Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 256
Filtrar
1.
J Neuroendocrinol ; 35(11): e13354, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37946684

RESUMO

Pituitary adenylate cyclase-activating polypeptide (PACAP) and the homologous peptide, vasoactive intestinal peptide (VIP), participate in glucose homeostasis using insulinotropic and counterregulatory processes. The role of VIP receptor 2 (VPAC2R) in these opposing actions needs further characterization. In this study, we examined the participation of VPAC2R on basal glycemia, fasted levels of glucoregulatory hormones and on glycemia responses during metabolic and psychogenic stress using gene-deleted (Vipr2-/- ) female mice. The mean basal glycemia was significantly greater in Vipr2-/- in the fed state and after an 8-h overnight fast as compared to wild-type (WT) mice. Insulin tolerance testing following a 5-h fast (morning fast, 0.38 U/kg insulin) indicated no effect of genotype. However, during a more intense metabolic challenge (8 h, ON fast, 0.25 U/kg insulin), Vipr2-/- females displayed significantly impaired insulin hypoglycemia. During immobilization stress, the hyperglycemic response and plasma epinephrine levels were significantly elevated above basal in Vipr2-/- , but not WT mice, in spite of similar stress levels of plasma corticosterone. Together, these results implicate participation of VPAC2R in upregulated counterregulatory processes influenced by enhanced sympathoexcitation. Moreover, the suppression of plasma GLP-1 levels in Vipr2-/- mice may have removed the inhibition on hepatic glucose production and the promotion of glucose disposal by GLP-1. qPCR analysis indicated deregulation of central gene markers of PACAP/VIP signaling in Vipr2-/- , upregulated medulla tyrosine hydroxylase (Th) and downregulated hypothalamic Vip transcripts. These results demonstrate a physiological role for VPAC2R in glucose metabolism, especially during insulin challenge and psychogenic stress, likely involving the participation of sympathoadrenal activity and/or metabolic hormones.


Assuntos
Receptores do Hormônio Hipofisário , Receptores de Peptídeo Intestinal Vasoativo , Camundongos , Feminino , Animais , Receptores de Peptídeo Intestinal Vasoativo/genética , Receptores de Peptídeo Intestinal Vasoativo/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Deleção de Genes , Peptídeo Intestinal Vasoativo/metabolismo , Insulina/metabolismo , Glucose , Peptídeo 1 Semelhante ao Glucagon , Receptores do Hormônio Hipofisário/genética , Receptores Tipo II de Peptídeo Intestinal Vasoativo/genética
2.
J Mol Neurosci ; 73(9-10): 724-737, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37646964

RESUMO

Previous evidence shows that rapid changes occur in the brain following spinal cord injury (SCI). Here, we interrogated the expression of the neuropeptides pituitary adenylyl cyclase-activating peptide (PACAP), vasoactive intestinal peptides (VIP), and their binding receptors in the rat brain 24 h following SCI. Female Sprague-Dawley rats underwent thoracic laminectomy; half of the rats received a mild contusion injury at the level of the T10 vertebrate (SCI group); the other half underwent sham surgery (sham group). Twenty-four hours post-surgery, the hypothalamus, thalamus, amygdala, hippocampus (dorsal and ventral), prefrontal cortex, and periaqueductal gray were collected. PACAP, VIP, PAC1, VPAC1, and VPAC2 mRNA and protein levels were measured by real-time quantitative polymerase chain reaction and Western blot. In SCI rats, PACAP expression was increased in the hypothalamus (104-141% vs sham) and amygdala (138-350%), but downregulated in the thalamus (35-95%) and periaqueductal gray (58-68%). VIP expression was increased only in the thalamus (175-385%), with a reduction in the amygdala (51-68%), hippocampus (40-75%), and periaqueductal gray (74-76%). The expression of the PAC1 receptor was the least disturbed by SCI, with decrease expression in the ventral hippocampus (63-68%) only. The expression levels of VPAC1 and VPAC2 receptors were globally reduced, with more prominent reductions of VPAC1 vs VPAC2 in the amygdala (21-70%) and ventral hippocampus (72-75%). In addition, VPAC1 downregulation also extended to the dorsal hippocampus (69-70%). These findings demonstrate that as early as 24 h post-SCI, there are region-specific disruptions of PACAP, VIP, and related receptor transcript and protein levels in supraspinal regions controlling higher cognitive functions.


Assuntos
Receptores do Hormônio Hipofisário , Traumatismos da Medula Espinal , Feminino , Ratos , Animais , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Ratos Sprague-Dawley , Receptores do Hormônio Hipofisário/genética , Receptores do Hormônio Hipofisário/metabolismo , Peptídeo Intestinal Vasoativo/genética , Peptídeo Intestinal Vasoativo/metabolismo , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/genética , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/metabolismo , Receptores Tipo II de Peptídeo Intestinal Vasoativo/genética , Receptores Tipo II de Peptídeo Intestinal Vasoativo/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Traumatismos da Medula Espinal/metabolismo , Encéfalo/metabolismo
3.
Endocrinol Nutr ; 63(6): 274-84, 2016.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-27091627

RESUMO

Pituitary adenomas are uncommon, difficult to diagnose tumors whose heterogeneity and low incidence complicate large-scale studies. The Molecular Registry of Pituitary Adenomas (REMAH) was promoted by the Andalusian Society of Endocrinology and Nutrition (SAEN) in 2008 as a cooperative clinical-basic multicenter strategy aimed at improving diagnosis and treatment of pituitary adenomas by combining clinical, pathological, and molecular information. In 2010, the Spanish Society of Endocrinology and Nutrition (SEEN) extended this project to national level and established 6 nodes with common protocols and methods for sample and clinical data collection, molecular analysis, and data recording in a common registry (www.remahnacional.com). The registry combines clinical data with molecular phenotyping of the resected pituitary adenoma using quantitative real-time PCR of expression of 26 genes: Pituitary hormones (GH-PRL-LH-FSH-PRL-ACTH-CGA), receptors (somatostatin, dopamine, GHRH, GnRH, CRH, arginine-vasopressin, ghrelin), other markers (Ki67, PTTG1), and control genes. Until 2015, molecular information has been collected from 704 adenomas, out of 1179 patients registered. This strategy allows for comparative and relational analysis between the molecular profile of the different types of adenoma and the clinical phenotype of patients, which may provide a better understanding of the condition and potentially help in treatment selection. The REMAH is therefore a unique multicenter, interdisciplinary network founded on a shared database that provides a far-reaching translational approach for management of pituitary adenomas, and paves the way for the conduct of combined clinical-basic innovative studies on large patient samples.


Assuntos
Adenoma/epidemiologia , Endocrinologia/organização & administração , Neoplasias Hipofisárias/epidemiologia , Medicina de Precisão/tendências , Sistema de Registros , Pesquisa Translacional Biomédica/tendências , Adenoma/química , Adenoma/genética , Adolescente , Adulto , Idoso , Criança , Bases de Dados Factuais , Endocrinologia/tendências , Feminino , Perfilação da Expressão Gênica , Estudos de Associação Genética , Humanos , Masculino , Pessoa de Meia-Idade , Biologia Molecular , Proteínas de Neoplasias/análise , Proteínas de Neoplasias/genética , Hormônios Hipofisários/análise , Hormônios Hipofisários/genética , Neoplasias Hipofisárias/química , Neoplasias Hipofisárias/genética , RNA Neoplásico/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores do Hormônio Hipofisário/análise , Receptores do Hormônio Hipofisário/genética , Sociedades Médicas , Espanha/epidemiologia , Adulto Jovem
4.
Nat Commun ; 6: 8842, 2015 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-26548314

RESUMO

Understanding of the genetic regulation of puberty timing has come largely from studies of rare disorders and population-based studies in women. Here, we report the largest genomic analysis for puberty timing in 55,871 men, based on recalled age at voice breaking. Analysis across all genomic variants reveals strong genetic correlation (0.74, P=2.7 × 10(-70)) between male and female puberty timing. However, some loci show sex-divergent effects, including directionally opposite effects between sexes at the SIM1/MCHR2 locus (Pheterogeneity=1.6 × 10(-12)). We find five novel loci for puberty timing (P<5 × 10(-8)), in addition to nine signals in men that were previously reported in women. Newly implicated genes include two retinoic acid-related receptors, RORB and RXRA, and two genes reportedly disrupted in rare disorders of puberty, LEPR and KAL1. Finally, we identify genetic correlations that indicate shared aetiologies in both sexes between puberty timing and body mass index, fasting insulin levels, lipid levels, type 2 diabetes and cardiovascular disease.


Assuntos
Doenças Cardiovasculares/genética , Diabetes Mellitus Tipo 2/genética , Síndrome do Ovário Policístico/genética , Puberdade/genética , Adolescente , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Estatura/genética , Índice de Massa Corporal , Densidade Óssea/genética , Criança , HDL-Colesterol/sangue , Proteínas da Matriz Extracelular/genética , Feminino , Estudo de Associação Genômica Ampla , Humanos , Insulina/sangue , Vértebras Lombares/diagnóstico por imagem , Masculino , Proteínas do Tecido Nervoso/genética , Membro 2 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Radiografia , Receptores Acoplados a Proteínas G/genética , Receptores para Leptina/genética , Receptores do Hormônio Hipofisário/genética , Proteínas Repressoras/genética , Receptor X Retinoide alfa/genética , Fatores Sexuais , Fatores de Tempo , Triglicerídeos/sangue , População Branca/genética , Adulto Jovem
5.
Endocrinology ; 155(1): 81-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24169555

RESUMO

Melanin-concentrating hormone (MCH) is an orexigenic neuropeptide that is a ligand for two subtypes of MCH receptors, MCHR1 and MCHR2. MCHR1 is universally expressed in mammals ranging from rodents to humans, but the expression of MCHR2 is substantially restricted. In mammals, MCHR2 has been defined in primates as well as other species such as cats and dogs but is not seen in rodents. Although the role of MCHR1 in mediating the actions of MCH on energy balance is clearly defined using mouse models, the role of MCHR2 is harder to characterize because of its limited expression. To determine any potential role of MCHR2 in energy balance, we generated a transgenic MCHR1R2 mouse model, where human MCHR2 is coexpressed in MCHR1-expressing neurons. As shown previously, control wild-type mice expressing only native MCHR1 developed diet-induced obesity when fed a high-fat diet. In contrast, MCHR1R2 mice had lower food intake, leading to their resistance to diet-induced obesity. Furthermore, we showed that MCH action is altered in MCHR1R2 mice. MCH treatment in wild-type mice inhibited the activation of the immediate-early gene c-fos, and coexpression of MCHR2 reduced the inhibitory actions of MCHR1 on this pathway. In conclusion, we developed an experimental animal model that can provide insight into the action of MCHR2 in the central nervous system and suggest that some actions of MCHR2 oppose the endogenous actions of MCHR1.


Assuntos
Dieta , Regulação da Expressão Gênica , Obesidade/genética , Receptores do Hormônio Hipofisário/metabolismo , Animais , Dieta Hiperlipídica , Modelos Animais de Doenças , Expressão Gênica , Glucose/metabolismo , Insulina/metabolismo , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/metabolismo , Obesidade/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores do Hormônio Hipofisário/genética , Receptores de Somatostatina/metabolismo , Transdução de Sinais
6.
J Clin Oncol ; 31(21): 2654-61, 2013 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-23775959

RESUMO

PURPOSE: Previous studies have demonstrated the prognostic importance of the immune microenvironment in follicular lymphoma (FL). To investigate the molecular mechanisms during which tumor-infiltrating T cells (TILs) are altered in the FL microenvironment, we studied highly purified CD4 and CD8 TILs from lymph node biopsies at diagnosis in treatment-naive patients with FL compared with reactive tonsils and the peripheral blood of healthy donors. PATIENTS AND METHODS: Gene expression profiling of highly purified CD4 and CD8 TILs was performed on the Affymetrix platform. Diagnostic tissue microarrays from an independent patient set (n = 172) were used to verify protein expression and analyze any impact of TIL-expressed genes on outcome. Time-lapse imaging was used to assess T-cell motility. RESULTS: The most upregulated genes in both CD4 and CD8 TILs were PMCH, ETV1, and TNFRSF9. PMCH is not expressed in peripheral blood T cells, but expression is highly induced on culture with FL. Both CD4 and CD8 TILs from patients with FL have significantly impaired motility compared with those of healthy TILs from reactive tonsils and this can be induced on healthy T cells by FL cells. During multivariate analysis, a model incorporating the number and location of T cells expressing PMCH, NAMPT, and ETV1 showed prognostic significance for overall survival and for time to transformation. CONCLUSION: We showed altered gene expression in TILs in FL and demonstrated that altering the immune microenvironment in FL affects overall survival and time to transformation in this disease.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos do Interstício Tumoral/imunologia , Linfoma Folicular/genética , Linfoma Folicular/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/patologia , Movimento Celular/imunologia , Expressão Gênica , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Linfócitos do Interstício Tumoral/patologia , Linfoma Folicular/patologia , Receptores Acoplados a Proteínas G/biossíntese , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/imunologia , Receptores do Hormônio Hipofisário/biossíntese , Receptores do Hormônio Hipofisário/genética , Receptores do Hormônio Hipofisário/imunologia , Fatores de Risco , Análise de Sobrevida , Análise Serial de Tecidos , Microambiente Tumoral/imunologia , Regulação para Cima
7.
J Neuroendocrinol ; 25(6): 570-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23414264

RESUMO

Previous studies have shown that oestradiol (E2) decreases the orexigenic effect of melanin-concentrating hormone (MCH). In the present study, we examined whether this action of E2 is mediated by its ability to decrease the expression of MCH or its receptor (MCHR1). Using immunocytochemistry and western blotting, we examined whether E2 decreases MCH-immunoreactive neurones or MCHR1 protein content in the hypothalamus of female rats. We found that both MCH and MCHR1 protein expression was decreased by acute E2 treatment in ovariectomised rats, and by the peri-ovulatory increase in circulating E2 in pro-oestrous rats, relative to rats at other cycle stages. To determine whether these changes in MCH/MCHR1 protein expression may be mediated by E2's ability to directly regulate the transcription of MCH and MCHR1 genes, the effect of E2 treatment on MCH and MCHR1 mRNA expression in a neuronal hypothalamic cell line was examined using real-time reverse transcriptase-polymerase chain reaction. We also determined whether MCH and oestrogen receptor (ER)α are co-expressed in the hypothalamus of female rats. E2 treatment did not decrease MCH or MCHR1 mRNA expression in vitro, and no hypothalamic neurones were identified that co-expressed MCH and ERα. We conclude that E2-dependent decreases in hypothalamic MCH/MCHR1 protein expression mediate the ability of E2 to decrease MCH-induced feeding. The current findings suggest, however, that E2 exerts these actions indirectly, most likely though interactions with other neuronal systems that provide afferent input to MCH and MCHR1 neurones.


Assuntos
Estradiol/farmacologia , Hormônios Hipotalâmicos/metabolismo , Hipotálamo/efeitos dos fármacos , Melaninas/metabolismo , Hormônios Hipofisários/metabolismo , Receptores do Hormônio Hipofisário/metabolismo , Animais , Sequência de Bases , Primers do DNA , Receptor alfa de Estrogênio/fisiologia , Estro , Feminino , Hormônios Hipotalâmicos/genética , Hipotálamo/metabolismo , Melaninas/genética , Hormônios Hipofisários/genética , Ratos , Ratos Long-Evans , Reação em Cadeia da Polimerase em Tempo Real , Receptores do Hormônio Hipofisário/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Mol Biol Rep ; 39(12): 10823-32, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23053978

RESUMO

More than 99 % of follicles in mammalian ovaries undergo a degenerative process known as atresia, and thus only a limited number of ovarian follicles actually ovulate after full growth and development. The endocrinological regulatory mechanisms involved in follicular development have been studied extensively, but the precise and systematic molecular mechanisms of steroidogenesis enzymes involved in atresia are unclear. In the present study, we examined whether and how the steroidogenesis enzymes are involved in porcine ovary follicular atresia. Expression of steroidogenic acute regulatory protein, CYP11, CYP17, 3ß-hydroxysteroid dehydrogenase (3ß-HSD), CYP19, as well as related pituitary and ovarian hormone receptors were quantified in ovaries. During porcine follicular atresia, expressions of P450 cholesterol side chain cleavage enzyme, progesterone and androgen receptors increased significantly during the late atretic stage, while the expression of aromatase and follicle-stimulating hormone receptors decreased significantly in the early stage. These data suggested that the regulation of aromatase by follicle-stimulating hormone might induce follicular atresia, and that progesterone and androgen production further promoted follicular atresia. Additionally, a correlation analysis indicated a large and complex interactive network among these genes and the endocrinological microenvironment of the follicles. Significant correlations were observed between expression of steroidogenic enzymes and their receptors, and also between progesterone and 17ß-estradiol (E2) levels in follicular fluid. Taken together, these results suggest that CYP19 plays a role during early atresia by regulating the production of E2, whereas CYP11 and 3ß-HSD increase atresia progression by increasing progesterone levels.


Assuntos
Atresia Folicular/genética , Perfilação da Expressão Gênica , Estudos de Associação Genética , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Esteroides/biossíntese , Sus scrofa/genética , Animais , Vias Biossintéticas/genética , Feminino , Regulação da Expressão Gênica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Receptores do Hormônio Hipofisário/genética , Receptores do Hormônio Hipofisário/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
9.
Bioorg Med Chem Lett ; 22(11): 3781-5, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22542010

RESUMO

An initial SAR study resulted in the identification of the novel, potent MCHR1 antagonist 2. After further profiling, compound 2 was discovered to be a potent inhibitor of the hERG potassium channel, which prevented its further development. Additional optimization of this structure resulted in the discovery of the potent MCHR1 antagonist 11 with a dramatically reduced hERG liability. The decrease in hERG activity was confirmed by several in vivo preclinical cardiovascular studies examining QT prolongation. This compound demonstrated good selectivity for MCHR1 and possessed good pharmacokinetic properties across preclinical species. Compound 11 was also efficacious in reducing body weight in two in vivo mouse models. This compound was selected for clinical evaluation and was given the code AMG 076.


Assuntos
Carbazóis/química , Ácidos Cicloexanocarboxílicos/química , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Receptores do Hormônio Hipofisário/antagonistas & inibidores , Animais , Peso Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Carbazóis/síntese química , Carbazóis/farmacocinética , Ácidos Cicloexanocarboxílicos/síntese química , Ácidos Cicloexanocarboxílicos/farmacocinética , Dieta Hiperlipídica , Cães , Avaliação Pré-Clínica de Medicamentos , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Receptores do Hormônio Hipofisário/genética , Receptores do Hormônio Hipofisário/metabolismo , Relação Estrutura-Atividade
10.
Peptides ; 30(11): 2014-24, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19540893

RESUMO

Melanin-concentrating hormone (MCH) peptide plays a major role in energy homeostasis regulation. Little is known about cellular functions engaged by endogenous MCH receptor (MCH-R1). Here, MCH-R1 mRNA and cognate protein were found expressed in human neuroblastoma SH-SY5Y cells. Electrophysiological experiments demonstrated that MCH modulated K(+) currents, an effect depending upon the time of cellular growth. MCH treatments induced a transient phosphorylation of MAPKinases, abolished by PD98059, and partially blocked by PTX, suggesting a Galphai/Galphao protein contribution. MCH stimulated expression and likely nuclear localization of phosphorylated p53 proteins, an effect fully dependent upon MAPKinase activities. MCH treatment also increased phosphorylation of Elk-1 and up-regulated Egr-1, two transcriptional factors targeted by the MAPKinase pathway. Finally, MCH provoked neurite outgrowth after 24h-treatment of neuroblastoma cells. This effect and transcriptional factors activation were partly prevented by PD98059. Collectively, our results provide the first evidence for a role of MCH in neuronal differentiation of endogenously MCH-R1-expressing cells via non-exclusive MAPKinase and p53 signaling pathways.


Assuntos
Hormônios Hipotalâmicos/farmacologia , Melaninas/farmacologia , Neuritos/efeitos dos fármacos , Neuroblastoma/metabolismo , Hormônios Hipofisários/farmacologia , Transdução de Sinais/efeitos dos fármacos , Southern Blotting , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Flavonoides/farmacologia , Humanos , Imuno-Histoquímica , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Técnicas de Patch-Clamp , Fosforilação/efeitos dos fármacos , Receptores do Hormônio Hipofisário/genética , Receptores do Hormônio Hipofisário/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p53/metabolismo , Proteínas Elk-1 do Domínio ets/metabolismo
11.
Peptides ; 30(11): 2071-5, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19442695

RESUMO

To date, there is a dearth of evidence to support functions for melanin-concentrating hormone (MCH) and melanin-concentrating hormone receptors (MCH-R) in mammalian skin physiology including pigmentation, inflammation and immune responses and skin cell proliferation. Much research is therefore still needed to define the roles of the hormone and its receptors in mammalian skin. This will be a crucial step to identifying pathogenic mechanisms that may involve the MCH/MCH-R system in the context of inflammatory and autoimmune skin diseases as well as skin cancers. The following review summarizes the studies which have been carried out to examine the expression and function of MCH and MCH-R in mammalian skin. Recent findings with regard to humoral immune responses to the MCH-R1 in patients with the skin depigmenting disease vitiligo are also discussed.


Assuntos
Hormônios Hipotalâmicos/fisiologia , Mamíferos/metabolismo , Melaninas/fisiologia , Hormônios Hipofisários/fisiologia , Receptores do Hormônio Hipofisário/imunologia , Receptores do Hormônio Hipofisário/metabolismo , Pele/metabolismo , Pele/patologia , Animais , Humanos , Hormônios Hipotalâmicos/genética , Hormônios Hipotalâmicos/metabolismo , Melaninas/genética , Melaninas/metabolismo , Hormônios Hipofisários/genética , Hormônios Hipofisários/metabolismo , Receptores do Hormônio Hipofisário/genética , Pigmentação da Pele/genética , Pigmentação da Pele/fisiologia
12.
Domest Anim Endocrinol ; 37(1): 45-54, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19339131

RESUMO

The transformation of the dominant follicle into a functional corpus luteum is accompanied by a profound molecular and morphological reorganization of somatic cell layers. Several studies have focused on gene expression during early processes of follicular differentiation as it relates to recruitment and selection of dominant follicles. However, little information exists on changes of gene expression profiles in late preovulatory follicles. This lack of information is addressed here to elucidate molecular mechanisms behind the LH-induced transition from the large dominant estrogen-active to the preovulatory follicle, an intermediate stage toward full luteinization. Transcripts encoding key molecules for the biosynthesis of steroid hormones and prostaglandins, as well as receptors for gonadotropic and growth hormones (Star, Cyp11a1, Hsd3b, Cyp17, Cyp19, Ptgs2, Fshr, Lhr, and Ghr), were quantified by real-time polymerase chain reaction (PCR) in the granulosa and theca of large dominant and late preovulatory follicles. The steroid hormones progesterone (P4) and estradiol-17beta (E2) were monitored to distinguish estrogen-active and estrogen-inactive follicles. We found that (1) independent of the follicular stage, the gene expression profile was very different in granulosa and theca; (2) the abundance of several key transcripts was lower in estrogen-inactive, compared with estrogen-active, dominant follicles; (3) in the granulosa of late preovulatory follicles, transcripts encoding steroidogenic enzymes and hormone receptors were largely down-regulated, whereas (4) progesterone and E2 were found at high concentrations in the follicular fluid. Collectively, our data show that late preovulatory follicles have a transient and unique gene expression profile and are clearly different from both the preceding and subsequent (follicular and luteal, respectively) stages.


Assuntos
Bovinos/metabolismo , Regulação para Baixo/fisiologia , Folículo Ovariano/enzimologia , Folículo Ovariano/metabolismo , Receptores do Hormônio Hipofisário/genética , Esteroides/biossíntese , Animais , Ciclo-Oxigenase 2/genética , Sistema Enzimático do Citocromo P-450/genética , Estradiol/análise , Estradiol/biossíntese , Estradiol/genética , Feminino , Líquido Folicular/química , Perfilação da Expressão Gênica , Folículo Ovariano/anatomia & histologia , Reação em Cadeia da Polimerase , Progesterona/análise , Progesterona/biossíntese , Progesterona/genética , RNA Mensageiro/análise , Receptores da Gonadotropina/genética , Receptores da Somatotropina/genética
13.
J Dermatol Sci ; 52(3): 186-92, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18657399

RESUMO

BACKGROUND: Alpha-melanocyte-stimulating hormone receptor 1 (MC1R) has an important role in skin pigmentation and variants of the gene have been established as independent risk factors for susceptibility to cutaneous malignant melanoma. OBJECTIVE: To explore whether variants of the gene also influence the onset of the disease. METHODS: We analyzed 285 melanoma patients of European ancestry for common variation in codon 84 (D84E) of the alpha-MSH receptor 1 gene, which is known to have functional consequences in MC1R protein activity. RESULTS: The mean age difference at diagnosis between MC1R 84E carriers and non-carriers was 9 years (95% confidence interval [CI]: 2-17; p=0.012), with 84E non-carrier patients being older. After adjusting for gender, Clark's level, phototype, eyes and hair colour, the risk for cutaneous malignant melanoma at any age was 2.07 times higher (95% CI: 1.21-3.52; p=0.008) among MC1R 84E carriers. Enrolment criteria, geographical origin, Clark's levels and Breslow's indexes were similar between MC1R 84E carriers and non-carriers. Further analyses based on the Clark level and Breslow's index, both indicative for cancer invasion, reasonably supported an unbiased selection of patients during the study enrolment. Additional exon re-sequencing of the cyclin-dependent kinase inhibitor 2A (CDKN2A) gene in MC1R 84E carriers ruled out the presence of high penetrance mutations that have previously been associated with early onset of the disease. CONCLUSION: Although our findings need to be confirmed by independent and larger studies we have described for the first time the association of D84E variant of the alpha-MSH receptor 1 gene as an independent risk factor for an earlier onset of cutaneous malignant melanoma.


Assuntos
Melanoma/genética , Receptores do Hormônio Hipofisário/genética , Neoplasias Cutâneas/genética , Adulto , Idade de Início , Idoso , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Mutação
14.
Gene ; 412(1-2): 71-83, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18302976

RESUMO

The adenohypophysis of vertebrates receives peptide hormones from the hypothalamus and secretes hormones that regulate diverse physiologic processes in peripheral organs. The adenohypophysis-mediated endocrine system is widely conserved across vertebrates but not invertebrates. Phylogenetic analysis indicates that the emergence of this system coincided with two rounds of whole-genome duplication (2R-WGD) in early vertebrates, but direct evidence linking these events has been unavailable. We detected all human paralogons (series of paralogous regions) formed in early vertebrates as traces of 2R-WGD, and examined the relationship between 2R-WGD and the evolution of genes essential to the adenohypophysis-mediated endocrine system. Regarding genes encoding transcription factors (TFs) involved in the terminal differentiation into hormone-secreting cells in adenohypophyseal development, we showed that most pairs of these genes and their paralogs were part of paralogons. In addition, our analysis also indicated that most of the paralog pairs in families of adenohypophyseal hormones and their receptors were part of paralogons. These results suggest that 2R-WGD played an important role in generating genes encoding adenohypophyseal TFs, hormones, and their receptors for increasing the diversification of hormone repertoire in the adenohypophysis-mediated endocrine system of vertebrates.


Assuntos
Evolução Biológica , Adeno-Hipófise/fisiologia , Vertebrados/genética , Vertebrados/fisiologia , Animais , Mapeamento Cromossômico , Duplicação Gênica , Humanos , Modelos Genéticos , Filogenia , Adeno-Hipófise/crescimento & desenvolvimento , Hormônios Adeno-Hipofisários/genética , Hormônios Adeno-Hipofisários/fisiologia , Receptores do Hormônio Hipofisário/genética , Receptores do Hormônio Hipofisário/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Vertebrados/classificação , Vertebrados/crescimento & desenvolvimento
15.
Biol Reprod ; 75(1): 34-44, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16554413

RESUMO

Body growth during critical periods is known to be an important factor in determining the age of maturity and fecundity in fish. However, the endocrine mechanisms controlling oogenesis in fish and the effects of growth on this process are poorly understood. In this study interactions between the growth and reproductive systems were examined by monitoring changes in various components of the FSH-ovary axis, plasma insulin-like growth factor 1 (Igf1), and ovarian gene expression in relation to body and previtellogenic oocyte growth in coho salmon. Samples were collected from females during two hypothesized critical periods when growth influences maturation in this species. Body growth during the fall-spring months was strongly related to the degree of oocyte development, with larger fish possessing more advanced oocytes than smaller, slower growing fish. The accumulation of cortical alveoli in the oocytes was associated with increases in plasma and pituitary FSH, plasma estradiol-17beta, and ovarian steroidogenic acute regulatory protein (star) gene expression, whereas ovarian transcripts for growth hormone receptor and somatolactin receptor decreased. As oocytes accumulated lipid droplets, a general increase occurred in plasma Igf1 and components of the FSH-ovary axis, including plasma FSH, estradiol-17beta, and ovarian mRNAs for gonadotropin receptors, star, igf1, and igf2. A consistent positive relationship between plasma Igf1, estradiol-17beta, and pituitary FSH during growth in the spring suggests that these factors are important links in the mechanism by which body growth influences the rate of oocyte development.


Assuntos
Oncorhynchus kisutch/fisiologia , Oócitos/citologia , Ovário/crescimento & desenvolvimento , Animais , Estradiol/sangue , Feminino , Hormônio Foliculoestimulante/análise , Hormônio Foliculoestimulante/sangue , Fator de Crescimento Insulin-Like I/análise , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like II , Masculino , Oncorhynchus kisutch/crescimento & desenvolvimento , Oogênese , Ovário/anatomia & histologia , Ovário/fisiologia , Fosfoproteínas/genética , Proteínas/genética , Receptores do FSH/genética , Receptores do LH/genética , Receptores do Hormônio Hipofisário/genética , Receptores da Somatotropina/genética , Estações do Ano
16.
Eur J Endocrinol ; 152(6): 851-62, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15941924

RESUMO

OBJECTIVE: The importance of the melanin-concentrating hormone (MCH) system for regulation of energy homeostasis and body weight has been demonstrated in rodents. We analysed the human MCH receptor 1 gene (MCHR1) with respect to human obesity. DESIGN: This consisted of genomic screening of 13.4 kb encompassing the MCHR1 in extremely obese German children and adolescents and association analyses for two coding single nucleotide polymorphisms (SNPs). To confirm initial positive association results, additional association studies and transmission disequilibrium tests in further German, Danish, French and American samples were conducted. Selected SNPs were investigated using functional in vitro studies and reporter gene assays. METHODS: Single-stranded conformation polymorphism analysis, re-sequencing, PCR-restriction fragment length polymorphism analyses, tetra-primer amplification refractory mutation systems, matrix-assisted laser desorption/ionization time of flight mass spectrometry and reporter gene assays were carried out as well as measuring inositol phosphate formation, inhibition of cAMP formation and activation of p42/44 MAP kinase. RESULTS: We identified 11 infrequent variations and two SNPs in the MCHR1 coding sequence and 18 SNPs (eight novel) in the flanking sequence. Association and transmission disequilibrium with obesity were detected for several SNPs in independent study groups of German obese children and adolescents and controls. In two German samples, encompassing 4056 and 295 individuals, trends towards association with obesity were detected. Findings in a second epidemiological German sample and in Danish, French and American samples were negative. Functional in vitro studies as well as reporter gene assays revealed no significant results. CONCLUSION: Our initial association of MCHR1 alleles/haplotype detected might be related to juvenile-onset obesity, conditional on a particular genetic and/or environmental background. Alternatively, we could not exclude the possibility that the initially detected association represented a false positive finding.


Assuntos
Obesidade/genética , Receptores do Hormônio Hipofisário/genética , Adolescente , Adulto , Animais , Células COS , Chlorocebus aethiops , AMP Cíclico/antagonistas & inibidores , DNA/química , DNA/genética , Feminino , Humanos , Fosfatos de Inositol/metabolismo , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Reação em Cadeia da Polimerase , Polimorfismo de Fragmento de Restrição , Polimorfismo de Nucleotídeo Único , Polimorfismo Conformacional de Fita Simples , Análise de Sequência de DNA
17.
Endocrinology ; 146(5): 2354-61, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15718271

RESUMO

Somatolactin (SL) is a pituitary hormone of the GH/prolactin (PRL) family that so far has been found only in fish. Compared with GH and PRL, the primary structure of SL is highly conserved among divergent fish species, suggesting it has an important function and a discriminating receptor that constrains structural change. However, SL functions are poorly understood, and receptors for SL have not yet been identified. During cloning of GH receptor cDNA from salmon, we found a variant with relatively high (38-58%) sequence identity to vertebrate GH receptors and low (28-33%) identity to PRL receptors; however, the recombinant protein encoding the extracellular domain showed only weak binding of GH. Ligand binding of the recombinant extracellular domain for this receptor confirmed that the cDNA encoded a specific receptor for SL. The SL receptor (SLR) has common features of a GH receptor including FGEFS motif, six cysteine residues in the extracellular domain, a single transmembrane region, and Box 1 and 2 regions in the intracellular domain. These structural characteristics place the SLR in the cytokine receptor type I homodimeric group, which includes receptors for GH, PRL, erythropoietin, thrombopoietin, granulocyte-colony stimulating factor, and leptin. Transcripts for SLR were found in 11 tissues with highest levels in liver and fat, supporting the notion that a major function of SL is regulation of lipid metabolism. Cloning SLR cDNA opens the way for discovery of new SL functions and target tissues in fish, and perhaps novel members of this receptor family in other vertebrates.


Assuntos
Glicoproteínas , Oncorhynchus , Hormônios Hipofisários , Receptores do Hormônio Hipofisário/análise , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , DNA Complementar/química , DNA Complementar/genética , Escherichia coli , Feminino , Proteínas de Peixes , Expressão Gênica , Masculino , Dados de Sequência Molecular , Filogenia , Receptores do Hormônio Hipofisário/química , Receptores do Hormônio Hipofisário/genética , Receptores da Prolactina/química , Receptores da Somatotropina/química , Proteínas Recombinantes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência , Distribuição Tecidual
18.
J Biol Chem ; 279(49): 51226-33, 2004 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-15358786

RESUMO

Protection against UV-mediated DNA damage and the onset of oncogenesis is afforded by the tanning response in which UV irradiation triggers melanocytes to increase production of melanin that is then transferred to keratinocytes. A key component of the tanning process is the UV-mediated induction of the pro-opiomelanocortin (POMC) and MC1R genes encoding the alpha-melanocyte-stimulating hormone and its receptor, respectively, which play a crucial role in pigmentation by regulating the intracellular levels of cAMP. How these genes are regulated in response to UV irradiation is not known. Here we have shown that UV-induced activation of the POMC and MC1R promoters is mediated by p38 stress-activated kinase signaling to the transcription factor, upstream stimulating factor-1 (USF-1). Importantly, melanocytes derived from USF-1 -/- mice exhibit a defective UV response and fail to activate POMC and MC1R expression in response to UV irradiation. The results define USF-1 as a critical UV-responsive activator of genes implicated in protection from solar radiation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Pró-Opiomelanocortina/biossíntese , Receptores do Hormônio Hipofisário/biossíntese , Fatores de Transcrição/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Motivos de Aminoácidos , Animais , Western Blotting , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , DNA/metabolismo , Dano ao DNA , DNA Complementar/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Humanos , Luciferases/metabolismo , Melaninas/metabolismo , Melanócitos/metabolismo , Camundongos , Modelos Biológicos , Pigmentação , Pró-Opiomelanocortina/genética , Regiões Promotoras Genéticas , RNA/metabolismo , Receptores do Hormônio Hipofisário/genética , Transdução de Sinais , Espectrofotometria , Luz Solar , Fatores de Tempo , Transcrição Gênica , Transfecção , Transgenes , Raios Ultravioleta , Fatores Estimuladores Upstream
19.
J Neuroendocrinol ; 16(9): 758-66, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15344914

RESUMO

Ageing alters fundamental aspects of circadian rhythmicity in mammals; the effects include reduced rhythm amplitude and alterations in period length and in entrainment to the light/dark cycle. Such changes may reflect disruptions in cellular function within the suprachiasmatic nucleus (SCN), the site of the predominant circadian pacemaker. In the SCN, vasoactive intestinal peptide (VIP)-synthesizing neurones receive various inputs, including retinohypothalamic projections containing pituitary adenylate cyclase activating peptide (PACAP). SCN VIP cells establish connections with local neurones and send efferents beyond the nucleus. Considerable evidence implicates VIP and PACAP in circadian rhythm maintenance and/or entrainment to photic Zeitgebers. These actions involve members of a distinct family of receptors; mRNAs for two such receptors, VPAC2 and PAC1, are present in the SCN. This study used isotopic in situ hybridization to examine the effects of ageing on expression of mRNAs for VIP, VPAC2 and PAC1 in the SCN of male rats under a 12 : 12 h light/dark cycle. Analysis of film autoradiographs from young adult (2-3 months) or aged (19-20 months) rats, at eight time points across the light/dark cycle, showed loss of diurnal rhythmicity and reduced levels for VIP mRNA in the aged group. A diurnal rhythm of VPAC2 receptor mRNA was present in both groups, but its levels were reduced in the aged rats. There were no differences between the two groups for PAC1 receptor mRNA expression. The present results indicate that ageing reduces VIP and VPAC2 receptor mRNA and eliminates diurnal expression of VIP mRNA within the SCN of aged male rats.


Assuntos
Envelhecimento/fisiologia , Ritmo Circadiano/fisiologia , Receptores do Hormônio Hipofisário/metabolismo , Receptores de Peptídeo Intestinal Vasoativo/metabolismo , Núcleo Supraquiasmático/metabolismo , Peptídeo Intestinal Vasoativo/metabolismo , Animais , Masculino , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Receptores do Hormônio Hipofisário/genética , Receptores de Peptídeo Intestinal Vasoativo/genética , Receptores Tipo II de Peptídeo Intestinal Vasoativo
20.
J Neurosci Res ; 77(1): 26-34, 2004 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15197736

RESUMO

We have demonstrated previously in primary cultures of mouse cerebellar granule cells (CGCs) that endogenously synthesized pituitary adenylate cyclase-activating polypeptide (PACAP) contributes at least in part to the activity-dependent survival of CGCs (Tabuchi et al. [2001] Neurosci. Res. 39:85-93). In this study, we have demonstrated that expression of vasoactive intestinal polypeptide (VIP), a member of the same VIP/secretin/glucagon family as PACAP, was activated markedly by Ca(2+) influx through L-type voltage-dependent Ca(2+) channels (L-VDCCs), which could be induced under the depolarizing condition induced by high concentration of potassium (K(+)) in the medium. The activation of VIP mRNA expression, different from that of PACAP, was dependent partly on de novo protein synthesis. On the other hand, mRNA expression of secretin and PACAP/VIP receptors (PAC(1), VPAC(1), and VPAC(2)) was not activated by the Ca(2+) influx; rather, PAC(1) mRNA expression was reduced. Exogenously added VIP prevented apoptosis of CGCs under nondepolarizing conditions, the effect of which was mediated specifically through the VPAC(1) receptor. Furthermore, the survival of CGCs under depolarizing conditions could be mediated partly through VPAC(1), the contribution of which was much less than that of PAC(1). These findings indicate that PACAP and VIP genes are coordinately activated by the Ca(2+) signals in CGCs, but the contribution of VIP to the activity-dependent survival of CGCs is quite small.


Assuntos
Potenciais de Ação/genética , Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio/genética , Córtex Cerebelar/metabolismo , Neurônios/metabolismo , Peptídeo Intestinal Vasoativo/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo L/genética , Membrana Celular/efeitos dos fármacos , Membrana Celular/genética , Membrana Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Córtex Cerebelar/citologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Camundongos , Camundongos Endogâmicos ICR , Neurônios/citologia , Potássio/metabolismo , Potássio/farmacologia , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G , Receptores dos Hormônios Gastrointestinais/genética , Receptores dos Hormônios Gastrointestinais/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Receptores do Hormônio Hipofisário/genética , Receptores do Hormônio Hipofisário/metabolismo , Receptores de Peptídeo Intestinal Vasoativo/genética , Receptores de Peptídeo Intestinal Vasoativo/metabolismo , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo , Peptídeo Intestinal Vasoativo/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA