Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
2.
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
3.
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
4.
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
5.
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
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.
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
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.
Avian Dis ; 41(1): 36-44, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9087318

RESUMO

The VP2 gene is part of the genomic segment A of infectious bursal disease virus (IBDV). It has been identified as the major host-protective antigen of IBDV and is known to contain conformationally dependent protective epitopes. A 643-base pair segment covering the hypervariable region of this gene from three recent serologic variant IBDV isolates from the southeastern United States, two variants from the Delmarva Peninsula, and three serologic standard viruses were amplified and sequenced using the reverse transcription polymerase chain reaction and cycle sequencing techniques. This was done to determine the molecular similarity among isolates that differ antigenically and pathologically. Sequence analysis suggested that the Arkansas (Ark) and Mississippi (Miss) isolates evolved closely and separately from the Delmarva variants (GLS and DELE), in contrast to the other southeastern variant Georgia (Ga), which is more closely related (98.32%) to Delaware E (DELE). All variants, except for Miss, underwent a shift in amino acid number 222 from proline to threonine. The sequence of Univax BD virus, a commercially available intermediate vaccine, was markedly different, evolving from a separate lineage than the others. Restriction enzyme sites could differentiate most isolates. Except for Miss, variants do not have EcoRII site at the larger hydrophilic domain. All variants lost their HaeIII, StuI, and StyI cutting sites with a change in base number 856. The TaqI site is in DELE, whereas the SpeI site is absent in the standard vaccine viruses. The SWASASGS heptapeptide is conserved in all virulent viruses, including APHIS, but not in the attenuated (Univax BD and Bursa Vac 3) and published (D78 and PBG98) vaccines.


Assuntos
Infecções por Birnaviridae/veterinária , Capsídeo/química , Vírus da Doença Infecciosa da Bursa/classificação , Doenças das Aves Domésticas , Sequência de Aminoácidos , Animais , Arkansas , Sequência de Bases , Infecções por Birnaviridae/virologia , Capsídeo/genética , Proteínas do Capsídeo , Galinhas , Delaware , Variação Genética , Georgia , Vírus da Doença Infecciosa da Bursa/genética , Vírus da Doença Infecciosa da Bursa/isolamento & purificação , Maryland , Mississippi , Dados de Sequência Molecular , Filogenia , Mapeamento por Restrição , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , Virginia
10.
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
11.
J Bacteriol ; 173(14): 4263-70, 1991 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-1829726

RESUMO

Porphyromonas (Bacteroides) gingivalis W12 binds and degrades human plasma fibronectin. In the presence of the protease inhibitor N-alpha-p-tosyl-L-lysyl chloromethyl ketone, P. gingivalis cells accumulated substantial amounts of 125I-fibronectin as a function of incubation time. Fibronectin binding was specific, reversible, and saturable. The Kd for the reaction was estimated to be on the order of 100 nM, and there was an average of 3.5 x 10(3) fibronectin binding sites per cell. Unlabeled fibronectin inhibited the binding of 125I-fibronectin to bacteria; however, fibrinogen was an even more efficient inhibitor of 125I-fibronectin binding. Unrelated proteins were without effect on fibronectin binding. A fibronectin-binding component (Mr, 150,000) was identified in sodium dodecyl sulfate-solubilized P. gingivalis. Fibronectin was degraded into discrete peptides by P. gingivalis W12. The degradation of fibronectin was inhibited by N-alpha-p-tosyl-L-lysyl chloromethyl ketone. Two P. gingivalis components (Mrs, 120,000 and 150,000) degraded fibronectin in substrate-containing gels following sodium dodecyl sulfate-polyacrylamide gel electrophoresis. In a previous study (M. S. Lantz, R. D. Allen, T. A. Vail, L. M. Switalski, and M. Hook, J. Bacteriol. 173:495-504, 1991), we found that the same strain of P. gingivalis bound and subsequently degraded human fibrinogen via apparently distinct cell surface components of molecular sizes similar to those of components now implicated in the binding and degradation of fibronectin. These results raise the possibility that the two ligands are recognized and modified by the same components on P. gingivalis W12. In support of this hypothesis, unlabeled fibrinogen effectively inhibited the binding of 125I-fibronectin to bacteria and blocked 125I-fibronectin binding to a P. gingivalis ligand-binding component (Mr, 150,000 immobilized on a nitrocellulose membrane.


Assuntos
Bacteroides/metabolismo , Proteínas de Transporte/metabolismo , Fibronectinas/metabolismo , Receptores Imunológicos/metabolismo , Sítios de Ligação , Eletroforese em Gel de Poliacrilamida , Radioisótopos do Iodo , Cinética , Peso Molecular , Inibidores de Proteases , Receptores de Fibronectina , Receptores Imunológicos/isolamento & purificação , Especificidade por Substrato , Tosilina Clorometil Cetona/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa