Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Endocrinol ; 53(3): 417-27, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25324488

RESUMO

During the development of diabetes ß-cells are exposed to elevated concentrations of proinflammatory cytokines, TNFα and IL1ß, which in vitro induce ß-cell death. The class B G-protein-coupled receptors (GPCRs): corticotropin-releasing factor receptor 1 (CRFR1) and CRFR2 are expressed in pancreatic islets. As downstream signaling by other class B GPCRs can protect against cytokine-induced ß-cell apoptosis, we evaluated the protective potential of CRFR activation in ß-cells in a pro-inflammatory setting. CRFR1/CRFR2 ligands activated AKT and CRFR1 signaling and reduced apoptosis in human islets. In rat and mouse insulin-secreting cell lines (INS-1 and MIN6), CRFR1 agonists upregulated insulin receptor substrate 2 (IRS2) expression, increased AKT activation, counteracted the cytokine-mediated decrease in BAD phosphorylation, and inhibited apoptosis. The anti-apoptotic signaling was dependent on prolonged exposure to corticotropin-releasing factor family peptides and followed PKA-mediated IRS2 upregulation. This indicates that CRFR signaling counteracts proinflammatory cytokine-mediated apoptotic pathways through upregulation of survival signaling in ß-cells. Interestingly, CRFR signaling also counteracted basal apoptosis in both cultured INS-1 cells and intact human islets.


Assuntos
Apoptose/efeitos dos fármacos , Hormônio Liberador da Corticotropina/farmacologia , Citocinas/efeitos adversos , Citoproteção/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Receptores de Hormônio Liberador da Corticotropina/agonistas , Animais , Morte Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Células Secretoras de Insulina/fisiologia , Interleucina-1beta/efeitos adversos , Camundongos , Ratos , Fator de Necrose Tumoral alfa/efeitos adversos
2.
Gen Comp Endocrinol ; 202: 69-75, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24769042

RESUMO

Corticotropin-releasing factor-binding protein (CRF-BP) is considered a key determinant for CRF receptor (CRF-R) activation by CRF and several related peptides. Earlier studies have shown that the CRF system is highly conserved in gene structures throughout evolution, yet little is known about the evolutionary conservation of its biological functions. Therefore, we address the functional properties of CRF-BP and CRF-Rs in a teleost fish (common carp; Cyprinus carpio L.). We report the finding of two similar, yet distinct, genes for both CRF-R1 and CRF-R2 in this species. The four receptors are differentially responsive to CRF, urotensin-I (UI), sauvagine, and urocortin-2 (Ucn-2) and -3 (Ucn-3) as shown by luciferase assays. In vitro, carp CRF-BP inhibits CRF- and UI-mediated activation of the newfound CRF-Rs, but its potency to do so varies between receptor and peptide ligand. This is the first paper to establish the functionality and physiological interplay between CRF-BP, CRF-Rs and CRF-family peptides in a teleostean species.


Assuntos
Carpas/metabolismo , Proteínas de Transporte/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Urotensinas/metabolismo , Proteínas de Anfíbios/metabolismo , Animais , AMP Cíclico/farmacologia , Células HEK293 , Humanos , Luciferases/metabolismo , Hormônios Peptídicos/metabolismo , Isoformas de Proteínas/metabolismo , Receptores de Hormônio Liberador da Corticotropina/genética , Proteínas Recombinantes/metabolismo , Urocortinas/metabolismo
3.
PLoS One ; 9(1): e84013, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24465390

RESUMO

Corticotropin-Releasing Factor Receptors (CRFRs) are class B1 G-protein-coupled receptors, which bind peptides of the corticotropin releasing factor family and are key mediators in the stress response. In order to dissect the receptors' binding specificity and enable structural studies, full-length human CRFR1α and mouse CRFR2ß as well as fragments lacking the N-terminal extracellular domain, were overproduced in E. coli. The characteristics of different CRFR2ß-PhoA gene fusion products expressed in bacteria were found to be in agreement with the predicted ones in the hepta-helical membrane topology model. Recombinant histidine-tagged CRFR1α and CRFR2ß expression levels and bacterial subcellular localization were evaluated by cell fractionation and Western blot analysis. Protein expression parameters were assessed, including the influence of E. coli bacterial hosts, culture media and the impact of either PelB or DsbA signal peptide. In general, the large majority of receptor proteins became inserted in the bacterial membrane. Across all experimental conditions significantly more CRFR2ß product was obtained in comparison to CRFR1α. Following a detergent screen analysis, bacterial membranes containing CRFR1α and CRFR2ß were best solubilized with the zwitterionic detergent FC-14. Binding of different peptide ligands to CRFR1α and CRFR2ß membrane fractions were similar, in part, to the complex pharmacology observed in eukaryotic cells. We suggest that our E. coli expression system producing functional CRFRs will be useful for large-scale expression of these receptors for structural studies.


Assuntos
Membrana Celular/metabolismo , Escherichia coli/metabolismo , Mamíferos/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Proteínas de Anfíbios/metabolismo , Animais , Western Blotting , Membrana Celular/efeitos dos fármacos , Hormônio Liberador da Corticotropina/metabolismo , Meios de Cultura/farmacologia , Detergentes/farmacologia , Vetores Genéticos , Humanos , Cinética , Ligantes , Camundongos , Fragmentos de Peptídeos/metabolismo , Hormônios Peptídicos/metabolismo , Ligação Proteica/efeitos dos fármacos , Sinais Direcionadores de Proteínas , Estrutura Terciária de Proteína , Receptores de Hormônio Liberador da Corticotropina/química , Proteínas Recombinantes de Fusão/metabolismo , Solubilidade
4.
Obesity (Silver Spring) ; 22(2): 380-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23804489

RESUMO

OBJECTIVE: Ghrelin is known to regulate appetite control and cellular metabolism. The corticotropin-releasing factor (CRF) family is also known to regulate energy balance. In this study, the links between ghrelin and the CRF family in C2C12 cells, a mouse myoblast cell line was investigated. DESIGN AND METHODS: C2C12 cells were treated with ghrelin in the presence or absence of CRF receptor antagonists and then subjected to different metabolic analyses. RESULTS: Ghrelin enhanced glucose uptake by C2C12 cells, induced GLUT4 translocation to the cell surface and decreased RBP4 expression. A CRF-R2 selective antagonist, anti-sauvagine-30, blocked ghrelin-induced glucose uptake, Ghrelin upregulated CRF-R2 but not CRF-R1 levels. Moreover, ghrelin-treated C2C12 cells displayed a cAMP and pERK activation in response to Ucn3, a CRF-R2 specific ligand, but not in response to CRF or stressin, CRF-R1 specific ligands. Ghrelin also induced UCP2 and UCP3 expression, which were blocked by anti- sauvagine-30. Ghrelin did not induce fatty acids uptake by C2C12 cells or ACC expression. Even though C2C12 cells clearly exhibited responses to ghrelin, the known ghrelin receptor, GHSR1a, was not detectable in C2C12 cells. CONCLUSION: The results suggest that, ghrelin plays a role in regulating muscle glucose and, raise the possibility that suppression of the CRF-R2 pathway might provide benefits in high ghrelin states.


Assuntos
Regulação da Expressão Gênica , Grelina/metabolismo , Glucose/metabolismo , Mioblastos/metabolismo , Receptores de Hormônio Liberador da Corticotropina/agonistas , Receptores de Grelina/metabolismo , Transdução de Sinais , Animais , Anticorpos Bloqueadores/farmacologia , Transporte Biológico/efeitos dos fármacos , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Regulação da Expressão Gênica/efeitos dos fármacos , Transportador de Glucose Tipo 4/metabolismo , Canais Iônicos/agonistas , Canais Iônicos/genética , Canais Iônicos/metabolismo , Camundongos , Proteínas Mitocondriais/agonistas , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Mioblastos/efeitos dos fármacos , Mioblastos/ultraestrutura , Transporte Proteico/efeitos dos fármacos , Pirimidinas/farmacologia , Pirróis/farmacologia , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Receptores de Hormônio Liberador da Corticotropina/genética , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Receptores de Grelina/genética , Proteínas Plasmáticas de Ligação ao Retinol/antagonistas & inibidores , Proteínas Plasmáticas de Ligação ao Retinol/genética , Proteínas Plasmáticas de Ligação ao Retinol/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Desacopladora 2 , Proteína Desacopladora 3 , Urocortinas/metabolismo
5.
J Biol Chem ; 288(32): 23128-40, 2013 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-23788641

RESUMO

Recent studies have shown that the pyruvate-isocitrate cycling pathway, involving the mitochondrial citrate/isocitrate carrier and the cytosolic NADP-dependent isocitrate dehydrogenase (ICDc), is involved in control of glucose-stimulated insulin secretion (GSIS). Here we demonstrate that pyruvate-isocitrate cycling regulates expression of the voltage-gated potassium channel family member Kv2.2 in islet ß-cells. siRNA-mediated suppression of ICDc, citrate/isocitrate carrier, or Kv2.2 expression impaired GSIS, and the effect of ICDc knockdown was rescued by re-expression of Kv2.2. Moreover, chronic exposure of ß-cells to elevated fatty acids, which impairs GSIS, resulted in decreased expression of Kv2.2. Surprisingly, knockdown of ICDc or Kv2.2 increased rather than decreased outward K(+) current in the 832/13 ß-cell line. Immunoprecipitation studies demonstrated interaction of Kv2.1 and Kv2.2, and co-overexpression of the two channels reduced outward K(+) current compared with overexpression of Kv2.1 alone. Also, siRNA-mediated knockdown of ICDc enhanced the suppressive effect of the Kv2.1-selective inhibitor stromatoxin1 on K(+) currents. Our data support a model in which a key function of the pyruvate-isocitrate cycle is to maintain levels of Kv2.2 expression sufficient to allow it to serve as a negative regulator of Kv channel activity.


Assuntos
Regulação da Expressão Gênica/fisiologia , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Isocitratos/metabolismo , Ácido Pirúvico/metabolismo , Canais de Potássio Shab/biossíntese , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/genética , Secreção de Insulina , Células Secretoras de Insulina/citologia , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/fisiologia , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/metabolismo , Masculino , Modelos Biológicos , Peptídeos/farmacologia , Potássio/metabolismo , Ratos , Ratos Sprague-Dawley , Canais de Potássio Shab/antagonistas & inibidores , Canais de Potássio Shab/genética , Venenos de Aranha/farmacologia
6.
Endocrinology ; 154(4): 1553-64, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23493376

RESUMO

Mouse (m) and human (h) urocortin 2 (Ucn 2) were identified by molecular cloning strategies and the primary sequence of their mature forms postulated by analogy to closely related members of the corticotropin-releasing factor (CRF) neuropeptide family. Because of the paucity of Ucn 2 proteins in native tissues, skin, muscle, and pancreatic cell lines were transduced with lentiviral constructs and secretion media were used to isolate and characterize Ucn 2 products and study processing. Primary structures were assigned using a combination of Edman degradation sequencing and mass spectrometry. For mUcn 2, transduced cells secreted a 39 amino acid peptide and the glycosylated prohormone lacking signal peptide; both forms were C-terminally amidated and highly potent to activate the type 2 CRF receptor. Chromatographic profiles of murine tissue extracts were consistent with cleavage of mUcn 2 prohormone to a peptidic form. By contrast to mUcn 2, mammalian cell lines transduced with hUcn 2 constructs secreted significant amounts of an 88 amino acid glycosylated hUcn 2 prohormone but were unable to further process this molecule. Similarly, WM-266-4 melanoma cells that express endogenous hUcn 2 secreted only the glycosylated prohormone lacking the signal peptide and unmodified at the C terminus. Although not amidated, hUcn 2 prohormone purified from overexpressing lines activated CRF receptor 2. Hypoxia and glycosylation, paradigms that might influence secretion or processing of gene products, did not significantly impact hUcn 2 prohormone cleavage. Our findings identify probable Ucn 2 processing products and should expedite the characterization of these proteins in mammalian tissues.


Assuntos
Hormônio Liberador da Corticotropina , Processamento de Proteína Pós-Traducional , Urocortinas , Sequência de Aminoácidos , Animais , Hipóxia Celular , Células Cultivadas , Hormônio Liberador da Corticotropina/química , Hormônio Liberador da Corticotropina/metabolismo , Glicosilação , Humanos , Camundongos , Dados de Sequência Molecular , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Transdução Genética , Urocortinas/química , Urocortinas/metabolismo
7.
Psychoneuroendocrinology ; 38(8): 1349-61, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23267723

RESUMO

We have previously demonstrated that viral-mediated overexpression of corticotropin-releasing factor (CRF) within the central nucleus of the amygdala (CeA) reproduces many of the behavioral and endocrine consequences of chronic stress. The present experiment sought to determine whether administration of the selective serotonin reuptake inhibitor (SSRI) escitalopram reverses the adverse effects of CeA CRF overexpression. In a 2×2 design, adult male rats received bilateral infusions of a control lentivirus or a lentivirus in which a portion of the CRF promoter is used to drive increased expression of CRF peptide. Four weeks later, rats were then implanted with an Alzet minipump to deliver vehicle or 10mg/kg/day escitalopram for a 4-week period of time. The defensive withdrawal (DW) test of anxiety and the sucrose-preference test (SPT) of anhedonia were performed both before and after pump implantation. Additional post-implant behavioral tests included the elevated plus maze (EPM) and social interaction (SI) test. Following completion of behavioral testing, the dexamethasone/CRF test was performed to assess HPA axis reactivity. Brains were collected and expression of HPA axis-relevant transcripts were measured using in situ hybridization. Amygdalar CRF overexpression increased anxiety-like behavior in the DW test at week eight, which was only partially prevented by escitalopram. In both CRF-overexpressing and control groups, escitalopram decreased hippocampal CRF expression while increasing hypothalamic and hippocampal expression of the glucocorticoid receptor (GR). These gene expression changes were associated with a significant decrease in HPA axis reactivity in rats treated with escitalopram. Interestingly, escitalopram increased the rate of weight gain only in rats overexpressing CRF. Overall these data support our hypothesis that amygdalar CRF is critical in anxiety-like behavior; because the antidepressant was unable to reverse behavioral manifestations of CeA CRF-OE. This may be a potential animal model to study treatment-resistant psychopathologies.


Assuntos
Tonsila do Cerebelo/metabolismo , Citalopram/farmacologia , Hormônio Liberador da Corticotropina/metabolismo , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Ansiedade/genética , Ansiedade/metabolismo , Comportamento Animal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Peso Corporal/genética , Hormônio Liberador da Corticotropina/genética , Dexametasona , Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Masculino , Testes de Função Adreno-Hipofisária/métodos , Sistema Hipófise-Suprarrenal/metabolismo , Ratos , Receptores de Glucocorticoides/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia
8.
PLoS One ; 7(12): e52181, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23251699

RESUMO

The peptide hormone Urocortin 3 (Ucn 3) is abundantly and exclusively expressed in mouse pancreatic beta cells where it regulates insulin secretion. Here we demonstrate that Ucn 3 first appears at embryonic day (E) 17.5 and, from approximately postnatal day (p) 7 and onwards throughout adult life, becomes a unifying and exclusive feature of mouse beta cells. These observations identify Ucn 3 as a potential beta cell maturation marker. To determine whether Ucn 3 is similarly restricted to beta cells in humans, we conducted comprehensive immunohistochemistry and gene expression experiments on macaque and human pancreas and sorted primary human islet cells. This revealed that Ucn 3 is not restricted to the beta cell lineage in primates, but is also expressed in alpha cells. To substantiate these findings, we analyzed human embryonic stem cell (hESC)-derived pancreatic endoderm that differentiates into mature endocrine cells upon engraftment in mice. Ucn 3 expression in hESC-derived grafts increased robustly upon differentiation into mature endocrine cells and localized to both alpha and beta cells. Collectively, these observations confirm that Ucn 3 is expressed in adult beta cells in both mouse and human and appears late in beta cell differentiation. Expression of Pdx1, Nkx6.1 and PC1/3 in hESC-derived Ucn 3(+) beta cells supports this. However, the expression of Ucn 3 in primary and hESC-derived alpha cells demonstrates that human Ucn 3 is not exclusive to the beta cell lineage but is a general marker for both the alpha and beta cell lineages. Ucn 3(+) hESC-derived alpha cells do not express Nkx6.1, Pdx1 or PC1/3 in agreement with the presence of a separate population of Ucn 3(+) alpha cells. Our study highlights important species differences in Ucn 3 expression, which have implications for its utility as a marker to identify mature beta cells in (re)programming strategies.


Assuntos
Células-Tronco Embrionárias/metabolismo , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Insulina/metabolismo , Urocortinas/metabolismo , Animais , Diferenciação Celular/genética , Linhagem da Célula , Células-Tronco Embrionárias/citologia , Expressão Gênica , Células Secretoras de Glucagon/citologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica/métodos , Células Secretoras de Insulina/citologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Transativadores/genética , Transativadores/metabolismo , Urocortinas/genética
9.
Biol Psychiatry ; 72(6): 437-47, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22704666

RESUMO

BACKGROUND: The corticotropin-releasing factor type 2 receptor (CRFR2) is suggested to play an important role in aiding recovery from acute stress, but any chronic effects of CRFR2 activation are unknown. CRFR2 in the midbrain raphé nuclei modulate serotonergic activity of this key source of serotonin (5-HT) forebrain innervation. METHODS: Transgenic mice overexpressing the highly specific CRFR2 ligand urocortin 3 (UCN3OE) were analyzed for stress-related behaviors and hypothalamic-pituitary-adrenal axis responses. Responses to 5-HT receptor agonist challenge were assessed by local cerebral glucose utilization, while 5-HT and 5-hydroxyindoleacetic acid content were quantified in limbic brain regions. RESULTS: Mice overexpressing urocortin 3 exhibited increased stress-related behaviors under basal conditions and impaired retention of spatial memory compared with control mice. Following acute stress, unlike control mice, they exhibited no further increase in these stress-related behaviors and showed an attenuated adrenocorticotropic hormone response. 5-HT and 5-hydroxyindoleacetic acid content of limbic nuclei were differentially regulated by stress in UCN3OE mice as compared with control mice. Responses to 5-HT type 1A receptor challenge were significantly and specifically reduced in UCN3OE mice. The distribution pattern of local cerebral glucose utilization and 5-HT type 1A receptor messenger RNA expression levels suggested this effect was mediated in the raphé nuclei. CONCLUSIONS: Chronic activation of CRFR2 promotes an anxiety-like state, yet with attenuated behavioral and hypothalamic-pituitary-adrenal axis responses to stress. This is reminiscent of stress-related atypical psychiatric syndromes such as posttraumatic stress disorder, chronic fatigue, and chronic pain states. This new understanding indicates CRFR2 antagonism as a potential novel therapeutic target for such disorders.


Assuntos
Ansiedade/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Receptor 5-HT1A de Serotonina/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Agonistas do Receptor 5-HT1 de Serotonina/metabolismo , Urocortinas/genética , Análise de Variância , Animais , Ansiedade/genética , Encéfalo/metabolismo , Cromatografia Líquida , Corticosterona/metabolismo , Ácido Hidroxi-Indolacético/análise , Hibridização In Situ , Camundongos , Camundongos Transgênicos , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptor 5-HT1A de Serotonina/genética , Receptores de Hormônio Liberador da Corticotropina/genética , Serotonina/análise , Estresse Fisiológico , Estresse Psicológico , Urocortinas/metabolismo
10.
Mol Cell Endocrinol ; 359(1-2): 43-52, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22330643

RESUMO

Activins are multifunctional proteins and members of the TGF-ß superfamily. Activins are expressed locally in most tissues and, analogous to the actions of other members of this large family of pleiotropic factors, play prominent roles in the regulation of diverse biological processes in both differentiated and embryonic stem cells. They have an essential role in maintaining tissue homeostasis in the adult and are known to contribute to the developmental programs in the embryo. Activins are further implicated in the growth and metastasis of tumor cells. Through distinct modes of action, inhibins and follistatins function as antagonists of activin and several other TGF-ß family members, including a subset of BMPs/GDFs, and modulate cellular responses and the signaling cascades downstream of these ligands. In the pituitary, the activin pathway is known to regulate key aspects of gonadotrope functions and also exert effects on other pituitary cell types. As in other tissues, activin is produced locally by pituitary cells and acts locally by exerting cell-type specific actions on gonadotropes. These local actions of activin on gonadotropes are modulated by the autocrine/paracrine actions of locally secreted follistatin and by the feedback actions of gonadal inhibin. Knowledge about the mechanism of activin, inhibin and follistatin actions is providing information about their importance for pituitary function as well as their contribution to the pathophysiology of pituitary adenomas. The aim of this review is to highlight recent findings and summarize the evidence that supports the important functions of activin, inhibin and follistatin in the pituitary.


Assuntos
Ativinas/fisiologia , Folistatina/fisiologia , Gonadotrofos/metabolismo , Inibinas/fisiologia , Ativinas/metabolismo , Animais , Folistatina/metabolismo , Proteína Forkhead Box L2 , Fatores de Transcrição Forkhead/metabolismo , Fatores de Transcrição Forkhead/fisiologia , Humanos , Inibinas/metabolismo , Hipófise/citologia , Hipófise/metabolismo , Transdução de Sinais , Proteínas da Superfamília de TGF-beta/metabolismo , Proteínas da Superfamília de TGF-beta/fisiologia
11.
Open Neuroendocrinol J ; 4: 90-101, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21927629

RESUMO

The pituitary gland plays a prominent role in the control of many physiological processes. This control is achieved through the actions and interactions of hormones and growth factors that are produced and secreted by the endocrine cell types and the non-endocrine constituents that collectively and functionally define this complex organ. The five endocrine cell types of the anterior lobe of the pituitary, somatotropes, lactotropes, corticotropes, thyrotropes and gonadotropes, are defined by their primary product, growth hormone (GH), prolactin (PRL), adrenocorticotropic hormone (ACTH), thyroid-stimulating hormone (TSH) and follicle stimulating hormone (FSH)/luteinizing hormone (LH). They are further distinguishable by the presence of cell surface receptors that display high affinity and selectivity for specific hypothalamic hormones and couple to appropriate downstream signaling pathways involved in the control of cell type specific responses, including the release and/or synthesis of pituitary hormones. Central control of the pituitary via the hypothalamus is further fine-tuned by the positive or negative actions of peripheral feedback signals and of a variety of factors that originate from sources within the pituitary. The focus of this review is the latter category of intrinsic factors that exert local control. Special emphasis is given to the TGF-ß family of growth factors, in particular activin effects on the gonadotrope population, because a considerable body of evidence supports their contribution to the local modulation of the embryonic and postnatal pituitary as well as pituitary pathogenesis. A number of other substances, including members of the cytokine and FGF families, VEGF, IGF1, PACAP, Ghrelin, adenosine and nitric oxide have also been shown or implicated to function as autocrine/paracrine factors, though, definitive proof remains lacking in some cases. The ever-growing list of putative autocrine/paracrine factors of the pituitary nevertheless has highlighted the complexity of the local network and its impact on pituitary functions.

12.
Angew Chem Int Ed Engl ; 50(35): 8077-81, 2011 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-21751313

RESUMO

Capturing the right ligand at the right spot: a well-balanced system for non-natural amino acid mutagenesis allows the ligand binding sites of a class II G-protein coupled receptor to be mapped and distinct binding domains to be identified for different ligands in the native environment of mammalian cells.


Assuntos
Aminoácidos/química , Ligantes , Receptores Acoplados a Proteínas G/química , Animais , Sítios de Ligação , Células HEK293 , Humanos , Mutagênese Sítio-Dirigida , Marcadores de Fotoafinidade , Ratos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
13.
Vet J ; 188(3): 318-24, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20554462

RESUMO

Urocortin (Ucn) peptides are the endogenous ligands for the corticotropin-releasing factor type 2 receptor (CRFR2). They have potentially important roles in cardiovascular physiology in health and disease, and show promise as therapeutics for congestive heart failure. Analysis of canine heart tissue showed mRNA expression of Ucn 1, Ucn 3 and CRFR2 in all heart chambers. Immunohistochemistry also demonstrated Ucns 1 and 3 expression in cardiomyocytes. To assess the potential usefulness of circulating Ucns as markers of heart disease, plasma samples from 45 dogs with cardiac disease and 15 controls were analysed by radioimmunoassay. Both Ucns 1 and 3 were measurable but the presence of cardiac disease did not alter their concentrations. Therefore, whilst Ucns are expressed in canine myocardium (where they may play a role in the endogenous neurohumoral response to cardiac disease or failure) they do not appear to be sensitive biomarkers of cardiac disease in our canine patient population.


Assuntos
Doenças Cardiovasculares/veterinária , Doenças do Cão/metabolismo , Miocárdio/metabolismo , Urocortinas/metabolismo , Animais , Biomarcadores/sangue , Biomarcadores/metabolismo , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/metabolismo , Estudos de Casos e Controles , Doenças do Cão/sangue , Cães , Feminino , Masculino , Miócitos Cardíacos/metabolismo , RNA Mensageiro/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Urocortinas/sangue
14.
Proc Natl Acad Sci U S A ; 107(44): 19020-5, 2010 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-20937857

RESUMO

Responding to stressful events requires numerous adaptive actions involving integrated changes in the central nervous and neuroendocrine systems. Numerous studies have implicated dysregulation of stress-response mechanisms in the etiology of stress-induced psychopathophysiologies. The urocortin neuropeptides are members of the corticotropin-releasing factor family and are associated with the central stress response. In the current study, a triple-knockout (tKO) mouse model lacking all three urocortin genes was generated. Intriguingly, these urocortin tKO mice exhibit increased anxiety-like behaviors 24 h following stress exposure but not under unstressed conditions or immediately following exposure to acute stress. The inability of these mutants to recover properly from the exposure to an acute stress was associated with robust alterations in the expression profile of amygdalar genes and with dysregulated serotonergic function in stress-related neurocircuits. These findings position the urocortins as essential factors in the stress-recovery process and suggest the tKO mouse line as a useful stress-sensitive mouse model.


Assuntos
Transtornos de Ansiedade/genética , Comportamento Animal , Modelos Animais de Doenças , Estresse Psicológico/genética , Urocortinas , Animais , Camundongos , Camundongos Knockout
15.
J Biol Chem ; 285(49): 38580-9, 2010 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-20843795

RESUMO

The corticotropin-releasing factor (CRF) peptide hormone family members coordinate endocrine, behavioral, autonomic, and metabolic responses to stress and play important roles within the cardiovascular, gastrointestinal, and central nervous systems, among others. The actions of the peptides are mediated by activation of two G-protein-coupled receptors of the B1 family, CRF receptors 1 and 2 (CRF-R1 and CRF-R2α,ß). The recently reported three-dimensional structures of the first extracellular domain (ECD1) of both CRF-R1 and CRF-R2ß (Pioszak, A. A., Parker, N. R., Suino-Powell, K., and Xu, H. E. (2008) J. Biol. Chem. 283, 32900-32912; Grace, C. R., Perrin, M. H., Gulyas, J., Digruccio, M. R., Cantle, J. P., Rivier, J. E., Vale, W. W., and Riek, R. (2007) Proc. Natl. Acad. Sci. U.S.A. 104, 4858-4863) complexed with peptide antagonists provided a starting point in understanding the binding between CRF ligands and receptors at a molecular level. We now report the three-dimensional NMR structure of the ECD1 of human CRF-R1 complexed with a high affinity agonist, α-helical cyclic CRF. In the structure of the complex, the C-terminal residues (23-41) of α-helical cyclic CRF bind to the ECD1 of CRF-R1 in a helical conformation mainly along the hydrophobic face of the peptide in a manner similar to that of the antagonists in their corresponding ECD1 complex structures. Unique to this study is the observation that complex formation between an agonist and the ECD1-CRF-R1 promotes the helical conformation of the N terminus of the former, important for receptor activation (Gulyas, J., Rivier, C., Perrin, M., Koerber, S. C., Sutton, S., Corrigan, A., Lahrichi, S. L., Craig, A. G., Vale, W., and Rivier, J. (1995) Proc. Natl. Acad. Sci. U.S.A. 92, 10575-10579).


Assuntos
Hormônio Liberador da Corticotropina/química , Receptores de Hormônio Liberador da Corticotropina/agonistas , Receptores de Hormônio Liberador da Corticotropina/química , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Humanos , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptores de Hormônio Liberador da Corticotropina/genética , Receptores de Hormônio Liberador da Corticotropina/metabolismo
16.
Eur J Heart Fail ; 12(8): 797-804, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20388649

RESUMO

AIMS: To test acute effects of the corticotropin-releasing factor-related peptide urocortin 2 (Ucn2) on left ventricular (LV) function and the propensity for ventricular arrhythmias in the isolated heart of an animal model of hypertension-induced heart failure. METHODS AND RESULTS: Hearts from Dahl salt-sensitive rats with severe LV dysfunction were perfused according to Langendorff. Left ventricular developed pressure and the positive and negative derivatives of LV pressure were analysed before and after perfusion with Ucn2 (n = 15) or normal perfusion solution (control, n = 9). Intracellular calcium cycling parameters were assessed by surface fluorometry. Furthermore, monophasic action potential duration (MAPD) and ventricular fibrillation threshold (VFT) were determined, the latter by a train-of-pulses method at increasing voltage to scan the vulnerable period of repolarization. Urocortin 2 significantly affected intracellular calcium cycling and improved LV contractile function and relaxation. Compared with baseline values, Ucn2 significantly decreased MAPD at 30, 50, and 90% repolarization and significantly increased VFT compared with baseline values. No changes were observed in control experiments. CONCLUSION: Administration of Ucn2 rapidly improves LV function and increases VF threshold in failing, isolated rat hearts with increased propensity for ventricular arrhythmias. These observations suggest a potential use of Ucn2 as a safe and novel agent for the treatment of acute heart failure.


Assuntos
Arritmias Cardíacas/fisiopatologia , Insuficiência Cardíaca/fisiopatologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Miocárdio/patologia , Urocortinas/farmacologia , Doença Aguda , Animais , Modelos Animais de Doenças , Fluorometria , Insuficiência Cardíaca/diagnóstico por imagem , Hemodinâmica , Hipertrofia Ventricular Esquerda/diagnóstico por imagem , Proteínas Sensoras de Cálcio Intracelular , Masculino , Perfusão , Ratos , Ratos Endogâmicos Dahl , Sódio na Dieta , Ultrassonografia , Urocortinas/análise , Urocortinas/uso terapêutico , Disfunção Ventricular Esquerda/fisiopatologia , Fibrilação Ventricular/diagnóstico por imagem , Fibrilação Ventricular/fisiopatologia
17.
Proc Natl Acad Sci U S A ; 107(2): 912-7, 2010 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-20080775

RESUMO

Corticotropin-releasing factor (CRF), originally characterized as the principal neuroregulator of the hypothalamus-pituitary-adrenal axis, has broad central and peripheral distribution and actions. We demonstrate the presence of CRF receptor type 1 (CRFR1) on primary beta cells and show that activation of pancreatic CRFR1 promotes insulin secretion, thus contributing to the restoration of normoglycemic equilibrium. Stimulation of pancreatic CRFR1 initiates a cAMP response that promotes insulin secretion in vitro and in vivo and leads to the phosphorylation of cAMP response element binding and the induction of the expression of several immediate-early genes. Thus, the insulinotropic actions of pancreatic CRFR1 oppose the activation of CRFR1 on anterior pituitary corticotropes, leading to the release of glucocorticoids that functionally antagonize the actions of insulin. Stimulation of the MIN6 insulinoma line and primary rat islets with CRF also activates the MAPK signaling cascade leading to rapid phosphorylation of Erk1/2 in response to CRFR1-selective ligands, which induce proliferation in primary rat neonatal beta cells. Importantly, CRFR1 stimulates insulin secretion only during conditions of intermediate to high ambient glucose, and the CRFR1-dependent phosphorylation of Erk1/2 is greater with elevated glucose concentrations. This response is reminiscent of the actions of the incretins, which potentiate insulin secretion only during elevated glucose conditions. The presence of CRFR1 on beta cells adds another layer of complexity to the intricate network of paracrine and autocrine factors and their cognate receptors whose coordinated efforts can dictate islet hormone output and regulate beta cell proliferation.


Assuntos
Glucose/farmacologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/fisiologia , Insulina/metabolismo , Receptores de Hormônio Liberador da Corticotropina/genética , Adrenalectomia , Animais , Divisão Celular , Linhagem Celular Tumoral , AMP Cíclico/metabolismo , DNA Complementar/genética , Citometria de Fluxo , Teste de Tolerância a Glucose , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Insulinoma , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Período Pós-Prandial , Ratos , Receptores de Hormônio Liberador da Corticotropina/deficiência
18.
Hypertension ; 53(4): 739-44, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19204182

RESUMO

Recently, novel corticotropin-releasing factor-related peptides, named urocortin 1, 2, and 3, and a distinct cardiac and peripheral vascular receptor (corticotropin-releasing factor receptor 2) were described being part of a peripheral corticotropin-releasing factor system modulating cardiovascular function in response to stress. Vasorelaxation and blood pressure lowering have been reported after acute administration of these peptides. No data are available on the acute and chronic effects of urocortin 2 on blood pressure in models of arterial hypertension. To test these effects, hypertensive salt-sensitive and normotensive salt-resistant Dahl rats were randomly assigned to twice-daily applications of urocortin 2 or vehicle for 5 weeks. Blood pressure, heart rate, and left ventricular dimension and function were recorded at baseline, after initial application, and, together with cardiac and aortic expression of urocortin 2 and its receptor, after 5 weeks of treatment. Urocortin 2 significantly reduced blood pressure in hypertensive rats without affecting heart rate. Long-term urocortin 2 treatment in hypertensive rats induced sustained blood pressure reduction and diminished the development of hypertension-induced left ventricular hypertrophy and the deterioration of left ventricular contractile function. Corticotropin-releasing factor receptor 2 expression was preserved despite chronic stimulation by urocortin 2. In conclusion, our study shows that, in an animal model of arterial hypertension, urocortin 2 has immediate and sustained blood pressure-lowering effects. Beneficial effects on blood pressure, left ventricular dimension, and function, together with preserved receptor expression, suggest that corticotropin-releasing factor receptor 2 stimulation by urocortin 2 may represent a novel approach to the treatment of arterial hypertension.


Assuntos
Anti-Hipertensivos/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Hormônio Liberador da Corticotropina/farmacologia , Hipertensão/tratamento farmacológico , Hipertensão/metabolismo , Urocortinas/farmacologia , Animais , Hormônio Liberador da Corticotropina/genética , Modelos Animais de Doenças , Ecocardiografia , Expressão Gênica/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Hipertrofia Ventricular Esquerda/diagnóstico por imagem , Hipertrofia Ventricular Esquerda/tratamento farmacológico , Hipertrofia Ventricular Esquerda/metabolismo , Masculino , Ratos , Ratos Endogâmicos Dahl , Receptores de Hormônio Liberador da Corticotropina/genética , Urocortinas/genética
19.
J Biol Chem ; 284(12): 7631-45, 2009 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-19106105

RESUMO

Follistatin is a transcriptional target and a modulator of activin action. Through an autocrine/paracrine loop, activin controls follistatin levels and thus regulates its own bioavailability. In gonadotropic alphaT3-1 cells, activin induces follistatin transcription primarily through the action of Smad3 at an intronic Smad-binding element (SBE1). Using a proteomics approach, we searched for endogenous alphaT3-1 proteins that participate in SBE1-mediated transcription. We identified FoxL2, a member of the forkhead family, as a candidate modulator of SBE1 function. Mutations of FoxL2 are associated with the blepharophimosis/ptosis/epicanthus inversus syndrome characterized with craniofacial defects and premature ovarian failure. FoxL2 localizes to alpha-glycoprotein subunit- and follicle-stimulating hormone beta-positive cells of the adult mouse pituitary and is present in alphaT3-1 and LbetaT2 cells, but its pituitary actions remain largely unknown. We have determined that FoxL2 binds to a forkhead-binding element (FKHB) located just downstream of the SBE1 site of the follistatin gene and functions as a Smad3 partner to drive SBE1-mediated transcription in alphaT3-1 cells treated with activin. Chromatin immunoprecipitation assays confirm that endogenous FoxL2 and Smad3 are recruited to the intronic enhancer of the follistatin gene where the SBE1 and FKHB sites are located. Exogenous FoxL2 enhances SBE1-mediated transcription, and short hairpin RNA-mediated knockdown of endogenous FoxL2 protein compromises this effect in alphaT3-1 cells. FoxL2 directly associates with Smad3 but not Smad2 or Smad4. This association between Smad3 and FoxL2 is mediated by the MH2 domain of Smad3 and is dependent on an intact forkhead domain in FoxL2. Altogether, these observations highlight a novel role for FoxL2 and suggest that it may function as a transcriptional regulator and a coordinator of Smad3 targets.


Assuntos
Folistatina/biossíntese , Fatores de Transcrição Forkhead/metabolismo , Hipófise/metabolismo , Proteína Smad3/metabolismo , Ativinas/farmacologia , Animais , Blefaroptose/genética , Blefaroptose/metabolismo , Linhagem Celular , Folistatina/genética , Proteína Forkhead Box L2 , Fatores de Transcrição Forkhead/genética , Humanos , Íntrons/fisiologia , Camundongos , Camundongos Knockout , Mutação , Elementos de Resposta/fisiologia , Proteína Smad3/genética , Transcrição Gênica
20.
Proc Natl Acad Sci U S A ; 105(10): 3939-44, 2008 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-18308934

RESUMO

The corticotropin-releasing factor (CRF) receptor CRFR2 is expressed widely in peripheral tissues and in the vasculature, although its functional roles in those tissues have only recently begun to be elucidated. Previously we found that genetic deletion of CRFR2 resulted in profound postnatal hypervascularization in mice, characterized by both an increase in total vessel number and a dramatic increase in vessel diameter. These data strongly suggested that ligands for CRFR2 act to limit tissue vascularity, perhaps as a counterbalance to factors that promote neovascularization. Urocortin 2 (Ucn2) is a specific ligand for the CRFR2. We hypothesized that activation of CRFR2 by Ucn2 might thus suppress tumor vascularization and consequently limit tumor growth. Here, we show that viral-mediated expression of Ucn2 strikingly inhibits the growth and vascularization of Lewis Lung Carcinoma Cell (LLCC) tumors in vivo. Further, we found that this effect on tumor growth inhibition was independent of whether exposure to Ucn2 occurred before or after establishment of measurable tumors. In vitro, Ucn2 directly inhibited the proliferation of LLCC, suggesting that the tumor-suppressing effects of CRFR2 activation involve a dual mechanism of both a direct inhibition of tumor cell cycling and the suppression of tumor vascularization. These results establish that Ucn2 inhibits tumor growth, suggesting a potential therapeutic role for CRFR2 ligands in clinical malignancies.


Assuntos
Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/patologia , Neovascularização Patológica/patologia , Urocortinas/metabolismo , Animais , Carcinoma Pulmonar de Lewis/genética , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Hormônio Liberador da Corticotropina/genética , Receptores de Hormônio Liberador da Corticotropina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...