Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Biol Chem ; 294(14): 5521-5535, 2019 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-30737285

RESUMO

AMPA-type glutamate receptors (AMPARs) mediate fast excitatory neurotransmission in the mammalian central nervous system. Preferential AMPAR subunit assembly favors heteromeric GluA1/GluA2 complexes. The presence of the GluA2 subunit generates Ca2+-impermeable (CI) AMPARs that have linear current-voltage (I-V) relationships. However, diverse forms of synaptic plasticity and pathophysiological conditions are associated with shifts from CI to inwardly rectifying, GluA2-lacking, Ca2+-permeable (CP) AMPARs on time scales ranging from minutes to days. These shifts have been linked to GluA1 phosphorylation at Ser-845, a protein kinase A (PKA)-targeted site within its intracellular C-terminal tail, often in conjunction with protein kinase A anchoring protein 79 (AKAP79; AKAP150 in rodents), which targets PKA to GluA1. However, AKAP79 may impact GluA1 phosphorylation at other sites by interacting with other signaling enzymes. Here, we evaluated the ability of AKAP79, its signaling components, and GluA1 phosphorylation sites to induce CP-AMPARs under conditions in which CI-AMPARs normally predominate. We found that GluA1 phosphorylation at Ser-831 is sufficient for the appearance of CP-AMPARs and that AKAP79-anchored protein kinase C (PKC) primarily drives the appearance of these receptors via this site. In contrast, other AKAP79-signaling components and C-terminal tail GluA1 phosphorylation sites exhibited a permissive role, limiting the extent to which AKAP79 promotes CP-AMPARs. This may reflect the need for these sites to undergo active phosphorylation/dephosphorylation cycles that control their residency within distinct subcellular compartments. These findings suggest that AKAP79, by orchestrating phosphorylation, represents a key to a GluA1 phosphorylation passcode, which allows the GluA1 subunit to escape GluA2 dominance and promote the appearance of CP-AMPARs.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Proteína Quinase C/metabolismo , Receptores de AMPA/metabolismo , Transdução de Sinais , Proteínas de Ancoragem à Quinase A/genética , Células HEK293 , Humanos , Fosforilação , Proteína Quinase C/genética , Receptores de AMPA/genética
2.
Mol Cell Neurosci ; 79: 45-52, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27998718

RESUMO

Some forms of long-term synaptic plasticity require docking of Ca2+/calmodulin-dependent protein kinase II α (CaMKIIα) to residues 1290-1309 within the intracellular C-terminal tail of the N-methyl-d-aspartate (NMDA) receptor GluN2B subunit. The phosphorylation of Ser1303 within this region destabilizes CaMKII binding. Interestingly, Ser1303 is a substrate for CaMKII itself, as well as PKC and DAPK1, but these kinases have been reported to have contradictory effects on the activity of GluN2B-containing NMDA receptors. Here, we re-assessed the effect of CaMKII on NMDA receptor desensitization in heterologous cells, as measured by the ratio of steady-state to peak currents induced during 3s agonist applications. CaMKIIα co-expression or infusion of constitutively active CaMKII limits the extent of desensitization and preserves current amplitude with repeated stimulation of recombinant GluN1A/GluN2B when examined using low intracellular chloride (Cl-) levels, characteristic of neurons beyond the first postnatal week. In contrast, CaMKIIα enhances the acute rate and extent of desensitization when intracellular Cl- concentrations are high. The apparent dependence of CaMKIIα effects on NMDA receptor desensitization on Cl- concentrations is consistent with the presence of a Ca2+-activated Cl- conductance endogenous to HEK 293 cells, which was confirmed by photolysis of caged-Ca2+. However, Ca2+-activated Cl- conductances are unaffected by CaMKIIα expression, indicating that CaMKII affects agonist-induced whole cell currents via modulation of the NMDA receptor. In support of this idea, CaMKIIα modulation of GluN2B-NMDA receptors is abrogated by the phospho-null mutation of Ser1303 in GluN2B to alanine and occluded by phospho-mimetic mutation of Ser1303 to aspartate regardless of intracellular Cl- concentration. Thus, CaMKII-mediated phosphorylation of GluN2B-containing NMDA receptors reduces desensitization at physiological (low) intracellular Cl-, perhaps serving as a feed-forward mechanism to sustain NMDA-mediated Ca2+ entry and continued CaMKII activation during learning and memory.


Assuntos
Potenciais de Ação , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Processamento de Proteína Pós-Traducional , Receptores de N-Metil-D-Aspartato/metabolismo , Sinalização do Cálcio , Cloretos/metabolismo , Células HEK293 , Humanos , Fosforilação , Ligação Proteica
3.
Mol Pharmacol ; 88(4): 728-35, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26199377

RESUMO

PKMζ is generated via an alternative transcriptional start site in the atypical protein kinase C (PKC)ζ isoform, which removes N-terminal regulatory elements, including the inhibitory pseudosubstrate domain, consequently rendering the kinase constitutively active. Persistent PKMζ activity has been proposed as a molecular mechanism for the long-term maintenance of synaptic plasticity underlying some forms of memory. Many studies supporting a role for PKMζ in synaptic plasticity and memory have relied on the PKCζ pseudosubstrate-derived ζ-inhibitory peptide (ZIP). However, recent studies have demonstrated that ZIP-induced impairments to synaptic plasticity and memory occur even in the absence of PKCζ, suggesting that ZIP exerts its actions via additional cellular targets. In this study, we demonstrated that ZIP interacts with conventional and novel PKC, in addition to atypical PKC isoforms. Moreover, when brain abundance of each PKC isoform and affinity for ZIP are taken into account, the signaling capacity of ZIP-responsive pools of conventional and novel PKCs may match or exceed that for atypical PKCs. Pseudosubstrate-derived peptides, like ZIP, are thought to exert their cellular action primarily by inhibiting PKC catalytic activity; however, the ZIP-sensitive catalytic core of PKC is known to participate in the enzyme's subcellular targeting, suggesting an additional mode of ZIP action. Indeed, we have demonstrated that ZIP potently disrupts PKCα interaction with the PKC-targeting protein A-kinase anchoring protein (AKAP) 79 and interferes with ionomycin-induced translocation of conventional PKC to the plasma membrane. Thus, ZIP exhibits broad-spectrum action toward the PKC family of enzymes, and this action may contribute to its unique ability to impair memory.


Assuntos
Encéfalo/metabolismo , Proteínas da Gravidez/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Proteínas da Gravidez/farmacologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Sprague-Dawley , Especificidade por Substrato/efeitos dos fármacos , Especificidade por Substrato/fisiologia
4.
J Neurosci ; 33(9): 4151-64, 2013 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-23447623

RESUMO

Selective control of receptor trafficking provides a mechanism for remodeling the receptor composition of excitatory synapses, and thus supports synaptic transmission, plasticity, and development. GluN3A (formerly NR3A) is a nonconventional member of the NMDA receptor (NMDAR) subunit family, which endows NMDAR channels with low calcium permeability and reduced magnesium sensitivity compared with NMDARs comprising only GluN1 and GluN2 subunits. Because of these special properties, GluN3A subunits act as a molecular brake to limit the plasticity and maturation of excitatory synapses, pointing toward GluN3A removal as a critical step in the development of neuronal circuitry. However, the molecular signals mediating GluN3A endocytic removal remain unclear. Here we define a novel endocytic motif (YWL), which is located within the cytoplasmic C-terminal tail of GluN3A and mediates its binding to the clathrin adaptor AP2. Alanine mutations within the GluN3A endocytic motif inhibited clathrin-dependent internalization and led to accumulation of GluN3A-containing NMDARs at the cell surface, whereas mimicking phosphorylation of the tyrosine residue promoted internalization and reduced cell-surface expression as shown by immunocytochemical and electrophysiological approaches in recombinant systems and rat neurons in primary culture. We further demonstrate that the tyrosine residue is phosphorylated by Src family kinases, and that Src-activation limits surface GluN3A expression in neurons. Together, our results identify a new molecular signal for GluN3A internalization that couples the functional surface expression of GluN3A-containing receptors to the phosphorylation state of GluN3A subunits, and provides a molecular framework for the regulation of NMDAR subunit composition with implications for synaptic plasticity and neurodevelopment.


Assuntos
Endocitose/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/química , Receptores de N-Metil-D-Aspartato/metabolismo , Tirosina/metabolismo , Trifosfato de Adenosina/farmacocinética , Motivos de Aminoácidos/efeitos dos fármacos , Motivos de Aminoácidos/genética , Análise de Variância , Animais , Biofísica , Biotinilação , Células Cultivadas , Córtex Cerebral/citologia , Chlorocebus aethiops , Clatrina/farmacologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Estimulação Elétrica , Embrião de Mamíferos , Endocitose/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Ácido Glutâmico/farmacologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/citologia , Humanos , Imunoprecipitação , Mutagênese/fisiologia , Mutação/fisiologia , Neurônios/efeitos dos fármacos , Neurotransmissores/farmacologia , Técnicas de Patch-Clamp , Isótopos de Fósforo/farmacocinética , Fosforilação/efeitos dos fármacos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Conformação Proteica , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Receptores de N-Metil-D-Aspartato/genética , Transfecção , Transferrina/metabolismo
5.
J Biol Chem ; 286(8): 6697-706, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21156788

RESUMO

GluA1 (formerly GluR1) AMPA receptor subunit phosphorylation at Ser-831 is an early biochemical marker for long-term potentiation and learning. This site is a substrate for Ca(2+)/calmodulin (CaM)-dependent protein kinase II (CaMKII) and protein kinase C (PKC). By directing PKC to GluA1, A-kinase anchoring protein 79 (AKAP79) facilitates Ser-831 phosphorylation and makes PKC a more potent regulator of GluA1 than CaMKII. PKC and CaM bind to residues 31-52 of AKAP79 in a competitive manner. Here, we demonstrate that common CaMKII inhibitors alter PKC and CaM interactions with AKAP79(31-52). Most notably, the classical CaMKII inhibitors KN-93 and KN-62 potently enhanced the association of CaM to AKAP79(31-52) in the absence (apoCaM) but not the presence of Ca(2+). In contrast, apoCaM association to AKAP79(31-52) was unaffected by the control compound KN-92 or a mechanistically distinct CaMKII inhibitor (CaMKIINtide). In vitro studies demonstrated that KN-62 and KN-93, but not the other compounds, led to apoCaM-dependent displacement of PKC from AKAP79(31-52). In the absence of CaMKII activation, complementary cellular studies revealed that KN-62 and KN-93, but not KN-92 or CaMKIINtide, inhibited PKC-mediated phosphorylation of GluA1 in hippocampal neurons as well as AKAP79-dependent PKC-mediated augmentation of recombinant GluA1 currents. Buffering cellular CaM attenuated the ability of KN-62 and KN-93 to inhibit AKAP79-anchored PKC regulation of GluA1. Therefore, by favoring apoCaM binding to AKAP79, KN-62 and KN-93 derail the ability of AKAP79 to efficiently recruit PKC for regulation of GluA1. Thus, AKAP79 endows PKC with a pharmacological profile that overlaps with CaMKII.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Receptores de AMPA/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas de Ancoragem à Quinase A/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Ativação Enzimática/efeitos dos fármacos , Células HEK293 , Hipocampo/metabolismo , Humanos , Neurônios/metabolismo , Fosforilação/efeitos dos fármacos , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Receptores de AMPA/genética
6.
Biochemistry ; 50(23): 5279-91, 2011 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-21561082

RESUMO

The ubiquitously expressed and highly promiscuous protein phosphatase 1 (PP1) regulates many cellular processes. Targeting PP1 to specific locations within the cell allows for the regulation of PP1 by conferring substrate specificity. In the present study, we identified AKAP79 as a novel PP1 regulatory subunit. Immunoprecipitaiton of the AKAP from rat brain extract found that the PP1 catalytic subunit copurified with the anchoring protein. This is a direct interaction, demonstrated by pulldown experiments using purified proteins. Interestingly, the addition of AKAP79 to purified PP1 catalytic subunit decreased phosphatase activity with an IC(50) of 811 ± 0.56 nM of the anchoring protein. Analysis of AKAP79 identified a PP1 binding site that conformed to a consensus PP1 binding motif (FxxR/KxR/K) in the first 44 amino acids of the anchoring protein. This was confirmed when a peptide mimicking this region of AKAP79 was able to bind PP1 by both pulldown assay and surface plasmon resonance. However, PP1 was still able to bind to AKAP79 upon deletion of this region, suggesting additional sites of contact between the anchoring protein and the phosphatase. Importantly, this consensus PP1 binding motif was found not to be responsible for PP1 inhibition, but rather enhanced phosphatase activity, as deletion of this domain resulted in an increased inhibition of PP1 activity. Instead, a second interaction domain localized to residues 150-250 of AKAP79 was required for the inhibition of PP1. However, the inhibitory actions of AKAP79 on PP1 are substrate dependent, as the anchoring protein did not inhibit PP1 dephosphorylation of phospho-PSD-95, a substrate found in AKAP79 complexes in the brain. These combined observations suggest that AKAP79 acts as a PP1 regulatory subunit that can direct PP1 activity toward specific targets in the AKAP79 complex.


Assuntos
Proteínas de Ancoragem à Quinase A/química , Proteína Fosfatase 1/química , Proteínas de Ancoragem à Quinase A/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Catálise , Células Cultivadas , Dados de Sequência Molecular , Proteína Fosfatase 1/metabolismo , Estrutura Terciária de Proteína , Ratos , Ressonância de Plasmônio de Superfície
7.
J Biol Chem ; 285(2): 923-34, 2010 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-19858198

RESUMO

Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) promotes trafficking and activation of the GluR1 subunit of alpha-amino- 3-hydroxy-5-methyl-4-isoxazolepropionic acid-type glutamate receptors (AMPARs) during synaptic plasticity. GluR1 is also modulated in parallel by multiprotein complexes coordinated by synapse-associated protein 97 (SAP97) that contain A-kinase anchoring protein 79/150 (AKAP79/150), protein kinase A, and protein phosphatase 2B. Here we show that SAP97 is present in CaMKII immune complexes isolated from rodent brain as well as from HEK293 cells co-expressing CaMKIIalpha and SAP97. CaMKIIalpha phosphorylated recombinant SAP97 within immune complexes in vitro and in intact cells. Four alternative mRNA splice variants of SAP97 expressing combinations of four inserts (I2, I3, I4, I5) in the U5 region between Src homology 3 (SH3) and guanylyl kinase-like (GK) domains were identified in rat brain at postnatal day 21. CaMKIIalpha preferentially phosphorylated a full-length SAP97 and a glutathione S-transferase (GST) fusion protein containing the I3 and I5 inserts (SAP97-I3I5 and GST-SH3-I3I5-GK, respectively) and also specifically interacted with GST-SH3-I3I5-GK compared with GST proteins containing other naturally occurring insert combinations. AKAP79/150 also directly and specifically bound only to GST-SH3-I3I5-GK, but CaMKII phosphorylation of GST-SH3-I3I5-GK prevented this interaction. AKAP79-dependent down-regulation of GluR1 AMPAR currents was ablated by overexpression of SAP97-I2I5 (which does not bind AKAP79) or by infusion of active CaMKIIalpha. Collectively, the data suggest that CaMKIIalpha targets a specific SAP97 splice variant to disengage AKAP79/150 from regulating GluR1 AMPARs, providing new insight into protein-protein interactions and phosphorylation events that are required for normal regulation of glutamatergic synaptic transmission, learning, and memory.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Proteínas de Membrana/metabolismo , Receptores de AMPA/metabolismo , Sinapses/metabolismo , Transmissão Sináptica/fisiologia , Proteínas de Ancoragem à Quinase A/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Encéfalo/metabolismo , Calcineurina/genética , Calcineurina/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Linhagem Celular , Proteína 1 Homóloga a Discs-Large , Guanilato Quinases , Humanos , Aprendizagem/fisiologia , Proteínas de Membrana/genética , Camundongos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Fosforilação/fisiologia , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína/fisiologia , Ratos , Receptores de AMPA/genética , Sinapses/genética
8.
Elife ; 102021 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-34787081

RESUMO

De novo protein synthesis is required for synapse modifications underlying stable memory encoding. Yet neurons are highly compartmentalized cells and how protein synthesis can be regulated at the synapse level is unknown. Here, we characterize neuronal signaling complexes formed by the postsynaptic scaffold GIT1, the mechanistic target of rapamycin (mTOR) kinase, and Raptor that couple synaptic stimuli to mTOR-dependent protein synthesis; and identify NMDA receptors containing GluN3A subunits as key negative regulators of GIT1 binding to mTOR. Disruption of GIT1/mTOR complexes by enhancing GluN3A expression or silencing GIT1 inhibits synaptic mTOR activation and restricts the mTOR-dependent translation of specific activity-regulated mRNAs. Conversely, GluN3A removal enables complex formation, potentiates mTOR-dependent protein synthesis, and facilitates the consolidation of associative and spatial memories in mice. The memory enhancement becomes evident with light or spaced training, can be achieved by selectively deleting GluN3A from excitatory neurons during adulthood, and does not compromise other aspects of cognition such as memory flexibility or extinction. Our findings provide mechanistic insight into synaptic translational control and reveal a potentially selective target for cognitive enhancement.


Assuntos
Memória/fisiologia , Biossíntese de Proteínas/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Feminino , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução de Sinais
9.
Nat Neurosci ; 9(5): 611-21, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16617342

RESUMO

A key step in glutamatergic synapse maturation is the replacement of developmentally expressed N-methyl-D-aspartate receptors (NMDARs) with mature forms that differ in subunit composition, electrophysiological properties and propensity to elicit synaptic plasticity. However, the mechanisms underlying the removal and replacement of synaptic NMDARs are poorly understood. Here we demonstrate that NMDARs containing the developmentally regulated NR3A subunit undergo rapid endocytosis from the dendritic plasma membrane in cultured rat hippocampal neurons. This endocytic removal is regulated by PACSIN1/syndapin1, which directly and selectively binds the carboxy-terminal domain of NR3A through its NPF motifs and assembles a complex of proteins including dynamin and clathrin. Endocytosis of NR3A by PACSIN1 is activity dependent, and disruption of PACSIN1 function causes NR3A accumulation at synaptic sites. Our results reveal a new activity-dependent mechanism involved in the regulation of NMDAR expression at synapses during development, and identify a brain-specific endocytic adaptor that confers spatiotemporal and subunit specificity to NMDAR endocytosis.


Assuntos
Endocitose/fisiologia , Glicoproteínas de Membrana/fisiologia , Neurônios/citologia , Proteoglicanas/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/metabolismo , Animais , Western Blotting/métodos , Células Cultivadas , Clonagem Molecular/métodos , Eletrofisiologia/métodos , Embrião de Mamíferos , Endocitose/efeitos dos fármacos , Imunofluorescência/métodos , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/citologia , Humanos , Hibridização In Situ/métodos , Glicoproteínas de Membrana/genética , Microscopia Imunoeletrônica/métodos , Mutação/fisiologia , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Proteoglicanas/genética , Ratos , Receptores de N-Metil-D-Aspartato/genética , Sinapses/efeitos dos fármacos , Sinapses/ultraestrutura , Sindecanas , Transfecção/métodos , Técnicas do Sistema de Duplo-Híbrido
10.
Neuron ; 97(2): 368-377.e3, 2018 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-29346754

RESUMO

Preservation of a balance between synaptic excitation and inhibition is critical for normal brain function. A number of homeostatic cellular mechanisms have been suggested to play a role in maintaining this balance, including long-term plasticity of GABAergic inhibitory synapses. Many previous studies have demonstrated a coupling of postsynaptic spiking with modification of perisomatic inhibition. Here, we demonstrate that activation of NMDA-type glutamate receptors leads to input-specific long-term potentiation of dendritic inhibition mediated by somatostatin-expressing interneurons. This form of plasticity is expressed postsynaptically and requires both CaMKIIα and the ß2 subunit of the GABA-A receptor. Importantly, this process may function to preserve dendritic inhibition, as genetic deletion of NMDAR signaling results in a selective weakening of dendritic inhibition. Overall, our results reveal a new mechanism for linking excitatory and inhibitory input in neuronal dendrites and provide novel insight into the homeostatic regulation of synaptic transmission in cortical circuits.


Assuntos
Dendritos/fisiologia , Potenciação de Longa Duração/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Inibição Neural/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Ácido gama-Aminobutírico/fisiologia , Animais , Sinalização do Cálcio/fisiologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Piramidais/fisiologia , Receptores de GABA-A/fisiologia
11.
J Neurosci ; 22(8): 3044-51, 2002 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11943807

RESUMO

Second messengers regulate synaptic plasticity by influencing the balance between kinase and phosphatase activity. One target of this balance is the phosphorylation state of the AMPA receptor glutamate receptor 1 (GluR1) subunit. Hippocampal long-term depression (LTD) is a calcium-dependent downregulation of synaptic AMPA receptor currents associated with dephosphorylation of Ser845, a cAMP-dependent protein kinase (PKA) site on GluR1. Recruitment of kinases and phosphatases to the AMPA receptor might enable modulation of AMPA receptor function. The neuronal A-kinase anchoring protein AKAP79/150 interacts with PKA and the calcium-dependent protein phosphatase PP2B and is linked to the AMPA receptor GluR1 subunit by synapse-associated protein 97 (SAP97), a membrane-associated guanylate kinase family protein. Here we demonstrate that AKAP79 not only promotes basal phosphorylation of Ser845 but also confers a calcium- and PP2B-mediated downregulation to GluR1 receptor currents. This AKAP79-dependent downregulation is contingent on the local presence of PKA, Ser845 of GluR1, and a PDZ (postsynaptic density 95/Discs large/zona occludens 1)-domain interaction between GluR1 and SAP97, all of which support basal phosphorylation of the receptor. These findings suggest that the AKAP79 signaling complex is sufficient to couple intracellular calcium levels to the PKA phosphorylation state of GluR1. Thus, the integration of intracellular signals relevant for LTD may be transduced to GluR1 by the AKAP79 signaling complex.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte/metabolismo , Inibição Neural/fisiologia , Receptores de AMPA/metabolismo , Transdução de Sinais/fisiologia , Proteínas de Ancoragem à Quinase A , Animais , Sítios de Ligação/fisiologia , Calcineurina/metabolismo , Cálcio/metabolismo , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ácido Glutâmico/farmacologia , Rim/citologia , Rim/metabolismo , Substâncias Macromoleculares , Proteínas de Membrana , Proteínas do Tecido Nervoso/metabolismo , Plasticidade Neuronal/fisiologia , Neurônios/citologia , Neurônios/metabolismo , Técnicas de Patch-Clamp , Fosforilação , Testes de Precipitina , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Sistemas do Segundo Mensageiro/fisiologia , Tempo
12.
J Biol Chem ; 283(17): 11445-52, 2008 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-18305116

RESUMO

Enhancement of AMPA receptor activity in response to synaptic plasticity inducing stimuli may arise, in part, through phosphorylation of the GluR1 AMPA receptor subunit at Ser-831. This site is a substrate for both Ca(2+)-calmodulin-dependent protein kinase II (CaMKII) and protein kinase C (PKC). However, neuronal protein levels of CaMKII may exceed those of PKC by an order of magnitude. Thus, it is unclear how PKC could effectively regulate this common target site. The multivalent neuronal scaffold A-kinase-anchoring protein 79 (AKAP79) is known to bind PKC and is linked to GluR1 by synapse-associated protein 97 (SAP97). Here, biochemical studies demonstrate that AKAP79 localizes PKC activity near the receptor, thus accelerating Ser-831 phosphorylation. Complementary electrophysiological studies indicate that AKAP79 selectively shifts the dose-dependence for PKC modulation of GluR1 receptor currents approximately 20-fold, such that low concentrations of PKC are as effective as much higher CaMKII concentrations. By boosting PKC activity near a target substrate, AKAP79 provides a mechanism to overcome limitations in kinase abundance thereby ensuring faithful signal propagation and efficient modification of AMPA receptor-mediated responses.


Assuntos
Proteínas de Ancoragem à Quinase A/fisiologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Cálcio/metabolismo , Regulação da Expressão Gênica , Proteína Quinase C/metabolismo , Proteína Quinase C/fisiologia , Receptores de AMPA/metabolismo , Proteínas de Ancoragem à Quinase A/metabolismo , Linhagem Celular , Eletrofisiologia/métodos , Hipocampo/metabolismo , Humanos , Modelos Biológicos , Neurônios/metabolismo , Fosforilação , Fatores de Tempo
13.
Channels (Austin) ; 1(2): 102-12, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18690020

RESUMO

L-type calcium channels (Ca(v)1.1-Ca(v)1.4) link Ca(2+) influx to membrane depolarization and serve a critical role in regulating membrane excitability, muscle contraction, hormone secretion, and gene transcription. In many tissues, L-type calcium channel activity (Ca(v)1.1 and Ca(v)1.2) is enhanced by transmitters and hormones that activate the cAMP-dependent protein kinase (PKA), which is largely thought to be mediated via phosphorylation of the pore forming alpha subunit. However, the ability of PKA to regulate Ca(v)1.3 and the sites contributing to effective modulation of channel activity remains to be established. Using HEK 293 cells, we demonstrate that currents carried by the long C-terminal splice variant of Ca(v)1.3 (Ca(v)1.3L) are selectively enhanced compared to the short C-terminal splice variant (Ca(v)1.3S) when the catalytic subunit of PKA is introduced into the cell via the whole-cell recording electrode. However, the persistence of this regulation is dependent on the identity of the auxiliary beta subunit, such that PKA produces only a transient increase in the presence of beta(3) while a persistent increase is observed in the presence of the beta(2a) subunit. Site-directed mutagenesis of consensus PKA phosphorylation sites revealed that Ser1964 and Ser1743 in Ca(v)1.3L were the predominant sites controlling PKA modulation in the presence of the beta(3) and beta(2a) auxiliary subunits, respectively. Therefore, beta subunits determine the contribution of distinct sites within Ca(v)1.3 towards PKA-mediated enhancement of channel activity. These data suggest that auxiliary beta subunits govern the access of signaling enzymes to L-type calcium channels.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Subunidades Proteicas/metabolismo , Animais , Encéfalo/metabolismo , Canais de Cálcio Tipo L/genética , Linhagem Celular , DNA Complementar/genética , Eletrofisiologia , Proteínas de Fluorescência Verde/metabolismo , Átrios do Coração/química , Ventrículos do Coração/química , Humanos , Rim/citologia , Miocárdio/metabolismo , Técnicas de Patch-Clamp , Fosforilação , Plasmídeos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , Subunidades Proteicas/genética , Ratos , Transfecção
14.
J Biol Chem ; 281(44): 33537-53, 2006 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-16940053

RESUMO

Resensitization of G protein-coupled receptors (GPCR) following prolonged agonist exposure is critical for restoring the responsiveness of the receptor to subsequent challenges by agonist. The 3'-5' cyclic AMP-dependent protein kinase (PKA) and serine 312 in the third intracellular loop of the human beta(1)-adrenergic receptor (beta(1)-AR) were both necessary for efficient recycling and resensitization of the agonist-internalized beta(1)-AR (Gardner, L. A., Delos Santos, N. M., Matta, S. G., Whitt, M. A., and Bahouth, S. W. (2004) J. Biol. Chem. 279, 21135-21143). Because PKA is compartmentalized near target substrates by interacting with protein kinase A anchoring proteins (AKAPs), the present study was undertaken to identify the AKAP involved in PKA-mediated phosphorylation of the beta(1)-AR and in its recycling and resensitization. Here, we report that Ht-31 peptide-mediated disruption of PKA/AKAP interactions prevented the recycling and functional resensitization of heterologously expressed beta(1)-AR in HEK-293 cells and endogenously expressed beta(1)-AR in SK-N-MC cells and neonatal rat cortical neurons. Whereas several endogenous AKAPs were identified in HEK-293 cells, small interfering RNA-mediated down-regulation of AKAP79 prevented the recycling of the beta(1)-AR in this cell line. Co-immunoprecipitations and fluorescence resonance energy transfer (FRET) microscopy experiments in HEK-293 cells revealed that the beta(1)-AR, AKAP79, and PKA form a ternary complex at the carboxyl terminus of the beta(1)-AR. This complex was involved in PKA-mediated phosphorylation of the third intracellular loop of the beta(1)-AR because disruption of PKA/AKAP interactions or small interfering RNA-mediated down-regulation of AKAP79 both inhibited this response. Thus, AKAP79 provides PKA to phosphorylate the beta(1)-AR and thereby dictate the recycling and resensitization itineraries of the beta(1)-AR.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Proteínas de Ancoragem à Quinase A , Proteínas Adaptadoras de Transdução de Sinal/genética , Agonistas de Receptores Adrenérgicos beta 1 , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/genética , Humanos , Microscopia de Fluorescência , Fosforilação , Ligação Proteica , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , RNA Interferente Pequeno , Receptores Adrenérgicos beta 1/genética
15.
J Biol Chem ; 277(50): 48796-802, 2002 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-12354762

RESUMO

Compartmentalization of protein kinases and phosphatases with substrates is a means to increase the efficacy of signal transduction events. The A-kinase anchoring protein, AKAP79, is a multivalent anchoring protein that maintains the cAMP-dependent protein kinase, protein kinase C, and protein phosphatase-2B (PP2B/calcineurin) at the postsynaptic membrane of excitatory synapses where it is recruited into complexes with N-methyl-d-aspartic acid or alpha-amino-3-hydroxy-5-methyl-isoxazole-4-propionic acid (AMPA)-subtype glutamate receptors. We have used cellular targeting of AKAP79 truncation and deletion mutants as an assay to map the PP2B-binding site on AKAP79. We demonstrate that residues 315-360 are necessary and sufficient for AKAP79-PP2B anchoring in cells. Multiple determinants contained within this region bind directly to the A subunit of PP2B and inhibit phosphatase activity. Peptides spanning the 315-360 region of AKAP79 can antagonize PP2B anchoring in vitro and targeting in transfected cells. Electrophysiological experiments further emphasize this point by demonstrating that a peptide encompassing residues 330-357 of AKAP79 attenuates PP2B-dependent down-regulation of GluR1 receptor currents when perfused into HEK293 cells. We propose that the structural features of this AKAP79-PP2B-binding domain may share similarities with other proteins that serve to coordinate PP2B localization and activity.


Assuntos
Calcineurina/metabolismo , Proteínas de Transporte/metabolismo , Animais , Sítios de Ligação , Células COS , Inibidores de Calcineurina , Proteínas de Transporte/química , Membrana Celular/enzimologia , Camundongos
16.
J Neurophysiol ; 92(2): 824-37, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15056682

RESUMO

The influx of calcium (Ca(2+)) ions through L-type channels underlies many cellular processes, ranging from initiation of gene transcription to activation of Ca(2+)-activated potassium channels. L-type channels possess a diagnostic pharmacology, being enhanced by the dihydropyridine BAY K 8644 and benzoylpyrrole FPL 64176. It is assumed that the action of these compounds is independent of the ion conducted through the channel. In contrast to this assumption, modulation of L-type channel activity in acutely dissociated rat CA1 hippocampal neurons depended on the divalent ion identity. BAY K 8644 and FPL 64176 substantially increased single-channel open time only when barium (Ba(2+)) was the permeant ion. BAY K 8644 increased single-channel conductance when either Ba(2+) or Ca(2+) ions were the charge carrier, an effect not observed with FPL 64176. BAY K 8644 enhanced the whole cell L-type channel Ca(2+)- or Ba(2+)-carried current without a change in deactivation tail kinetics. In contrast, enhancement by FPL 64176 was associated with a dramatic slowing of deactivation kinetics only when Ba(2+) and not Ca(2+) was the charge carrier. Current activation was slowed by FPL 64176 with either charge carrier, an effect arising from a clustering of agonist-modified long-duration openings toward the end of the voltage step. These data indicate that agonists enhanced L-type current by distinct mechanisms dependent on the permeant ion, indicating that care must be considered when used as diagnostic tools.


Assuntos
Bário/metabolismo , Agonistas dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo L/fisiologia , Cálcio/metabolismo , Hipocampo/metabolismo , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Animais , Canais de Cálcio Tipo L/metabolismo , Condutividade Elétrica , Técnicas In Vitro , Permeabilidade , Pirróis/farmacologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA