 1,4,5-Trifosfato/biossíntese , Hormônio Luteinizante/metabolismo , Dados de Sequência Molecular , Adeno-Hipófise/química , Proteínas/genética , RNA Mensageiro/análise , Ratos , Receptores LHRH/genética , Proteínas Recombinantes de Fusão , Transfecção
16.
Endocrinology ; 137(9): 3942-7, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8756570

RESUMO

G protein-coupled receptor kinases (GRK 1-6) stimulate short-term desensitization (< 5 min) by phosphorylating G-protein coupled receptors, and also participate in receptor sequestration, which may relate to intermediate-term desensitization (30-60 min). The existence of such kinases and hence a potential role for them in gonadotrope/GnRH receptor desensitization was investigated using the PCR to identify GRKs in messenger RNA (mRNA) from the mouse alpha T3-1 gonadotrope cell line. The 150-bp complementary DNAs amplified by PCR from the kinase catalytic domain were cloned and sequenced. Seventeen of 42 clones were receptor kinases based on high nucleotide identities of 85-100% and amino acid identities of 97-100% with rat GRK2 and 3, and with human GRK6. Among the eight GRK3 clones was one differing from rat GRK3 by a single nucleotide and seven differing by six; no amino acid difference resulted from the nucleotide differences. Of the five GRK2 clones, one sequence was identical with rat GRK2, but four sequences differed by three nucleotides and one amino acid. Among four GRK6 sequences, one showed 15 nucleotide differences from human GRK6 (with no amino acid differences), and three had 16 nucleotide and one amino acid differences. For each of the three GRKs found, the most closely related isoform is assumed to be the mouse homolog of rat GRK2 and GRK3, and human GRK6, whereas the others are assumed to be previously undescribed isoforms or subtypes of GRK2, 3, and 6. Immunocytochemical staining using antibodies to GRK2, 3, and 6 confirmed their presence in alpha T3-1 cells. The function of these GRKs in alpha T3-1 cells is unknown, but they may be involved in short-term desensitization of the gonadotrope/GnRH receptor or perhaps, more likely, the sequestration of this receptor during intermediate-term desensitization.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/análise , Gonadotropinas Hipofisárias/metabolismo , Adeno-Hipófise/química , Proteínas Serina-Treonina Quinases , Receptores Proteína Tirosina Quinases/análise , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/genética , Quinase 3 de Receptor Acoplado a Proteína G , Quinases de Receptores Acoplados a Proteína G , Proteínas de Ligação ao GTP/análise , Humanos , Imuno-Histoquímica , Camundongos , Dados de Sequência Molecular , Sondas de Oligonucleotídeos/genética , Adeno-Hipófise/citologia , Reação em Cadeia da Polimerase , Ratos , Receptores Proteína Tirosina Quinases/genética , Transcrição Gênica , Quinases de Receptores Adrenérgicos beta
17.
Endocrinology ; 137(3): 1057-62, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8603574

RESUMO

The LH secretory response of gonadotropes to GnRH varies during the estrous cycle of the rat. The increased secretion of estrogens during the 24-48 h before the preovulatory surge of LH secretion and the enhanced quantities of progesterone secreted acutely during the surge elevate the responsiveness of hypophysial gonadotropes to GnRH. However, the cause of the massive decline in GnRH responsiveness that occurs during or after the surge remains unknown. In the present studies, we investigated the possibility that it is due to GnRH-induced desensitization of gonadotropes. Dispersed pituitary cells from proestrous and estrous rats were preincubated with GnRH (3 or 6 h, 10(- 10) or 10(-9) M), progesterone (13 h, 100 or 200 nM), GnRH plus progesterone, or medium alone. Then, the cells were retrypsinized to permit performance of the reverse hemolytic plaque assay for measurement of LH secretin, during which they were treated with GnRH(0,10(-11),10(-10), and 10(-8)M) for 2 h. The cells from estrous animals showed the large decline in GnRH responsiveness typical of that day of the cycle compared to those from proestrous animals (the total amount of LH secreted decreased by 50-70%). Preincubation of cells from proestrous rat pituitary glands with GnRH in concentrations and for durations that were designed to mimic the physiological situation induced a decline in GnRH responsiveness similar to that observed at estrus. Preincubation with progesterone also reduced the pituitary responsiveness to GnRH in a dose-dependent manner, but did not show additive effects with GnRH. Our results suggest that the major increase in GnRH secretion that induces the preovulatory surge of LH secretion may also participate in inducing the major decrease in pituitary responsiveness to GnRH that occurs from proestrus to estrus.


Assuntos
Estro/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Luteinizante/biossíntese , Hipófise/metabolismo , Animais , Células Cultivadas , Feminino , Ratos
18.
Mol Cell Endocrinol ; 114(1-2): 127-35, 1995 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-8674837

RESUMO

Enhanced responsiveness of the pituitary gland to GnRH is a fundamental step required for secretion of the proestrous LH surge, and is achieved primarily by the actions of ovarian steroid hormones on the gonadotropes. The mechanisms involved are still unclear but the cAMP second messenger pathway can mediate some of these activities. The present study was undertaken to determine the effects of in vitro cholera toxin (CTX; increases cAMP) pretreatment of pituitary cells from proestrous and diestrous 1 rats relative to their LH secretory response to GnRH using a reverse hemolytic plaque assay. The number of gonadotropes that secrete LH in the group treated with CTX increased at low doses of GnRH and also in the absence of the peptide, but decreased at high doses, showing a dual effect: stimulation of some gonadotropes and inhibition of others. The inhibition was achieved within 3 h of pretreatment in proestrous cells but it was not seen until 20 h in diestrous 1 cells. This suggests the existence of at least two subpopulations of gonadotropes, one of which is stimulated by cAMP and another which is inhibited.


Assuntos
AMP Cíclico/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Hormônio Luteinizante/metabolismo , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/fisiologia , Animais , Toxina da Cólera/farmacologia , Diestro/fisiologia , Feminino , Técnica de Placa Hemolítica , Técnicas In Vitro , Proestro/fisiologia , Ratos , Ratos Sprague-Dawley
19.
Virus Res ; 37(1): 1-12, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7483817

RESUMO

Caliciviruses were isolated from feces of skunks imported from the north central United States to Canada. Virus isolation was accomplished using adenovirus-transformed human kidney (293) cells, swine testes and Vero cells. Plaque size variants were presented, but there was no apparent difference in virus morphology by negative stain or immune electron microscopy. Pigs infected with skunk calicivirus had a slightly elevated body temperature at 3 days postinfection. Although the infected animals seroconverted, no overt clinical signs were observed. Purified infectious genomic skunk calicivirus RNA behaved exactly as San Miguel sea lion virus (SMSV) 1 and 4 genomic RNA in cell culture transfection studies. Of the cell types examined, only primary porcine kidney, 293 and Vero cells supported viral replication. No viral replication was detected in cells of bovine, equine, ovine, caprine or feline origin. The skunk caliciviruses contained a single capsid protein with a relative mobility similar to SMSV virus 1 and 4 capsid proteins. The capsid protein was positive by Western blot analysis with SMSV and vesicular exanthema of swine virus (VESV) antisera. Purified RNA from skunk calicivirus infected cells was subjected to reverse transcription followed by polymerase chain reaction. Nucleotide sequences were identified that had greater than 85% similarity to the 2C and RNA polymerase gene regions of SMSV 1 and 4 and VESV A48. Predicted amino acid sequences of these regions were greater than 95% similar and the partial coding sequence of the polymerase gene contained the YGDD sequence common to positive-strand RNA virus polymerases.


Assuntos
Antígenos Virais/imunologia , Caliciviridae/isolamento & purificação , Mephitidae/virologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Western Blotting , Caliciviridae/classificação , Caliciviridae/ultraestrutura , Infecções por Caliciviridae/veterinária , Infecções por Caliciviridae/virologia , Capsídeo/análise , Células Cultivadas , Chlorocebus aethiops , Genótipo , Humanos , Microscopia Imunoeletrônica , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , RNA Viral/análise , Suínos , Transfecção , Células Vero , Replicação Viral
20.
Cancer Res ; 55(4): 817-22, 1995 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-7850795

RESUMO

Recently we reported the presence of specific high affinity binding sites for luteinizing hormone-releasing hormone (LHRH) and its analogues (Kd = 1.5 or 1.7 nM) in the human epithelial ovarian cancer cell lines EFO-21 and EFO-27. The proliferation of these cell lines was inhibited by nM concentrations of a LHRH agonist. This study was performed to ascertain whether these ovarian cancer cell lines produce LHRH and whether the high affinity LHRH binding site found previously was identical to the pituitary LHRH receptor. Significant amounts of immunoreactive LHRH were found in the extracts of both the EFO-21 cell line (449 +/- 56 fmol/10(6) cells) and the EFO-27 line (409 +/- 76 fmol/10(6) cells). LHRH bioactivity of these extracts, assessed in terms of release of luteinizing hormone by rat pituitary cells, was comparable to that of authentic LHRH. EFO-21 and EFO-27 cells expressed the mRNAs for both human LHRH and human LHRH receptor as assessed by reverse transcriptase-PCR using oligonucleotide primers according to published sequences. In addition, in eight of eight biopsy samples of human epithelial ovarian cancers we detected mRNA for LHRH, six of these specimens expressed the mRNA representing the LHRH receptor. These data support the concept that human epithelial ovarian cancers might have a local system based on LHRH to regulate cell proliferation. It is still obscure at present whether LHRH produced locally has a stimulatory, inhibitory, or no impact on the proliferation of ovarian cancer cells. However, exogenous LHRH agonists seem to have clear antiproliferative activity, probably mediated through LHRH receptors. This finding might provide the base for novel approaches in the therapy of epithelial ovarian cancer.


Assuntos
Hormônio Liberador de Gonadotropina/genética , Neoplasias Ovarianas/metabolismo , RNA Mensageiro/metabolismo , Receptores LHRH/genética , Animais , Sequência de Bases , Sítios de Ligação , Biópsia , DNA de Neoplasias/genética , Epitélio/patologia , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Dados de Sequência Molecular , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/ultraestrutura , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , Ratos , Receptores LHRH/metabolismo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA