Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Int J Obes (Lond) ; 39(1): 105-13, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24732144

RESUMO

BACKGROUND: In the central nervous system (CNS), thyrotropin-releasing hormone (TRH) has an important role in regulating energy balance. We previously showed that dietary deprivation of leucine in mice increases energy expenditure through CNS-dependent regulation. However, the involvement of central TRH in this regulation has not been reported. METHODS: Male C57J/B6 mice were maintained on a control or leucine-deficient diet for 7 days. Leucine-deprived mice were either third intracerebroventricular (i.c.v.) injected with a TRH antibody followed by intraperitoneal (i.p.) injection of triiodothyronine (T3) or i.c.v. administrated with an adenovirus of shCREB (cAMP-response element binding protein) followed by i.c.v. injection of TRH. Food intake and body weight were monitored daily. Oxygen consumption, physical activity and rectal temperature were assessed after the treatment. After being killed, the hypothalamus and the brown adipose tissue were collected and the expression of related genes and proteins related was analyzed. In other experiments, control or leucine-deficient medium incubated primary cultured neurons were either infected with adenovirus-mediated short hairpin RNA targeting extracellular signal-regulated kinases 1 and 2 (Ad-shERK1/2) or transfected with plasmid-overexpressing protein phosphatase 1 regulatory subunit 3C (PPP1R3C). RESULTS: I.c.v. administration of anti-TRH antibodies significantly reduced leucine deprivation-stimulated energy expenditure. Furthermore, the effects of i.c.v. TRH antibodies were reversed by i.p. injection of T3 during leucine deprivation. Moreover, i.c.v. injection of Ad-shCREB (adenovirus-mediated short hairpin RNA targeting CREB) significantly suppressed leucine deprivation-stimulated energy expenditure via modulation of TRH expression. Lastly, TRH expression was regulated by CREB, which was phosphorylated by ERK1/2 and dephosphorylated by PPP1R3C-containing protein Ser/Thr phosphatase type 1 (PP1) under leucine deprivation in vitro. CONCLUSIONS: Our data indicate a novel role for TRH in regulating energy expenditure via T3 during leucine deprivation. Furthermore, our findings reveal that TRH expression is activated by CREB, which is phosphorylated by ERK1/2 and dephosphorylated by PPP1R3C-containing PP1. Collectively, our studies provide novel insights into the regulation of energy homeostasis by the CNS in response to an essential amino-acid deprivation.


Assuntos
Sistema Nervoso Central/metabolismo , Metabolismo Energético/efeitos dos fármacos , Hipotálamo/metabolismo , Leucina/deficiência , Receptores do Hormônio Liberador da Tireotropina/metabolismo , Hormônio Liberador de Tireotropina/metabolismo , Animais , Anticorpos/farmacologia , Western Blotting , Sistema Nervoso Central/fisiopatologia , Hipotálamo/fisiopatologia , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Transdução de Sinais
2.
Arq. bras. endocrinol. metab ; Arq. bras. endocrinol. metab;58(9): 962-966, 12/2014. tab, graf
Artigo em Inglês | LILACS | ID: lil-732181

RESUMO

Reduced sensitivity to thyroid hormones (RSTH) is a rare disease that affects about 3,000 individuals, belonging to about 1,000 families. It results from reduced intracellular action of thyroid hormones (TH) genetically determined and manifests as persistent hyperthyroxinemia with non-suppressed thyroid-stimulating hormone (TSH). We describe a 67-years old, Caucasian woman, with past history of subtotal thyroidectomy due to diffuse goiter, who presents with a recurrence of goiter. Although she is clinically euthyroid, laboratory evaluation shows persistent hyperthyroxinemia with non-suppressed TSH. Response to thyrotropin releasing hormone (TRH) test was normal and TSH concentrations were not suppressed during oral administration of suprafisiologic doses of levothyroxine (L-T4). Peripheral blood DNA was extracted from the patient and a mutation was found localized in cluster one, at codon 346 of the ligand binding domain of the THRB gene. The patient’s son underwent thyroid function testing (TFT) and genetic study, both negative, suggesting a sporadic mutation. RSTH should be considered in all hyperthyroxinemic patients who are clinically euthyroid. Mutations interfering with three major steps required for TH action on target tissues have been, so far, identified (TR-β, TR-α, MCT8, SPB2). Each mutation is associated with a distinctive syndrome. Goal of management is to maintain a normal serum TSH level and a eumetabolic state and offer appropriate genetic counselling and prenatal diagnosis. Inappropriate treatment of eumetabolic patients results in hypothyroidism and need for TH replacement.


A sensibilidade reduzida aos hormônios tiroidianos (RSTH) é uma doença rara que afeta cerca de 3.000 indivíduos em 1.000 famílias. Ela resulta de uma ação intracelular reduzida de hormônios tiroidianos (TH), é geneticamente determinada e se manifesta como hipertiroxinemia persistente com hormônio tireoestimulante (TSH) não suprimido. Descrevemos o caso de uma mulher caucasiana de 67 anos de idade com histórico de tiroidectomia subtotal por bócio difuso e que apresentou recorrência do bócio. Embora ela fosse clinicamente eutiroide, a avaliação laboratorial mostrou hipertiroxinemia persistente com TSH não suprimido. A resposta ao hormônio liberador da tireotrofina (TRH) foi normal e as concentrações de TSH não foram suprimidas durante a administração oral de doses suprafisiológicas de levotiroxina (L-T4). Foi extraído DNA de sangue periférico da paciente e encontrada uma mutação no cluster um do códon 346 do domínio de ligação do ligante do gene THRB. O filho da paciente foi submetido a um teste de função da tiroide e a um estudo genético, ambos negativos, o que sugeriu uma mutação esporádica. O RSTH deve ser considerado em todos os pacientes hipertiroxinêmicos que sejam clinicamente eutiroides. Foram identificadas, até hoje, mutações que interferem com os três passos principais necessários para a ação do TH sobre os tecidos-alvo (TR-b, TR-α, MCT8, SPB2). Cada mutação está associada com uma síndrome distinta. O objetivo do manejo é manter o nível sérico normal de TSH e um estado eumetabólico, além de se oferecer aconselhamento genético adequado e diagnóstico pré-natal. O tratamento inadequado de pacientes eumetabólicos leva ao hipotireoidismo e requer reposição de TH.


Assuntos
Idoso , Feminino , Humanos , Mutação , Doenças Raras/genética , Síndrome da Resistência aos Hormônios Tireóideos/genética , DNA , Éxons , Genes erbA , Bócio/genética , Hipertireoxinemia/sangue , Reação em Cadeia da Polimerase , Recidiva , Receptores do Hormônio Liberador da Tireotropina/sangue , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Testes de Função Tireóidea , Tireotropina/sangue , Tireotropina/efeitos dos fármacos , Tiroxina/farmacologia
3.
Arq Bras Endocrinol Metabol ; 58(9): 962-6, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25627054

RESUMO

Reduced sensitivity to thyroid hormones (RSTH) is a rare disease that affects about 3,000 individuals, belonging to about 1,000 families. It results from reduced intracellular action of thyroid hormones (TH) genetically determined and manifests as persistent hyperthyroxinemia with non-suppressed thyroid-stimulating hormone (TSH). We describe a 67-years old, Caucasian woman, with past history of subtotal thyroidectomy due to diffuse goiter, who presents with a recurrence of goiter. Although she is clinically euthyroid, laboratory evaluation shows persistent hyperthyroxinemia with non-suppressed TSH. Response to thyrotropin releasing hormone (TRH) test was normal and TSH concentrations were not suppressed during oral administration of suprafisiologic doses of levothyroxine (L-T4). Peripheral blood DNA was extracted from the patient and a mutation was found localized in cluster one, at codon 346 of the ligand binding domain of the THRB gene. The patient's son underwent thyroid function testing (TFT) and genetic study, both negative, suggesting a sporadic mutation. RSTH should be considered in all hyperthyroxinemic patients who are clinically euthyroid. Mutations interfering with three major steps required for TH action on target tissues have been, so far, identified (TR-ß, TR-α, MCT8, SPB2). Each mutation is associated with a distinctive syndrome. Goal of management is to maintain a normal serum TSH level and a eumetabolic state and offer appropriate genetic counselling and prenatal diagnosis. Inappropriate treatment of eumetabolic patients results in hypothyroidism and need for TH replacement.


Assuntos
Mutação , Doenças Raras/genética , Síndrome da Resistência aos Hormônios Tireóideos/genética , Idoso , DNA/análise , Éxons , Feminino , Genes erbA , Bócio/genética , Humanos , Hipertireoxinemia/sangue , Reação em Cadeia da Polimerase , Receptores do Hormônio Liberador da Tireotropina/sangue , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Recidiva , Testes de Função Tireóidea , Tireotropina/sangue , Tireotropina/efeitos dos fármacos , Tiroxina/farmacologia
4.
Menopause ; 19(11): 1253-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22713859

RESUMO

OBJECTIVE: Thyroid gland dysfunction is associated with menstrual cycle disturbances, infertility, and increased risk of miscarriage, but the mechanisms are poorly understood. However, little is known about the regulation of these receptors in the uterus. The aim of this study was to determine the effects of long-term treatment with steroid hormones on the expression, distribution, and regulation of the receptors for thyrotropin-releasing hormone (TRHR) and thyroid-stimulating hormone (TSHR), thyroid hormone receptor α1/α2 (THRα1/α2), and THRß1 in the uterus of surgically menopausal monkeys. METHODS: Eighty-eight cynomolgus macaques were ovariectomized and treated orally with conjugated equine estrogens (CEE; n = 20), a combination of CEE and medroxyprogesterone acetate (MPA; n = 20), or tibolone (n = 28) for 2 years. The control group (OvxC; n = 20) received no treatment. Immunohistochemistry was used to evaluate the protein expression and distribution of the receptors in luminal epithelium, glands, stroma, and myometrium of the uterus. RESULTS: Immunostaining of TRHR, TSHR, and THRs was detected in all uterine compartments. Epithelial immunostaining of TRHR was down-regulated in the CEE + MPA group, whereas in stroma, both TRHR and TSHR were increased by CEE + MPA treatment as compared with OvxC. TRHR immunoreactivity was up-regulated, but THRα and THRß were down-regulated, in the myometrium of the CEE and CEE + MPA groups. The thyroid-stimulating hormone level was higher in the CEE and tibolone groups as compared with OvxC, but the level of free thyroxin did not differ between groups. CONCLUSIONS: All receptors involved in thyroid hormone function are expressed in monkey uterus, and they are all regulated by long-term steroid hormone treatment. These findings suggest that there is a possibility of direct actions of thyroid hormones, thyroid-stimulating hormone and thyrotropin-releasing hormone on uterine function.


Assuntos
Hormônios Esteroides Gonadais/farmacologia , Macaca fascicularis , Receptores dos Hormônios Tireóideos/efeitos dos fármacos , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Receptores da Tireotropina/efeitos dos fármacos , Útero/química , Animais , Estrogênios Conjugados (USP)/administração & dosagem , Feminino , Imuno-Histoquímica/veterinária , Acetato de Medroxiprogesterona/administração & dosagem , Norpregnenos/administração & dosagem , Receptores dos Hormônios Tireóideos/análise , Receptores da Tireotropina/análise , Receptores do Hormônio Liberador da Tireotropina/análise , Receptores alfa dos Hormônios Tireóideos/análise , Receptores alfa dos Hormônios Tireóideos/efeitos dos fármacos , Receptores beta dos Hormônios Tireóideos/análise , Receptores beta dos Hormônios Tireóideos/efeitos dos fármacos , Útero/fisiologia
5.
Expert Opin Ther Pat ; 21(11): 1673-91, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22017410

RESUMO

INTRODUCTION: The potential therapeutic applications of thyrotropin-releasing hormone (TRH) have attracted attention, based on its broad-spectrum neuropharmacological action rather than its endocrine properties. These central nervous system (CNS)-mediated effects provide the rationale for use of TRH and its analogs in the treatment of brain and spinal injury, and CNS disorders like schizophrenia, Alzheimer's disease, epilepsy, amyotrophic lateral sclerosis, Parkinson's disease, depression, shock and ischemia. AREAS COVERED: This review summarizes the patent literature and advances in the discovery and development of novel TRH analogs over the past 20 years. It provides a comprehensive overview of the development of new TRH analogs, giving emphasis to their pharmaceutical profile. EXPERT OPINION: The use of TRH in the treatment of various CNS disorders has been proven clinically. However, TRH itself is a poor drug candidate due to its short plasma half-life (5 min), poor biopharmaceutical properties (low intestinal and CNS permeability) and endocrine side effect. Nevertheless, researchers have come up with metabolically stable, more potent and selective TRH analogs and prodrugs. Taltirelin, one of the TRH analogs, has been approved under the trade name of Ceredist(®) in Japan for the treatment of spinocerebellar degeneration. Several other TRH analogs are in various stages of preclinical or clinical development.


Assuntos
Doenças do Sistema Nervoso Central/tratamento farmacológico , Desenho de Fármacos , Hormônio Liberador de Tireotropina/farmacologia , Animais , Doenças do Sistema Nervoso Central/fisiopatologia , Meia-Vida , Humanos , Patentes como Assunto , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Receptores do Hormônio Liberador da Tireotropina/metabolismo , Hormônio Liberador de Tireotropina/análogos & derivados , Hormônio Liberador de Tireotropina/farmacocinética , Distribuição Tecidual
6.
Cell Tissue Res ; 334(2): 283-94, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18795335

RESUMO

Thyrotropin-releasing hormone (TRH) was initially discovered as a neuropeptide synthesized in the hypothalamus. Receptors for this hormone include TRH-receptor-1 (TRH-R1) and -2 (TRH-R2). Previous studies have shown that TRH-R1 and TRH-R2 are localized exclusively in adult Leydig cells (ALCs). We have investigated TRH-R1 and TRH-R2 expression in the testes of postnatal 8-, 14-, 21- 35-, 60-, and 90-day-old rats and in ethane dimethane sulfonate (EDS)-treated adult rats by using Western blotting, immunohistochemistry, and immunofluorescence. The effects of TRH on testosterone secretion of primary cultured ALCs from 90-day-old rats and DNA synthesis in Leydig cells from 21-day-old rats have also been examined. Western blotting and immunohistochemistry demonstrated that TRH-R1 and TRH-R2 were expressed in fetal Leydig cells (in 8-day-old rats) and in all stages of adult-type Leydig cells during development. Immunofluorescence double-staining revealed that newly regenerated Leydig cells in post-EDS 21-day rats expressed TRH-R1 and TRH-R2 on their first reappearance. Incubation with various doses of TRH affected testosterone secretion of primary cultured ALCs. Low concentrations of TRH (0.001, 0.01, and 0.1 ng/ml) inhibited basal and human chorionic gonadotrophin (hCG)-stimulated testosterone secretion of isolated ALCs, whereas relatively high doses of TRH (1 and 10 ng/ml) increased hCG-stimulated testosterone secretion. As detected by a 5-bromo-2'-deoxyuridine incorporation test, the DNA synthesis of Leydig cells from 21-day-old rats was promoted by low TRH concentrations. Thus, we have clarified the effect of TRH on testicular function: TRH might regulate the development of Leydig cells before maturation and the secretion of testosterone after maturation.


Assuntos
Células Intersticiais do Testículo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores do Hormônio Liberador da Tireotropina/metabolismo , Testosterona/biossíntese , Animais , Hormônios/farmacologia , Células Intersticiais do Testículo/efeitos dos fármacos , Células Intersticiais do Testículo/ultraestrutura , Masculino , Mesilatos/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Testículo/efeitos dos fármacos , Testículo/metabolismo , Testículo/ultraestrutura , Hormônio Liberador de Tireotropina/farmacologia
7.
Neurosci Lett ; 431(1): 26-30, 2008 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-18069127

RESUMO

Glp-Asn-Pro-D-Tyr-D-TrpNH(2) is a novel synthetic peptide that mimics and amplifies central actions of thyrotropin-releasing hormone (TRH) in rat without releasing TSH. The aim of this study was to compare the binding properties of this pentapeptide and its all-L counterpart (Glp-Asn-Pro-Tyr-TrpNH(2)) to TRH receptors in native rat brain tissue and cells expressing the two TRH receptor subtypes identified in rat to date, namely TRHR1 and TRHR2. Radioligand binding studies were carried out using [(3)H][3-Me-His(2)]TRH to label receptors in hippocampal, cortical and pituitary tissue, GH4 pituitary cells, as well as CHO cells expressing TRHR1 and/or TRHR2. In situ hybridization studies suggest that cortex expresses primarily TRHR2 mRNA, hippocampus primarily TRHR1 mRNA and pituitary exclusively TRHR1 mRNA. Competition experiments showed [3-Me-His(2)]TRH potently displaced [(3)H][3-Me-His(2)]TRH binding from all tissues/cells investigated. Glp-Asn-Pro-D-Tyr-D-TrpNH(2) in concentrations up to 10(-5)M did not displace [(3)H][3-Me-His(2)]TRH binding to membranes derived from GH4 cells or CHO-TRHR1 cells, consistent with its lack of binding to pituitary membranes and TSH-releasing activity. Similar results were obtained for the corresponding all-L peptide. In contrast, both pentapeptides displaced binding from rat hippocampal membranes (pIC(50) Glp-Asn-Pro-D-Tyr-D-TrpNH(2): 7.7+/-0.2; pIC(50) Glp-Asn-Pro-Tyr-TrpNH(2): 6.6+/-0.2), analogous to cortical membranes (pIC(50) Glp-Asn-Pro-D-Tyr-D-TrpNH(2): 7.8+/-0.2; pIC(50) Glp-Asn-Pro-Tyr-TrpNH(2): 6.6+/-0.2). Neither peptide, however, displaced [(3)H][3-Me-His(2)]TRH binding to CHO-TRHR2. Thus, this study reveals for the first time significant differences in the binding properties of native and heterologously expressed TRH receptors. Also, the results raise the possibility that Glp-Asn-Pro-D-Tyr-D-TrpNH(2) is not displacing [(3)H][3-Me-His(2)]TRH from a known TRH receptor in rat cortex, but rather a hitherto unidentified TRH receptor.


Assuntos
Córtex Cerebral/metabolismo , Hipocampo/metabolismo , Oligopeptídeos/metabolismo , Hipófise/metabolismo , Receptores do Hormônio Liberador da Tireotropina/metabolismo , Hormônio Liberador de Tireotropina/análogos & derivados , Hormônio Liberador de Tireotropina/metabolismo , Sequência de Aminoácidos/fisiologia , Animais , Sítios de Ligação/fisiologia , Ligação Competitiva/efeitos dos fármacos , Ligação Competitiva/fisiologia , Células CHO , Córtex Cerebral/efeitos dos fármacos , Cricetinae , Cricetulus , Hipocampo/efeitos dos fármacos , Oligopeptídeos/síntese química , Oligopeptídeos/farmacologia , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Hormônios Peptídicos/metabolismo , Hormônios Peptídicos/farmacologia , Hipófise/efeitos dos fármacos , Ensaio Radioligante , Ratos , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Hormônio Liberador de Tireotropina/síntese química , Hormônio Liberador de Tireotropina/farmacologia
8.
Cell Biochem Funct ; 26(2): 264-74, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18041110

RESUMO

The molecular mechanisms involved in GPCR-initiated signaling cascades where the two receptors share the same signaling cascade, such as thyrotropin-releasing hormone (TRH) and angiotensin II (ANG II), are still far from being understood. Here, we analyzed hormone-induced Ca(2+) responses and the process of desensitization in HEK-293 cells, which express endogenous ANG II receptors. These cells were transfected to express exogenously high levels of TRH receptors (clone E2) or both TRH receptors and G(11)alpha protein (clone E2M11). We observed that the characteristics of the Ca(2+) response, as well as the process of desensitization, were both strongly dependent on receptor number and G(11)alpha protein level. Whereas treatment of E2 cells with TRH or ANG II led to significant desensitization of the Ca(2+) response to subsequent addition of either hormone, the response was not desensitized in E2M11 cells expressing high levels of G(11)alpha. In addition, stimulation of both cell lines with THR elicited a clear heterologous desensitization to subsequent stimulation with ANG II. On the other hand, ANG II did not affect a subsequent response to TRH. ANG II-mediated signal transduction was strongly dependent on plasma membrane integrity modified by cholesterol depletion, but signaling through TRH receptors was altered only slightly under these conditions. It may be concluded that the level of expression of G-protein-coupled receptors and their cognate G-proteins strongly influences not only the magnitude of the Ca(2+) response but also the process of desensitization and resistance to subsequent hormone addition.


Assuntos
Angiotensina II/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Membrana Celular/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/biossíntese , Hormônio Liberador de Tireotropina/farmacologia , Cálcio/farmacologia , Sinalização do Cálcio/fisiologia , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Colesterol/análise , Colesterol/metabolismo , Relação Dose-Resposta a Droga , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/efeitos dos fármacos , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Humanos , Receptores do Hormônio Liberador da Tireotropina/biossíntese , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Temperatura , Fatores de Tempo , Transfecção
9.
Mol Cell Biol ; 26(6): 2247-61, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16508001

RESUMO

In pituitary GH3B6 cells, signaling involving the protein kinase C (PKC) multigene family can self-organize into a spatiotemporally coordinated cascade of isoform activation. Indeed, thyrotropin-releasing hormone (TRH) receptor activation sequentially activated green fluorescent protein (GFP)-tagged or endogenous PKCbeta1, PKCalpha, PKCepsilon, and PKCdelta, resulting in their accumulation at the entire plasma membrane (PKCbeta and -delta) or selectively at the cell-cell contacts (PKCalpha and -epsilon). The duration of activation ranged from 20 s for PKCalpha to 20 min for PKCepsilon. PKCalpha and -epsilon selective localization was lost in the presence of Gö6976, suggesting that accumulation at cell-cell contacts is dependent on the activity of a conventional PKC. Constitutively active, dominant-negative PKCs and small interfering RNAs showed that PKCalpha localization is controlled by PKCbeta1 activity and is calcium independent, while PKCepsilon localization is dependent on PKCalpha activity. PKCdelta was independent of the cascade linking PKCbeta1, -alpha, and -epsilon. Furthermore, PKCalpha, but not PKCepsilon, is involved in the TRH-induced beta-catenin relocation at cell-cell contacts, suggesting that PKCepsilon is not the unique functional effector of the cascade. Thus, TRH receptor activation results in PKCbeta1 activation, which in turn initiates a calcium-independent but PKCbeta1 activity-dependent sequential translocation of PKCalpha and -epsilon. These results challenge the current understanding of PKC signaling and raise the question of a functional dependence between isoforms.


Assuntos
Proteína Quinase C/metabolismo , Transdução de Sinais , Animais , Cálcio/metabolismo , Carbazóis/farmacologia , Comunicação Celular , Linhagem Celular , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Indóis/farmacologia , Ionomicina/farmacologia , Isoenzimas/genética , Isoenzimas/metabolismo , Hipófise/citologia , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/genética , Proteína Quinase C beta , Transporte Proteico , Ratos , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Receptores do Hormônio Liberador da Tireotropina/metabolismo , Hormônio Liberador de Tireotropina/metabolismo , Hormônio Liberador de Tireotropina/farmacologia , beta Catenina/efeitos dos fármacos , beta Catenina/metabolismo
10.
Mol Endocrinol ; 19(11): 2859-70, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16020481

RESUMO

To investigate the function of dimerization of the TRH receptor, a controlled dimerization system was developed. A variant FK506 binding protein (FKBP) domain was fused to the receptor C terminus and dimerization induced by incubating cells with dimeric FKBP ligand, AP20187. The TRH receptor-fusion bound hormone and signaled normally. Addition of dimerizer to cells expressing the receptor-FKBP fusion dramatically increased the fraction of receptor running as dimer on SDS-PAGE. AP20187 caused dimerization in a time- and concentration-dependent manner, acting within 1 min. Dimerizer had no effect on TRH receptors lacking the FKBP domain, and its effects were blocked by excess monomeric FKBP ligand. AP20187-induced dimerization did not cause receptor phosphorylation, inositol phosphate production, or ERK1/2 activation, and dimerizer did not alter signaling by TRH. Induced dimerization did, however, alter TRH receptor trafficking. TRH promoted greater receptor internalization in cells treated with AP20187 but not monomeric ligand, based on loss of surface binding sites and immunostaining. Dimerization increased the rate of internalization of TRH receptors and decreased the apparent rate of receptor recycling. AP20187 enhanced the small amount of TRH-induced receptor internalization when the receptor-FKBP fusion protein was expressed in cells lacking beta-arrestins. The results show that controlled dimerization of the TRH receptor potentiates hormone-induced receptor trafficking.


Assuntos
Receptores do Hormônio Liberador da Tireotropina/metabolismo , Hormônio Liberador de Tireotropina/metabolismo , Animais , Arrestinas/metabolismo , Células CHO , Cálcio/metabolismo , Membrana Celular/química , Cricetinae , Dimerização , Ativação Enzimática/efeitos dos fármacos , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Transporte Proteico/efeitos dos fármacos , Receptores do Hormônio Liberador da Tireotropina/análise , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tacrolimo/análogos & derivados , Tacrolimo/farmacologia , Proteínas de Ligação a Tacrolimo/análise , Proteínas de Ligação a Tacrolimo/efeitos dos fármacos , Proteínas de Ligação a Tacrolimo/metabolismo , Hormônio Liberador de Tireotropina/farmacologia , Fosfolipases Tipo C/metabolismo , beta-Arrestinas
11.
Mol Pharmacol ; 66(5): 1192-200, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15306657

RESUMO

A binding pocket for thyrotropin-releasing hormone (TRH) within the transmembrane helices of the TRH receptor type 1 (TRH-R1) has been identified based on experimental evidence and computer simulations. To determine the binding site for a competitive inverse agonist, midazolam, three of the four residues that directly contact TRH and other residues that restrain TRH-R1 in an inactive conformation were screened by mutagenesis and binding assays. We found that two residues that directly contact TRH, Asn-110 in transmembrane helix 3 (3.37) and Arg-306 in transmembrane helix 7 (7.39), were important for midazolam binding but another, Tyr-282 in transmembrane helix 6 (6.51), was not. A highly conserved residue, Trp-279 in transmembrane helix 6 (6.48), which was reported to be critical in stabilizing TRH-R1 in an inactive state but not for TRH binding, was critical for midazolam binding. We used our previous model of the unoccupied TRH-R1 to generate a model of the TRH-R1/midazolam complex. The experimental results and the molecular model of the complex suggest that midazolam binds to TRH-R1 within a transmembrane helical pocket that partially overlaps the TRH binding pocket. This result is consistent with the competitive antagonism of midazolam binding. We suggest that the mechanism of inverse agonism effected by midazolam involves its direct interaction with Trp-279, which contributes to the stabilization of the inactive conformation of TRH-R1.


Assuntos
Midazolam/farmacologia , Receptores do Hormônio Liberador da Tireotropina/metabolismo , Triptofano/metabolismo , Benzodiazepinas/farmacologia , Células Cultivadas , Humanos , Modelos Moleculares , Estrutura Secundária de Proteína , Receptores do Hormônio Liberador da Tireotropina/química , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Hormônio Liberador de Tireotropina/metabolismo , Transfecção
12.
Radiats Biol Radioecol ; 43(3): 334-6, 2003.
Artigo em Russo | MEDLINE | ID: mdl-12881991

RESUMO

The prolonged effect of thyroliberin in ULD after single intramuscular injection on contractility of lymphatic vessels directly was investigated. The controlled group of animals received injection of 0.2 ml of physiological solution. The experimental group was injected by 0.2 ml of thyroliberin in concentrations of 10(-10) or 10(-16) mol/l (1 x 10(-4) and 1 x 10(-10) micrograms/kg of the body weight respectively). During the experiment the animals were grouped in the following way: 1) directly after the injection; 2) 3 hours later; 3) on the 1st day and then every day during 2 weeks. Lymphatic vessels reactivity of the experimental animals as well as controlled was studied by application of thyroliberin and noradrenalin (in concentrations of 1 x 10(-16) and 1 x 10(-6) mol/l respectively) directly on mesentery lymphatic vessels. The lymphatic vessels reaction in control group of animals on the noradrenalin and thyroliberin was the same during the period of observation. Thyroliberin stimulated contractility at concentration of 1 x 10(-16) mol/l. The reaction of experimental group was dramatically decreased to 10(-4) mol/l on the 1st and the 3rd day (in the case i.m. injected concentration 1 x 10(-10) mol/l) and to 10(-10) mol/l (in the case of i.m. injected concentration 10(-16) mol/l). The lymphatic vessels reactivity to exogenous thyroliberin gradually established at the 6-7th days till 12th day from the moment of thyroliberin injection. The mechanisms of the action of thyroliberin in ULD are discussed.


Assuntos
Sistema Linfático/efeitos dos fármacos , Sistema Linfático/fisiologia , Mesentério/efeitos dos fármacos , Hormônio Liberador de Tireotropina/farmacologia , Animais , Relação Dose-Resposta a Droga , Endotélio Linfático/efeitos dos fármacos , Endotélio Linfático/fisiologia , Masculino , Mesentério/fisiologia , Contração Muscular/efeitos dos fármacos , Norepinefrina/farmacologia , Ratos , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Receptores do Hormônio Liberador da Tireotropina/metabolismo
13.
Life Sci ; 72(4-5): 601-7, 2002 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-12467901

RESUMO

Thyrotropin-releasing hormone (TRH) and some of its stable analogues have been shown to improve neurologic dysfunctions such as brain trauma in both animals and humans. Our previous study revealed that taltirelin, a novel orally active TRH analogue, binds to rat brain TRH receptors in vivo. The present study was undertaken to investigate whether taltirelin has neuroprotective effects in transient brain ischemia of C57BL/6J mice induced by bilateral carotid artery occlusion (2VO). Neuronal cell density in the hippocampal CA1 region of C57BL/6J mice was significantly (39.9%) decreased 1 week after 2VO-reperfusion, compared to the case of the sham group, and this reduction of hippocampal neuronal density was significantly suppressed by an intravenous (i.v.) injection of taltirelin (0.3 mg/kg). The i.v. injection of taltirelin at this dosage produced a significant increase in the dissociation constant (Kd) of specific [3H]MeTRH binding in sham and 2VO-reperfusion groups (33.6 and 51.4%, respectively) compared with the vehicle-treated group. These results indicate that the intravenously injected taltirelin bound to TRH receptors in the ischemic brain. There was little difference in the brain-to-plasma concentration ratio (Kp) of [14C]sucrose between the sham and 2VO groups of C57BL/6J mice, indicating that the tight junction of the blood-brain barrier may be intact in the ischemic brain. In conclusion, the study has shown that taltirelin may have a significant neuroprotective effect on the ischemic brain.


Assuntos
Ataque Isquêmico Transitório/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Nootrópicos/uso terapêutico , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Hormônio Liberador de Tireotropina/análogos & derivados , Hormônio Liberador de Tireotropina/uso terapêutico , Animais , Encéfalo/patologia , Química Encefálica/efeitos dos fármacos , Contagem de Células , Relação Dose-Resposta a Droga , Hipocampo/patologia , Ataque Isquêmico Transitório/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/patologia , Sacarose/metabolismo
14.
Bioorg Med Chem ; 10(2): 291-302, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11741778

RESUMO

A modified synthetic route has been developed so that the steric size of constraints added to the pyroglutamate region of TRH (pGluHisProNH(2)) can be varied. Both an analogue with a smaller ethylene bridge and a larger, more flexible propane bridge in this region have been synthesized. These analogues were synthesized in order to probe why the initial incorporation of an ethane bridge into this region of the molecule had led to an analogue with a binding constant and potency three times lower than that of an directly analogous unconstrained analogue. The data for both analogues indicated that the fall off in activity caused by the ethane bridge in the initial analogue was not caused by the size of the bridge.


Assuntos
Ácido Pirrolidonocarboxílico/química , Hormônio Liberador de Tireotropina/análogos & derivados , Hormônio Liberador de Tireotropina/química , Bioquímica/métodos , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos/métodos , Etano/química , Humanos , Conformação Proteica , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Receptores do Hormônio Liberador da Tireotropina/metabolismo , Relação Estrutura-Atividade
15.
Planta Med ; 67(3): 236-9, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11345694

RESUMO

The influence of two plant coumarins, osthol and xanthotoxin, on intracellular Ca2+ ([Ca2+]i) transients evoked by TRH were studied in clonal rat pituitary GH4C1 cells. Osthol, but not xanthotoxin, decreased the TRH-induced transient increase in [Ca2+]i in Fluo-3 loaded cells incubated in Ca(2+)-free buffer. Binding experiments with [3H]TRH showed that osthol decreased the binding of TRH to its receptor, whereas the affinity of the receptor for TRH increased. This resulted in a decreased TRH-evoked production of IP3 in cells treated with osthol, and a decreased mobilization of sequestered calcium. Osthol did not inhibit the release of calcium evoked by exogenous IP3 in permeabilized cells. Furthermore, osthol decreased the uptake of 45Ca2+ in response to high K+. Xanthotoxin had no effects in these experiments. The results show that osthol modulates TRH-evoked responses by interacting with the TRH receptor.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Cálcio/metabolismo , Cumarínicos/farmacologia , Metoxaleno/farmacologia , Hormônio Liberador de Tireotropina/metabolismo , Animais , Apiaceae/uso terapêutico , Bloqueadores dos Canais de Cálcio/química , Bloqueadores dos Canais de Cálcio/isolamento & purificação , Radioisótopos de Cálcio , Células Cultivadas , Cumarínicos/química , Cumarínicos/isolamento & purificação , Marcação por Isótopo , Metoxaleno/isolamento & purificação , Fosfatidilinositóis/análise , Fitoterapia , Hipófise/efeitos dos fármacos , Potássio/farmacocinética , Ratos , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Receptores do Hormônio Liberador da Tireotropina/metabolismo , Trítio
16.
J Biol Chem ; 276(20): 16720-30, 2001 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-11279027

RESUMO

G protein-coupled inwardly rectifying K+ (GIRK) channels can be activated or inhibited by distinct classes of receptor (G(alpha)i/o- and G(alpha)q-coupled), providing dynamic regulation of cellular excitability. Receptor-mediated activation involves direct effects of G(beta)gamma subunits on GIRK channels, but mechanisms involved in GIRK channel inhibition have not been fully elucidated. An HEK293 cell line that stably expresses GIRK1/4 channels was used to test G protein mechanisms that mediate GIRK channel inhibition. In cells transiently or stably cotransfected with 5-HT1A (G(alpha)i/o-coupled) and TRH-R1 (G(alpha)q-coupled) receptors, 5-HT (5-hydroxytryptamine; serotonin) enhanced GIRK channel currents, whereas thyrotropin-releasing hormone (TRH) inhibited both basal and 5-HT-activated GIRK channel currents. Inhibition of GIRK channel currents by TRH primarily involved signaling by G(alpha)q family subunits, rather than G(beta)gamma dimers: GIRK channel current inhibition was diminished by Pasteurella multocida toxin, mimicked by constitutively active members of the G(alpha)q family, and reduced by minigene constructs that disrupt G(alpha)q signaling, but was completely preserved in cells expressing constructs that interfere with signaling by G(beta)gamma subunits. Inhibition of GIRK channel currents by TRH and constitutively active G(alpha)q was reduced by, an inhibitor of phospholipase C (PLC). Moreover, TRH- R1-mediated GIRK channel inhibition was diminished by minigene constructs that reduce membrane levels of the PLC substrate phosphatidylinositol bisphosphate, further implicating PLC. However, we found no evidence for involvement of protein kinase C, inositol trisphosphate, or intracellular calcium. Although these downstream signaling intermediaries did not contribute to receptor-mediated GIRK channel inhibition, bath application of TRH decreased GIRK channel activity in cell-attached patches. Together, these data indicate that receptor-mediated inhibition of GIRK channels involves PLC activation by G(alpha) subunits of the G(alpha)q family and suggest that inhibition may be communicated at a distance to GIRK channels via unbinding and diffusion of phosphatidylinositol bisphosphate away from the channel.


Assuntos
Proteínas de Bactérias , Proteínas Heterotriméricas de Ligação ao GTP/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização , Canais de Potássio/fisiologia , Receptores de Serotonina/fisiologia , Receptores do Hormônio Liberador da Tireotropina/fisiologia , Serotonina/farmacologia , Hormônio Liberador de Tireotropina/farmacologia , Fosfolipases Tipo C/metabolismo , Toxinas Bacterianas/farmacologia , Linhagem Celular , Membrana Celular/fisiologia , Dimerização , Inibidores Enzimáticos/farmacologia , Estrenos/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Proteínas Heterotriméricas de Ligação ao GTP/química , Humanos , Indóis/farmacologia , Inositol 1,4,5-Trifosfato/metabolismo , Maleimidas/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Pasteurella multocida , Ésteres de Forbol/farmacologia , Bloqueadores dos Canais de Potássio , Subunidades Proteicas , Pirrolidinonas/farmacologia , Receptores de Serotonina/efeitos dos fármacos , Receptores 5-HT1 de Serotonina , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Transfecção
17.
Endocrinology ; 141(10): 3717-22, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11014227

RESUMO

Juxtamembrane residues in the putative third intracellular (I3) loops of a number of G protein-coupled receptors (GPCRs) have been shown to be important for coupling to G proteins. According to standard hydropathy analysis, the I3 loop of the mouse TRH receptor type 1 (mTRH-R1) is composed of 51 amino acids from position-213 to position-263. We constructed deletion and site-specific I3 loop TRH-R mutants and studied their binding and TRH-stimulated signaling activities. As expected, the effects of these mutations on TRH binding were small (less than 5-fold decreases in affinity). No effect on TRH-stimulated signaling activity was found in a mutant receptor in which the I3 loop was shortened to 16 amino acids by deleting residues from Asp-226 to Ser-260. In contrast, mutants with deletions from Asp-222 to Ser-260 or from Asp-226 to Gln-263 exhibited reduced TRH-stimulated signaling. In the region near transmembrane helix 6, single site-specific substitution of either Arg-261 or Lys-262 by neutral glutamine had little effect on signaling, but mutant TRH-Rs that were substituted by glutamine at both basic residues exhibited reduced TRH-stimulated activity. The reduced signaling activity of this doubly substituted mutant was reversed by over expressing the a subunit of Gq. These data demonstrate that the juxtamembrane regions in the TRH-R I3 loop are important for coupling to Gq.


Assuntos
Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Receptores do Hormônio Liberador da Tireotropina/genética , Receptores do Hormônio Liberador da Tireotropina/metabolismo , Sequência de Aminoácidos/genética , Substituição de Aminoácidos , Animais , Células COS , Feminino , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Ligantes , Conformação Molecular , Dados de Sequência Molecular , Oócitos , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Receptores do Hormônio Liberador da Tireotropina/fisiologia , Transdução de Sinais/efeitos dos fármacos , Hormônio Liberador de Tireotropina/farmacologia , Xenopus laevis
18.
J Am Vet Med Assoc ; 211(1): 79-81, 1997 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-9215417

RESUMO

OBJECTIVE: To evaluate results of a combined dexamethasone suppression/thyrotropin-releasing hormone (TRH) stimulation test in horses suspected clinically to have a pars intermedia pituitary adenoma (PIPA). DESIGN: Case-control study. ANIMALS: 7 healthy adult horses and 5 horses suspected to have a PIPA. PROCEDURE: A baseline blood sample was collected, and dexamethasone (40 micrograms/kg [18 micrograms/lb] of body weight, IV) was administered; a second blood sample was collected 3 hours later, and TRH (1.1 mg, IV) was administered; serial blood samples were collected 15, 30, 45, 60, and 90 minutes and 21 hours after TRH administration (24 hours after dexamethasone injection). Cortisol concentration was determined for all blood samples. RESULTS: Baseline cortisol concentration was significantly lower in horses suspected to have a PIPA than in healthy horses. Cortisol concentration was suppressed by dexamethasone in both groups; however, after TRH administration, cortisol concentration returned to baseline values in horses suspected to have a PIPA, but not in healthy horses. Concentration was still less than the baseline value 24 hours after dexamethasone administration in healthy horses. CLINICAL IMPLICATIONS: The combined dexamethasone suppression/TRH stimulation test may be a useful diagnostic test in horses suspected to have a PIPA. For clinical application, collection of a blood sample 30 minutes after TRH administration is recommended.


Assuntos
Adenoma/veterinária , Doenças dos Cavalos/fisiopatologia , Cavalos/fisiologia , Hidrocortisona/sangue , Neoplasias Hipofisárias/veterinária , Adenoma/diagnóstico , Adenoma/fisiopatologia , Animais , Estudos de Casos e Controles , Dexametasona , Feminino , Glucocorticoides , Doenças dos Cavalos/diagnóstico , Hipófise/fisiopatologia , Neoplasias Hipofisárias/diagnóstico , Neoplasias Hipofisárias/fisiopatologia , Receptores do Hormônio Liberador da Tireotropina/antagonistas & inibidores , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Hormônio Liberador de Tireotropina
19.
FEBS Lett ; 407(3): 257-60, 1997 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-9175863

RESUMO

Wild-type and palmitoylation defective mutants of the murine G protein G11alpha were transfected into HEK293 cells. Wild-type G11alpha was membrane associated, Cys9Ser Cys10Ser G11alpha was present in the soluble fraction whilst both Cys9Ser G11alpha and Cys10Ser G11alpha were distributed between the fractions. Expression of the rat TRH receptor resulted in agonist stimulation of inositol phosphate accumulation. The degree of stimulation produced by TRH following co-transfection of the palmitoylation-resistant forms of G11alpha compared to the wild-type protein correlated with the amount of membrane-associated G protein.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Receptores do Hormônio Liberador da Tireotropina/metabolismo , Animais , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Proteínas de Ligação ao GTP/química , Proteínas de Ligação ao GTP/genética , Humanos , Camundongos , Mutação , Ácidos Palmíticos/química , Ácidos Palmíticos/metabolismo , Ratos , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Receptores do Hormônio Liberador da Tireotropina/genética , Hormônio Liberador de Tireotropina/farmacologia , Transfecção
20.
Mol Pharmacol ; 51(5): 785-93, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9145916

RESUMO

The localization of an epitope-tagged receptor for thyrotropin-releasing hormone (TRH) expressed in different cell contexts was studied with immunofluorescence microscopy. In pituitary lactotrophs, which normally express TRH receptors, and in AtT20 pituitary corticotrophs, TRH receptor immunoreactivity was primarily confined to the plasma membrane. In HEK 293 and COS7 cells, TRH receptors were predominantly intracellular. In transiently transfected COS7 cells, the TRH receptor colocalized with endoplasmic reticulum and Golgi markers. The pattern of TRH receptor immunofluorescence was the same over a wide range of receptor expression in transiently transfected COS7 cells, and all cell lines bound similar amounts of 3H- and rhodamine-labeled TRH analogs, suggesting that cell-specific differences in TRH receptor localization were not simply the result of overexpression. In all cell contexts, TRH receptors on the plasma membrane underwent extensive ligand-driven endocytosis. Inhibitors of glycosylation did not alter the subcellular distribution of receptors. In HEK 293 cells expressing the transfected TRH receptor, protein synthesis inhibitors caused translocation of intracellular receptors to the cell surface, as shown by a marked increase in cell surface immunofluorescence and [3H][N3-methyl-His2]TRH binding. These results demonstrate that the subcellular localization of the TRH receptor depends on the cell context in which it is expressed and that intracellular receptors are capable of translocation to the plasma membrane.


Assuntos
Receptores do Hormônio Liberador da Tireotropina/metabolismo , Frações Subcelulares/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Linhagem Celular , Cicloeximida/farmacologia , Humanos , Microscopia de Fluorescência , Dados de Sequência Molecular , Inibidores da Síntese de Proteínas/farmacologia , Puromicina/farmacologia , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA