Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int Immunopharmacol ; 98: 107872, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34182241

RESUMO

It is quite difficult to generate monoclonal antibodies that recognize the three-dimensional structures of the antigens of interest. To address this limitation, we developed a new hybridoma technology termed "optimized stereospecific targeting (SST)". Here we aimed at generating stereospecific monoclonal antibodies against a G protein-coupled receptor (GPCR). The optimized SST technique enabled the efficient production of conformation-specific monoclonal antibodies against human corticotropin-releasing hormone receptor 1 (huCRHR1). Hybridoma cells secreting stereospecific monoclonal antibodies were selectively cloned by a limiting dilution method and the target monoclonal antibodies were purified by protein A column chromatography. They specifically cross-reacted with native huCRHR1 expressed on the surface of CHO cells, whereas they showed no affinity for MDA-MB-231 cancer cells, which abundantly express EphA2 on the cell surface. Furthermore, immunofluorescence analysis revealed that treatment of huCRHR1-expressing CHO cells with 4% paraformaldehyde led to a decrease in the affinity of purified monoclonal antibodies for intact huCRHR1 on the cell surface. In addition, purified monoclonal antibodies showed no cross-reactivity with huCRHR1 expressed on Sf9 insect cells. These results strongly suggest that monoclonal antibodies generated by the optimized SST technique feature specific binding to the intact form of the target GPCR on mammalian cells.


Assuntos
Anticorpos Monoclonais/farmacologia , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Animais , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/uso terapêutico , Especificidade de Anticorpos , Células CHO , Linhagem Celular Tumoral , Cricetulus , Reações Cruzadas , Feminino , Humanos , Camundongos , Receptor EphA2/metabolismo , Receptores de Hormônio Liberador da Corticotropina/genética , Receptores de Hormônio Liberador da Corticotropina/ultraestrutura , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Células Sf9 , Spodoptera
2.
Front Endocrinol (Lausanne) ; 12: 792912, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35095763

RESUMO

The first intracellular loop (ICL1) of G protein-coupled receptors (GPCRs) has received little attention, although there is evidence that, with the 8th helix (H8), it is involved in early conformational changes following receptor activation as well as contacting the G protein ß subunit. In class B1 GPCRs, the distal part of ICL1 contains a conserved R12.48KLRCxR2.46b motif that extends into the base of the second transmembrane helix; this is weakly conserved as a [R/H]12.48KL[R/H] motif in class A GPCRs. In the current study, the role of ICL1 and H8 in signaling through cAMP, iCa2+ and ERK1/2 has been examined in two class B1 GPCRs, using mutagenesis and molecular dynamics. Mutations throughout ICL1 can either enhance or disrupt cAMP production by CGRP at the CGRP receptor. Alanine mutagenesis identified subtle differences with regard elevation of iCa2+, with the distal end of the loop being particularly sensitive. ERK1/2 activation displayed little sensitivity to ICL1 mutation. A broadly similar pattern was observed with the glucagon receptor, although there were differences in significance of individual residues. Extending the study revealed that at the CRF1 receptor, an insertion in ICL1 switched signaling bias between iCa2+ and cAMP. Molecular dynamics suggested that changes in ICL1 altered the conformation of ICL2 and the H8/TM7 junction (ICL4). For H8, alanine mutagenesis showed the importance of E3908.49b for all three signal transduction pathways, for the CGRP receptor, but mutations of other residues largely just altered ERK1/2 activation. Thus, ICL1 may modulate GPCR bias via interactions with ICL2, ICL4 and the Gß subunit.


Assuntos
Motivos de Aminoácidos/fisiologia , Receptores de Peptídeo Relacionado com o Gene de Calcitonina/ultraestrutura , Receptores de Hormônio Liberador da Corticotropina/ultraestrutura , Receptores de Glucagon/ultraestrutura , Proteína Semelhante a Receptor de Calcitonina/metabolismo , Proteína Semelhante a Receptor de Calcitonina/fisiologia , Proteína Semelhante a Receptor de Calcitonina/ultraestrutura , Sinalização do Cálcio , AMP Cíclico/metabolismo , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases , Simulação de Dinâmica Molecular , Domínios Proteicos , Estrutura Terciária de Proteína , Proteína 1 Modificadora da Atividade de Receptores/metabolismo , Proteína 1 Modificadora da Atividade de Receptores/fisiologia , Proteína 1 Modificadora da Atividade de Receptores/ultraestrutura , Receptores de Peptídeo Relacionado com o Gene de Calcitonina/metabolismo , Receptores de Peptídeo Relacionado com o Gene de Calcitonina/fisiologia , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Receptores de Hormônio Liberador da Corticotropina/fisiologia , Receptores Acoplados a Proteínas G , Receptores de Glucagon/metabolismo , Receptores de Glucagon/fisiologia
3.
Mol Cell ; 77(3): 656-668.e5, 2020 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-32004469

RESUMO

Class B G protein-coupled receptors (GPCRs) are important therapeutic targets for major diseases. Here, we present structures of peptide and Gs-bound pituitary adenylate cyclase-activating peptide, PAC1 receptor, and corticotropin-releasing factor (CRF), (CRF1) receptor. Together with recently solved structures, these provide coverage of the major class B GPCR subfamilies. Diverse orientations of the extracellular domain to the receptor core in different receptors are at least partially dependent on evolutionary conservation in the structure and nature of peptide interactions. Differences in peptide interactions to the receptor core also influence the interlinked TM2-TM1-TM6/ECL3/TM7 domain, and this is likely important in their diverse signaling. However, common conformational reorganization of ECL2, linked to reorganization of ICL2, modulates G protein contacts. Comparison between receptors reveals ICL2 as a key domain forming dynamic G protein interactions in a receptor- and ligand-specific manner. This work advances our understanding of class B GPCR activation and Gs coupling.


Assuntos
Receptores de Hormônio Liberador da Corticotropina/ultraestrutura , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/ultraestrutura , Sequência de Aminoácidos , Microscopia Crioeletrônica/métodos , Encefalinas , Humanos , Ligantes , Modelos Moleculares , Peptídeos , Precursores de Proteínas , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/ultraestrutura , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Transdução de Sinais
4.
Mol Cell ; 77(3): 669-680.e4, 2020 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-32004470

RESUMO

Corticotropin-releasing factor (CRF) and the three related peptides urocortins 1-3 (UCN1-UCN3) are endocrine hormones that control the stress responses by activating CRF1R and CRF2R, two members of class B G-protein-coupled receptors (GPCRs). Here, we present two cryoelectron microscopy (cryo-EM) structures of UCN1-bound CRF1R and CRF2R with the stimulatory G protein. In both structures, UCN1 adopts a single straight helix with its N terminus dipped into the receptor transmembrane bundle. Although the peptide-binding residues in CRF1R and CRF2R are different from other members of class B GPCRs, the residues involved in receptor activation and G protein coupling are conserved. In addition, both structures reveal bound cholesterol molecules to the receptor transmembrane helices. Our structures define the basis of ligand-binding specificity in the CRF receptor-hormone system, establish a common mechanism of class B GPCR activation and G protein coupling, and provide a paradigm for studying membrane protein-lipid interactions for class B GPCRs.


Assuntos
Receptores de Hormônio Liberador da Corticotropina/ultraestrutura , Sequência de Aminoácidos , Sítios de Ligação , Hormônio Liberador da Corticotropina , Microscopia Crioeletrônica/métodos , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Humanos , Peptídeos/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Urocortinas/metabolismo
5.
Artigo em Inglês | MEDLINE | ID: mdl-25149913

RESUMO

Understanding the neurobiological bases for sex differences in alcohol dependence is needed to help guide the development of individualized therapies for alcohol abuse disorders. In the present study, alcohol-induced adaptations in (1) anxiety-like behavior, (2) patterns of c-Fos activation and (3) subcellular distribution of corticotropin releasing factor receptor in locus coeruleus (LC) neurons was investigated in male and female Sprague-Dawley rats that were chronically exposed to ethanol using a liquid diet. Results confirm and extend reports by others showing that chronic ethanol exposure produces an anxiogenic-like response in both male and female subjects. Ethanol-induced sex differences were observed with increased c-Fos expression in LC neurons of female ethanol-treated subjects compared to controls or male subjects. Results also reveal sex differences in the subcellular distribution of the CRFr in LC-noradrenergic neurons with female subjects exposed to ethanol exhibiting a higher frequency of plasmalemmal CRFrs. These adaptations have implications for LC neuronal activity and its neural targets across the sexes. Considering the important role of the LC in ethanol-induced activation of the hypothalamo-pituitary-adrenal (HPA) axis, the present results indicate important sex differences in feed-forward regulation of the HPA axis that may render alcohol dependent females more vulnerable to subsequent stress exposure.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Locus Cerúleo/citologia , Neurônios/ultraestrutura , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Caracteres Sexuais , Frações Subcelulares/metabolismo , Análise de Variância , Animais , Feminino , Locomoção/efeitos dos fármacos , Locus Cerúleo/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Microscopia Eletrônica de Transmissão , Neurônios/efeitos dos fármacos , Postura , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Receptores de Hormônio Liberador da Corticotropina/ultraestrutura , Frações Subcelulares/efeitos dos fármacos , Fatores de Tempo
6.
Biol Psychiatry ; 73(11): 1087-94, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23452664

RESUMO

BACKGROUND: Social stress is a risk factor for affective disorders for certain vulnerable individuals. Stress and depression are linked in part through regulation of the dorsal raphe (DR)-serotonin (5-HT) system by the stress-related neuropeptide, corticotropin-releasing factor (CRF). We used a rat social stress model that shows individual differences in coping strategies to determine whether differences in CRF-5-HT interactions underlie individual differences in the vulnerability to social stress. METHODS: Rats were exposed to the resident-intruder model of social stress for 5 days. In vivo single-unit recordings assessed DR-5-HT neuronal responses to CRF and immunoelectron microscopy assessed CRF1 and CRF2 cellular localization 24 hours after the last stress. RESULTS: Rats responded to social stress passively, assuming defeat with short latencies (48%), or actively, with proactive behaviors and longer defeat latencies (LL, 52%). Whereas CRF (30 ng, intra-DR) inhibited 5-HT neuronal activity of control and SL rats, it activated 5-HT neurons of LL rats, an effect that was CRF2-mediated. Consistent with this, social stress promoted CRF1 internalization together with CRF2 recruitment to the plasma membrane of DR neurons selectively in LL rats. CONCLUSIONS: These data suggest that a proactive coping strategy toward social stress is associated with a redistribution of CRF1 and CRF2 in DR-5-HT neurons that primes the system to be activated by subsequent stress. The lack of this adaptation in passive coping rats may contribute to their depressive-like phenotype. These studies provide a cellular mechanism for individual differences in stress responses and consequences.


Assuntos
Adaptação Biológica/fisiologia , Núcleos da Rafe/patologia , Neurônios Serotoninérgicos/fisiologia , Estresse Psicológico/patologia , Estresse Psicológico/psicologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Adaptação Biológica/efeitos dos fármacos , Análise de Variância , Animais , Arrestinas/metabolismo , Biotina/análogos & derivados , Biotina/metabolismo , Hormônio Liberador da Corticotropina/farmacologia , Modelos Animais de Doenças , Individualidade , Masculino , Microscopia Imunoeletrônica , Fragmentos de Peptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Tempo de Reação , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Receptores de Hormônio Liberador da Corticotropina/ultraestrutura , Neurônios Serotoninérgicos/efeitos dos fármacos , Neurônios Serotoninérgicos/ultraestrutura , Serotonina/metabolismo , beta-Arrestinas
7.
Neuroscience ; 159(2): 526-39, 2009 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-19166913

RESUMO

The anxiolytic effects of opiates active at the mu-opioid receptor (mu-OR) may be ascribed, in part, to suppression of neurons that are responsive to the stress-associated peptide, corticotropin releasing factor (CRF), in the central amygdala (CeA) and bed nucleus of the stria terminalis (BNST). The corticotropin releasing factor receptor (CRFr) and mu-OR are expressed in both the CeA and BNST, but their subcellular relationship to each other is not known in either region. To address this question, we used dual electron microscopic immunolabeling of mu-OR and CRFr in the mouse lateral CeA and anterolateral BNST. Immunolabeling for each receptor was detected in the same as well as in separate somatic, dendritic and axonal profiles of neurons in each region. CRFr had a plasmalemmal or cytoplasmic distribution in many dendrites, including those co-expressing mu-OR. The co-expression of CRFr and mu-OR also was seen near excitatory-type synapses on dendritic spines. In both the CeA and BNST, over 50% of the CRFr-labeled dendritic profiles (dendrites and spines) contained immunoreactivity for the mu-OR. However, less than 25% of the dendritic profiles containing the mu-OR were labeled for CRFr in either region, suggesting that opiate activation of the mu-OR affects many neurons in addition to those responsive to CRF. The dendritic profiles containing CRFr and/or mu-OR received asymmetric, excitatory-type synapses from unlabeled or CRFr-labeled axon terminals. In contrast, the mu-OR was identified in terminals forming symmetric, inhibitory-type synapses. Thus, in both the CeA and BNST, mu-OR and CRFr have strategic locations for mediation of CRF and opioid effects on the postsynaptic excitability of single neurons, and on the respective presynaptic release of excitatory and inhibitory neurotransmitters. The commonalities in the synaptic location of both receptors in the CeA and BNST suggest that this is a fundamental cellular association of relevance to both drug addiction and stress-induced disorders.


Assuntos
Tonsila do Cerebelo/citologia , Neurônios/citologia , Terminações Pré-Sinápticas/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Receptores Opioides mu/metabolismo , Núcleos Septais/citologia , Sinapses/metabolismo , Animais , Axônios/metabolismo , Axônios/ultraestrutura , Dendritos/metabolismo , Dendritos/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão/métodos , Microscopia Imunoeletrônica/métodos , Neurônios/metabolismo , Neurônios/ultraestrutura , Terminações Pré-Sinápticas/ultraestrutura , Receptores de Hormônio Liberador da Corticotropina/ultraestrutura , Sinapses/ultraestrutura
8.
J Comp Neurol ; 512(3): 323-35, 2009 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-19003957

RESUMO

Corticotrophin-releasing factor (CRF) is expressed in the central nucleus of the amygdala (CeA), where the CRF receptor (CRFr) plays an important role in anxiety- and stress-related behaviors. To determine the subcellular sites of CRFr activation in this region, we examined the electron microscopic immunolabeling of antisera recognizing CRF or CRFr. The ultrastructural analysis was principally conducted in the lateral subdivision of the rat CeA, with comparisons being made in mice so as to optimally utilize mutant mice in control experiments. The CRFr labeling was seen in many small dendrites and dendritic spines as well as in a few somata, large dendrites, axons, and axon terminals or more rarely in glial processes. Approximately 35% of the CRFr-labeled dendrites contained CRF immunoreactivity, which was distributed diffusely throughout the cytoplasm, or specifically affiliated with either endomembranes or large dense-core vesicles. The CRF-immunoreactive vesicles also were present in somata and axon terminals with or without CRFr labeling. The CRF immunoreactivity was usually absent from both terminals and dendrites joined by asymmetric, excitatory-type synapses, where a postsynaptic location of the CRFr was commonly observed. Numerous terminals containing both CRF and CRFr were seen, however, within the neuropil and sometimes apposing the excitatory synapses. These results provide ultrastructural evidence for a primary involvement of CRF receptors in modulation of the postsynaptic excitability of CeA neurons, an effect that may be limited by the availability of CRF. The findings have important implications for understanding CRF mediation of rapid responses to stress.


Assuntos
Tonsila do Cerebelo , Hormônio Liberador da Corticotropina/análise , Receptores de Hormônio Liberador da Corticotropina/ultraestrutura , Tonsila do Cerebelo/química , Tonsila do Cerebelo/ultraestrutura , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/química , Neurônios/ultraestrutura , Ratos , Sinapses/química , Sinapses/ultraestrutura
9.
Eur J Neurosci ; 23(11): 2991-8, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16819988

RESUMO

Corticotropin-releasing factor (CRF) acts within the locus coeruleus (LC), to modulate activity of the LC-norepinephrine (NE) system. Combining molecular and cellular approaches, we demonstrate CRF receptor (CRFr) mRNA expression in Sprague-Dawley rat LC and provide the first in vivo evidence for agonist-induced internalization of CRFr. CRFr mRNA was detected in LC micropunches by RT-PCR. In dual labelling immunofluorescence studies, tyrosine hydroxylase (TH) containing neurons exhibited CRFr labelling. At the ultrastructural level, immunogold-silver labelling for CRFr was localized to the plasma membrane of TH-immunoperoxidase labelled dendrites. CRF (100 ng) injection into the LC produced a robust neuronal activation that peaked 10-15 min after injection and was maintained for the duration of the recording. This was associated with CRFr internalization in LC neurons that was apparent at 5 and 30 min after injection. By 5 min after injection the ratio of cytoplasmic to total dendritic CRFr-labelling was 0.81 +/- 0.01 in rats injected with CRF and 0.59 +/- 0.02 in rats injected with artificial cerebrospinal fluid (ACSF; P < 0.0001). Enhanced internalization of CRFr was maintained at 30 min after CRF injection, with the ratio being 0.86 +/- 0.02 for CRF-injected cases and 0.57 +/- 0.03 for ACSF-injected cases (P < 0.0001). Internalized CRFr was associated with early endosomes, indicative of degradation or recycling. Agonist-induced CRFr internalization in LC neurons may underlie acute desensitization to CRF or stress. This process may be a pivotal target by which stressors or pharmacological agents regulate the sensitivity of the LC-NE system to CRF and subsequent stressors.


Assuntos
Hormônio Liberador da Corticotropina/farmacologia , Locus Cerúleo/citologia , Neurônios/efeitos dos fármacos , Norepinefrina/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Imuno-Histoquímica/métodos , Masculino , Microscopia Imunoeletrônica/métodos , Neurônios/metabolismo , Neurônios/ultraestrutura , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Hormônio Liberador da Corticotropina/genética , Receptores de Hormônio Liberador da Corticotropina/ultraestrutura , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/metabolismo
10.
Neuroscience ; 126(3): 533-40, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15183503

RESUMO

Neuropeptides modulate neuronal function in hippocampus, but the organization of hippocampal sites of peptide release and actions is not fully understood. The stress-associated neuropeptide corticotropin releasing hormone (CRH) is expressed in inhibitory interneurons of rodent hippocampus, yet physiological and pharmacological data indicate that it excites pyramidal cells. Here we aimed to delineate the structural elements underlying the actions of CRH, and determine whether stress influenced hippocampal principal cells also via actions of this endogenous peptide. In hippocampal pyramidal cell layers, CRH was located exclusively in a subset of GABAergic somata, axons and boutons, whereas the principal receptor mediating the peptide's actions, CRH receptor 1 (CRF1), resided mainly on dendritic spines of pyramidal cells. Acute 'psychological' stress led to activation of principal neurons that expressed CRH receptors, as measured by rapid phosphorylation of the transcription factor cyclic AMP responsive element binding protein. This neuronal activation was abolished by selectively blocking the CRF1 receptor, suggesting that stress-evoked endogenous CRH release was involved in the activation of hippocampal principal cells.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Hipocampo/metabolismo , Terminações Pré-Sinápticas/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Estresse Psicológico/fisiopatologia , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Imuno-Histoquímica , Interneurônios/metabolismo , Interneurônios/ultraestrutura , Microscopia Eletrônica , Terminações Pré-Sinápticas/ultraestrutura , Ratos , Ratos Sprague-Dawley , Receptores de Hormônio Liberador da Corticotropina/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...