Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 298
Filtrar
1.
Pharm Biol ; 60(1): 689-698, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35298359

RESUMEN

CONTEXT: The mammalian circadian clock system regulates physiological function. Crude drugs, containing Polygalae Radix, and Kampo, combining multiple crude drugs, have been used to treat various diseases, but few studies have focussed on the circadian clock. OBJECTIVE: We examine effective crude drugs, which cover at least one or two of Kampo, for the shortening effects on period length of clock gene expression rhythm, and reveal the mechanism of shortening effects. MATERIALS AND METHODS: We prepared 40 crude drugs. In the in vitro experiments, we used mouse embryonic fibroblasts from PERIOD2::LUCIFERASE knock-in mice (background; C57BL/6J mice) to evaluate the effect of crude drugs on the period length of core clock gene, Per2, expression rhythm by chronic treatment (six days) with distilled water or crude drugs (100 µg/mL). In the in vivo experiments, we evaluated the free-running period length of C57BL/6J mice fed AIN-93M or AIN-93M supplemented with 1% crude drug (6 weeks) that shortened the period length of the PERIOD2::LUCIFERASE expression rhythm in the in vitro experiments. RESULTS: We found that Polygalae Radix (ED50: 24.01 µg/mL) had the most shortened PERIOD2::LUCIFERASE rhythm period length in 40 crude drugs and that the CaMKII pathway was involved in this effect. Moreover, long-term feeding with AIN-93M+Polygalae Radix slightly shortened the free-running period of the mouse locomotor activity rhythm. DISCUSSION AND CONCLUSIONS: Our results indicate that Polygalae Radix may be regarded as a new therapy for circadian rhythm disorder and that the CaMKII pathway may be regarded as a target pathway for circadian rhythm disorders.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Relojes Circadianos/efectos de los fármacos , Extractos Vegetales/farmacología , Polygala , Animales , Relación Dosis-Respuesta a Droga , Masculino , Medicina Kampo , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Extractos Vegetales/uso terapéutico , Transducción de Señal/efectos de los fármacos
2.
Mol Brain ; 14(1): 152, 2021 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-34607601

RESUMEN

The glutamatergic signaling pathway is involved in molecular learning and human cognitive ability. Specific single variants (SNVs, formerly single-nucleotide polymorphisms) in the genes encoding N-methyl-D-aspartate receptor subunits have been associated with neuropsychiatric disorders by altering glutamate transmission. However, these variants associated with cognition and mental activity have rarely been explored in healthy adolescents. In this study, we screened for SNVs in the glutamatergic signaling pathway to identify genetic variants associated with cognitive ability. We found that SNVs in the subunits of ionotropic glutamate receptors, including GRIA1, GRIN1, GRIN2B, GRIN2C, GRIN3A, GRIN3B, and calcium/calmodulin-dependent protein kinase IIα (CaMK2A) are associated with cognitive function. Plasma CaMK2A level was correlated positively with the cognitive ability of Taiwanese senior high school students. We demonstrated that elevating CaMK2A increased its autophosphorylation at T286 and increased the expression of its downstream targets, including GluA1 and phosphor- GluA1 in vivo. Additionally, methyl-CpG binding protein 2 (MeCP2), a downstream target of CaMK2A, was found to activate the expression of CaMK2A, suggesting that MeCP2 and CaMK2A can form a positive feedback loop. In summary, two members of the glutamatergic signaling pathway, CaMK2A and MeCP2, are implicated in the cognitive ability of adolescents; thus, altering the expression of CaMK2A may affect cognitive ability in youth.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Cognición/fisiología , Proteína 2 de Unión a Metil-CpG/fisiología , Psicología del Adolescente , Receptores Ionotrópicos de Glutamato/genética , Transducción de Señal/fisiología , Adolescente , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/sangre , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Línea Celular Tumoral , Activación Enzimática , Retroalimentación Fisiológica/fisiología , Femenino , Ácido Glutámico/fisiología , Células HEK293 , Humanos , Masculino , Neuroblastoma , Fosforilación , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , Receptores Ionotrópicos de Glutamato/fisiología , Valores de Referencia , Taiwán
3.
Int J Mol Sci ; 22(5)2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33800907

RESUMEN

BACKGROUND: In the present study, we examined superoxide-mediated excitatory nociceptive transmission on at-level neuropathic pain following spinal thoracic 10 contusion injury (SCI) in male Sprague Dawley rats. METHODS: Mechanical sensitivity at body trunk, neuronal firing activity, and expression of superoxide marker/ionotropic glutamate receptors (iGluRs)/CamKII were measured in the T7/8 dorsal horn, respectively. RESULTS: Topical treatment of superoxide donor t-BOOH (0.4 mg/kg) increased neuronal firing rates and pCamKII expression in the naïve group, whereas superoxide scavenger Tempol (1 mg/kg) and non-specific ROS scavenger PBN (3 mg/kg) decreased firing rates in the SCI group (* p < 0.05). SCI showed increases of iGluRs-mediated neuronal firing rates and pCamKII expression (* p < 0.05); however, t-BOOH treatment did not show significant changes in the naïve group. The mechanical sensitivity at the body trunk in the SCI group (6.2 ± 0.5) was attenuated by CamKII inhibitor KN-93 (50 µg, 3.9 ± 0.4) or Tempol (1 mg, 4 ± 0.4) treatment (* p < 0.05). In addition, the level of superoxide marker Dhet showed significant increase in SCI rats compared to the sham group (11.7 ± 1.7 vs. 6.6 ± 1.5, * p < 0.05). CONCLUSIONS: Superoxide and the pCamKII pathway contribute to chronic at-level neuropathic pain without involvement of iGluRs following SCI.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Hiperalgesia/tratamiento farmacológico , Proteínas del Tejido Nervioso/fisiología , Neuralgia/tratamiento farmacológico , Nocicepción/efectos de los fármacos , Traumatismos de la Médula Espinal/tratamiento farmacológico , Superóxidos/metabolismo , Animales , Bencilaminas/farmacología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Contusiones/fisiopatología , Óxidos N-Cíclicos/farmacología , Depuradores de Radicales Libres/uso terapéutico , Hiperalgesia/etiología , Masculino , Modelos Animales , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Neuralgia/etiología , Umbral del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Receptores Ionotrópicos de Glutamato/efectos de los fármacos , Marcadores de Spin , Asta Dorsal de la Médula Espinal/efectos de los fármacos , Traumatismos de la Médula Espinal/fisiopatología , Sulfonamidas/farmacología , Transmisión Sináptica
4.
Brain Res ; 1750: 147154, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33068634

RESUMEN

Activation of Ca2+/calmodulin kinase II (CaMKII) and the N-Methyl D-aspartate receptor (NMDAR), particularly its GluN2B subunit, contribute to the central sensitization of nociceptive pathways and persistent pain. Using mutant mice wherein the activity-driven binding of CaMKII to S1303 in GluN2B is abrogated (GluN2BKI), this study investigated the importance of this interaction for acute and persistent inflammatory nociception. GluN2BKI, wild type and heterozygote mice did not differ in responses to acute noxious heat stimuli as measured with tail flick, paw flick, or hot plate assays, nor did they differ in their responses to mechanical stimulation with von Frey filaments. Surprisingly, the three genotypes exhibited similar spontaneous pain behaviors and hypersensitivity to heat or mechanical stimuli induced by intraplantar injection of capsaicin; however, GluN2BKI mice did not immediately attend to the paw. WT and GluN2BKI mice also did not differ in the nociceptive behaviors elicited by intraplantar injection of formalin, even though MK801 greatly reduced these behaviors in both genotypes concordant with NMDAR dependence. CaMKII binding to GluN2B at S1303 therefore does not appear to be critical for the development of inflammatory nociception. Finally, intrathecal KN93 reduced formalin-induced nociceptive behaviors in GluN2BKI mice. KN93 does not inhibit CaKMII, but rather binds Ca2+/calmodulin. It has multiple other targets including Ca2+-, Na+- and K+-channels, as well as various kinases. Therefore, the use of GluN2BKI mice provided genetic specificity in assessing the role of CaMKII in inflammatory pain signaling cascades. These results challenge current thinking on the involvement of the CaMKII-NMDAR interaction in inflammatory pain.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Inflamación/metabolismo , Dolor/metabolismo , Animales , Señalización del Calcio/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Calmodulina/metabolismo , Capsaicina/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Nocicepción/fisiología , Dolor/fisiopatología , Unión Proteica , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de N-Metil-D-Aspartato/fisiología
5.
Neurobiol Learn Mem ; 175: 107299, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32853813

RESUMEN

This study investigated the role of calcium2+/calmodulin-dependent protein kinase II (CaMKII), a protein in the second messenger pathway of NMDA receptors, in the ventral tegmental area (VTA) in the acquisition and performance of conditioned approach learning. Male Long-Evans rats (N = 79) were exposed to 3 (to test acquisition) or 7 (to test performance) conditioning sessions in which they received 30 paired presentations of a light stimulus (CS) and a food pellet (US) on a random time schedule. These conditioning sessions were then followed by one 30-min session without the CS or US and lastly by a CS-only test session, where only the light stimulus was presented (without food) according to the same schedule as the conditioning sessions. Bilateral intra-VTA injections of KN93 (vehicle, 3.0, 4.5 or 6.0 µg/0.5 µL), a CaMKII inhibitor, were administered prior to each conditioning session to test effects on the acquisition of conditioned approach or prior to the CS-only test session to test effects on the performance of conditioned approach. KN93, when given prior to conditioning sessions, significantly reduced the number of conditioned approach responses emitted during CS presentations in the CS-only test. When KN93 was given prior to the CS-only test it had no effect. These results suggest that CaMKII activation in the VTA is necessary for the acquisition, but not the performance, of reward-related learning.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Conducta de Elección/fisiología , Condicionamiento Clásico/fisiología , Área Tegmental Ventral/fisiología , Animales , Conducta Animal/efectos de los fármacos , Bencilaminas/farmacología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Condicionamiento Clásico/efectos de los fármacos , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Masculino , Microinyecciones , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Sulfonamidas/farmacología , Área Tegmental Ventral/efectos de los fármacos
6.
Cell Signal ; 75: 109734, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32791339

RESUMEN

Contrast-induced nephropathy (CIN), refers to acute kidney injury observed after administration of contrast media during angiographic or other medical procedures such as urography, and accounting for 12% of all causes of acute renal failure, but no specific prevention or treatment strategy exists for its obscure pathophysiology. The aim of our study was to explore the influence of calcium/calmodulin-dependent protein kinase II (CaMKII) in CIN by using HK-2 cells. Knockdown of CypD was achieved by lentivirus, and CaMKII overexpression by transfection with the plasmid. In this study, we have demonstrated that CypD-mediated mPTP opening triggered mitochondrial dysfunction and tubule cells apoptosis in CIN. We also found that iohexol treatment was associated with mitochondrial ROS overloading, ATP depletion and LDH release. Inhibition of CypD with the pharmacologic inhibitor or knockdown of CypD abrogated mPTP opening, oxidative stress, mitochondria damage, and cell apoptosis induced by iohexol. In addition, we found that inhibition of the CaMKII activity alleviated iohexol-induced CypD expression, whereas also decreased mPTP opening, oxidative stress, mitochondria damage, and cell apoptosis, similarly to the inhibition of CypD did. Moreover, CaMKII overexpression enhanced iohexol-induced mPTP opening, mitochondrial damage and renal tubular epithelial cells apoptosis. These findings first identified the novel role of CaMKII in iohexol-induced tubular cells apoptosis and delineated the CaMKII-CypD/mPTP pathway during contrast-induced tubular cell damage. Hence, these results could provide a new strategy for CIN protection.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Enfermedades Renales/inducido químicamente , Riñón/lesiones , Enfermedad Aguda , Apoptosis , Línea Celular , Medios de Contraste/efectos adversos , Humanos , Mitocondrias/metabolismo
7.
PLoS Comput Biol ; 16(7): e1008078, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32701987

RESUMEN

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.


Asunto(s)
Señalización del Calcio , Cuerpo Estriado/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Dopamina/fisiología , Aprendizaje , Plasticidad Neuronal , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Simulación por Computador , Dendritas/fisiología , Espinas Dendríticas/fisiología , Cinética , Ratones , Neuronas/fisiología , Receptores de Dopamina D2 , Recompensa
8.
Sci Rep ; 10(1): 10707, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32612162

RESUMEN

Synapse-Associated Protein 97 (SAP97) is an anchoring protein that in cardiomyocytes targets to the membrane and regulates Na+ and K+ channels. Here we compared the electrophysiological effects of native (WT) and p.P888L SAP97, a common polymorphism. Currents were recorded in cardiomyocytes from mice trans-expressing human WT or p.P888L SAP97 and in Chinese hamster ovary (CHO)-transfected cells. The duration of the action potentials and the QT interval were significantly shorter in p.P888L-SAP97 than in WT-SAP97 mice. Compared to WT, p.P888L SAP97 significantly increased the charge of the Ca-independent transient outward (Ito,f) current in cardiomyocytes and the charge crossing Kv4.3 channels in CHO cells by slowing Kv4.3 inactivation kinetics. Silencing or inhibiting Ca/calmodulin kinase II (CaMKII) abolished the p.P888L-induced Kv4.3 charge increase, which was also precluded in channels (p.S550A Kv4.3) in which the CaMKII-phosphorylation is prevented. Computational protein-protein docking predicted that p.P888L SAP97 is more likely to form a complex with CaMKII than WT. The Na+ current and the current generated by Kv1.5 channels increased similarly in WT-SAP97 and p.P888L-SAP97 cardiomyocytes, while the inward rectifier current increased in WT-SAP97 but not in p.P888L-SAP97 cardiomyocytes. The p.P888L SAP97 polymorphism increases the Ito,f, a CaMKII-dependent effect that may increase the risk of arrhythmias.


Asunto(s)
Potenciales de Acción/fisiología , Arritmias Cardíacas/fisiopatología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Homólogo 1 de la Proteína Discs Large/metabolismo , Miocitos Cardíacos/metabolismo , Canales de Potasio Shal/fisiología , Animales , Arritmias Cardíacas/genética , Células CHO , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Línea Celular , Cricetulus , Homólogo 1 de la Proteína Discs Large/genética , Humanos , Canal de Potasio Kv1.5/fisiología , Ratones , Técnicas de Placa-Clamp , Fosforilación/fisiología , Polimorfismo de Nucleótido Simple/genética
9.
Theranostics ; 10(7): 3049-3063, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194854

RESUMEN

Simultaneously targeted treatment of tumor cells and their surrounding growth-supporting immune cells is a promising strategy to reshape immunosuppressive tumor microenvironment (TME) and potentiate host innate and adaptive antitumor immune responses. Methods: We designed a series of melittin-(RADA)n hybrid peptide sequences with varying self-assembling motifs of RADA and screened out a melittin-(RADA)6 peptide that has an optimal gel-formation ability and in vitro antitumor activity. Results: The formed melittin-(RADA)6 (MR52) hydrogel scaffold could be loaded with a specific Ca2+/calmodulin-dependent protein kinase II (CAMKII) inhibitor, KN93, originally found to have both direct tumoricidal activity and macrophages-reprogramming ability, for potent immunotherapy against melanoma and hepatoma ascites in mice models. Our MR52 hydrogel has an interweaving nanofiber-like structure, possesses direct antitumor and controlled drug release properties, and promotes the enhanced intracellular uptake of loaded cargo. Compared to free KN93, the MR52-KN93 hydrogel (MRK) improved the killing effects and levels of immunogenic cell death (ICD) on tumor cells significantly. Due to the dual role of KN93, the injection of the MRK hydrogel retarded the growth of subcutaneous melanoma tumors dramatically and resulted in a high number of mature dendritic cells of draining lymph nodes, significantly enhancing the portion of cytotoxic T cells and reduced number of M2-like tumor-associated macrophages (TAMs) in tumors. Using a mouse model of malignant ascites (MAs), where traditional therapy was ineffective, we demonstrated that the MRK hydrogel treatment offered a significantly prolonged survival compared to controls. Following treatment with the MRK hydrogel, macrophages had elevated programmed cell death protein ligand-1 (PD-L1) expression, promising follow-up combined anti-PD-1 therapy that confers a cure rate of approximately 30% against MAs in mice models. Conclusion: Thus, the MRK hydrogel may serve as a prospective platform for antitumor applications.


Asunto(s)
Antineoplásicos/uso terapéutico , Ascitis/terapia , Bencilaminas/uso terapéutico , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Hidrogeles/administración & dosificación , Inmunoterapia/métodos , Neoplasias Hepáticas Experimentales/terapia , Melanoma Experimental/terapia , Meliteno/administración & dosificación , Terapia Molecular Dirigida/métodos , Proteínas de Neoplasias/antagonistas & inhibidores , Oligopéptidos/administración & dosificación , Inhibidores de Proteínas Quinasas/uso terapéutico , Sulfonamidas/uso terapéutico , Macrófagos Asociados a Tumores/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Antineoplásicos/administración & dosificación , Ascitis/etiología , Ascitis/inmunología , Antígeno B7-H1/biosíntesis , Bencilaminas/administración & dosificación , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Técnicas de Reprogramación Celular , Composición de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Inyecciones Intraperitoneales , Neoplasias Hepáticas Experimentales/complicaciones , Neoplasias Hepáticas Experimentales/inmunología , Activación de Macrófagos , Masculino , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas de Neoplasias/fisiología , Inhibidores de Proteínas Quinasas/administración & dosificación , Distribución Aleatoria , Proteínas Recombinantes de Fusión/administración & dosificación , Sulfonamidas/administración & dosificación , Escape del Tumor/efectos de los fármacos , Macrófagos Asociados a Tumores/clasificación , Macrófagos Asociados a Tumores/enzimología
10.
J Neurosci ; 40(11): 2228-2245, 2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32001612

RESUMEN

Sensory cortex exhibits receptive field plasticity throughout life in response to changes in sensory experience and offers the experimental possibility of aligning functional changes in receptive field properties with underpinning structural changes in synapses. We looked at the effects on structural plasticity of two different patterns of whisker deprivation in male and female mice: chessboard deprivation, which causes functional plasticity; and all deprived, which does not. Using 2-photon microscopy and chronic imaging through a cranial window over the barrel cortex, we found that layer 2/3 neurones exhibit robust structural plasticity, but only in response to whisker deprivation patterns that cause functional plasticity. Chessboard pattern deprivation caused dual-component plasticity in layer 2/3 by (1) increasing production of new spines that subsequently persisted for weeks and (2) enlarging spine head sizes in the preexisting stable spine population. Structural plasticity occurred on basal dendrites, but not apical dendrites. Both components of plasticity were absent in αCaMKII-T286A mutants that lack LTP and experience-dependent potentiation in barrel cortex, implying that αCaMKII autophosphorylation is not only important for stabilization and enlargement of spines, but also for new spine production. These studies therefore reveal the relationship between spared whisker potentiation in layer 2/3 neurones and the form and mechanisms of structural plasticity processes that underlie them.SIGNIFICANCE STATEMENT This study provides a missing link in a chain of reasoning that connects LTP to experience-dependent functional plasticity in vivo We found that increases in dendritic spine formation and spine enlargement (both of which are characteristic of LTP) only occurred in barrel cortex during sensory deprivation that produced potentiation of sensory responses. Furthermore, the dendritic spine plasticity did not occur during sensory deprivation in mice lacking LTP and experience-dependent potentiation (αCaMKII autophosphorylation mutants). We also found that the dual-component dendritic spine plasticity only occurred on basal dendrites and not on apical dendrites, thereby resolving a paradox in the literature suggesting that layer 2/3 neurones lack structural plasticity in response to sensory deprivation.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Espinas Dendríticas/fisiología , Plasticidad Neuronal/fisiología , Neuronas/enzimología , Privación Sensorial/fisiología , Corteza Somatosensorial/fisiopatología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/deficiencia , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Tamaño de la Célula , Espinas Dendríticas/ultraestructura , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/ultraestructura , Fosforilación , Procesamiento Proteico-Postraduccional , Técnica de Ventana Cutánea , Corteza Somatosensorial/citología , Trastornos Somatosensoriales/fisiopatología , Vibrisas/lesiones , Vibrisas/inervación
11.
J Neurosci ; 40(11): 2332-2342, 2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32005763

RESUMEN

Emotional disorders are common comorbid conditions that further exacerbate the severity and chronicity of chronic pain. However, individuals show considerable vulnerability to the development of chronic pain under similar pain conditions. In this study on male rat and mouse models of chronic neuropathic pain, we identify the histone deacetylase Sirtuin 1 (SIRT1) in central amygdala as a key epigenetic regulator that controls the development of comorbid emotional disorders underlying the individual vulnerability to chronic pain. We found that animals that were vulnerable to developing behaviors of anxiety and depression under the pain condition displayed reduced SIRT1 protein levels in central amygdala, but not those animals resistant to the emotional disorders. Viral overexpression of local SIRT1 reversed this vulnerability, but viral knockdown of local SIRT1 mimicked the pain effect, eliciting the pain vulnerability in pain-free animals. The SIRT1 action was associated with CaMKIIα downregulation and deacetylation of histone H3 lysine 9 at the CaMKIIα promoter. These results suggest that, by transcriptional repression of CaMKIIα in central amygdala, SIRT1 functions to guard against the emotional pain vulnerability under chronic pain conditions. This study indicates that SIRT1 may serve as a potential therapeutic molecule for individualized treatment of chronic pain with vulnerable emotional disorders.SIGNIFICANCE STATEMENT Chronic pain is a prevalent neurological disease with no effective treatment at present. Pain patients display considerably variable vulnerability to developing chronic pain, indicating individual-based molecular mechanisms underlying the pain vulnerability, which is hardly addressed in current preclinical research. In this study, we have identified the histone deacetylase Sirtuin 1 (SIRT1) as a key regulator that controls this pain vulnerability. This study reveals that the SIRT1-CaMKIIaα pathway in central amygdala acts as an epigenetic mechanism that guards against the development of comorbid emotional disorders under chronic pain, and that its dysfunction causes increased vulnerability to the development of chronic pain. These findings suggest that SIRT1 activators may be used in a novel therapeutic approach for individual-based treatment of chronic pain.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Núcleo Amigdalino Central/fisiopatología , Dolor Crónico/fisiopatología , Distrés Psicológico , Sirtuina 1/fisiología , Neuralgia del Trigémino/fisiopatología , Acetilación , Animales , Ansiedad/etiología , Ansiedad/fisiopatología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Núcleo Amigdalino Central/enzimología , Dolor Crónico/psicología , Depresión/etiología , Depresión/fisiopatología , Susceptibilidad a Enfermedades , Regulación hacia Abajo , Conducta Exploratoria , Neuronas GABAérgicas/enzimología , Vectores Genéticos , Histonas/metabolismo , Hiperalgesia/fisiopatología , Masculino , Ratones , Regiones Promotoras Genéticas , Ratas , Ratas Wistar , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/genética , Natación , Transcripción Genética , Neuralgia del Trigémino/psicología
12.
Mol Reprod Dev ; 87(5): 598-606, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32017318

RESUMEN

Extracellular calcium is required for intracellular Ca2+ oscillations needed for egg activation, but the regulatory mechanism is still poorly understood. The present study was designed to demonstrate the function of calcium-sensing receptor (CASR), which could recognize extracellular calcium as first messenger, during porcine egg activation. CASR expression was markedly upregulated following egg activation. Functionally, the addition of CASR agonist NPS R-568 significantly enhanced pronuclear formation rate, while supplementation of CASR antagonist NPS2390 compromised egg activation. There was no change in NPS R-568 group compared with control group when the egg activation was performed without extracellular calcium addition. The addition of NPS2390 precluded the activation-dependent [Ca2+ ]i rise. When egg activation was conducted in intracellular Ca2+ chelator BAPTA-AM and NPS R-568 containing medium, CASR function was abolished. Meanwhile, CASR activation increased the level of the [Ca2+ ]i effector p-CAMKII, and the presence of KN-93, an inhibitor of CAMKII, significantly reduced the CASR-mediated increasement of pronuclear formation rate. Furthermore, the increase of CASR expression following activation was reversed by inhibiting CAMKII activity, supporting a positive feedback loop between CAMKII and CASR. Altogether, these findings provide a new pathway of egg activation about CASR, as the extracellular Ca2+ effector, promotes egg activation via its downstream effector and upstream regulator CAMKII.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Fertilización/fisiología , Receptores Sensibles al Calcio/fisiología , Porcinos/fisiología , Adamantano/análogos & derivados , Adamantano/farmacología , Animales , Bencilaminas/farmacología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Células Cultivadas , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Femenino , Fertilización/efectos de los fármacos , Masculino , Fenetilaminas/farmacología , Propilaminas/farmacología , Quinoxalinas/farmacología , Receptores Sensibles al Calcio/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Interacciones Espermatozoide-Óvulo/efectos de los fármacos , Sulfonamidas/farmacología
13.
Neuron ; 105(2): 334-345.e9, 2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31786014

RESUMEN

Electrical events in neurons occur on the order of milliseconds, but the brain can process and reproduce intervals millions of times longer. We present what we believe to be the first neuronal mechanism for timing intervals longer than a few seconds. The activation and gradual relaxation of calcium-independent CaMKII measure a 6-min time window to coordinate two male-specific events during Drosophila mating: sperm transfer and a simultaneous decrease in motivation. We localize these functions to four neurons whose electrical activity is necessary only to report the conclusion of the decline in CaMKII's activity, not for the measurement of the interval. The computation of elapsed time is therefore largely invisible to standard methods of monitoring neuronal activity. Its broad conservation, ubiquitous expression, and tunable duration of activity suggest that CaMKII may time a wide variety of behavioral and cognitive processes.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Motivación/fisiología , Conducta Sexual Animal/fisiología , Factores de Tiempo , Animales , Animales Modificados Genéticamente , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Drosophila , Femenino , Masculino , Neuronas/fisiología
14.
Brain Res Bull ; 155: 11-18, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31743748

RESUMEN

Curtailment of sleep in modern society leads to a spectrum of neuropsychiatric disorders. However, the molecular mechanisms underlying the effects of sleep deprivation (SD) remain elusive and currently there is no effective therapy to alleviate these effects. Here, we aimed to examine SD-induced cellular and molecular alterations in mouse prefrontal cortex (PFC) and whether subchronic citalopram (CTM) treatment can negate these alterations. Three-month-old C57BL/6 J mice were divided into control (Ctrl), SD, CTM alone and CTM + SD groups. CTM and CTM + SD group mice were treated with CTM for five consecutive days at a dose of 10 mg/kg per day before the experimental procedure. SD and CTM + SD group mice were sleep-deprived for 24 h using an automated treadmill method. We found that 24 h SD causes a marked reduction in the levels of phosphorylated calcium/calmodulin kinase II (pCaMKII), phosphorylated cyclic AMP-responsive element binding protein (pCREB) and brain-derived neurotrophic factor (BDNF) in mouse PFC. Patch clamp recording of pyramidal neurons from acute PFC slices revealed that SD decreases the amplitude of miniature excitatory postsynaptic currents (mEPSCs), suggesting a SD-induced postsynaptic alteration. Interestingly, subchronic CTM treatment prevents such SD-induced reductions in pCaMKII, pCREB and BDNF levels, and in mEPSC amplitude. These data suggest that CTM offers neuroprotection against SD-induced molecular and electrophysiological alterations.


Asunto(s)
Antidepresivos de Segunda Generación/administración & dosificación , Citalopram/administración & dosificación , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiopatología , Transducción de Señal/efectos de los fármacos , Privación de Sueño/fisiopatología , Animales , Factor Neurotrófico Derivado del Encéfalo/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Femenino , Masculino , Ratones Endogámicos C57BL , Potenciales Postsinápticos Miniatura , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología
15.
EMBO J ; 38(23): e101230, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31625188

RESUMEN

Tunneling nanotubes (TNTs) are actin-based transient tubular connections that allow direct communication between distant cells. TNTs play an important role in several physiological (development, immunity, and tissue regeneration) and pathological (cancer, neurodegeneration, and pathogens transmission) processes. Here, we report that the Wnt/Ca2+ pathway, an intracellular cascade that is involved in actin cytoskeleton remodeling, has a role in TNT formation and TNT-mediated transfer of cargoes. Specifically, we found that Ca2+ /calmodulin-dependent protein kinase II (CaMKII), a transducer of the Wnt/Ca2+ pathway, regulates TNTs in a neuronal cell line and in primary neurons. We identified the ß isoform of CaMKII as a key molecule in modulating TNT formation and transfer, showing that this depends on the actin-binding activity of the protein. Finally, we found that the transfer of vesicles and aggregated α-synuclein between primary neurons can be regulated by the activation of the Wnt/Ca2+ pathway. Our findings suggest that Wnt/Ca2+ pathway could be a novel promising target for therapies designed to impair TNT-mediated propagation of pathogens.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Calcio/metabolismo , Comunicación Celular , Membrana Celular/metabolismo , Nanotubos/química , Neuronas/fisiología , Proteínas Wnt/metabolismo , Actinas/metabolismo , Animales , Señalización del Calcio , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Transducción de Señal
16.
Neuron ; 103(3): 380-394, 2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-31394063

RESUMEN

The Ca2+/calmodulin (CaM)-dependent protein kinase II (CaMKII) was touted as a memory molecule, even before its involvement in long-term potentiation (LTP) was shown. The enzyme has not disappointed, with subsequent demonstrations of remarkable structural and regulatory properties. Its neuronal functions now extend to long-term depression (LTD), and last year saw the first direct evidence for memory storage by CaMKII. Although CaMKII may have taken the spotlight, it is a member of a large family of diverse and interesting CaM kinases. Our aim is to place CaMKII in context of the other CaM kinases and then review certain aspects of this kinase that are of current interest.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Secuencia de Aminoácidos , Animales , Encéfalo/enzimología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/química , Cognición/fisiología , Humanos , Potenciación a Largo Plazo/fisiología , Memoria/fisiología , Modelos Moleculares , Familia de Multigenes , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/fisiología , Fosforilación , Conformación Proteica , Dominios Proteicos , Mapeo de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/clasificación , Proteínas Serina-Treonina Quinasas/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Transmisión Sináptica
17.
FASEB J ; 33(9): 10193-10206, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31199885

RESUMEN

The ion pump Na+, K+-ATPase (NKA) is a receptor for the cardiotonic steroid ouabain. Subsaturating concentration of ouabain triggers intracellular calcium oscillations, stimulates cell proliferation and adhesion, and protects from apoptosis. However, it is controversial whether ouabain-bound NKA is considered a signal transducer. To address this question, we performed a global analysis of protein phosphorylation in COS-7 cells, identifying 2580 regulated phosphorylation events on 1242 proteins upon 10- and 20-min treatment with ouabain. Regulated phosphorylated proteins include the inositol triphosphate receptor and stromal interaction molecule, which are essential for initiating calcium oscillations. Hierarchical clustering revealed that ouabain triggers a structured phosphorylation response that occurs in a well-defined, time-dependent manner and affects specific cellular processes, including cell proliferation and cell-cell junctions. We additionally identify regulation of the phosphorylation of several calcium and calmodulin-dependent protein kinases (CAMKs), including 2 sites of CAMK type II-γ (CAMK2G), a protein known to regulate apoptosis. To verify the significance of this result, CAMK2G was knocked down in primary kidney cells. CAMK2G knockdown impaired ouabain-dependent protection from apoptosis upon treatment with high glucose or serum deprivation. In conclusion, we establish NKA as the coordinator of a broad, tightly regulated phosphorylation response in cells and define CAMK2G as a downstream effector of NKA.-Panizza, E., Zhang, L., Fontana, J. M., Hamada, K., Svensson, D., Akkuratov, E. E., Scott, L., Mikoshiba, K., Brismar, H., Lehtiö, J., Aperia, A. Ouabain-regulated phosphoproteome reveals molecular mechanisms for Na+, K+-ATPase control of cell adhesion, proliferation, and survival.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Ouabaína/farmacología , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , ATPasa Intercambiadora de Sodio-Potasio/fisiología , Secuencia de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Células COS , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , División Celular/efectos de los fármacos , División Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Chlorocebus aethiops , Regulación hacia Abajo/efectos de los fármacos , Glucosa/farmacología , Receptores de Inositol 1,4,5-Trifosfato/fisiología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/enzimología , Proteínas Quinasas Activadas por Mitógenos/biosíntesis , Proteínas Quinasas Activadas por Mitógenos/genética , Modelos Moleculares , Fosforilación , Conformación Proteica , Proteínas Quinasas/efectos de los fármacos , Proteoma , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Ratas , ATPasa Intercambiadora de Sodio-Potasio/efectos de los fármacos
18.
Neuron ; 103(3): 473-488.e6, 2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-31202540

RESUMEN

Innate defensive responses are essential for animal survival and are conserved across species. The ventral tegmental area (VTA) plays important roles in learned appetitive and aversive behaviors, but whether it plays a role in mediating or modulating innate defensive responses is currently unknown. We report that VTAGABA+ neurons respond to a looming stimulus. Inhibition of VTAGABA+ neurons reduced looming-evoked defensive flight behavior, and photoactivation of these neurons resulted in defense-like flight behavior. Using viral tracing and electrophysiological recordings, we show that VTAGABA+ neurons receive direct excitatory inputs from the superior colliculus (SC). Furthermore, we show that glutamatergic SC-VTA projections synapse onto VTAGABA+ neurons that project to the central nucleus of the amygdala (CeA) and that the CeA is involved in mediating the defensive behavior. Our findings demonstrate that aerial threat-related visual information is relayed to VTAGABA+ neurons mediating innate behavioral responses, suggesting a more general role of the VTA.


Asunto(s)
Reacción de Fuga/fisiología , Miedo/fisiología , Neuronas GABAérgicas/fisiología , Área Tegmental Ventral/fisiología , Vías Aferentes/fisiología , Animales , Señalización del Calcio , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Núcleo Amigdalino Central/fisiología , Genes Reporteros , Ácido Glutámico/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/fisiología , Optogenética , Estimulación Luminosa , Proteínas Proto-Oncogénicas c-fos/análisis , Ácido gamma-Aminobutírico/fisiología
19.
Sci Rep ; 9(1): 9291, 2019 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-31243295

RESUMEN

Reactive oxygen species (ROS) contribute to myocardial death during ischemia-reperfusion (I/R) injury, but detailed knowledge of molecular pathways connecting ROS to cardiac injury is lacking. Activation of the Ca2+/calmodulin-dependent protein kinase II (CaMKIIδ) is implicated in myocardial death, and CaMKII can be activated by ROS (ox-CaMKII) through oxidation of regulatory domain methionines (Met281/282). We examined I/R injury in mice where CaMKIIδ was made resistant to ROS activation by knock-in replacement of regulatory domain methionines with valines (MMVV). We found reduced myocardial death, and improved left ventricular function 24 hours after I/R injury in MMVV in vivo and in vitro compared to WT controls. Loss of ATP sensitive K+ channel (KATP) current contributes to I/R injury, and CaMKII promotes sequestration of KATP from myocardial cell membranes. KATP current density was significantly reduced by H2O2 in WT ventricular myocytes, but not in MMVV, showing ox-CaMKII decreases KATP availability. Taken together, these findings support a view that ox-CaMKII and KATP are components of a signaling axis promoting I/R injury by ROS.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Miocardio/patología , Oxígeno/química , Daño por Reperfusión/patología , Adenosina Trifosfato/química , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Supervivencia Celular , Ecocardiografía , Prueba de Tolerancia a la Glucosa , Peróxido de Hidrógeno/química , Insulina/metabolismo , Masculino , Metionina/química , Ratones , Isquemia Miocárdica , Miocitos Cardíacos/metabolismo , Oxidación-Reducción , Condicionamiento Físico Animal , Potasio/química , Dominios Proteicos , Especies Reactivas de Oxígeno/metabolismo
20.
Nat Neurosci ; 22(7): 1110-1121, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31160741

RESUMEN

Learning to predict rewards based on environmental cues is essential for survival. The orbitofrontal cortex (OFC) contributes to such learning by conveying reward-related information to brain areas such as the ventral tegmental area (VTA). Despite this, how cue-reward memory representations form in individual OFC neurons and are modified based on new information is unknown. To address this, using in vivo two-photon calcium imaging in mice, we tracked the response evolution of thousands of OFC output neurons, including those projecting to VTA, through multiple days and stages of cue-reward learning. Collectively, we show that OFC contains several functional clusters of neurons distinctly encoding cue-reward memory representations, with only select responses routed downstream to VTA. Unexpectedly, these representations were stably maintained by the same neurons even after extinction of the cue-reward pairing, and supported behavioral learning and memory. Thus, OFC neuronal activity represents a long-term cue-reward associative memory to support behavioral adaptation.


Asunto(s)
Adaptación Psicológica/fisiología , Aprendizaje por Asociación/fisiología , Señalización del Calcio , Condicionamiento Clásico/fisiología , Memoria a Largo Plazo/fisiología , Neuronas/fisiología , Corteza Prefrontal/fisiología , Recompensa , Estimulación Acústica , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Señales (Psicología) , Conducta de Ingestión de Líquido/fisiología , Extinción Psicológica , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/fisiología , Neuronas/enzimología , Optogenética , Técnicas de Placa-Clamp , Corteza Prefrontal/citología , Análisis de la Célula Individual , Área Tegmental Ventral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...