Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.010
Filtrar
1.
J Neurosci ; 43(30): 5559-5573, 2023 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-37419689

RESUMO

Widespread release of norepinephrine (NE) throughout the forebrain fosters learning and memory via adrenergic receptor (AR) signaling, but the molecular mechanisms are largely unknown. The ß2 AR and its downstream effectors, the trimeric stimulatory Gs-protein, adenylyl cyclase (AC), and the cAMP-dependent protein kinase A (PKA), form a unique signaling complex with the L-type Ca2+ channel (LTCC) CaV1.2. Phosphorylation of CaV1.2 by PKA on Ser1928 is required for the upregulation of Ca2+ influx on ß2 AR stimulation and long-term potentiation induced by prolonged theta-tetanus (PTT-LTP) but not LTP induced by two 1-s-long 100-Hz tetani. However, the function of Ser1928 phosphorylation in vivo is unknown. Here, we show that S1928A knock-in (KI) mice of both sexes, which lack PTT-LTP, express deficiencies during initial consolidation of spatial memory. Especially striking is the effect of this mutation on cognitive flexibility as tested by reversal learning. Mechanistically, long-term depression (LTD) has been implicated in reversal learning. It is abrogated in male and female S1928A knock-in mice and by ß2 AR antagonists and peptides that displace ß2 AR from CaV1.2. This work identifies CaV1.2 as a critical molecular locus that regulates synaptic plasticity, spatial memory and its reversal, and LTD.SIGNIFICANCE STATEMENT We show that phosphorylation of the Ca2+ channel CaV1.2 on Ser1928 is important for consolidation of spatial memory and especially its reversal, and long-term depression (LTD). Identification of Ser1928 as critical for LTD and reversal learning supports the model that LTD underlies flexibility of reference memory.


Assuntos
Plasticidade Neuronal , Memória Espacial , Camundongos , Masculino , Feminino , Animais , Plasticidade Neuronal/fisiologia , Potenciação de Longa Duração/fisiologia , Transdução de Sinais , Fosforilação , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Hipocampo/fisiologia
2.
Handb Exp Pharmacol ; 279: 59-82, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36592229

RESUMO

Calcium influx through voltage-gated calcium channels, Cav1.2, in cardiomyocytes initiates excitation-contraction coupling in the heart. The force and rate of cardiac contraction are modulated by the sympathetic nervous system, mediated substantially by changes in intracellular calcium. Norepinephrine released from sympathetic neurons innervating the heart and epinephrine secreted by the adrenal chromaffin cells bind to ß-adrenergic receptors on the sarcolemma of cardiomyocytes initiating a signaling cascade that generates cAMP and activates protein kinase A, the targets of which control calcium influx. For decades, the mechanisms by which PKA regulated calcium channels in the heart were not known. Recently, these mechanisms have been elucidated. In this chapter, we will review the history of the field and the studies that led to the identification of the evolutionarily conserved process.


Assuntos
Canais de Cálcio , Cálcio , Humanos , Canais de Cálcio/metabolismo , Cálcio/metabolismo , Receptores Adrenérgicos beta/metabolismo , Miócitos Cardíacos/metabolismo , Sistema Nervoso Simpático/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Canais de Cálcio Tipo L/metabolismo , Fosforilação
3.
Learn Mem ; 29(12): 435-446, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36446603

RESUMO

Empirical and computational methods were combined to examine whether individual or dual-drug treatments can restore the deficit in long-term synaptic facilitation (LTF) of the Aplysia sensorimotor synapse observed in a cellular model of Coffin-Lowry syndrome (CLS). The model was produced by pharmacological inhibition of p90 ribosomal S6 kinase (RSK) activity. In this model, coapplication of an activator of the mitogen-activated protein kinase (MAPK) isoform ERK and an activator of protein kinase A (PKA) resulted in enhanced phosphorylation of RSK and enhanced LTF to a greater extent than either drug alone and also greater than their additive effects, which is termed synergism. The extent of synergism appeared to depend on another MAPK isoform, p38 MAPK. Inhibition of p38 MAPK facilitated serotonin (5-HT)-induced RSK phosphorylation, indicating that p38 MAPK inhibits activation of RSK. Inhibition of p38 MAPK combined with activation of PKA synergistically activated both ERK and RSK. Our results suggest that cellular models of disorders that affect synaptic plasticity and learning, such as CLS, may constitute a useful strategy to identify candidate drug combinations, and that combining computational models with empirical tests of model predictions can help explain synergism of drug combinations.


Assuntos
Síndrome de Coffin-Lowry , Proteínas Quinases Dependentes de AMP Cíclico , Plasticidade Neuronal , Proteínas Quinases p38 Ativadas por Mitógeno , Humanos , Síndrome de Coffin-Lowry/fisiopatologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Plasticidade Neuronal/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Serotonina/farmacologia
4.
Int J Mol Sci ; 23(19)2022 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-36232721

RESUMO

During vascular development, endothelial cAMP-dependent protein kinase A (PKA) regulates angiogenesis by controlling the number of tip cells, and PKA inhibition leads to excessive angiogenesis. Whether this role of endothelial PKA is restricted to embryonic and neonatal development or is also required for vascular homeostasis later on is unknown. Here, we show that perinatal (postnatal days P1-P3) of later (P28-P32) inhibition of endothelial PKA using dominant-negative PKA expressed under the control of endothelial-specific Cdh5-CreERT2 recombinase (dnPKAiEC mice) leads to severe subcutaneous edema, hypoalbuminemia, hypoglycemia and premature death. These changes were accompanied by the local hypersprouting of blood vessels in fat pads and the secondary enlargement of subcutaneous lymphatic vessels. Most noticeably, endothelial PKA inhibition caused a dramatic disorganization of the liver vasculature. Hepatic changes correlated with decreased gluconeogenesis, while liver albumin production seems to be unaffected and hypoalbuminemia is rather a result of increased leakage into the interstitium. Interestingly, the expression of dnPKA only in lymphatics using Prox1-CreERT2 produced no phenotype. Likewise, the mosaic expression in only endothelial subpopulations using Vegfr3-CreERT2 was insufficient to induce edema or hypoglycemia. Increased expression of the tip cell marker ESM1 indicated that the inhibition of PKA induced an angiogenic response in the liver, although tissue derived pro- and anti-angiogenic factors were unchanged. These data indicate that endothelial PKA is a gatekeeper of endothelial cell activation not only in development but also in adult homeostasis, preventing the aberrant reactivation of the angiogenic program.


Assuntos
Vasos Sanguíneos , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico , Células Endoteliais , Fígado , Albuminas , Animais , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/fisiologia , AMP Cíclico , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Homeostase , Hipoalbuminemia , Hipoglicemia , Fígado/metabolismo , Fígado/fisiologia , Camundongos , Recombinases
5.
Sci China Life Sci ; 65(3): 500-514, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34505970

RESUMO

The Hedgehog (Hh) signaling is one of the essential signaling pathways during embryogenesis and in adults. Hh signal transduction relies on primary cilium, a specialized cell surface organelle viewed as the hub of cell signaling. Protein kinase A (PKA) has been recognized as a potent negative regulator of the Hh pathway, raising the question of how such a ubiquitous kinase specifically regulates one signaling pathway. We reviewed recent genetic, molecular and biochemical studies that have advanced our mechanistic understanding of PKA's role in Hh signaling in vertebrates, focusing on the compartmentalized PKA at the centrosome and in the primary cilium. We outlined the recently developed genetic and optical tools that can be harvested to study PKA activities during the course of Hh signal transduction.


Assuntos
Cílios/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Proteínas Hedgehog/fisiologia , Animais , Centrossomo/fisiologia , Humanos , Transdução de Sinais/fisiologia , Proteína GLI1 em Dedos de Zinco/fisiologia
6.
Int J Mol Sci ; 22(16)2021 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-34445723

RESUMO

Lipid droplets (LDs) are ubiquitous organelles that fulfill essential roles in response to metabolic cues. The identification of several neutral lipid synthesizing and regulatory protein complexes have propelled significant advance on the mechanisms of LD biogenesis in the endoplasmic reticulum (ER). However, our understanding of signaling networks, especially transcriptional mechanisms, regulating membrane biogenesis is very limited. Here, we show that the nutrient-sensing Target of Rapamycin Complex 1 (TORC1) regulates LD formation at a transcriptional level, by targeting DGA1 expression, in a Sit4-, Mks1-, and Sfp1-dependent manner. We show that cytosolic pH (pHc), co-regulated by the plasma membrane H+-ATPase Pma1 and the vacuolar ATPase (V-ATPase), acts as a second messenger, upstream of protein kinase A (PKA), to adjust the localization and activity of the major transcription factor repressor Opi1, which in turn controls the metabolic switch between phospholipid metabolism and lipid storage. Together, this work delineates hitherto unknown molecular mechanisms that couple nutrient availability and pHc to LD formation through a transcriptional circuit regulated by major signaling transduction pathways.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Gotículas Lipídicas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Concentração de Íons de Hidrogênio , Gotículas Lipídicas/fisiologia , Metabolismo dos Lipídeos/fisiologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/fisiologia , Proteínas de Membrana/metabolismo , Proteína Fosfatase 2/metabolismo , Proteínas Repressoras/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia , Transdução de Sinais , Fatores de Transcrição/fisiologia
7.
Mol Nutr Food Res ; 65(20): e2100167, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34268878

RESUMO

SCOPE: Adiponectin (ADPN), a kind of adipokines, plays an important role in the regulation of lipid metabolism. The objective of this study is focused on the ADPN to investigate the functional mechanisms of pectin oligosaccharide (POS) from hawthorn fruit in the improvement of hepatic fatty acid oxidation. METHOD AND RESULTS: High-fat fed mice are used in this experiment. POS is administrated with the doses of 0.25, 0.75, and 1.5 g kg-1 diet, respectively. The results demonstrate that gene and protein expressions of ADPN synthesis regulators involved in PKA/ERK/CREB and C/EBPα/PPARγ pathways are upregulated by POS administration. POS also activates the AdiopR1/AMPKα/PGC1 and AdipoR2/PPARα signaling pathways to improve the fatty acid oxidation in the liver, which is further accelerated by the enhancement of mitochondrial functions. CONCLUSION: POS can act as an ADPN activator to improve lipid metabolism, leading it to the applications of biomedical and functional foods for ameliorating chronic liver diseases resulted from a high-energy diet.


Assuntos
Adiponectina/biossíntese , Crataegus/química , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/metabolismo , Pectinas/farmacologia , Proteínas Quinases Ativadas por AMP/fisiologia , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Masculino , Camundongos , Oxirredução , PPAR gama/fisiologia , Receptores de Adiponectina/fisiologia , Transdução de Sinais/fisiologia
8.
Curr Genet ; 67(5): 807-821, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34086083

RESUMO

The cAMP-dependent protein kinase (Pka1) regulates many cellular events, including sexual development and glycogenesis, and response to the limitation of glucose, in Schizosaccharomyces pombe. Despite its importance in many cellular events, the targets of the cAMP/PKA pathway have not been fully investigated. Here, we demonstrate that the expression of mug14 is induced by downregulation of the cAMP/PKA pathway and limitation of glucose. This regulation is dependent on the function of Rst2, a transcription factor that regulates transition from mitosis to meiosis. The loss of the C2H2-type zinc finger domain in Rst2, termed Rst2 (C2H2∆), abolished the induction of Mug14 expression. Upon deletion of the stress starvation response element of the S. pombe (STREP: CCCCTC) sequence, which is a potential binding site of Rst2 on mug14, in the pka1∆ strain, its induction was abolished. The expression of Mug14 was significantly reduced and delayed by the limitation of glucose and also by nitrogen starvation in the rst2∆ strain. Mug14 is known to share a common function with Mde1 and Mta3 in the methionine salvage pathway, but the expression of mde1 and mta3 mRNAs was not enhanced by pka1 deletion and limitation of glucose. We conclude that the expression of Mug14 is upregulated by Rst2 under the control of the cAMP/PKA signaling pathway, which senses the limitation of glucose.


Assuntos
Proteínas de Ciclo Celular/genética , Regulação Fúngica da Expressão Gênica , Proteínas de Schizosaccharomyces pombe/genética , Schizosaccharomyces/genética , Fatores de Transcrição/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Glucose/metabolismo , Proteínas de Fluorescência Verde/genética , Sistema de Sinalização das MAP Quinases , Nitrogênio/metabolismo , Conformação de Ácido Nucleico , Regiões Promotoras Genéticas , RNA Fúngico , RNA Mensageiro , Proteínas Recombinantes de Fusão/genética , Schizosaccharomyces/enzimologia , Proteínas de Schizosaccharomyces pombe/fisiologia , Estresse Fisiológico
9.
J Pharmacol Sci ; 145(3): 253-261, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33602505

RESUMO

Facilitation of cardiac function in response to signals from the sympathetic nervous system is initiated by the phosphorylation of L-type voltage-dependent Ca2+ channels (VDCCs) by protein kinase A (PKA), which in turn is activated by ß-adrenoceptors. Among the five subunits (α1, ß, α2/δ, and γ) of VDCCs, the α1 subunit and the family of ß subunits are substrates for PKA-catalyzed phosphorylation; however, the subunit responsible for ß-adrenergic augmentation of Ca2+ channel function has yet to be specifically identified. Here we show that the VDCC ß2 subunit is required for PKA phosphorylation upon sympathetic acceleration. In mice with ß2 subunit-null mutations, cardiac muscle contraction in response to isoproterenol was reduced and there was no significant increase in Ca2+ channel currents upon PKA-dependent phosphorylation. These findings indicate that within the sympathetic nervous system the ß2 subunit of VDCCs is required for physiological PKA-dependent channel phosphorylation.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Receptores Adrenérgicos beta/fisiologia , Sistema Nervoso Simpático/metabolismo , Sistema Nervoso Simpático/fisiologia , Animais , Catálise , Células Cultivadas , Isoproterenol/farmacologia , Camundongos , Mutação , Contração Miocárdica/efeitos dos fármacos , Fosforilação , Receptores Adrenérgicos beta/genética
10.
Nat Commun ; 11(1): 5318, 2020 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-33087709

RESUMO

Synaptic vesicles (SVs) can be pooled across multiple synapses, prompting questions about their dynamic allocation for neurotransmission and plasticity. We find that the axonal traffic of recycling vesicles is not supported by ubiquitous microtubule-based motility but relies on actin instead. Vesicles freed from synaptic clusters undergo ~1 µm bouts of active transport, initiated by nearby elongation of actin filaments. Long distance translocation arises when successive bouts of active transport were linked by periods of free diffusion. The availability of SVs for active transport can be promptly increased by protein kinase A, a key player in neuromodulation. Vesicle motion is in turn impeded by shutting off axonal actin polymerization, mediated by nitric oxide-cyclic GMP signaling leading to inhibition of RhoA. These findings provide a potential framework for coordinating post-and pre-synaptic strength, using retrograde regulation of axonal actin dynamics to mobilize and recruit presynaptic SV resources.


Assuntos
Citoesqueleto de Actina/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Óxido Nítrico/fisiologia , Vesículas Sinápticas/fisiologia , Animais , Transporte Axonal/fisiologia , Transporte Biológico Ativo , Células Cultivadas , GMP Cíclico/fisiologia , Feminino , Hipocampo/citologia , Hipocampo/fisiologia , Proteínas Luminescentes/metabolismo , Masculino , Neurônios/fisiologia , Nocodazol/farmacologia , Ratos , Ratos Sprague-Dawley , Transmissão Sináptica/fisiologia , Vesículas Sinápticas/efeitos dos fármacos
11.
Mol Cell Endocrinol ; 518: 110992, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32853743

RESUMO

The 3', 5'-cyclic adenosine monophosphate (cAMP) dependent protein kinase (PKA) is a tetrameric holoenzyme comprising a set of two regulatory subunits (PKA-R) and two catalytic (PKA-C) subunits. The PKA-R subunits act as sensors of cAMP and allow PKA-C activity. One of the first signaling events observed during mammalian sperm capacitation is PKA activation. Thus, understanding how PKA activity is restricted in space and time is crucial to decipher the critical steps of sperm capacitation. It is widely accepted that PKA specificity depends on several levels of regulation. Anchoring proteins play a pivotal role in achieving proper localization signaling, subcellular targeting and cAMP microdomains. These multi-factorial regulation steps are necessary for a precise spatio-temporal activation of PKA. Here we discuss recent understanding of regulatory mechanisms of PKA in mammalian sperm, such as post-translational modifications, in the context of its role as the master orchestrator of molecular events conducive to capacitation.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Capacitação Espermática/fisiologia , Reação Acrossômica/fisiologia , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Humanos , Masculino , Mamíferos , Processamento de Proteína Pós-Traducional/fisiologia , Transdução de Sinais/fisiologia , Espermatozoides/metabolismo
12.
Proc Natl Acad Sci U S A ; 117(35): 21740-21746, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32817533

RESUMO

The Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) anion channel is essential for epithelial salt-water balance. CFTR mutations cause cystic fibrosis, a lethal incurable disease. In cells CFTR is activated through the cAMP signaling pathway, overstimulation of which during cholera leads to CFTR-mediated intestinal salt-water loss. Channel activation is achieved by phosphorylation of its regulatory (R) domain by cAMP-dependent protein kinase catalytic subunit (PKA). Here we show using two independent approaches--an ATP analog that can drive CFTR channel gating but is unsuitable for phosphotransfer by PKA, and CFTR mutants lacking phosphorylatable serines--that PKA efficiently opens CFTR channels through simple binding, under conditions that preclude phosphorylation. Unlike when phosphorylation happens, CFTR activation by PKA binding is completely reversible. Thus, PKA binding promotes release of the unphosphorylated R domain from its inhibitory position, causing full channel activation, whereas phosphorylation serves only to maintain channel activity beyond termination of the PKA signal. The results suggest two levels of CFTR regulation in cells: irreversible through phosphorylation, and reversible through R-domain binding to PKA--and possibly also to other members of a large network of proteins known to interact with the channel.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Canais de Ânion Dependentes de Voltagem/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Ânions/metabolismo , Fenômenos Biofísicos , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Ativação do Canal Iônico/fisiologia , Mutagênese Sítio-Dirigida , Nucleotídeos/metabolismo , Oócitos/metabolismo , Técnicas de Patch-Clamp/métodos , Fosforilação , Ligação Proteica/fisiologia , Serina/metabolismo , Canais de Ânion Dependentes de Voltagem/fisiologia , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo
13.
PLoS One ; 15(8): e0231806, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32817622

RESUMO

The cAMP-dependent protein kinase (PKA) signaling pathway is the primary means by which the heart regulates moment-to-moment changes in contractility and metabolism. We have previously found that PKA signaling is dysfunctional in the diabetic heart, yet the underlying mechanisms are not fully understood. The objective of this study was to determine if decreased insulin signaling contributes to a dysfunctional PKA response. To do so, we isolated adult cardiomyocytes (ACMs) from wild type and Akita type 1 diabetic mice. ACMs were cultured in the presence or absence of insulin and PKA signaling was visualized by immunofluorescence microscopy using an antibody that recognizes proteins specifically phosphorylated by PKA. We found significant decreases in proteins phosphorylated by PKA in wild type ACMs cultured in the absence of insulin. PKA substrate phosphorylation was decreased in Akita ACMs, as compared to wild type, and unresponsive to the effects of insulin. The decrease in PKA signaling was observed regardless of whether the kinase was stimulated with a beta-agonist, a cell-permeable cAMP analog, or with phosphodiesterase inhibitors. PKA content was unaffected, suggesting that the decrease in PKA signaling may be occurring by the loss of specific PKA substrates. Phospho-specific antibodies were used to discern which potential substrates may be sensitive to the loss of insulin. Contractile proteins were phosphorylated similarly in wild type and Akita ACMs regardless of insulin. However, phosphorylation of the glycolytic regulator, PFK-2, was significantly decreased in an insulin-dependent manner in wild type ACMs and in an insulin-independent manner in Akita ACMs. These results demonstrate a defect in PKA activation in the diabetic heart, mediated in part by deficient insulin signaling, that results in an abnormal activation of a primary metabolic regulator.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Diabetes Mellitus/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Diabetes Mellitus Experimental/metabolismo , Modelos Animais de Doenças , Insulina/metabolismo , Insulina/farmacologia , Insulina/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/fisiologia , Inibidores de Fosfodiesterase/farmacologia , Fosforilação/efeitos dos fármacos , Cultura Primária de Células , Transdução de Sinais/efeitos dos fármacos
14.
PLoS Comput Biol ; 16(7): e1008078, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32701987

RESUMO

Animals remember temporal links between their actions and subsequent rewards. We previously discovered a synaptic mechanism underlying such reward learning in D1 receptor (D1R)-expressing spiny projection neurons (D1 SPN) of the striatum. Dopamine (DA) bursts promote dendritic spine enlargement in a time window of only a few seconds after paired pre- and post-synaptic spiking (pre-post pairing), which is termed as reinforcement plasticity (RP). The previous study has also identified underlying signaling pathways; however, it still remains unclear how the signaling dynamics results in RP. In the present study, we first developed a computational model of signaling dynamics of D1 SPNs. The D1 RP model successfully reproduced experimentally observed protein kinase A (PKA) activity, including its critical time window. In this model, adenylate cyclase type 1 (AC1) in the spines/thin dendrites played a pivotal role as a coincidence detector against pre-post pairing and DA burst. In particular, pre-post pairing (Ca2+ signal) stimulated AC1 with a delay, and the Ca2+-stimulated AC1 was activated by the DA burst for the asymmetric time window. Moreover, the smallness of the spines/thin dendrites is crucial to the short time window for the PKA activity. We then developed a RP model for D2 SPNs, which also predicted the critical time window for RP that depended on the timing of pre-post pairing and phasic DA dip. AC1 worked for the coincidence detector in the D2 RP model as well. We further simulated the signaling pathway leading to Ca2+/calmodulin-dependent protein kinase II (CaMKII) activation and clarified the role of the downstream molecules of AC1 as the integrators that turn transient input signals into persistent spine enlargement. Finally, we discuss how such timing windows guide animals' reward learning.


Assuntos
Sinalização do Cálcio , Corpo Estriado/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Dopamina/fisiologia , Aprendizagem , Plasticidade Neuronal , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/fisiologia , Simulação por Computador , Dendritos/fisiologia , Espinhas Dendríticas/fisiologia , Cinética , Camundongos , Neurônios/fisiologia , Receptores de Dopamina D2 , Recompensa
15.
Biol Pharm Bull ; 43(8): 1196-1201, 2020 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-32475934

RESUMO

Oxidative stress plays an essential role in obstructive sleep apnea-hypopnea syndrome-induced cognitive dysfunction in children. This study investigated the effects of edaravone, a potent free radical scavenger, on intermittent hypoxia (IH)-induced oxidative damage and cognition impairment in a young rat model of IH. IH rats were treated with edaravone for 4 weeks. Behavioral testing was performed using the Morris water maze, and hippocampal tissues were harvested for further analyses. Edaravone attenuated IH-induced cognitive impairment, reduced morphological and structural abnormalities, and increased the number of mitochondria in the IH rats. Furthermore, edaravone significantly increased the inhibition of hydroxyl free radicals; reduced expressions of superoxide anion, malondialdehyde, and 8-hydroxy-2'-deoxyguanosine; and upregulated the expression of manganese superoxide dismutase, catalase, cAMP, protein kinase A, phosphorylated-cAMP response element-binding (p-CREB), B-cell lymphoma 2, and brain-derived neurotrophic factor in the hippocampal tissue of IH rats. Our findings suggest that edaravone attenuated IH-induced cognitive impairment and hippocampal damage by upregulating p-CREB in young rats.


Assuntos
Disfunção Cognitiva/tratamento farmacológico , Edaravone/uso terapêutico , Hipocampo/efeitos dos fármacos , Hipóxia/complicações , Animais , AMP Cíclico/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Edaravone/farmacologia , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Teste do Labirinto Aquático de Morris , Estresse Oxidativo , Ratos , Ratos Wistar
16.
Neurobiol Learn Mem ; 172: 107244, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32376452

RESUMO

Fear extinction is a form of new learning that inhibits expression of the original fear memory without erasing the conditioned stimulus-unconditioned stimulus association. Much is known about the mechanisms that underlie the acquisition of extinction, but the way in which fear extinction is maintained has been scarcely explored. Evidence suggests that protein kinase A (PKA) in the frontal cortex might be related to the persistence of extinction. Phosphodiesterase-4 (PDE4) specifically hydrolyzes cyclic adenosine monophosphate (cAMP). The present study evaluated the effect of the selective PDE4 inhibitor roflumilast (ROF; 0.01, 0.03, and 0.1 mg/kg given i.p.) on acquisition and consolidation of the extinction of fear memory in male Wistar rats in a contextual fear conditioning paradigm. When administered before acquisition, 0.1 mg/kg ROF disrupted short-term (1 day) extinction recall. In contrast, 0.03 mg/kg ROF administration in the late consolidation phase (3 h after extinction learning) but not in the early phase immediately after learning improved long-term extinction recall at 11 days, suggesting potentiation of the persistence of extinction. This effect of ROF requires the first (day 1) exposure to the context. A similar effect was observed when 9 ng ROF or 30 µM 8-bromoadenosine 3',5'-cAMP (PKA activator) was directly infused in the infralimbic cortex (IL), a brain region necessary for memory extinction. The PKA activity-dependent ROF-induced effect in the IL was correlated with an increase in its brain-derived neurotrophic factor (BDNF) protein expression, while blockade of PKA with 10 µM H89 in the IL abolished the ROF-induced increase in BDNF expression and prevented the effect of ROF on extinction recall. These effects were not associated with changes in anxiety-like behavior or general exploratory behavior. Altogether, these findings suggest that cAMP-PKA activity in the IL during the late consolidation phase after extinction learning underlies the persistence of extinction.


Assuntos
Extinção Psicológica/fisiologia , Medo/fisiologia , Memória/fisiologia , Córtex Pré-Frontal/fisiologia , Transdução de Sinais , Aminopiridinas/administração & dosagem , Animais , Benzamidas/administração & dosagem , AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Ciclopropanos/administração & dosagem , Extinção Psicológica/efeitos dos fármacos , Medo/efeitos dos fármacos , Masculino , Memória/efeitos dos fármacos , Consolidação da Memória/efeitos dos fármacos , Consolidação da Memória/fisiologia , Inibidores da Fosfodiesterase 4/administração & dosagem , Córtex Pré-Frontal/efeitos dos fármacos , Ratos Wistar
17.
Elife ; 92020 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-32324136

RESUMO

Germ cells are vulnerable to stress. Therefore, how organisms protect their future progeny from damage in a fluctuating environment is a fundamental question in biology. We show that in Caenorhabditis elegans, serotonin released by maternal neurons during stress ensures the viability and stress resilience of future offspring. Serotonin acts through a signal transduction pathway conserved between C. elegans and mammalian cells to enable the transcription factor HSF1 to alter chromatin in soon-to-be fertilized germ cells by recruiting the histone chaperone FACT, displacing histones, and initiating protective gene expression. Without serotonin release by maternal neurons, FACT is not recruited by HSF1 in germ cells, transcription occurs but is delayed, and progeny of stressed C. elegans mothers fail to complete development. These studies uncover a novel mechanism by which stress sensing by neurons is coupled to transcription response times of germ cells to protect future offspring.


Assuntos
Desenvolvimento Embrionário/fisiologia , Células Germinativas/fisiologia , Neurônios/fisiologia , Serotonina/fisiologia , Estresse Fisiológico/fisiologia , Animais , Caenorhabditis elegans/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Proteínas de Ligação a DNA/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição de Choque Térmico/fisiologia , Proteínas de Grupo de Alta Mobilidade/fisiologia , Temperatura Alta , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/fisiologia , Fatores de Elongação da Transcrição/fisiologia
18.
Invest Ophthalmol Vis Sci ; 61(2): 24, 2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-32068794

RESUMO

Purpose: O-GlcNAcylation of cellular proteins contributes to the pathophysiology of diabetes and evidence supports a role for augmented O-GlcNAcylation in diabetic retinopathy. The aim of this study was to investigate the impact of the renin-angiotensin system on retinal protein O-GlcNAcylation. Methods: Mice fed a high-fat diet were treated chronically with the angiotensin-converting enzyme inhibitor captopril or captopril plus the angiotensin-(1-7) Mas receptor antagonist A779. Western blotting and quantitative polymerase chain reaction were used to analyze retinal homogenates. Similar analyses were performed on lysates from human MIO-M1 retinal Müller cell cultures exposed to media supplemented with angiotensin-(1-7). Culture conditions were manipulated to influence the hexosamine biosynthetic pathway and/or signaling downstream of the Mas receptor. Results: In the retina of mice fed a high-fat diet, captopril attenuated protein O-GlcNAcylation in a manner dependent on Mas receptor activation. In MIO-M1 cells, angiotensin-(1-7) or adenylate cyclase activation were sufficient to enhance cyclic AMP (cAMP) levels and inhibit O-GlcNAcylation. The repressive effect of cAMP on O-GlcNAcylation was dependent on exchange protein activated by cAMP (EPAC), but not protein kinase A, and was recapitulated by a constitutively active variant of the small GTPase Rap1. We provide evidence that cAMP and angiotensin-(1-7) act to suppress O-GlcNAcylation by inhibition of O-GlcNAc transferase (OGT) activity. In cells exposed to an O-GlcNAcase inhibitor or hyperglycemic culture conditions, mitochondrial superoxide levels were elevated; however, angiotensin-(1-7) signaling prevented the effect. Conclusions: Angiotensin-(1-7) inhibits retinal protein O-GlcNAcylation via an EPAC/Rap1/OGT signaling axis.


Assuntos
Angiotensina I/farmacologia , N-Acetilglucosaminiltransferases/metabolismo , Fragmentos de Peptídeos/farmacologia , Retina/metabolismo , Animais , Captopril/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Retinopatia Diabética/metabolismo , Camundongos , Sistema Renina-Angiotensina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
19.
Nutrients ; 12(1)2020 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-31906276

RESUMO

A high fat diet (HFD) intake is crucial for the development and progression of metabolic syndrome (MtS). Increasing evidence links gut dysbiosis with the metabolic and vascular alterations associated with MtS. Here we studied the use of a combination of various probiotic strains together with a prebiotic (synbiotic) in a commercially available Prodefen® Plus. MtS was induced by HFD (45%) in male Wistar rats. Half of the MtS animals received Prodefen® Plus for 4 weeks. At 12 weeks, we observed an increase in body weight, together with the presence of insulin resistance, liver steatosis, hypertriglyceridemia and hypertension in MtS rats. Prodefen® Plus supplementation did not affect the body weight gain but ameliorated all the MtS-related symptoms. Moreover, the hypertension induced by HFD is caused by a diminished both nitric oxide (NO) functional role and release probably due to a diminished neuronal nitric oxide synthase (nNOS) activation by protein kinase A (PKA) pathway. Prodefen® Plus supplementation for 4 weeks recovered the NO function and release and the systolic blood pressure was returned to normotensive values as a result. Overall, supplementation with Prodefen® Plus could be considered an interesting non-pharmacological approach in MtS.


Assuntos
Vasos Sanguíneos/fisiopatologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Síndrome Metabólica/enzimologia , Óxido Nítrico Sintase/fisiologia , Simbióticos/administração & dosagem , Animais , Pressão Sanguínea/fisiologia , Vasos Sanguíneos/enzimologia , Peso Corporal , Dieta Hiperlipídica , Suplementos Nutricionais , Modelos Animais de Doenças , Hipertensão/enzimologia , Hipertensão/etiologia , Hipertensão/fisiopatologia , Resistência à Insulina , Masculino , Síndrome Metabólica/etiologia , Síndrome Metabólica/fisiopatologia , Probióticos/administração & dosagem , Ratos , Ratos Wistar
20.
Mol Biol Cell ; 31(1): 45-58, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31721649

RESUMO

Dynamic subcellular regulation of protein kinase A (PKA) activity is important for the motile behavior of many cell types, yet the mechanisms governing PKA activity during cell migration remain largely unknown. The motility of SKOV-3 epithelial ovarian cancer (EOC) cells has been shown to be dependent both on localized PKA activity and, more recently, on mechanical reciprocity between cellular tension and extracellular matrix rigidity. Here, we investigated the possibility that PKA is regulated by mechanical signaling during migration. We find that localized PKA activity in migrating cells rapidly decreases upon inhibition of actomyosin contractility (specifically, of myosin ATPase, Rho kinase, or myosin light-chain kinase activity). Moreover, PKA activity is spatially and temporally correlated with cellular traction forces in migrating cells. Additionally, PKA is rapidly and locally activated by mechanical stretch in an actomyosin contractility-dependent manner. Finally, inhibition of PKA activity inhibits mechanically guided migration, also known as durotaxis. These observations establish PKA as a locally regulated effector of cellular mechanotransduction and as a regulator of mechanically guided cell migration.


Assuntos
Actomiosina/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Mecanotransdução Celular/fisiologia , Citoesqueleto de Actina/metabolismo , Actomiosina/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proteínas Contráteis/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Matriz Extracelular/metabolismo , Feminino , Humanos , Contração Muscular , Miosinas/metabolismo , Fosforilação , Quinases Associadas a rho/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...