Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Neurochem Int ; 148: 105098, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34129896

RESUMEN

Obesity is characterized by a condition of low-grade chronic inflammation that facilitates development of numerous comorbidities and dysregulation of brain homeostasis. It is reported that obesity can lead to behavioral alterations such as cognitive decline and depression-like behaviors both in humans and rodents. Saponins from panax japonicus (SPJ) have been reported to exhibit anti-inflammatory action in mouse model of diet-induced obesity. We evaluated the neuroprotection of SPJ on high fat diet (HFD) induced impaired behaviors such as memory deficit and depressive-like behaviors, and explored the underlying mechanisms. 6-week male Balb/c mice were divided into normal control group (NC, 17% total calories from fat), HFD group (60% total calories from fat), and HFD treated with SPJ groups (orally gavaged with dosages of 15 mg/kg and 45 mg/kg), respectively. After treatment for 16 weeks, behavioral tests were performed to evaluate the cognition and depression-like behaviors of the mice. The underling mechanisms of SPJ on HFD-induced impaired behaviors were investigated through histopathological observation, Western blot analysis and immunofluorescence. Our results showed that HFD-fed mice caused behavioral disorders, neuronal degeneration as well as elevated neuroinflammation, which was partly involved in NLRP3 inflammasome that finally resulted in decreased protein levels of AMPA receptors and down-regulated phosphorylated levels of CaMKII and CREB in cortex and hippocampus. All the above changes in cortex and hippocampus induced by HFD were mitigated by SPJ treatment. SPJ treatment alleviated HFD-induced recognitive impairment and depression-like behaviors of mice, which could be partly due to the capacity of SPJ to mitigate neuroinflammation through inhibition of NLRP3 inflammasome and upregulation of AMPA receptors signaling pathway.


Asunto(s)
Conducta Animal/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Inflamasomas/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Panax/química , Receptores AMPA/biosíntesis , Receptores AMPA/efectos de los fármacos , Saponinas/farmacología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/biosíntesis , Depresión/inducido químicamente , Depresión/psicología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/psicología , Ratones , Ratones Endogámicos BALB C , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
2.
Int J Dev Neurosci ; 69: 44-48, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29966740

RESUMEN

BACKGROUND: Frankincense is an oleo gum resin derived from trees of genus Boswellia. It has favorable effects on memory formation. However, the probable underlying molecular mechanisms have not been assessed. Frankincense exerts some of its effects via activation of protein kinases. Calcium/calmodulin kinaseII (CaMKII) and CaMKIV are crucial mediators of learning and memory. We studied the effect of maternal injection of the aqueous extract of frankincense during gestation and lactation periods on spatial memory performance and the mRNA expression levels of the hippocampal CaMKIIand CaMKIV in the offspring rats. METHODS: Aqueous extract of Frankincense (50 and 100 mg/kg) or tap water was gavaged to distinct female rats during gestation and lactation periods. Memory performance was assessed in groups of male offspring using Morris water maze. In other groups of the offspring (with no memory test), the hippocampi of the juvenile rats were removed 30 days after labor. A real-time PCR method was used to measure the mRNA levels of CaMKII and CaMKIV. RESULTS: Frankincense improved spatial memory retrieval in the offspring rats in a dose-dependent manner. The mRNA expression of hippocampal CaMKIV was unchanged between groups. However, the mRNA expression of hippocampal CaMKII was dose-dependently upregulated in the rats, whose mothers had received frankincense. CONCLUSIONS: Due to the crucial role of the CaMKII in memory formation, the results provide a molecular basis for the effect of administration of frankincense to mother rats on improvement of the memory in the offspring.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Olíbano/química , Hipocampo/metabolismo , Memoria/efectos de los fármacos , Extractos Vegetales/farmacología , Desempeño Psicomotor/efectos de los fármacos , Animales , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/biosíntesis , ADN Complementario/biosíntesis , ADN Complementario/genética , Relación Dosis-Respuesta a Droga , Femenino , Hipocampo/efectos de los fármacos , Lactancia , Aprendizaje por Laberinto/efectos de los fármacos , Embarazo , Ratas , Ratas Wistar , Regulación hacia Arriba/efectos de los fármacos
3.
Neuromolecular Med ; 17(2): 121-36, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25680935

RESUMEN

Stress is any condition that impairs the balance of the organism physiologically or psychologically. The response to stress involves several neurohormonal consequences. Glutamate is the primary excitatory neurotransmitter in the central nervous system, and its release is increased by stress that predisposes to excitotoxicity in the brain. Memantine is an uncompetitive N-methyl D-aspartate glutamatergic receptors antagonist and has shown beneficial effect on cognitive function especially in Alzheimer's disease. The aim of the work was to investigate memantine effect on memory and behavior in animal models of acute and repeated restraint stress with the evaluation of serum markers of stress and the expression of hippocampal markers of synaptic plasticity. Forty-two male rats were divided into seven groups (six rats/group): control, acute restraint stress, acute restraint stress with Memantine, repeated restraint stress, repeated restraint stress with Memantine and Memantine groups (two subgroups as positive control). Spatial working memory and behavior were assessed by performance in Y-maze. We evaluated serum cortisol, tumor necrotic factor, interleukin-6 and hippocampal expression of brain-derived neurotrophic factor, synaptophysin and calcium-/calmodulin-dependent protein kinase II. Our results revealed that Memantine improved spatial working memory in repeated stress, decreased serum level of stress markers and modified the hippocampal synaptic plasticity markers in both patterns of stress exposure; in ARS, Memantine upregulated the expression of synaptophysin and brain-derived neurotrophic factor and downregulated the expression of calcium-/calmodulin-dependent protein kinase II, and in repeated restraint stress, it upregulated the expression of synaptophysin and downregulated calcium-/calmodulin-dependent protein kinase II expression.


Asunto(s)
Conducta Animal/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/uso terapéutico , Hipocampo/efectos de los fármacos , Memantina/uso terapéutico , Plasticidad Neuronal/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Restricción Física/efectos adversos , Memoria Espacial/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Estrés Psicológico/tratamiento farmacológico , Enfermedad Aguda , Animales , Ansiedad/sangre , Ansiedad/tratamiento farmacológico , Ansiedad/etiología , Conducta Animal/fisiología , Biomarcadores/sangre , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Evaluación Preclínica de Medicamentos , Antagonistas de Aminoácidos Excitadores/farmacología , Reacción Cataléptica de Congelación/efectos de los fármacos , Reacción Cataléptica de Congelación/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Aseo Animal/efectos de los fármacos , Aseo Animal/fisiología , Hipocampo/química , Hipocampo/fisiopatología , Hidrocortisona/sangre , Interleucina-6/sangre , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Memantina/farmacología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Neurogénesis/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Wistar , Memoria Espacial/fisiología , Estrés Fisiológico/fisiología , Estrés Psicológico/sangre , Estrés Psicológico/etiología , Estrés Psicológico/fisiopatología , Sinaptofisina/biosíntesis , Sinaptofisina/genética , Factor de Necrosis Tumoral alfa/sangre
4.
Pharmacol Rep ; 67(2): 370-5, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25712666

RESUMEN

BACKGROUND: Neuroinflammation is considered to be a major factor in several neurodegenerative diseases. Recently, the polyunsaturated fatty acid omega-3 has been shown to have anti-inflammatory effects and might play an effective role in improving memory impairment due to inflammation. In order to test this, we stimulated neuroinflammation in an animal model and induced memory dysfunction as measured by reduced retention of passive avoidance learning (PAL) and altered expression of CaMKII-α, a gene known to be crucial for memory formation. We then investigated whether treatment with dietary omega-3 prevents inflammation-induced memory dysfunction in this model. METHODS: Male wistar rats (200-220 g) were fed either a control diet or a diet containing omega-3 (400mg/kg, po) for 1 month prior. Rats then received injection of either saline or LPS (500 µg/kg, ip) and were subjected to the PAL acquisition task. The retention test was performed 24h later, and animals were sacrificed immediately. Hippocampi were dissected and stored at -80°C. Finally, TNF-α levels and CaMKII-α gene expression were measured by ELISA and qRT-PCR, respectively. RESULTS: We found that LPS treatment significantly impaired PAL and memory, increased TNF-α levels and impaired CaMKII-α gene expression. In control and LPS-injected animals, pre-treatment with omega-3 improved performance on the PAL task and increased CAMKII-α gene expression. CONCLUSION: Taken together, these data suggest that dietary omega-3 may improve cognitive function and provide a potential therapy for memory impairment due to neuroinflammation.


Asunto(s)
Reacción de Prevención/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Ácidos Grasos Omega-3/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Lipopolisacáridos/antagonistas & inhibidores , Trastornos de la Memoria/prevención & control , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Dieta , Expresión Génica/efectos de los fármacos , Lipopolisacáridos/farmacología , Masculino , Trastornos de la Memoria/inducido químicamente , Ratas , Factor de Necrosis Tumoral alfa/metabolismo
5.
Neuroscience ; 218: 216-25, 2012 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-22626645

RESUMEN

In the present study, withdrawal symptoms induced by morphine or ß-endorphin administered intracerebroventricularly (i.c.v.) were compared in ICR mice. Naloxone (10mg/kg) was post-treated intraperitoneally (i.p.) 3h after either a single or repeated (1 time/day for 3 days) i.c.v. injections with opioids. Withdrawal symptoms such as jumping frequency, diarrhea, weight loss, rearing, penile licking and paw tremor were observed for 30 min immediately after naloxone treatment. Withdrawal symptoms (jumping, diarrhea, weight loss, rearing, penile licking and paw tremor) observed in the group treated with morphine was persistently increased during 3 days. On the other hand, withdrawal symptoms such as diarrhea, weight loss and rearing in ß-endorphin-treated group were increased after a single injection with ß-endorphin, but gradually decreased after the repeated injection. Furthermore, no jumping behavior, penile licking and paw tremor in ß-endorphin-treated group were observed throughout the whole period of time. In addition, the hypothalamic changes of several signal molecules such as pERK, pCaMK-IIα, c-FOS and pCREB expression were observed during the presence or absence of withdrawal responses induced by morphine or ß-endorphin administered once or repeatedly. Both hypothalamic pCaMK-IIα and c-FOS expressions were increased by naloxone treatment in acutely administered morphine group, whereas only pCaMK-IIα expression was elevated by naloxone treatment in repeatedly administered morphine group. In contrast with the findings in morphine-treated group, only pCaMK-IIα expression was decreased by naloxone treatment in repeatedly administered ß-endorphin group. Our results suggest that profiles of the withdrawal symptoms induced by morphine and ß-endorphin administered supraspinally appear to be differentially regulated. The pCaMK-IIα and the c-FOS protein expression may play important roles for the regulation of naloxone-precipitated withdrawal symptoms such as jumping, diarrhea, weight loss, rearing, penile licking and paw tremor induced by morphine-treated group, whereas the phosphorylation of hypothalamic pCaMK-IIα appears to be involved only in the regulation of naloxone-precipitated withdrawal symptoms such as diarrhea, weight loss and rearing in ß-endorphin-treated group.


Asunto(s)
Hipotálamo/efectos de los fármacos , Morfina/administración & dosificación , Narcóticos/administración & dosificación , Síndrome de Abstinencia a Sustancias , betaendorfina/administración & dosificación , Animales , Western Blotting , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Hipotálamo/metabolismo , Inyecciones Intraventriculares , Masculino , Ratones , Ratones Endogámicos ICR , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Síndrome de Abstinencia a Sustancias/metabolismo
6.
Toxicol Sci ; 118(1): 108-18, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20810541

RESUMEN

Calcium is an essential signaling molecule in developing B cells, thus altering calcium dynamics represents a potential target for toxicant effects. GW7845, a tyrosine analog and potent peroxisome proliferator-activated receptor γ agonist, induces rapid mitogen-activated protein kinase (MAPK)-dependent apoptosis in bone marrow B cells. Changes in calcium dynamics are capable of mediating rapid initiation of cell death; therefore, we investigated the contribution of calcium to GW7845-induced apoptosis. Treatment of a nontransformed murine pro/pre-B cell line (BU-11) with GW7845 (40 µM) resulted in intracellular calcium release. Multiple features of GW7845-induced cell death were suppressed by the calcium chelator BAPTA, including MAPK activation, loss of mitochondrial membrane potential, cytochrome c release, caspase-3 activation, and DNA fragmentation. A likely mechanism for the calcium-mediated effects is activation of CaMKII, a calcium-dependent MAP4K. We observed that three CaMKII isoforms (ß, γ, and δ) are expressed in lymphoid tissues and bone marrow B cells. Treatment with GW7845 increased CaMKII activity. All features of GW7845-induced cell death, except loss of mitochondrial membrane potential, were suppressed by CaMKII inhibitors (KN93 and AIP-II), suggesting the activation of multiple calcium-driven pathways. To determine if CaMKII activation is a common feature of early B cell death following perturbation of Ca(2+) flux, we dissected tributyltin (TBT)-induced death signaling. High-dose TBT (1 µM) is known to activate calcium-dependent death. TBT induced rapid apoptosis that was associated with intracellular calcium release, CaMKII activation and MAPK activation, and was inhibited by AIP-II. Thus, we show that early B cells are susceptible to calcium-triggered cell death through a CaMKII/MAPK-dependent pathway.


Asunto(s)
Apoptosis/fisiología , Linfocitos B/metabolismo , Células de la Médula Ósea/enzimología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Calcio/metabolismo , Animales , Apoptosis/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Línea Celular , Quelantes/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Isoenzimas , Ligandos , MAP Quinasa Quinasa 4/biosíntesis , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Oxazoles/farmacología , Compuestos de Trialquiltina/farmacología , Tirosina/análogos & derivados , Tirosina/farmacología
7.
Naunyn Schmiedebergs Arch Pharmacol ; 381(2): 137-45, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20052460

RESUMEN

Scutellarin is a flavonoid extracted from a traditional Chinese herb, Erigeron breviscapus Hand Mazz, which has been broadly used in treating various cardiovascular diseases. In this study, we investigated its effect on cardiac hypertrophy and the underlying mechanism. Both in vitro and in vivo cardiac hypertrophy models were employed to explore the anti-hypertrophic action of scutellarin. We found that scutellarin significantly suppressed the hypertrophic growth of neonatal cardiac myocytes exposed to phenylephrine (PE) and mouse heart subjected to pressure overload induced by aortic banding, accompanied with the decreased expression of hypertrophic markers beta-myosin heavy chain and atrial natriuretic peptide. We then measured the change of free intracellular calcium using laser scanning confocal microscope. We found that scutellarin alleviated the increment of free intracellular calcium during cardiac hypertrophy either induced by PE or aortic banding. The expression of calcium downstream effectors calcineurin and phosphorylated calmodulin kinase II (CaMKII) were significantly suppressed by scutellarin. Our study indicated that scutellarin exerts its anti-hypertrophic activity via suppressing the Ca(2+)-mediated calcineurin and CaMKII pathways, which supports the observation that clinical application of scutellarin is beneficial for cardiovascular disease patients.


Asunto(s)
Apigenina/farmacología , Calcineurina/biosíntesis , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Calcio/fisiología , Glucuronatos/farmacología , Vasodilatadores/farmacología , Animales , Animales Recién Nacidos , Apigenina/uso terapéutico , Señalización del Calcio , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/metabolismo , Cardiomegalia/patología , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Glucuronatos/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fenilefrina/farmacología , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Vasodilatadores/uso terapéutico
8.
Neuroscience ; 163(3): 741-9, 2009 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-19596052

RESUMEN

The senescence-accelerated mouse prone-8 (SAMP8) is characterized by early onset of learning and memory deficits along with spontaneous overproduction of soluble beta-amyloid peptide (Abeta) in the brain. In our study, 4 month old male SAMP8 mice were orally administered 0.05% and 0.1% green tea catechins (GTC, w/v) in drinking water for 6 months. We found that a supplementation with 0.05% or 0.1% GTC prevented spatial learning and memory impairments of mice in the Morris water maze. Better performance of GTC-treated mice was associated with decreased levels of Abeta(1-42) oligomers in the hippocampus. The activity of the protein kinase A/cAMP-response element binding protein (PKA/CREB) pathway, one of the molecular targets of Abeta oligomers which is crucial for late long-term potentiation and long-term memory formation, was significantly increased after GTC administration. We also found that chronic 0.05% or 0.1% GTC consumption prevented the reductions of three representative proteins of synaptic function and synaptic structure, including brain-derived neurotrophic factor(BDNF), post-synaptic density protein-95 (PSD95) and Ca(2+)/calmodulin-dependent protein kinase II (CaMKII). These results demonstrated that long-term 0.05% or 0.1% green tea catechin administration may prevent spatial learning and memory decline of SAMP8 mice by decreasing Abeta(1-42) oligomers and upregulating synaptic plasticity-related proteins in the hippocampus.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Catequina/farmacología , Hipocampo/efectos de los fármacos , Aprendizaje/efectos de los fármacos , Proteínas de la Membrana/biosíntesis , Fragmentos de Péptidos/metabolismo , Conducta Espacial/efectos de los fármacos , Envejecimiento , Animales , Camellia sinensis , Catequina/administración & dosificación , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Homólogo 4 de la Proteína Discs Large , Guanilato-Quinasas , Hipocampo/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos , Actividad Motora/efectos de los fármacos , Plasticidad Neuronal , Fosforilación , Sinapsis/fisiología , Factores de Tiempo , Regulación hacia Arriba
9.
Neurochem Res ; 33(10): 2092-8, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18408996

RESUMEN

The effects of acute and chronic morphine treatments on the expression of Ca2+/calmodulin dependent protein kinase II (CaMK II) gene in rat brain were investigated using in situ hybridization histochemistry. Our data showed that repeated, but not single morphine administration, resulted in significant up-regulation of the alpha-CaMK II gene expression in hippocampus and frontal cortex. We further studied the time courses of alpha-CaMK II gene expression in response to repeated morphine administration. After 3 days of consecutive morphine injections, the alpha-CaMK II mRNA levels exhibited a trend of up-regulation, and after 6 days of consecutive morphine injections it increased over 50-60% as compared with the control group. The alpha-CaMK II mRNA levels remained high 24 h after the cessation of chronic morphine treatment and returned to the control level 72 h later. However, changes of alpha-CaMK II gene levels mentioned above were not detected in amygdala or piriform cortex. Taken together, our data demonstrate that chronic morphine treatment region-specific up-regulates the levels of the alpha-CaMK II gene expression in hippocampus and frontal cortex.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Morfina/administración & dosificación , Animales , Esquema de Medicación , Lóbulo Frontal/enzimología , Hipocampo/enzimología , Masculino , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
10.
Arch Biochem Biophys ; 466(2): 234-41, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17706588

RESUMEN

Gonadotropin-releasing hormone (GnRH) is secreted from hypothalamic GnRH neurons. There is accumulating evidence that GnRH neurons have GnRH receptors and that the autocrine action of GnRH activates MAP kinase. In this study, we found that KN93, an inhibitor of Ca(2+)/calmodulin-dependent protein kinases (CaM kinases), inhibited the GnRH-induced activation of MAP kinase in immortalized GnRH neurons (GT1-7 cells). Immunoblot analysis indicated that the CaM kinase IIdelta2 isoform (CaM kinase IIdelta2) and synapsin I were expressed in GT1-7 cells. GnRH treatment rapidly increased phosphorylation of synapsin I at serine 603, a specific phosphorylation site for CaM kinase II, suggesting that GnRH treatment rapidly activated CaM kinase IIdelta2. In addition, when we stably overexpressed CaM kinase IIdelta2 in GT1-7 cells, the activation of MAP kinase was strongly enhanced. These results suggest that CaM kinase IIdelta2 was involved in the GnRH-induced activation of MAP kinase in GT1-7 cells.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Hormona Liberadora de Gonadotropina/fisiología , Hipotálamo/citología , Neuronas/enzimología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Bovinos , Línea Celular , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Ratones , Ratas , Transducción de Señal , Sinapsinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA