Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Biol Cell ; : mbcE24020083, 2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38758660

RESUMO

Chromaffin cells of the adrenal medulla transduce sympathetic nerve activity into stress hormone secretion. The two neurotransmitters principally responsible for coupling cell stimulation to secretion are acetylcholine and pituitary adenylate activating polypeptide (PACAP). In contrast to acetylcholine, PACAP evokes a persistent secretory response from chromaffin cells. However, the mechanisms by which PACAP acts are poorly understood. Here, it is shown that PACAP induces sustained increases in cytosolic Ca2+ which are disrupted when Ca2+ influx through L-type channels is blocked or internal Ca2+ stores are depleted. PACAP liberates stored Ca2+ via inositol trisphosphate receptors (IP3Rs) on the endoplasmic reticulum (ER), thereby functionally coupling Ca2+ mobilization to Ca2+ influx and supporting Ca2+-induced Ca2+-release. These Ca2+ influx and mobilization pathways are unified by an absolute dependence on phospholipase C epsilon (PLCε) activity. Thus, the persistent secretory response that is a defining feature of PACAP activity, in situ, is regulated by a signaling network that promotes sustained elevations in intracellular Ca2+ through multiple pathways.

2.
Mol Pharmacol ; 105(2): 104-115, 2024 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-38164584

RESUMO

Metabotropic glutamate receptors (mGluRs) are obligate dimer G protein coupled receptors that can all function as homodimers. Here, each mGluR homodimer was examined for its G protein coupling profile using a bioluminescence resonance energy transfer-based assay that detects the interaction between a split YFP-tagged Gß 1γ2 and a Nanoluciferase tagged free Gßγ sensor, MAS-GRK3-ct- nanoluciferase with 14 specific Gα proteins heterologously expressed, representing each family. Canonically, the group II and III mGluRs (2 and 3 and 4, 6, 7, and 8, respectively) are thought to couple to Gi/o exclusively. In addition, the group I mGluRs (1 and 5) are known to couple to the Gq/11 family and generally thought to also couple to the pertussis toxin-sensitive Gi/o family some reports have suggested Gs coupling is possible as cAMP elevations have been noted. In this study, coupling was observed with all eight mGluRs through the Gi/o proteins and only mGluR1 and mGluR5 through Gq/11, and, perhaps surprisingly, not G14 None activated any Gs protein. Interestingly, coupling was seen with the group I and II but not the group III mGluRs to G16 Slow but significant coupling to Gz was also seen with the group II receptors. SIGNIFICANCE STATEMENT: Metabotropic glutamate receptor (mGluR)-G protein coupling has not been thoroughly examined, and some controversy remains about whether some mGluRs can activate Gαs family members. Here we examine the ability of each mGluR to activate representative members of every Gα protein family. While all mGluRs can activate Gαi/o proteins, only the group I mGluRs couple to Gαq/11, and no members of the family can activate Gαs family members, including the group I receptors alone or with positive allosteric modulators.


Assuntos
Proteínas de Ligação ao GTP , Transdução de Sinais , Humanos , Proteínas de Ligação ao GTP/metabolismo , Toxina Pertussis , Proteínas de Transporte/metabolismo
3.
bioRxiv ; 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38260572

RESUMO

Chromaffin cells of the adrenal medulla transduce sympathetic nerve activity into stress hormone secretion. The two neurotransmitters principally responsible for coupling cell stimulation to secretion are acetylcholine and pituitary adenylate activating polypeptide (PACAP). In contrast to acetylcholine, PACAP evokes a persistent secretory response from chromaffin cells. However, the mechanisms by which PACAP acts are poorly understood. Here, it is shown that PACAP induces sustained increases in cytosolic Ca 2+ which are disrupted when Ca 2+ influx through L-type channels is blocked or internal Ca 2+ stores are depleted. PACAP liberates stored Ca 2+ via inositol trisphosphate receptors (IP3Rs) on the endoplasmic reticulum (ER), thereby functionally coupling Ca 2+ mobilization to Ca 2+ influx and supporting Ca 2+ -induced Ca 2+ -release. These Ca 2+ influx and mobilization pathways are unified by an absolute dependence on phospholipase C epsilon (PLCε) activity. Thus, the persistent secretory response that is a defining feature of PACAP activity, in situ , is regulated by a signaling network that promotes sustained elevations in intracellular Ca 2+ through multiple pathways.

4.
bioRxiv ; 2023 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-37546908

RESUMO

Metabotropic glutamate receptors (mGluRs) are obligate dimer G protein coupled receptors that can all function as homodimers. Here, each mGluR homodimer was examined for its G protein coupling profile using a BRET based assay that detects the interaction between a split YFP-tagged Gß1γ2 and a Nanoluc tagged free Gßγ sensor, MAS-GRK3-ct-NLuc with 14 specific Ga proteins heterologously expressed, representing each family. Canonically, the group II and III mGluRs (2&3, and 4, 6, 7&8, respectively) are thought to couple to Gi/o exclusively. In addition, the group I mGluRs (1&5) are known to couple to the Gq/11 family, and generally thought to also couple to the PTX-sensitive Gi/o family; some reports have suggested Gs coupling is possible as cAMP elevations have been noted. In this study, coupling was observed with all 8 mGluRs through the Gi/o proteins, and only mGluR1&5 through Gq/11, and perhaps surprisingly, not G14. None activated any Gs protein. Interestingly, coupling was seen with the group I and II, but not the group III mGluRs to G16. Slow but significant coupling to Gz was also seen with the group II receptors.

5.
J Gen Physiol ; 155(2)2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36538657

RESUMO

The adrenomedullary chromaffin cell transduces chemical messages into outputs that regulate end organ function throughout the periphery. At least two important neurotransmitters are released by innervating preganglionic neurons to stimulate exocytosis in the chromaffin cell-acetylcholine (ACh) and pituitary adenylate cyclase activating polypeptide (PACAP). Although PACAP is widely acknowledged as an important secretagogue in this system, the pathway coupling PACAP stimulation to chromaffin cell secretion is poorly understood. The goal of this study is to address this knowledge gap. Here, it is shown that PACAP activates a Gαs-coupled pathway that must signal through phospholipase C ε (PLCε) to drive Ca2+ entry and exocytosis. PACAP stimulation causes a complex pattern of Ca2+ signals in chromaffin cells, leading to a sustained secretory response that is kinetically distinct from the form stimulated by ACh. Exocytosis caused by PACAP is associated with slower release of peptide cargo than exocytosis stimulated by ACh. Importantly, only the secretory response to PACAP, not ACh, is eliminated in cells lacking PLCε expression. The data show that ACh and PACAP, acting through distinct signaling pathways, enable nuanced and variable secretory outputs from chromaffin cells.


Assuntos
Células Cromafins , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Acetilcolina/farmacologia , Acetilcolina/metabolismo , Cálcio/metabolismo , Catecolaminas/metabolismo , Células Cromafins/metabolismo
6.
Neuropharmacology ; 199: 108801, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34547332

RESUMO

Metabotropic glutamate receptors (mGluRs) are an essential component of the mammalian central nervous system. These receptors modulate neuronal excitability in response to extracellular glutamate through the activation of intracellular heterotrimeric G proteins. Like most other class C G protein-coupled receptors, mGluRs function as obligate dimer proteins, meaning they need to form dimer complexes before becoming functional receptors. All mGluRs possess the ability to homodimerize, but studies over the past ten years have demonstrated these receptors are also capable of forming heterodimers in specific patterns. These mGluR heterodimers appear to have their own unique biophysical behavior and pharmacology with both native and synthetic compounds with few rules having been identified that allow for prediction of the consequences of any particular mGluR pair forming heterodimers. Here, we review the relevant literature demonstrating the existence and consequences of mGluR heterodimerization. By collecting biophysical and pharmacological data of several mGluR heterodimers we demonstrate the lack of generalizable behavior of these complexes indicating that each individual dimeric pair needs to be investigated independently. Additionally, by combining sequence alignment and structural analysis, we propose that interactions between the ß4-A Helix Loop and the D Helix in the extracellular domain of these receptors are the structural components that dictate heterodimerization compatibility. Finally, we discuss the potential implications of mGluR heterodimerization from the viewpoints of further developing our understanding of neuronal physiology and leveraging mGluRs as a therapeutic target for the treatment of pathophysiology.


Assuntos
Multimerização Proteica , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Humanos
7.
Sci Rep ; 10(1): 14866, 2020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32913201

RESUMO

The composition, stoichiometry and interactions of supramolecular protein complexes are a critical determinant of biological function. Several techniques have been developed to study molecular interactions and quantify subunit stoichiometry at the single molecule level. However, these typically require artificially low expression levels or detergent isolation to achieve the low fluorophore concentrations required for single molecule imaging, both of which may bias native subunit interactions. Here we present an alternative approach where protein complexes are assembled at physiological concentrations and subsequently diluted in situ for single-molecule level observations while preserving them in a near-native cellular environment. We show that coupling this dilution strategy with fluorescence correlation spectroscopy permits quantitative assessment of cytoplasmic oligomerization, while stepwise photobleaching and single molecule colocalization may be used to study the subunit stoichiometry of membrane receptors. Single protein recovery after dilution (SPReAD) is a simple and versatile means of extending the concentration range of single molecule measurements into the cellular regime while minimizing potential artifacts and perturbations of protein complex stoichiometry.


Assuntos
Corantes Fluorescentes/química , Microscopia de Fluorescência/métodos , Complexos Multiproteicos/química , Imagem Individual de Molécula/métodos , Fusão Celular , Humanos , Complexos Multiproteicos/metabolismo , Fotodegradação
8.
PLoS One ; 15(2): e0219886, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32023253

RESUMO

Förster Resonance Energy Transfer (FRET) has become an immensely powerful tool to profile intra- and inter-molecular interactions. Through fusion of genetically encoded fluorescent proteins (FPs) researchers have been able to detect protein oligomerization, receptor activation, and protein translocation among other biophysical phenomena. Recently, two bright monomeric red fluorescent proteins, mRuby3 and mScarlet-I, have been developed. These proteins offer much improved physical properties compared to previous generations of monomeric red FPs that should help facilitate more general adoption of Green/Red FRET. Here we assess the ability of these two proteins, along with mCherry, to act as a FRET acceptor for the bright, monomeric, green-yellow FP mNeonGreen using intensiometric FRET and 2-photon Fluorescent Lifetime Imaging Microscopy (FLIM) FRET techniques. We first determined that mNeonGreen was a stable donor for 2-photon FLIM experiments under a variety of imaging conditions. We then tested the red FP's ability to act as FRET acceptors using mNeonGreen-Red FP tandem construct. With these constructs we found that mScarlet-I and mCherry are able to efficiently FRET with mNeonGreen in spectroscopic and FLIM FRET. In contrast, mNeonGreen and mRuby3 FRET with a much lower efficiency than predicted in these same assays. We explore possible explanations for this poor performance and determine mRuby3's protein maturation properties are a major contributor. Overall, we find that mNeonGreen is an excellent FRET donor, and both mCherry and mScarlet-I, but not mRuby3, act as practical FRET acceptors, with the brighter mScarlet-I out performing mCherry in intensiometric studies, but mCherry out performing mScarlet-I in instances where consistent efficiency in a population is critical.


Assuntos
Transferência Ressonante de Energia de Fluorescência/métodos , Proteínas de Fluorescência Verde/metabolismo , Proteínas Luminescentes/normas , Transferência Ressonante de Energia de Fluorescência/normas , Células HEK293 , Humanos , Microscopia Intravital/métodos , Microscopia de Fluorescência/métodos , Análise de Célula Única/métodos , Proteína Vermelha Fluorescente
9.
Pharmacol Res Perspect ; 7(3): e00471, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31065376

RESUMO

Metabotropic glutamate receptors (mGluRs) are class C G protein coupled receptors with widespread expression in the central nervous system. There are eight mGluRs in the mammalian genome. Research on mGluRs relies on the availability of selective compounds. While many selective allosteric compounds have been described, selectivity of orthosteric agonists and antagonists has been more difficult due to the similarity of the glutamate binding pocket across the mGluR family. LY341495 has been used for decades as a potent and selective group II mGluR antagonist. The selectivity of LY341495 was investigated here between mGluR2, a group II mGluR, and mGluR4, a group III receptor, heterologously expressed in adult rat sympathetic neurons from the superior cervical ganglion (SCG), which provides a null-mGluR background upon which mGluRs were examined in isolation. The compound does in fact selectively inhibit mGluR2 over mGluR4, but in such a way that it makes signaling of the two receptors more difficult to distinguish. The glutamate potency of mGluR2 is about 10-fold higher than mGluR4. 50 nmol L-1 LY341495 did not alter mGluR4 signaling but shifted the mGluR2 glutamate dose-response about 10-fold, such that it overlapped more closely with that of mGluR4. Increasing the LY341494 dose to 500 nmol L-1 further shifted the glutamate dose-response of mGluR2 by another ~10-fold, but also shifted that of mGluR4 similarly. Thus, while glutamate is a moderately selective agonist of mGluR2 over mGluR4 when applied alone, in the presence of increasing concentrations of LY341495, this selectivity of glutamate is lost.


Assuntos
Aminoácidos/farmacologia , Receptores de Glutamato Metabotrópico/metabolismo , Gânglio Cervical Superior/metabolismo , Xantenos/farmacologia , Animais , Relação Dose-Resposta a Droga , Regulação para Baixo , Gânglios Simpáticos/citologia , Gânglios Simpáticos/efeitos dos fármacos , Gânglios Simpáticos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Ratos , Receptores de Glutamato Metabotrópico/genética , Transdução de Sinais/efeitos dos fármacos , Gânglio Cervical Superior/citologia , Gânglio Cervical Superior/efeitos dos fármacos
10.
Channels (Austin) ; 11(6): 555-573, 2017 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-28837380

RESUMO

Type two voltage gated calcium (CaV2) channels are the primary mediators of neurotransmission at neuronal presynapses, but their function at neural soma is also important in regulating excitability. 1 Mechanisms that regulate CaV2 channel expression at synapses have been studied extensively, which motivated us to perform similar studies in the soma. Rat sympathetic neurons from the superior cervical ganglion (SCG) natively express CaV2.2 and CaV2.3. 2 We noted previously that heterologous expression of CaV2.1 but not CaV2.2 results in increased calcium current in SCG neurons. 3 In the present study, we extended these observations to show that both CaV2.1 and CaV2.3 expression resulted in increased calcium currents while CaV2.2 expression did not. Further, CaV2.1 could displace native CaV2.2 channels, but CaV2.3 expression could not. Heterologous expression of the individual accessory subunits α2δ-1, α2δ-2, α2δ-3, or ß4 alone failed to increase current density, suggesting that the calcium current ceiling when CaV2.2 was over-expressed was not due to lack of these subunits. Interestingly, introduction of recombinant α2δ subunits produced surprising effects on displacement of native CaV2.2 by recombinant channels. Both α2δ-1 and α2δ-2 seemed to promote CaV2.2 displacement by recombinant channel expression, while α2δ-3 appeared to protect CaV2.2 from displacement. Thus, we observe a selective prioritization of CaV channel functional expression in neurons by specific α2δ subunits. These data highlight a new function for α2δ subtypes that could shed light on subtype selectivity of CaV2 membrane expression.


Assuntos
Canais de Cálcio Tipo N/biossíntese , Neurônios/metabolismo , Subunidades Proteicas/metabolismo , Animais , Canais de Cálcio Tipo N/química , Canais de Cálcio Tipo N/genética , Canais de Cálcio Tipo N/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Masculino , Subunidades Proteicas/biossíntese , Subunidades Proteicas/genética , Ratos , Ratos Wistar
11.
Cold Spring Harb Protoc ; 2017(7): pdb.top092288, 2017 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-28679719

RESUMO

From patch-clamp techniques to recombinant DNA technologies, three-dimensional protein modeling, and optogenetics, diverse and sophisticated methods have been used to study ion channels and how they determine the electrical properties of cells.


Assuntos
Técnicas Citológicas/métodos , Variação Genética , Canais Iônicos/genética , Canais Iônicos/metabolismo , Técnicas Citológicas/história , História do Século XX , História do Século XXI
12.
J Pharmacol Exp Ther ; 361(1): 122-129, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28179473

RESUMO

In rat sympathetic neurons from the superior cervical ganglia (SCG) expressing metabotropic glutamate receptor mGluR1 or mGluR5, overexpression of scaffolding Homer proteins, which bind to a Homer ligand in their C termini, cause receptor clustering and uncoupling from ion channel modulation. In the absence of recombinant Homer protein overexpression, uncoupling of mGluRs from voltage-dependent channels can be induced by expression of Preso1, an adaptor of proline-directed kinases that phosphorylates the Homer ligand and recruits binding of endogenous Homer proteins. Here we show that in SCG neurons expressing mGluR1 and the tyrosine receptor kinase B, treatment with brain-derived neurotrophic factor (BDNF) produces a similar uncoupling of the receptors from calcium channels. We investigated the pathways that mediate this uncoupling and compared it with uncoupling observed with Preso1 expression. Both BDNF- and Preso1-induced uncoupling require residues T1151 and S1154 in the mGluR1 Homer ligand (TPPSPF). Uncoupling via Preso1 but not BDNF was prevented by expression of a dominant negative Cdk5, suggesting that endogenous Cdk5 mediates Preso1-dependent phosphorylation of mGluR1. Dominant negative Cdk5 did not block the BDNF effect but this was sensitive to inhibitors of the mitogen-activated protein kinase kinase/extracellular signal-regulated kinase cascade. Interestingly, the BDNF pathway appeared to require native Preso1 binding to mGluR, because overexpression of the Preso1 FERM domain, which mediates the Preso1-mGluR interaction, prevented BDNF-induced uncoupling. These data suggest that the BDNF/tyrosine receptor kinase B and Cdk5 pathways converge at the level of mGluR to similarly induce Homer ligand phosphorylation, recruit Homer binding, and uncouple mGluRs from channel regulation.


Assuntos
Proteínas de Arcabouço Homer/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas do Tecido Nervoso/biossíntese , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Ratos , Ratos Wistar
13.
BMC Neurosci ; 16: 17, 2015 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-25881041

RESUMO

BACKGROUND: Metabotropic glutamate receptors (mGluRs) are class C G protein coupled receptors with widespread central nervous system expression. mGluR7 is a member of this family that has been implicated in numerous physiological and pathological processes, but the very low potency of mGluR7 for glutamate, its natural ligand, raise questions about the nature of its physiological role. RESULTS: Here, evidence is presented using heterologous expression in sympathetic neurons from the rat superior cervical ganglion (SCG) and modulation of the native SCG calcium currents as an assay for receptor signaling, that mGluR7 exhibits constitutive activity. This activity is detectable as basal calcium channel modulation in the absence of ligand that is not observed in untransfected cells or those transfected with other members of the mGluR family. Further, this basal channel modulation was reversibly inhibited with the mGluR7 inverse agonist MMPIP. Surprisingly, MMPIP did not strongly inhibit agonist-induced mGluR7 activation. Finally, the selective mGluR8 agonist (R,S)-PPG was also able to act as an inverse agonist at mGluR7. CONCLUSIONS: These findings introduce a novel potential physiological role for mGluR7 in the nervous system, that of a constitutively active receptor, and thereby suggest a model in which mGluR7 signaling may be impactful without the need to invoke strong receptor activation by millimolar concentrations of extracellular glutamate. Constitutive activity of mGluR7 may be eliminated or reduced by the presence of other group III mGluRs, perhaps due to heterodimer formation. In addition, both MMPIP and PPG acted as inverse agonists at mGluR7, and agonists at mGluR8.


Assuntos
Neurônios/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Cálcio/metabolismo , Células Cultivadas , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Piridonas/farmacologia , Ratos Wistar , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Gânglio Cervical Superior/efeitos dos fármacos , Gânglio Cervical Superior/metabolismo
14.
Mol Pharmacol ; 86(5): 492-504, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25113912

RESUMO

Metabotropic glutamate receptors (mGluRs) function as dimers. Recent work suggests that mGluR1 and mGluR5 may physically interact, but the nature and functional consequences of this relationship have not been addressed. In this study, the functional and pharmacological consequences of this interaction were investigated. Using heterologous expression of mGluR cDNA in rat sympathetic neurons from the superior cervical ganglion and inhibition of the native calcium currents as an assay for receptor activation, a functional interdependence between mGluR1 and mGluR5 was demonstrated. In neurons coexpressing these receptors, combining a selective mGluR1 competitive antagonist with either an mGluR1- or mGluR5-selective negative allosteric modulator (NAM) BAY36-7620 [(3aS,6aS)-hexahydro-5-methylene-6a-(2-naphthalenylmethyl)-1H-cyclopenta[c]furan-1-one] or MPEP [2-methyl-6-(phenylethynyl)pyridine hydrochloride], respectively, strongly occluded signaling by both receptors to an approximately equal degree. By contrast, in cells coexpressing mGluR1 and mGluR2, combining the same mGluR1 competitive inhibitor with an mGluR1 or mGluR2 NAM yielded partial and full inhibition of the response, respectively, as expected for independently acting receptors. In neurons expressing mGluR1 and mGluR5, the selective NAMs each strongly inhibited the response to glutamate, suggesting that these receptors do not interact as heterodimers, which would not be inhibited by selective NAMs. Finally, evidence for a similar mGluR1/mGluR5 functional dependence is shown in medium spiny striatal neurons. Together, these data demonstrate cooperative signaling between mGluR1 and mGluR5 in a manner inconsistent with heterodimerization, and thus suggest an interaction between homodimers.


Assuntos
Receptor de Glutamato Metabotrópico 5/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Transdução de Sinais/fisiologia , Regulação Alostérica/fisiologia , Animais , Cálcio/metabolismo , Ácido Glutâmico/metabolismo , Masculino , Neurônios/metabolismo , Ratos , Ratos Wistar , Gânglio Cervical Superior/metabolismo
15.
Neuropharmacology ; 66: 302-10, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22659088

RESUMO

Group I metabotropic glutamate receptors (mGluR1 and 5) are G protein coupled receptors that regulate neuronal activity in a number of ways. Some of the most well studied functions of group I mGluRs, such as initiation of multiple forms of mGluR-dependent long-term depression, require receptor localization near the post-synaptic density (PSD). This localization is in turn dependent on the Homer family of scaffolding proteins which bind to a small motif on the distal C-termini of mGluR1 and 5, localize the receptors near the PSD, strengthen coupling to post-synaptic effectors and simultaneously uncouple the mGluRs from extra-synaptic effectors such as voltage dependent ion channels. Here the selectivity of this uncoupling process was examined by testing the ability of Homer-2b to uncouple mGluR1 from multiple voltage dependent calcium channels including Ca(V2.2) (N-type), Ca(V3.2) (T-type), and Ca(V2.1) (P/Q-type) expressed in rat sympathetic neurons from the superior cervical ganglion (SCG). Of these, only the mGluR1-Ca(V2.1) modulatory pathway was insensitive to Homer-2b expression. Uncoupling from this channel was achieved by co-expression of an mGluR1 C-terminal protein designed to disrupt a previously described direct interaction between these two proteins, suggesting that this interaction allows incorporation of Ca(V2.1) into the mGluR1/Homer signaling complex, thereby preserving modulation in the presence of scaffolding Homer proteins. This article is part of a Special Issue entitled 'Metabotropic Glutamate Receptors'.


Assuntos
Canais de Cálcio Tipo N/fisiologia , Proteínas de Transporte/fisiologia , Receptor Cross-Talk/fisiologia , Receptores de Glutamato Metabotrópico/fisiologia , Animais , Canais de Cálcio/fisiologia , Canais de Cálcio Tipo N/biossíntese , Proteínas de Transporte/biossíntese , Proteínas de Arcabouço Homer , Masculino , Potenciais da Membrana/fisiologia , Modelos Neurológicos , Neurônios/metabolismo , Neurônios/fisiologia , Cultura Primária de Células , Ratos , Ratos Wistar , Receptores de Glutamato Metabotrópico/biossíntese , Gânglio Cervical Superior/metabolismo , Gânglio Cervical Superior/fisiologia
16.
Mol Pharmacol ; 82(3): 438-47, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22653971

RESUMO

Metabotropic glutamate receptors (mGluRs) were thought until recently to function mainly as stable homodimers, but recent work suggests that heteromerization is possible. Despite the growth in available compounds targeting mGluRs, little is known about the pharmacological profile of mGluR heterodimers. Here, this question was addressed for the mGluR2/4 heterodimer, examined by coexpressing both receptors in isolated sympathetic neurons from the rat superior cervical ganglion (SCG), a native neuronal system with a null mGluR background. Under conditions that favor mGluR2/4 heterodimer formation, activation of the receptor was not evident with the mGluR2-selective agonist (2S,2'R,3'R)-2-(2',3'-dicarboxycyclopropyl)glycine (DCG-IV) or with the mGluR4 selective agonist L-(+)-2-amino-4-phosphonobutyric acid (L-AP4); however, full activation was apparent when both ligands were applied together, confirming that mGluR dimers require ligand binding in both subunits for full activation. Properties of allosteric modulators were also examined, including the findings that negative allosteric modulators (NAMs) have two binding sites per dimer and that positive allosteric modulators (PAMs) have only a single site per dimer. In SCG neurons, mGluR2/4 dimers were not inhibited by the mGluR2-selective NAM (Z)-1-[2-cycloheptyloxy-2-(2,6-dichlorophenyl)ethenyl]-1H-1,2,4-triazole (Ro 64-5229), supporting the two-site model. Furthermore, application of the mGluR4 selective PAMs N-(4-chloro-3-methoxyphenyl)-2-pyridinecarboxamide (VU0361737) or N-phenyl-7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxamide (PHCCC) and combined application of mGluR4 PAMs with the mGluR2 selective PAM biphenyl indanone-A failed to potentiate glutamate responses through mGluR2/4, suggesting that mGluR2/4 heterodimers are not modulatable by PAMs that are currently available.


Assuntos
Neurônios/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Gânglio Cervical Superior/metabolismo , Regulação Alostérica/efeitos dos fármacos , Animais , Sítios de Ligação , Ciclopropanos/farmacologia , Glicina/análogos & derivados , Glicina/farmacologia , Ligantes , Neurônios/efeitos dos fármacos , Propionatos/farmacologia , Multimerização Proteica , Subunidades Proteicas , Ratos , Ratos Wistar , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Gânglio Cervical Superior/efeitos dos fármacos
17.
BMC Pharmacol ; 12: 6, 2012 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-22642439

RESUMO

BACKGROUND: The efficacy, potency, and selectivity of the compound 2-Chloro-5-hydroxyphenylglycine (CHPG), a nominally selective agonist for metabotropic glutamate receptor 5 (mGluR5), were examined with select mGluRs by examining their ability to induce modulation of the native voltage dependent ion channels in isolated sympathetic neurons from the rat superior cervical ganglion (SCG). SCG neurons offer a null mGluR-background in which specific mGluR subtypes can be made to express via intranuclear cDNA injection. RESULTS: Consistent with previous reports, CHPG strongly activated mGluR5b expressed in SCG neurons with an apparent EC50 around 60 µM. Surprisingly, CHPG also activated two mGluR1 splice variants with a similar potency as at mGluR5 when calcium current inhibition was used as an assay for receptor function. No effect of 1 mM CHPG was seen in cells expressing mGluR2 or mGluR4, suggesting that CHPG only activates group I mGluRs (mGluR1 and 5). CHPG was also able to induce modulation of M-type potassium current through mGluR1, but not as consistently as glutamate. Since this channel is modulated through a Gq-dependent pathway, these data indicate that CHPG may exhibit some biased agonist properties on mGluR1. Closer examination of the voltage-independent, Gq-mediated component of mGluR-induced calcium current modulation data confirmed that some biased agonism was evident, but the effect was weak and inconsistent. CONCLUSIONS: These data contrast with the established literature which suggests that CHPG is a selective mGluR5 agonist. Instead, CHPG appears to act equally well as an agonist at mGluR1. While some weak biased agonism was observed with CHPG acting on mGluR1, but not mGluR5, favoring Gi/o signaling over Gq/11, this effect does not appear sufficient to fully explain the discrepancies in the literature.


Assuntos
Agonistas de Aminoácidos Excitatórios/farmacologia , Glicina/análogos & derivados , Fenilacetatos/farmacologia , Receptores de Glutamato Metabotrópico/agonistas , Animais , Cálcio/fisiologia , Glicina/farmacologia , Masculino , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Ratos , Ratos Wistar , Receptor de Glutamato Metabotrópico 5 , Receptores de Glutamato Metabotrópico/fisiologia , Gânglio Cervical Superior/citologia
18.
Nat Neurosci ; 15(6): 836-44, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22561452

RESUMO

Group I metabotropic glutamate receptors (mGluRs), including mGluR1 and mGluR5, are G protein­coupled receptors (GPCRs) that are expressed at excitatory synapses in brain and spinal cord. GPCRs are often negatively regulated by specific G protein­coupled receptor kinases and subsequent binding of arrestin-like molecules. Here we demonstrate an alternative mechanism in which group I mGluRs are negatively regulated by proline-directed kinases that phosphorylate the binding site for the adaptor protein Homer, and thereby enhance mGluR­Homer binding to reduce signaling. This mechanism is dependent on a multidomain scaffolding protein, Preso1, that binds mGluR, Homer and proline-directed kinases and that is required for their phosphorylation of mGluR at the Homer binding site. Genetic ablation of Preso1 prevents dynamic phosphorylation of mGluR5, and Preso1(−/−) mice exhibit sustained, mGluR5-dependent inflammatory pain that is linked to enhanced mGluR signaling. Preso1 creates a microdomain for proline-directed kinases with broad substrate specificity to phosphorylate mGluR and to mediate negative regulation.


Assuntos
Proteínas de Transporte/metabolismo , Neurônios/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Transdução de Sinais/fisiologia , Sequência de Aminoácidos , Animais , Western Blotting , Encéfalo/metabolismo , Proteínas de Transporte/química , Células HEK293 , Proteínas de Arcabouço Homer , Humanos , Imuno-Histoquímica , Imunoprecipitação , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Fosforilação , Densidade Pós-Sináptica , Proteínas Quinases Direcionadas a Prolina/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Ratos Wistar , Receptores de Glutamato Metabotrópico/química , Transfecção
19.
J Neurophysiol ; 104(1): 439-48, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20463192

RESUMO

Metabotropic glutamate receptors (mGluRs) form covalently linked homodimers and contain large, N-terminal extracellular ligand binding, "venus fly trap" (VFT) domains. These domains, when expressed separately, are secreted as disulfide linked dimers and can dimerize with full-length receptors. mGluR splice variants have been described that contain only this domain, but the consequences of their interaction on receptor signaling have not been explored. Here it is shown that an mGluR1 mutant containing only the VFT is retained on the cell surface when a full-length receptor is co-expressed. Further, when expressed in rat superior cervical ganglion (SCG) neurons and modulation of native calcium currents is used as an assay for receptor activity, the VFT acts as a dominant negative with respect to mGluR1 signaling. Although full-length mGluR1 and mGluR5 are not known to heterodimerize, the mGluR5 VFT partially occludes mGluR1 signaling and the mGluR1 VFT potently occludes mGluR5 signaling in SCG neurons. In addition, an mGluR1 point mutant, mGluR1 C140G, which cannot covalently dimerize, functions like the wild-type receptor when expressed alone. The C140G mutant is inhibited by the mGluR1 VFT construct but does not retain the mGluR1 VFT on the cell surface, suggesting that the loss of C140 renders the interaction reversible. Finally, a peptide designed to disrupt mGluR1 dimerization reduced signaling through the C140G mutant receptor, but only when applied intracellularly for several hours, indicating that loss of signaling requires disruption of dimerization prior to plasma membrane insertion.


Assuntos
Receptores de AMPA/fisiologia , Transdução de Sinais/fisiologia , Sequência de Aminoácidos , Animais , Membrana Celular/metabolismo , DNA/genética , DNA/isolamento & purificação , Dimerização , Espaço Extracelular/metabolismo , Espaço Extracelular/fisiologia , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Plasmídeos/genética , Mutação Puntual/fisiologia , Ligação Proteica , Conformação Proteica , Proteínas Proto-Oncogênicas c-myc/metabolismo , Ratos , Ratos Wistar , Receptor de Glutamato Metabotrópico 5 , Receptores de AMPA/antagonistas & inibidores , Receptores de AMPA/genética , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Gânglio Cervical Superior/citologia , Gânglio Cervical Superior/fisiologia
20.
Eur J Pharmacol ; 627(1-3): 63-8, 2010 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-19879871

RESUMO

Recent studies indicate that the intracellular C-terminus of Group I metabotropic glutamate receptors (mGlu(1) and mGlu(5) receptor) is important in G protein coupling. To determine the necessity of the C-tail, a deletion mutant of mGlu(1) receptor was constructed, which included the first 840 amino acids of the rat mGlu(1a) receptor (mGlu(1)-dCT). G protein coupling of the receptors was assessed by measuring glutamate mediated inhibition of native calcium currents when each receptor was expressed in isolated sympathetic neurons from the rat superior cervical ganglion. Wild type mGlu(1) receptor activates both the Galpha(i/o) and Galpha(q/11) protein families. Each pathway can be detected in superior cervical ganglion neurons as voltage dependent and voltage independent inhibition of the calcium currents, respectively. While wild type mGlu(1) receptor gave rise to a strong, mixed voltage dependent and independent calcium current inhibition, mGlu(1)-dCT exhibited a weaker inhibition that was strongly voltage dependent, indicating activation of Galpha(i/o) was predominant. Further, pertussis toxin treatment reduced the inhibition by wild type mGlu(1) receptor to a smaller, voltage independent inhibition as expected, but completely abolished signaling through mGlu(1)-dCT. Finally, to test whether mGlu(1)-dCT could produce any activation of Galpha(q/11), inhibition of the native superior cervical ganglion M-type potassium currents was examined. M-channels, inhibited by PIP(2) depletion, were strongly inhibited by glutamate in cells expressing wild type mGlu(1) receptor, but no inhibition was detectable in neurons expressing mGlu(1)-dCT. These data indicate that C-terminal deletion of mGlu(1) receptor selectively abolishes Galpha(q/11) coupling.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Receptores de Glutamato Metabotrópico/química , Receptores de Glutamato Metabotrópico/metabolismo , Deleção de Sequência , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo N/metabolismo , Condutividade Elétrica , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Toxina Pertussis/toxicidade , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/metabolismo , Ratos , Ratos Wistar , Receptores de Glutamato Metabotrópico/genética , Especificidade por Substrato , Gânglio Cervical Superior/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...