Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Psychiatry ; 28(2): 588-600, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36357671

RESUMO

Posttraumatic stress disorder (PTSD) after the pandemic has emerged as a major neuropsychiatric component of post-acute COVID-19 syndrome, yet the current pharmacotherapy for PTSD is limited. The use of adrenergic drugs to treat PTSD has been suggested; however, it is hindered by conflicting clinical results and a lack of mechanistic understanding of drug actions. Our studies, using both genetically modified mice and human induced pluripotent stem cell-derived neurons, reveal a novel α2A adrenergic receptor (α2AAR)-spinophilin-cofilin axis in the hippocampus that is critical for regulation of contextual fear memory reconsolidation. In addition, we have found that two α2 ligands, clonidine and guanfacine, exhibit differential abilities in activating this signaling axis to disrupt fear memory reconsolidation. Stimulation of α2AAR with clonidine, but not guanfacine, promotes the interaction of the actin binding protein cofilin with the receptor and with the dendritic spine scaffolding protein spinophilin to induce cofilin activation at the synapse. Spinophilin-dependent regulation of cofilin is required for clonidine-induced disruption of contextual fear memory reconsolidation. Our results inform the interpretation of differential clinical observations of these two drugs on PTSD and suggest that clonidine could provide immediate treatment for PTSD symptoms related to the current pandemic. Furthermore, our study indicates that modulation of dendritic spine morphology may represent an effective strategy for the development of new pharmacotherapies for PTSD.


Assuntos
COVID-19 , Células-Tronco Pluripotentes Induzidas , Animais , Humanos , Camundongos , Fatores de Despolimerização de Actina/farmacologia , Adrenérgicos/farmacologia , Clonidina/farmacologia , Medo/fisiologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas dos Microfilamentos/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo
2.
Hum Cell ; 33(3): 502-511, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32447572

RESUMO

Antipsychotic-induced weight gain is a well-established but poorly understood clinical phenomenon. New mechanistic insights into how antipsychotics modulate adipose physiology are sorely needed, in hopes of either devising a therapeutic intervention to ameliorate weight gain or contributing to improved design of future agents. In this study, we have hypothesized that the weight gain-associated tricyclic antipsychotics clozapine and chlorpromazine directly impact adipose tissue by potentiating adipogenic differentiation of preadipocytes. Utilizing a well-established in vitro model system (3T3-L1 preadipocyte cell line), we demonstrate that, when applied specifically during induction of adipogenic differentiation, both clozapine and chlorpromazine significantly potentiate in vitro adipogenesis, observed as morphological changes and increased intracellular lipid accumulation. These persistent effects, observed at endpoints well after the end of antipsychotic exposure, are accompanied by increased transcript- and protein-level expression of the mature adipocyte marker perilipin-1, as indicated by RT-qPCR and Western blotting, but not by further upregulation of pro-adipogenic transcription factors versus positive controls. Our findings point to a possible physiological mechanism of antipsychotic-induced hyperplasia, with potentiated expression of mature adipocyte markers enhancing the differentiation and maturation of preadipocytes.


Assuntos
Adipócitos/citologia , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Antidepressivos Tricíclicos/efeitos adversos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Células Cultivadas , Humanos , Aumento de Peso/efeitos dos fármacos
3.
PLoS One ; 12(7): e0180638, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28700667

RESUMO

Affective disorders impact nearly 10% of the adult population in the United States in a given year. Synaptic dysfunction has recently emerged as a key neurobiological mechanism underlying affective disorders such as anxiety and depression. In this study, we investigate the potential role of two synaptic scaffolding proteins, neurabin and spinophilin, in regulating anxiety- and depression-related behaviors at different ages using genetically deficient mice. Loss of the neurabin gene reduces anxiety-like behavior in the elevated zero maze in young adult mice (3-5 months old), but not in middle aged mice (11-13 months old), whereas loss of spinophilin decreases anxiety in middle-aged mice, but not in young adult mice. Neurabin knockout (KO) mice also show reduced immobility in the repeated force swim test (FST) at 3-5 months, but not 11-3 months, of age, compared to age- and strain-matched wild type (WT) controls. Conversely, spinophilin KO mice display a lower level of this behavioral despair than matched WT controls after repeated FST trials at the middle age (11-13 months) but not the young age (3-5 months). Together, these data indicate that, despite their structural similarities and overlapping function in regulating synaptic cytoskeleton, the two homologs neurabin and spinophilin play important yet distinct roles in the regulation of anxiety- and depression-like behaviors in an age-dependent manner. Our studies provide new insights into the complex neurobiology of affective disorders.


Assuntos
Envelhecimento/metabolismo , Ansiedade/metabolismo , Comportamento Animal , Depressão/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Análise de Variância , Animais , Imobilização , Aprendizagem em Labirinto , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Proteínas do Tecido Nervoso/genética , Natação
4.
Neuropharmacology ; 116: 38-49, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27956055

RESUMO

The therapeutic mechanism of action underlying many psychopharmacological agents remains poorly understood, due largely to the extreme molecular promiscuity exhibited by these agents with respect to potential central nervous system targets. Agents of the tricyclic chemical class, including both antidepressants and antipsychotics, exhibit a particularly high degree of molecular promiscuity; therefore, any clarification of how these agents interact with specific central nervous system targets is of great potential significance to the field. Here, we present evidence demonstrating that tricyclic antipsychotics appear to segregate into three distinct groups based upon their molecular interactions with the centrally-important α2A adrenergic receptor (AR). Specifically, while the α2AAR binds all antipsychotics tested with similar affinities, and none of the agents are able to induce classical heterotrimeric G protein-mediated α2AAR signaling, significant differences are observed with respect to arrestin3 recruitment and receptor endocytosis. All antipsychotics tested induce arrestin3 recruitment to the α2AAR, but with differing strengths. Both chlorpromazine and clozapine drive significant α2AAR endocytosis, but via differing clathrin-dependent and lipid raft-dependent pathways, while fluphenazine does not drive a robust response. Intriguingly, in silico molecular modeling suggests that each of the three exhibits unique characteristics in interacting with the α2AAR ligand-binding pocket. In addition to establishing these three antipsychotics as novel arrestin-biased ligands at the α2AAR, our findings provide key insights into the molecular actions of these clinically-important agents.


Assuntos
Antipsicóticos/farmacologia , Endocitose/efeitos dos fármacos , Receptores Adrenérgicos alfa 2/metabolismo , beta-Arrestina 1/metabolismo , beta-Arrestina 2/metabolismo , Animais , Caveolina 1/metabolismo , Clorpromazina/farmacologia , Clozapina/farmacologia , AMP Cíclico/metabolismo , Endocitose/fisiologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Flufenazina/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Knockout , Simulação de Acoplamento Molecular , Norepinefrina/farmacologia , beta-Arrestina 1/genética , beta-Arrestina 2/genética
5.
Prog Mol Biol Transl Sci ; 132: 207-25, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26055060

RESUMO

Antidepressant drugs remain poorly understood, especially with respect to pharmacological mechanisms of action. This lack of knowledge results from the extreme complexity inherent to psychopharmacology, as well as to a corresponding lack of knowledge regarding depressive disorder pathophysiology. While the final analysis is likely to be multifactorial and heterogeneous, compelling evidence exists for upregulation of brain α2 adrenergic receptors (ARs) in depressed patients. This evidence has sparked a line of research into actions of a particular antidepressant drug class, the tricyclic antidepressants (TCAs), as direct ligands at α(2A)ARs. Our findings, as outlined herein, demonstrate that TCAs function as arrestin-biased ligands at α(2A)ARs. Importantly, TCA-induced α(2A)AR/arrestin recruitment leads to receptor endocytosis and downregulation of α(2A)AR expression with prolonged exposure. These findings represent a novel mechanism linking α(2)AR trafficking with antidepressant pharmacology.


Assuntos
Antidepressivos Tricíclicos/uso terapêutico , Depressão/tratamento farmacológico , Receptores Adrenérgicos alfa 2/fisiologia , Animais , Antidepressivos/uso terapêutico , Antidepressivos Tricíclicos/química , Encéfalo/embriologia , Movimento Celular , Desenho de Fármacos , Endocitose , Humanos , Ligantes , Camundongos , Transporte Proteico , Transdução de Sinais , Regulação para Cima
6.
Biochem Biophys Res Commun ; 451(3): 461-6, 2014 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-25128275

RESUMO

Antidepressant mechanisms of action remain shrouded in mystery, greatly hindering our ability to develop therapeutics which can fully treat patients suffering from depressive disorders. In an attempt to shed new light on this topic, we have undertaken a series of studies investigating actions of tricyclic antidepressant drugs (TCAs) at the α2A adrenergic receptor (AR), a centrally important receptor, dysregulation of which has been linked to depression. Our previous work established a particular TCA, desipramine, as an arrestin-biased α2AAR ligand driving receptor endocytosis and downregulation but not canonical heterotrimeric G protein-mediated signaling. The present work is aimed at broadening our understanding of how members of the TCA drug class act at the α2AAR, as we have selected the closely related but subtly different TCAs imipramine and amitriptyline for evaluation. Our data demonstrate that these drugs do also function as direct arrestin-biased α2AAR ligands. However, these data reveal differences in receptor affinity and in the extent/nature of arrestin recruitment to and endocytosis of α2AARs. Specifically, amitriptyline exhibits an approximately 14-fold stronger interaction with the receptor, is a weaker driver of arrestin recruitment, and preferentially recruits a different arrestin subtype. Extent of endocytosis is similar for all TCAs studied so far, and occurs in an arrestin-dependent manner, although imipramine uniquely retains a slight ability to drive α2AAR endocytosis in arrestin-null cells. These findings signify an important expansion of our mechanistic understanding of antidepressant pharmacology, and provide useful insights for future medicinal chemistry efforts.


Assuntos
Antidepressivos Tricíclicos/farmacologia , Arrestinas/fisiologia , Endocitose/efeitos dos fármacos , Receptores Adrenérgicos alfa 2/efeitos dos fármacos , Amitriptilina/farmacologia , Animais , Desipramina/farmacologia , Células HEK293 , Humanos , Imipramina/farmacologia , Camundongos , Receptores Adrenérgicos alfa 2/metabolismo
7.
J Biol Chem ; 288(40): 29193-205, 2013 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-23965992

RESUMO

Inter-regulation of adrenergic receptors (ARs) via cross-talk is a long appreciated but mechanistically unclear physiological phenomenon. Evidence from the AR literature and our own extensive studies on regulation of α2AARs by the scaffolding protein spinophilin have illuminated a potential novel mechanism for cross-talk from ß to α2ARs. In the present study, we have characterized a mode of endogenous AR cross-talk in native adrenergic neurons whereby canonical ßAR-mediated signaling modulates spinophilin-regulated α2AAR endocytosis through PKA. Our findings demonstrate that co-activation of ß and α2AARs, either by application of endogenous agonist or by simultaneous stimulation with distinct selective agonists, results in acceleration of endogenous α2AAR endocytosis in native neurons. We show that receptor-independent PKA activation by forskolin is sufficient to accelerate α2AAR endocytosis and that α2AAR stimulation alone drives accelerated endocytosis in spinophilin-null neurons. Endocytic response acceleration by ß/α2AAR co-activation is blocked by PKA inhibition and lost in spinophilin-null neurons, consistent with our previous finding that spinophilin is a substrate for phosphorylation by PKA that disrupts its interaction with α2AARs. Importantly, we show that α2AR agonist-mediated α2AAR/spinophilin interaction is blocked by ßAR co-activation in a PKA-dependent fashion. We therefore propose a novel mechanism for cross-talk from ß to α2ARs, whereby canonical ßAR-mediated signaling coupled to PKA activation results in phosphorylation of spinophilin, disrupting its interaction with α2AARs and accelerating α2AAR endocytic responses. This mechanism of cross-talk has significant implications for endogenous adrenergic physiology and for therapeutic targeting of ß and α2AARs.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Endocitose , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Receptores Adrenérgicos alfa 2/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Arrestinas/metabolismo , Colforsina/farmacologia , Embrião de Mamíferos/citologia , Endocitose/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Epinefrina/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Isoproterenol/farmacologia , Camundongos , Proteínas dos Microfilamentos/deficiência , Modelos Biológicos , Proteínas Mutantes/metabolismo , Proteínas do Tecido Nervoso/deficiência , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sistema Nervoso Simpático/citologia
8.
Neurosci Biobehav Rev ; 36(10): 2214-25, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22910678

RESUMO

Dysfunction in noradrenergic neurotransmission has long been theorized to occur in depressive disorders. The α2 adrenergic receptor (AR) family, as a group of key players in regulating the noradrenergic system, has been investigated for involvement in the neurobiology of depression and mechanisms of antidepressant therapies. However, a clear picture of the α2ARs in depressive disorders has not been established due to the existence of apparently conflicting findings in the literature. In this article, we report that a careful accounting of methodological differences within the literature can resolve the present lack of consensus on involvement of α2ARs in depression. In particular, the pharmacological properties of the radioligand (e.g. agonist versus antagonist) utilized for determining receptor density are crucial in determining study outcome. Upregulation of α2AR density detected by radiolabeled agonists but not by antagonists in patients with depressive disorders suggests a selective increase in the density of high-affinity conformational state α2ARs, which is indicative of enhanced G protein coupling to the receptor. Importantly, this high-affinity state α2AR upregulation can be normalized with antidepressant treatments. Thus, depressive disorders appear to be associated with increased α2AR sensitivity and responsiveness, which may represent a physiological basis for the putative noradrenergic dysfunction in depressive disorders. In addition, we review changes in some key α2AR accessory proteins in depressive disorders and discuss their potential contribution to α2AR dysfunction.


Assuntos
Antidepressivos/uso terapêutico , Transtorno Depressivo/tratamento farmacológico , Transtorno Depressivo/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Modelos Biológicos , Neurobiologia , Receptores Adrenérgicos alfa 2/genética
9.
Biochem Biophys Res Commun ; 420(1): 161-5, 2012 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-22405824

RESUMO

The precise physiological effects of antidepressant drugs, and in particular their actions at non-monoamine transporter targets, are largely unknown. We have recently identified the tricyclic antidepressant drug desipramine (DMI) as a direct ligand at the α(2A) adrenergic receptor (AR) without itself driving heterotrimeric G protein/downstream effector activation [5]. In this study, we report our novel finding that DMI modulates α(2A)AR signaling in response to the endogenous agonist norepinephrine (NE). DMI acted as a signaling potentiator, selectively enhancing NE-induced α(2A)AR-mediated ERK1/2 MAPK signaling. This potentiation of ERK1/2 activation was observed as an increase in NE response sensitivity and a prolongation of the activation kinetics. DMI in a physiologically relevant ratio with NE effectively turned on ERK1/2 signaling that is lacking in response to physiological NE alone. Further, the DMI-induced ERK1/2 potentiation relied on heterotrimeric G(i/o) proteins and was arrestin-independent. This modulatory effect of DMI on NE signaling provides novel insight into the effects of this antidepressant drug on the noradrenergic system which it regulates, insight which enhances our understanding of the therapeutic mechanism for DMI.


Assuntos
Inibidores da Captação Adrenérgica/farmacologia , Antidepressivos Tricíclicos/farmacologia , Desipramina/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/biossíntese , Proteína Quinase 3 Ativada por Mitógeno/biossíntese , Receptores Adrenérgicos alfa 2/metabolismo , Animais , Arrestina/genética , Arrestina/metabolismo , Células Cultivadas , Sinergismo Farmacológico , Ativação Enzimática , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Norepinefrina/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
10.
Neuropharmacology ; 62(7): 2354-62, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22369787

RESUMO

Many antidepressant drugs, including the tricyclic antidepressant desipramine (DMI), are broadly understood to function by modulating central noradrenergic neurotransmission. α(2) adrenergic receptors (α(2)ARs) are key regulators of the noradrenergic system, and previous work has implicated α(2)ARs in mediating the antidepressant activity of DMI in the rodent forced swim test (FST). However, little is known about intracellular regulators of antidepressant drug action. α(2)AR function is tightly regulated by its intracellular interacting partners arrestin and the dendritic protein spinophilin. We have previously established the competitive and reciprocal nature of these interacting proteins at the α(2)AR in the context of classic agonist effects, and have shown DMI to be a direct arrestin-biased ligand at the receptor. In the present study, we report that mice deficient in the α(2A)AR subtype lack DMI-induced antidepressant behavioral effects in the FST. As well, mice deficient in arrestin3 lack antidepressant response to DMI, while spinophilin-null mice have enhanced antidepressant response to DMI compared with wild-type controls, indicating that this α(2A)AR-mediated response is reciprocally regulated by arrestin and spinophilin. The characteristic of α(2A)AR-dependence and arrestin3 involvement was shared by the antidepressant effect of the classic α(2)AR agonist clonidine but not the non-tricyclic norepinephrine reuptake inhibitor reboxetine, supporting a model whereby DMI exerts its antidepressant effect through direct engagement of the α(2A)AR and arrestin3. Our results implicate arrestin- and spinophilin-mediated regulation of the α(2A)AR in the pharmacology of the noradrenergic antidepressant DMI, and suggest that manipulation of this mode of receptor regulation may represent a novel and viable therapeutic strategy.


Assuntos
Neurônios Adrenérgicos/fisiologia , Antidepressivos/farmacologia , Arrestinas/fisiologia , Desipramina/farmacologia , Proteínas dos Microfilamentos/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Receptores Adrenérgicos alfa 2/fisiologia , Neurônios Adrenérgicos/efeitos dos fármacos , Inibidores da Captação Adrenérgica/farmacologia , Animais , Imobilização/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Natação/fisiologia
11.
J Neurosci ; 32(8): 2683-95, 2012 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-22357852

RESUMO

Endogenous adenosine is an essential protective agent against neural damage by various insults to the brain. However, the therapeutic potential of adenosine receptor-directed ligands for neuroprotection is offset by side effects in peripheral tissues and organs. An increase in adenosine receptor responsiveness to endogenous adenosine would enhance neuroprotection while avoiding the confounding effects of exogenous ligands. Here we report novel regulation of adenosine-evoked responses by a neural tissue-specific protein, neurabin. Neurabin attenuated adenosine A(1) receptor (A1R) signaling by assembling a complex between the A1R and the regulator of G-protein signaling 4 (RGS4), a protein known to turn off G-protein signaling. Inactivation of the neurabin gene enhanced A1R signaling and promoted the protective effect of adenosine against excitotoxic seizure and neuronal death in mice. Furthermore, administration of a small molecule inhibitor of RGS4 significantly attenuated seizure severity in mice. Notably, the dose of kainate capable of inducing an ∼50% rate of death in wild-type (WT) mice did not affect neurabin-null mice or WT mice cotreated with an RGS4 inhibitor. The enhanced anti-seizure and neuroprotective effect achieved by disruption of the A1R/neurabin/RGS4 complex is elicited by the on-site and on-demand release of endogenous adenosine, and does not require administration of A1R ligands. These data identify neurabin-RGS4 as a novel tissue-selective regulatory mechanism for fine-tuning adenosine receptor function in the nervous system. Moreover, these findings implicate the A1R/neurabin/RGS4 complex as a valid therapeutic target for specifically manipulating the neuroprotective effects of endogenous adenosine.


Assuntos
Adenosina/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas RGS/metabolismo , Convulsões/metabolismo , Antagonistas do Receptor A1 de Adenosina/farmacologia , Animais , Animais Recém-Nascidos , Morte Celular/efeitos dos fármacos , Células Cultivadas , Cricetinae , Cricetulus , Modelos Animais de Doenças , Eletroencefalografia , Fluoresceínas , Hipocampo/patologia , Marcação In Situ das Extremidades Cortadas , Ácido Caínico/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/deficiência , Modelos Biológicos , Proteínas do Tecido Nervoso/deficiência , Compostos Orgânicos/metabolismo , Fenilisopropiladenosina/farmacologia , Proteínas RGS/antagonistas & inibidores , Receptor A1 de Adenosina/genética , Receptor A1 de Adenosina/metabolismo , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico , Convulsões/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sulfonamidas/farmacologia , Fatores de Tempo , Transfecção , Xantinas/farmacologia , Xantinas/uso terapêutico
12.
J Biol Chem ; 286(41): 36063-36075, 2011 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-21859713

RESUMO

The neurobiological mechanisms of action underlying antidepressant drugs remain poorly understood. Desipramine (DMI) is an antidepressant classically characterized as an inhibitor of norepinephrine reuptake. Available evidence, however, suggests a mechanism more complex than simple reuptake inhibition. In the present study, we have characterized the direct interaction between DMI and the α(2A)-adrenergic receptor (α(2A)AR), a key regulator of noradrenergic neurotransmission with altered expression and function in depression. DMI alone was found to be sufficient to drive receptor internalization acutely and a robust down-regulation of α(2A)AR expression and signaling following prolonged stimulation in vitro. These effects are achieved through arrestin-biased regulation of the receptor, as DMI selectively induces recruitment of arrestin but not activation of heterotrimeric G proteins. Meanwhile, a physiologically relevant concentration of endogenous agonist (norepinephrine) was unable to sustain a down-regulation response. Prolonged in vivo administration of DMI resulted in significant down-regulation of synaptic α(2A)AR expression, a response that was lost in arrestin3-null animals. We contend that direct DMI-driven arrestin-mediated α(2A)AR down-regulation accounts for the therapeutically desirable but mechanistically unexplained adaptive alterations in receptor expression associated with this antidepressant. Our results provide novel insight into both the pharmacology of this antidepressant drug and the targeting of the α(2A)AR in depression.


Assuntos
Antidepressivos Tricíclicos/farmacologia , Arrestinas/metabolismo , Desipramina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Receptores Adrenérgicos alfa 2/biossíntese , Transmissão Sináptica/efeitos dos fármacos , Animais , Arrestinas/genética , Depressão/tratamento farmacológico , Depressão/genética , Depressão/metabolismo , Regulação para Baixo/genética , Sistemas de Liberação de Medicamentos , Células HEK293 , Proteínas Heterotriméricas de Ligação ao GTP/genética , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Humanos , Camundongos , Camundongos Knockout , Transmissão Sináptica/genética
14.
J Biol Chem ; 283(21): 14516-23, 2008 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-18367453

RESUMO

Spinophilin plays critical roles in regulating trafficking and signaling of the alpha(2)-adrenergic receptor (AR) both in vitro and in vivo (Wang, Q., Zhao, J., Brady, A. E., Feng, J., Allen, P. B., Lefkowitz, R. J., Greengard, P., and Limbird, L. E. (2004) Science 304, 1940-1944). In the present study, we demonstrate that protein kinase A (PKA) phosphorylation of spinophilin modulates the spinophilin-alpha(2A)AR interaction to regulate alpha(2A)AR internalization. Activation of PKA by forskolin abolishes the agonist-enhanced interaction between spinophilin and the alpha(2A)AR, and this event can be blocked by Ser --> Ala mutations at the PKA phosphorylation sites of spinophilin. In addition, a Ser --> Asp mutation that mimics the phosphorylated state at the PKA phosphorylation site Ser-177, which is located within the alpha(2A)AR binding region of spinophilin, is sufficient to block the spinophilin-alpha(2A)AR interaction in intact cells. In cells expressing mutant spinophilin carrying the S177D mutation, agonist-induced internalization of the alpha(2A)AR is accelerated and enhanced, as revealed by both intact cell enzyme-linked immunosorbent assay and quantitative immunofluorescent studies. Furthermore, activation of PKA by forskolin enhances agonist-induced internalization of the alpha(2A)AR in cells expressing wild type spinophilin, but not in cells lacking spinophilin or expressing the spinophilin mutant Sp177D. These results strongly support that PKA phosphorylation of spinophilin is functionally relevant in regulating alpha(2A)AR trafficking. Therefore, modulation of spinophilin-receptor interaction through phosphorylation of spinophilin may represent a novel mechanism whereby PKA regulates G protein-coupled receptor trafficking.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Animais , Membrana Celular/metabolismo , Células Cultivadas , Colforsina/farmacologia , Regulação da Expressão Gênica , Camundongos , Proteínas dos Microfilamentos/agonistas , Proteínas dos Microfilamentos/genética , Mutação/genética , Proteínas do Tecido Nervoso/agonistas , Proteínas do Tecido Nervoso/genética , Fosforilação/efeitos dos fármacos , Ligação Proteica , Receptores Adrenérgicos alfa 2/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...