Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
J Physiol ; 595(12): 4089-4108, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28105734

RESUMO

KEY POINTS: Spontaneous sarcoplasmic reticulum (SR) Ca2+ release events increased in fructose-rich diet mouse (FRD) myocytes vs. control diet (CD) mice, in the absence of significant changes in SR Ca2+ load. In HEK293 cells, hyperglycaemia significantly enhanced [3 H]ryanodine binding and Ca2+ /calmodulin-dependent protein kinase II (CaMKII) phosphorylation of RyR2-S2814 residue vs. normoglycaemia. These increases were prevented by CaMKII inhibition. FRD significantly augmented cardiac apoptosis in WT vs. CD-WT mice, which was prevented by co-treatment with the reactive oxygen species scavenger Tempol. Oxidative stress was also increased in FRD-SR-autocamide inhibitory peptide (AIP) mice, expressing the SR-targeted CaMKII inhibitor AIP, without any significant enhancement of apoptosis vs. CD-SR-AIP mice. FRD produced mitochondrial swelling and membrane depolarization in FRD-WT mice but not in FRD-S2814A mice, in which the CaMKII site on ryanodine receptor 2 was ablated. FRD decreased mitochondrial area, mean Feret diameter and the mean distance between SR and the outer mitochondrial membrane vs. CD hearts. This remodelling was prevented in AC3I mice, with cardiac-targeted CaMKII inhibition. ABSTRACT: The impact of cardiac apoptosis in pre-diabetic stages of diabetic cardiomyopathy is unknown. We show that myocytes from fructose-rich diet (FRD) animals exhibit arrhythmias produced by exacerbated Ca2+ /calmodulin-protein kinase (CaMKII) activity, ryanodine receptor 2 (RyR2) phosphorylation and sarcoplasmic reticulum (SR) Ca2+ leak. We tested the hypothesis that this mechanism also underlies cardiac apoptosis in pre-diabetes. We generated a pre-diabetic model in FRD mice. FRD mice showed an increase in oxidative stress, hypertrophy and systolic dysfunction. FRD myocytes exhibited enhanced SR Ca2+ spontaneous events in the absence of SR Ca2+ load alterations vs. control-diet (CD) myocytes. In HEK293 cells, hyperglycaemia significantly enhanced [3 H]ryanodine binding and CaMKII phosphorylation of RyR2-S2814 residue vs. normoglycaemia. CaMKII inhibition prevented hyperglycaemia-induced alterations. FRD also evoked cardiac apoptosis in WT mice vs. CD-WT mice. Co-treatment with the reactive oxygen species scavenger Tempol prevented FRD-induced apoptosis in WT mice. In contrast, FRD enhanced oxidative stress but not apoptosis in FRD-SR-AIP mice, in which a CaMKII inhibitor is targeted to the SR. FRD produced mitochondrial membrane depolarization in WT mice but not in S2814A mice, in which the CaMKII phosphorylation site on RyR2 was ablated. Furthermore, FRD decreased mitochondrial area, mean Feret diameter and mean SR-mitochondrial distance vs. CD-WT hearts. This remodelling was prevented in AC3I mice, with cardiac-targeted CaMKII inhibition. CaMKII phosphorylation of RyR2, SR Ca2+ leak and mitochondrial membrane depolarization are critically involved in the apoptotic pathway of the pre-diabetic heart. The FRD-induced decrease in SR-mitochondrial distance is likely to additionally favour Ca2+ transit between the two organelles.


Assuntos
Apoptose/fisiologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Cálcio/metabolismo , Calmodulina/metabolismo , Intolerância à Glucose/metabolismo , Transdução de Sinais/fisiologia , Animais , Arritmias Cardíacas/metabolismo , Sinalização do Cálcio/fisiologia , Proteínas de Ligação ao Cálcio/metabolismo , Cardiomiopatias/metabolismo , Linhagem Celular , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/metabolismo , Retículo Sarcoplasmático/metabolismo
2.
FASEB J ; 29(9): 3773-87, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26023182

RESUMO

Annexin A4 (AnxA4), a Ca(2+)- and phospholipid-binding protein, is up-regulated in the human failing heart. In this study, we examined the impact of AnxA4 on ß-adrenoceptor (ß-AR)/cAMP-dependent signal transduction. Expression of murine AnxA4 in human embryonic kidney (HEK)293 cells dose-dependently inhibited cAMP levels after direct stimulation of adenylyl cyclases (ACs) with forskolin (FSK), as determined with an exchange protein activated by cAMP-Förster resonance energy transfer (EPAC-FRET) sensor and an ELISA (control vs. +AnxA4: 1956 ± 162 vs. 1304 ± 185 fmol/µg protein; n = 8). Disruption of the anxA4 gene led to a consistent increase in intracellular cAMP levels in isolated adult mouse cardiomyocytes, with heart-directed expression of the EPAC-FRET sensor, stimulated with FSK, and as determined by ELISA, also in mouse cardiomyocytes stimulated with the ß-AR agonist isoproterenol (ISO) (anxA4a(+/+) vs. anxA4a(-/-): 5.1 ± 0.3 vs. 6.7 ± 0.6 fmol/µg protein) or FSK (anxA4a(+/+) vs. anxA4a(-/-): 1891 ± 238 vs. 2796 ± 343 fmol/µg protein; n = 9-10). Coimmunoprecipitation experiments in HEK293 cells revealed a direct interaction of murine AnxA4 with human membrane-bound AC type 5 (AC5). As a functional consequence of AnxA4-mediated AC inhibition, AnxA4 inhibited the FSK-induced transcriptional activation mediated by the cAMP response element (CRE) in reporter gene studies (10-fold vs. control; n = 4 transfections) and reduced the FSK-induced phosphorylation of the CRE-binding protein (CREB) measured on Western blots (control vs. +AnxA4: 150 ± 17% vs. 105 ± 10%; n = 6) and by the use of the indicator of CREB activation caused by phosphorylation (ICAP)-FRET sensor, indicating CREB phosphorylation. Inactivation of AnxA4 in anxA4a(-/-) mice was associated with an increased cardiac response to ß-AR stimulation. Together, these results suggest that AnxA4 is a novel direct negative regulator of AC5, adding a new facet to the functions of annexins.


Assuntos
Adenilil Ciclases/metabolismo , Anexina A4/metabolismo , Membrana Celular/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Adenilil Ciclases/genética , Animais , Anexina A4/genética , Proteína de Ligação a CREB/genética , Proteína de Ligação a CREB/metabolismo , Membrana Celular/genética , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Fosforilação/fisiologia
3.
J Mol Cell Cardiol ; 50(1): 230-8, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20971119

RESUMO

Cardiac myocyte overexpression of CaMKIIδ(C) leads to cardiac hypertrophy and heart failure (HF) possibly caused by altered myocyte Ca(2+) handling. A central defect might be the marked CaMKII-induced increase in diastolic sarcoplasmic reticulum (SR) Ca(2+) leak which decreases SR Ca(2+) load and Ca(2+) transient amplitude. We hypothesized that inhibition of CaMKII near the SR membrane would decrease the leak, improve Ca(2+) handling and prevent the development of contractile dysfunction and HF. To test this hypothesis we crossbred CaMKIIδ(C) overexpressing mice (CaMK) with mice expressing the CaMKII-inhibitor AIP targeted to the SR via a modified phospholamban (PLB)-transmembrane-domain (SR-AIP). There was a selective decrease in the amount of activated CaMKII in the microsomal (SR/membrane) fraction prepared from these double-transgenic mice (CaMK/SR-AIP) mice. In ventricular cardiomyocytes from CaMK/SR-AIP mice, SR Ca(2+) leak, assessed both as diastolic Ca(2+) shift into SR upon tetracaine in intact myocytes or integrated Ca(2+) spark release in permeabilized myocytes, was significantly reduced. The reduced leak was accompanied by enhanced SR Ca(2+) load and twitch amplitude in double-transgenic mice (vs. CaMK), without changes in SERCA expression or NCX function. However, despite the improved myocyte Ca(2+) handling, cardiac hypertrophy and remodeling was accelerated in CaMK/SR-AIP and cardiac function worsened. We conclude that while inhibition of SR localized CaMKII in CaMK mice improves Ca(2+) handling, it does not necessarily rescue the HF phenotype. This implies that a non-SR CaMKIIδ(C) exerts SR-independent effects that contribute to hypertrophy and HF, and this CaMKII pathway may be exacerbated by the global enhancement of Ca transients.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Cálcio/metabolismo , Retículo Sarcoplasmático/metabolismo , Animais , Western Blotting , Ecocardiografia , Ventrículos do Coração/citologia , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo , Fosforilação
4.
Neuron ; 52(2): 321-33, 2006 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-17046694

RESUMO

It is well established that ligand-gated chloride flux across the plasma membrane modulates neuronal excitability. We find that a voltage-dependent Cl(-) conductance increases neuronal excitability in immature rodents as well, enhancing the time course of NMDA receptor-mediated miniature excitatory postsynaptic potentials (mEPSPs). This Cl(-) conductance is activated by CaMKII, is electrophysiologically identical to the CaMKII-activated CLC-3 conductance in nonneuronal cells, and is absent in clc-3(-/-) mice. Systematically decreasing [Cl(-)](i) to mimic postnatal [Cl(-)](i) regulation progressively decreases the amplitude and decay time constant of spontaneous mEPSPs. This Cl(-)-dependent change in synaptic strength is absent in clc-3(-/-) mice. Using surface biotinylation, immunohistochemistry, electron microscopy, and coimmunoprecipitation studies, we find that CLC-3 channels are localized on the plasma membrane, at postsynaptic sites, and in association with NMDA receptors. This is the first demonstration that a voltage-dependent chloride conductance modulates neuronal excitability. By increasing postsynaptic potentials in a Cl(-) dependent fashion, CLC-3 channels regulate neuronal excitability postsynaptically in immature neurons.


Assuntos
Canais de Cloreto/metabolismo , Potenciais Pós-Sinápticos Excitadores/fisiologia , Neurônios/metabolismo , Sinapses/metabolismo , Transmissão Sináptica/fisiologia , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Diferenciação Celular/fisiologia , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Canais de Cloreto/genética , Cloretos/metabolismo , Regulação para Baixo/fisiologia , Ácido Glutâmico/metabolismo , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Hipocampo/ultraestrutura , Camundongos , Camundongos Knockout , Neurônios/ultraestrutura , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/ultraestrutura , Membranas Sinápticas/metabolismo , Sinaptossomos/metabolismo
5.
J Mol Cell Cardiol ; 48(6): 1298-306, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20060004

RESUMO

Ca(2+)-calmodulin-dependent protein kinase II (CaMKII) plays an important role mediating apoptosis/necrosis during ischemia-reperfusion (IR). We explored the mechanisms of this deleterious effect. Langendorff perfused rat and transgenic mice hearts with CaMKII inhibition targeted to sarcoplasmic reticulum (SR-AIP) were subjected to global IR. The onset of reperfusion increased the phosphorylation of Thr(17) site of phospholamban, without changes in total protein, consistent with an increase in CaMKII activity. Instead, there was a proportional decrease in the phosphorylation of Ser2815 site of ryanodine receptors (RyR2) and the amount of RyR2 at the onset of reperfusion, i.e. the ratio Ser2815/RyR2 did not change. Inhibition of the reverse Na(+)/Ca(2+)exchanger (NCX) mode (KBR7943) diminished phospholamban phosphorylation, reduced apoptosis/necrosis and enhanced mechanical recovery. CaMKII-inhibition (KN-93), significantly decreased phospholamban phosphorylation, infarct area, lactate dehydrogenase release (LDH) (necrosis), TUNEL positive nuclei, caspase-3 activity, Bax/Bcl-2 ratio and Ca(2+)-induced mitochondrial swelling (apoptosis), and increased contractile recovery when compared with non-treated IR hearts or IR hearts pretreated with the inactive analog, KN-92. Blocking SR Ca(2+) loading and release (thapsigargin/dantrolene), mitochondrial Ca(2+) uniporter (ruthenium red/RU360), or mitochondrial permeability transition pore (cyclosporine A), significantly decreased infarct size, LDH release and apoptosis. SR-AIP hearts failed to show an increase in the phosphorylation of Thr(17) of phospholamban at the onset of reflow and exhibited a significant decrease in infarct size, apoptosis and necrosis respect to controls. The results reveal an apoptotic-necrotic pathway mediated by CaMKII-dependent phosphorylations at the SR, which involves the reverse NCX mode and the mitochondria as trigger and end effectors, respectively, of the cascade.


Assuntos
Apoptose , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais , Animais , Caspase 3/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Modelos Biológicos , Necrose , Fosforilação , Ratos , Ratos Wistar
6.
Biochim Biophys Acta ; 1763(11): 1275-81, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17069901

RESUMO

Ca(2+) signaling through CaMKII is critical in regulating myocyte function with regard to excitation-contraction-relaxation cycles and excitation-transcription coupling. To investigate the role of nuclear CaMKII in cardiac function, transgenic mice were designed and generated to target the expression of a CaMKII inhibitory peptide, AIP (KKALRRQEAVDAL), to the nucleus. The transgenic construct consists of the murine alpha-myosin heavy chain promoter followed by the expression unit containing nucleotides encoding a four repeat concatemer of AIP (AIP(4)) and a nuclear localization signal (NLS). Western blot and immunohistochemical analyses demonstrate that AIP(4) is expressed only in the nucleus of cardiac myocytes of the transgenic mice (NLS-AIP(4)). The function of cytoplasmic CaMKII is not affected by the expression of AIP(4) in the nucleus. Inhibition of nuclear CaMKII activity resulted in reduced translocation of HDAC5 from nucleus to cytoplasm in NLS-AIP(4) mouse hearts. Loss of nuclear CaMKII activity causes NLS-AIP(4) mice to have smaller hearts than their nontransgenic littermates. Transcription factors including CREB and NFkappaB are not regulated by cardiac nuclear CaMKII. With physiological stresses such as pregnancy or aging (8 months), NLS-AIP(4) mice develop hypertrophy symptoms including enlarged atria, systemic edema, sedentariness, and morbidity. RT-PCR analyses revealed that the hypertrophic marker genes, such as ANF and beta-myosin heavy chain, were upregulated in pregnancy stressed mice. Our results suggest that absence of adequate Ca2+signaling through nuclear CaMKII regulated pathways leads to development of cardiac disease.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Cardiomegalia/genética , Coração/anatomia & histologia , Miocárdio/enzimologia , Sinais de Localização Nuclear/metabolismo , Sequência de Aminoácidos , Animais , Fator Natriurético Atrial/genética , Sinalização do Cálcio , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/análise , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Núcleo Celular/enzimologia , Marcadores Genéticos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Cadeias Pesadas de Miosina/genética , Sinais de Localização Nuclear/análise , Sinais de Localização Nuclear/genética , Tamanho do Órgão/genética , Peptídeos/farmacologia , Fosforilação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Miosinas Ventriculares/genética
7.
J Neurotrauma ; 24(4): 638-50, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17439347

RESUMO

Traumatic brain injury (TBI) can dramatically increase levels of intracellular calcium ([Ca(2+)](i)). One consequence of increased [Ca(2+)](i) would be altered activity and function of calcium-regulated proteins, including calcium-calmodulin-dependent protein kinase II (CaMKII), which is autophosphorylated on Thr(286)(pCaMKII(286)) in the presence of calcium and calmodulin. Therefore, we hypothesized that TBI would result in increased levels of pCaMKII(286), and that such increases would occur early after injury in brain regions known to be damaged following lateral fluid percussion TBI (i.e., hippocampus and cortex). In order to test this hypothesis, immunostaining of CaMKII was examined in rat hippocampus and cortex after lateral fluid percussion (LFP) injury using an antibody directed against pCaMKII(286). LFP injury produced a marked increase in pCaMKII(286) immunostaining in the hippocampus and overlying cortex 30 min after TBI. The pattern of increased immunostaining was uneven, and unexpectedly absent in some hippocampal CA3 pyramidal neurons. This suggests that phosphatase activity may also increase following TBI, resulting in dephosphorylation of pCaMKII(286) in subpopulations of CA3 pyramidal neurons. Western blotting confirmed a rapid increase in levels of pCaMKII(286) at 10 and 30 min after brain injury, and that it was transient and no longer significantly elevated when examined at 3, 8, and 24 h. These results demonstrate that TBI alters the autophosphorylation state of CaMKII, an important neuronal regulator of critical cell functions, including enzyme activities, cell structure, gene expression, and neuronal plasticity, and provide a molecular mechanism that is likely to contribute to cell injury and impaired plasticity after TBI.


Assuntos
Lesões Encefálicas/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Animais , Benzoxazinas , Western Blotting , Temperatura Corporal/fisiologia , Peso Corporal/fisiologia , Lesões Encefálicas/patologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Lateralidade Funcional/fisiologia , Imuno-Histoquímica , Masculino , Oxazinas , Fosforilação , Equilíbrio Postural/fisiologia , Ratos , Ratos Sprague-Dawley
8.
Int J Cardiol ; 202: 394-406, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26432489

RESUMO

BACKGROUND: Heart failure and arrhythmias occur more frequently in patients with type 2 diabetes (T2DM) than in the general population. T2DM is preceded by a prediabetic condition marked by elevated reactive oxygen species (ROS) and subclinical cardiovascular defects. Although multifunctional Ca2+ calmodulin-dependent protein kinase II (CaMKII) is ROS-activated and CaMKII hyperactivity promotes cardiac diseases, a link between prediabetes and CaMKII in the heart is unprecedented. OBJECTIVES: To prove the hypothesis that increased ROS and CaMKII activity contribute to heart failure and arrhythmogenic mechanisms in early stage diabetes. METHODS-RESULTS: Echocardiography, electrocardiography, biochemical and intracellular Ca2+ (Ca2+i) determinations were performed in fructose-rich diet-induced impaired glucose tolerance, a prediabetes model, in rodents. Fructose-rich diet rats showed decreased contractility and hypertrophy associated with increased CaMKII activity, ROS production, oxidized CaMKII and enhanced CaMKII-dependent ryanodine receptor (RyR2) phosphorylation compared to rats fed with control diet. Isolated cardiomyocytes from fructose-rich diet showed increased spontaneous Ca2+i release events associated with spontaneous contractions, which were prevented by KN-93, a CaMKII inhibitor, or addition of Tempol, a ROS scavenger, to the diet. Moreover, fructose-rich diet myocytes showed increased diastolic Ca2+ during the burst of spontaneous Ca2+i release events. Mice treated with Tempol or with sarcoplasmic reticulum-targeted CaMKII-inhibition by transgenic expression of the CaMKII inhibitory peptide AIP, were protected from fructose-rich diet-induced spontaneous Ca2+i release events, spontaneous contractions and arrhythmogenesis in vivo, despite ROS increases. CONCLUSIONS: RyR2 phosphorylation by ROS-activated CaMKII, contributes to impaired glucose tolerance-induced arrhythmogenic mechanisms, suggesting that CaMKII inhibition could prevent prediabetic cardiovascular complications and/or evolution.


Assuntos
Arritmias Cardíacas/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Cálcio/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Aminoácidos/metabolismo , Animais , Arritmias Cardíacas/patologia , Arritmias Cardíacas/prevenção & controle , Benzilaminas/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/química , Cromo/metabolismo , Diabetes Mellitus Tipo 2/patologia , Modelos Animais de Doenças , Frutose/administração & dosagem , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/prevenção & controle , Masculino , Camundongos , Miócitos Cardíacos/metabolismo , Ácidos Nicotínicos/metabolismo , Fosforilação , Estado Pré-Diabético/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Retículo Sarcoplasmático/metabolismo , Sulfonamidas/farmacologia
9.
J Gen Physiol ; 121(5): 413-25, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12695484

RESUMO

Annexin A4 (Anx4) belongs to a ubiquitous family of Ca2+-dependent membrane-binding proteins thought to be involved in membrane trafficking and membrane organization within cells. Anx4 localizes to the apical region in epithelia; however, its physiological role is unclear. We show that Anx4 exhibited binding to liposomes (phosphatidylcholine:phosphatidylserine, 1:1) in the presence of Ca2+ and binding was reversible with EDTA. Anx4 binding resulted in liposome aggregation and a reduction in membrane water permeability of 29% (P < 0.001) at 25 degrees C. These effects were not seen in the presence of Ca2+ or Anx4 alone and were reversible with EDTA. Measurements of membrane fluidity made by monitoring fluorescence anisotropy of 2-(12-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)dodecanoyl-1-hexadecanoyl-sn-glycero-3-phosphocholine (NBD-HPC) demonstrated that Anx4 binding rigidified the outer leaflet of the bilayer (P < 0.001), thus providing a molecular explanation for the inhibition of water flux. To determine whether Anx4 would produce similar effects on physiological membranes we constructed liposomes which recapitulated the lipid composition of the inner leaflet of the MDCK apical membrane. These membranes exhibited reductions to water permeability upon Anx4 binding (19.5% at 25 degrees C, 31% at 37 degrees C; P < 0.01 and P < 0.001, respectively) and to proton permeability (15% at 25 degrees C, 19.5% at 37 degrees C; P < 0.05). Since our in vitro experiments indicated an effect on membrane permeability, we examined localization of Anx4 in the kidney collecting duct, a region of the nephron responsible for concentrating urine through water reabsorbtion. Anx4 was shown to colocalize apically with aquaporin 2 (AQP2) in collecting duct epithelia. To test for the existence of a functional interaction between Anx4 and AQP2 we isolated AQP2-containing endosomes and exposed them to Anx4/Ca2+. Water flux rates were unchanged, indicating Anx4 does not directly regulate AQP2. We conclude that Anx4 can alter the physical properties of membranes by associating with them and regulate passive membrane permeability to water and protons. These properties represent important new functions for Anx4.


Assuntos
Anexina A4/metabolismo , Endossomos/metabolismo , Lipossomos/química , Prótons , Água/metabolismo , Animais , Anexina A4/química , Anexina A4/fisiologia , Aquaporina 2 , Aquaporina 6 , Aquaporinas/metabolismo , Cálcio/química , Cálcio/metabolismo , Permeabilidade da Membrana Celular , Feminino , Polarização de Fluorescência , Imunofluorescência , Técnicas In Vitro , Túbulos Renais Coletores/metabolismo , Masculino , Fluidez de Membrana , Osmose , Permeabilidade , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Água/química
10.
Cell Signal ; 15(11): 1049-57, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14499348

RESUMO

Calcium is a second messenger that is implicated in the regulation of cell cycle transitions. Calmodulin is a ubiquitous protein that translates intracellular calcium signals and activates several enzymes including calcium/calmodulin-dependent protein kinase II (CaMKII). Pharmacological inhibitors and constitutively active mutants have implicated CaMKII in cell cycle mediation. Specifically, constitutively active CaMKII impedes mitosis. In order to elucidate the molecular mechanisms underlying this phenomenon, the effect of constitutively active CaMKII gene expression on cdc2/cyclin B1 was investigated. As seen in previous studies with S. pombe, constitutively active CaMKII-hindered mitosis. However, this report shows that CaMKII does not cause permanent cell cycle arrest but delays progression into mitosis. Constitutive CaMKII expression also leads to elevations in cyclin B1 expression and cdc2 tyrosine-15 phosphorylation, analogous to observations in cells treated with hydroxyurea. Taken together, these data suggest that constitutive CaMKII may delay mitosis by activating a cell cycle checkpoint.


Assuntos
Sinalização do Cálcio/fisiologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Ciclina B/metabolismo , Mitose/fisiologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Calmodulina/metabolismo , Clonagem Molecular , Ciclina B1 , Ativação Enzimática/fisiologia , Citometria de Fluxo , Células HeLa , Humanos , Interfase/fisiologia , Mutagênese Sítio-Dirigida , Fosforilação , Fase S/fisiologia
11.
Circ Arrhythm Electrophysiol ; 4(6): 947-57, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22009705

RESUMO

BACKGROUND: Digitalis-induced Na(+) accumulation results in an increase in Ca(2+)(i) via the Na(+)/Ca(2+) exchanger, leading to enhanced sarcoplasmic reticulum (SR) Ca(2+) load, responsible for the positive inotropic and toxic arrhythmogenic effects of glycosides. A digitalis-induced increase in Ca(2+)(i) could also activate calcium-calmodulin kinase II (CaMKII), which has been shown to have proarrhythmic effects. Here, we investigate whether CaMKII underlies digitalis-induced arrhythmias and the subcellular mechanisms involved. METHODS AND RESULTS: In paced rat ventricular myocytes (0.5 Hz), 50 µmol/L ouabain increased contraction amplitude by 160 ± 5%. In the absence of electric stimulation, ouabain promoted spontaneous contractile activity and Ca(2+) waves. Ouabain activated CaMKII (p-CaMKII), which phosphorylated its downstream targets, phospholamban (PLN) (Thr17) and ryanodine receptor (RyR) (Ser2814). Ouabain-induced spontaneous activity was prevented by inhibiting CaMKII with 2.5 µmol/L KN93 but not by 2.5 µmol/L of the inactive analog, KN92. Similar results were obtained using the CaMKII inhibitor, autocamtide-2 related inhibitory peptide (AIP) (1 to 2.5 µmol/L), and in myocytes from transgenic mice expressing SR-targeted AIP. Consistently, CaMKII overexpression exacerbated ouabain-induced spontaneous contractile activity. Ouabain was associated with an increase in SR Ca(2+) content and Ca(2+) spark frequency, indicative of enhanced SR Ca(2+) leak. KN93 suppressed the ouabain-induced increase in Ca(2+) spark frequency without affecting SR Ca(2+) content. Similar results were obtained with digoxin. In vivo, ouabain-induced arrhythmias were prevented by KN93 and absent in SR-AIP mice. CONCLUSIONS: These results show for the first time that CaMKII mediates ouabain-induced arrhythmic/toxic effects. We suggest that CaMKII-dependent phosphorylation of the RyR, resulting in Ca(2+) leak from the SR, is the underlying mechanism involved.


Assuntos
Arritmias Cardíacas/induzido quimicamente , Sinalização do Cálcio/efeitos dos fármacos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Cardiotônicos/toxicidade , Frequência Cardíaca/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Ouabaína/toxicidade , Animais , Antiarrítmicos/farmacologia , Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/fisiopatologia , Arritmias Cardíacas/prevenção & controle , Benzilaminas/farmacologia , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Estimulação Cardíaca Artificial , Células Cultivadas , Eletrocardiografia , Ativação Enzimática , Ventrículos do Coração/enzimologia , Ventrículos do Coração/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Peptídeos/genética , Peptídeos/metabolismo , Peptídeos/farmacologia , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Wistar , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/enzimologia , Trocador de Sódio e Cálcio/metabolismo , Sulfonamidas/farmacologia , Fatores de Tempo , Transfecção
12.
Am J Physiol Renal Physiol ; 294(4): F919-27, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18256316

RESUMO

Annexin A4 (anxA4) is a member of the Ca(2+)-dependent membrane-binding family of proteins implicated in the regulation of ion conductances, Ca(2+) homeostasis, and membrane trafficking. We demonstrate, in mice, that annexins 1-6 are present in whole bladder and exhibit differential expression in the urothelium. An anxA4a-knockout (anxA4a(-/-)) mouse model shows no protein in the urothelium by immunofluorescence and immunoblotting. In wild-type bladders, anxA4a in umbrella cells showed uniform cytoplasmic staining and some association with the nuclear membrane. Application of a hydrostatic pressure to bladders mounted in Ussing chambers resulted in redistribution of anxA4a from cytoplasm to cellular boundaries in the basal and intermediate cells but not in superficial umbrella cells. We hypothesized that anxA4a might be important for barrier function or for stretch-activated membrane trafficking. To test these hypotheses, we conducted a series of functional and morphological analyses on bladders from control and anxA4a(-/-) animals. The transepithelial resistances, water permeabilities, and urea permeabilities of anxA4a(-/-) bladders were not different from controls, indicating that barrier function was intact. Membrane trafficking in response to hydrostatic pressure as measured by capacitance increases was also normal for anxA4a(-/-) bladders. Cystometrograms performed on live animals showed that voiding frequency and intrabladder pressures were also not different. There were no differences in bladder surface morphology or cellular architecture examined by scanning and transmission electron microscopy, respectively. We conclude that loss of anxA4 from the urothelium does not affect barrier function, membrane trafficking, or normal bladder-voiding behavior.


Assuntos
Anexina A4/deficiência , Anexina A4/genética , Bexiga Urinária/fisiologia , Urotélio/fisiologia , Animais , Anexina A4/fisiologia , Permeabilidade da Membrana Celular , Immunoblotting , Camundongos , Camundongos Knockout , Ureia/metabolismo , Bexiga Urinária/citologia , Bexiga Urinária/ultraestrutura , Urotélio/citologia
13.
J Mol Cell Cardiol ; 42(1): 196-205, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17052727

RESUMO

Cardiac Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) in heart has been implicated in Ca(2+) current (I(Ca)) facilitation, enhanced sarcoplasmic reticulum (SR) Ca(2+) release and frequency-dependent acceleration of relaxation (FDAR) via enhanced SR Ca(2+) uptake. However, questions remain about how CaMKII may work in these three processes. Here we tested the role of CaMKII in these processes using transgenic mice (SR-AIP) that express four concatenated repeats of the CaMKII inhibitory peptide AIP selectively in the SR membrane. Wild type mice (WT) and mice expressing AIP exclusively in the nucleus (NLS-AIP) served as controls. Increasing stimulation frequency produced typical FDAR in WT and NLS-AIP, but FDAR was markedly inhibited in SR-AIP. Quantitative analysis of cytosolic Ca(2+) removal during [Ca(2+)](i) decline revealed that FDAR is due to an increased apparent V(max) of SERCA. CaMKII-dependent RyR phosphorylation at Ser2815 and SR Ca(2+) leak was both decreased in SR-AIP vs. WT. This decrease in SR Ca(2+) leak may partly balance the reduced SERCA activity leading to relatively unaltered SR-Ca(2+) load in SR-AIP vs. WT myocytes. Surprisingly, CaMKII regulation of the L-type Ca(2+) channel (I(Ca) facilitation and recovery from inactivation) was abolished by the SR-targeted CaMKII inhibition in SR-AIP mice. Inhibition of CaMKII effects on I(Ca) and RyR function by the SR-localized AIP places physical constraints on the localization of these proteins at the junctional microdomain. Thus SR-targeted CaMKII inhibition can directly inhibit the activation of SR Ca(2+) uptake, SR Ca(2+) release and I(Ca) by CaMKII, effects which have all been implicated in triggered arrhythmias.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Miócitos Cardíacos/metabolismo , Peptídeos/metabolismo , Retículo Sarcoplasmático/metabolismo , Animais , Sinalização do Cálcio , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Citosol/metabolismo , Feminino , Técnicas In Vitro , Cinética , Masculino , Camundongos , Camundongos Transgênicos , Contração Miocárdica , Peptídeos/genética , Fosforilação , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Trocador de Sódio e Cálcio/metabolismo
14.
Biochem Biophys Res Commun ; 350(1): 179-84, 2006 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-16996475

RESUMO

Using the mouse Langendorff heart perfusion model, the signaling pathways that regulate cardiac CREB-S133 phosphorylation have been defined. In mouse hearts stimulated with isoproterenol (ISO) (10(-8) M), endothelin-1 (ET-1) (10(-8) M), and phorbol 12-myristate 13-acetate (TPA) (10(-7) M), CREB-S133 phosphorylation was attained only by TPA-treatment. Activation of protein kinase A (PKA) was achieved by ISO. ISO- and ET-1-stimulation activated Ca2+/calmodulin-dependent kinase II (CaMKII). Protein kinase C (PKC) and p90(RSK) were activated with all three stimuli. Inhibition of ERK1/2 with PD98059 (10(-5) M) completely inhibited the activation of p90(RSK), but did not block CREB-S133 phosphorylation in TPA-perfused heart, indicating that PKA, CaMKII, and p90(RSK) do not phosphorylate CREB-S133 in the murine heart. PKC activation is signal specific. Analyses of PKC isoforms suggest that CREB phosphorylation is mediated by PKC epsilon translocating into nucleus only with TPA stimulation. These results, unlike those reported in other tissues, demonstrate that cardiac CREB is not a multi-signal target.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Miocárdio/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Núcleo Celular/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/química , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Endotelina-1/farmacologia , Ativação Enzimática , Coração/efeitos dos fármacos , Isoenzimas/metabolismo , Isoproterenol/farmacologia , Camundongos , Dados de Sequência Molecular , Reperfusão Miocárdica , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Proteína Quinase C-épsilon/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
15.
Am J Physiol Heart Circ Physiol ; 290(2): H599-606, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16143658

RESUMO

Transgenic (TG) mice expressing a Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibitory peptide targeted to the cardiac myocyte longitudinal sarcoplasmic reticulum (LSR) display reduced phospholamban phosphorylation at Thr17 and develop dilated myopathy when stressed by gestation and parturition (Ji Y, Li B, Reed TD, Lorenz JN, Kaetzel MA, and Dedman JR. J Biol Chem 278: 25063-25071, 2003). In the present study, these animals (TG) are evaluated for the effect of inhibition of sarcoplasmic reticulum (SR) CaMKII activity on the contractile characteristics and Ca2+ cycling of myocytes. Analysis of isolated work-performing hearts demonstrated moderate decreases in the maximal rates of contraction and relaxation (+/-dP/dt) in TG mice. The response of the TG hearts to increases in load is reduced. The TG hearts respond to isoproterenol (Iso) in a dose-dependent manner; the contractile properties were reduced in parallel to wild-type hearts. Assessment of isolated cardiomyocytes from TG mice revealed 40-47% decrease in the maximal rates of myocyte shortening and relengthening under both basal and Iso-stimulated conditions. Although twitch Ca2+ transient amplitudes were not significantly altered, the rate of twitch intracellular Ca2+ concentration decline was reduced by approximately 47% in TG myocytes, indicating decreased SR Ca2+ uptake function. Caffeine-induced Ca2+ transients indicated unaltered SR Ca2+ content and Na+/Ca2+ exchange function. Phosphorylation assays revealed an approximately 30% decrease in the phosphorylation of ryanodine receptor Ser2809. Iso stimulation increased the phosphorylation of both phospholamban Ser16 and the ryanodine receptor Ser2809 but not phospholamban Thr17 in TG mice. This study demonstrates that inhibition of SR CaMKII activity at the LSR results in alterations in cardiac contractility and Ca2+ handling in TG hearts.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Cálcio/metabolismo , Homeostase/fisiologia , Contração Miocárdica/fisiologia , Miocárdio/metabolismo , Retículo Sarcoplasmático/enzimologia , Animais , Cafeína/farmacologia , Proteínas de Ligação ao Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Cardiotônicos/farmacologia , Feminino , Técnicas In Vitro , Isoproterenol/farmacologia , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo , Peptídeos/metabolismo , Peptídeos/farmacologia , Fosforilação , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
16.
Biochem Biophys Res Commun ; 322(4): 1171-7, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15336965

RESUMO

Annexins are a family of membrane binding proteins that are characterized by a hypervariable amino terminus followed by a series of highly conserved Ca2+-phospholipid binding domains. Annexins function by binding to anionic phospholipid surfaces in a Ca2+-dependent manner. They self-associate to form trimers which further assemble into sheets that cover the membrane surface and alter properties such as fluidity and permeability. This submembranous skeleton alters integral protein functions such as ion transport properties and shields the surface from phospholipid binding proteins such as phospholipases and protein kinase C. Transgenic mouse hearts overexpressing wild type annexin VI (AnxVI673), a dominant-negative truncated annexin VI (residues 1-129, Anx129) and an annexin VI-null mouse (AnxVI-/-) have implicated the protein as a regulator of intracellular Ca2+ homeostasis which affects cardiac function.


Assuntos
Anexina A6/fisiologia , Contração Miocárdica , Animais , Anexina A6/genética , Cálcio/metabolismo , Camundongos , Miocárdio/metabolismo
17.
J Biol Chem ; 278(44): 43276-83, 2003 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-12912993

RESUMO

Annexin IV (AIV), a Ca2+-dependent membrane-binding protein, is expressed in many epithelia. Annexin IV modifies membrane bilayers by increasing rigidity, reducing water and H+ permeability, promoting vesicle aggregation, and regulating ion conductances, all in a Ca2+-dependent manner. We have characterized a mouse in which a gene trap has been inserted into the first intron of annexin IV. Processing of the primary transcript is disrupted. Northern blot and immunoblot data indicated that annexin IV expression was eliminated in many but not all tissues. Immunohistochemical analysis, however, demonstrated that annexin IV expression was eliminated in some cell types, but was unaltered in others. 5'-Rapid amplification of cDNA ends analysis of intestinal and kidney RNA revealed three transcripts, AIVa, AIVb, and AIVc. AIVa is widely distributed. AIVb is expressed only in the digestive tract. AIVc expression is very restricted. A selected number of epithelial cells of unique morphology demonstrate high concentrations. All three transcripts produce an identical annexin IV protein. The different tissue and cell-specific expression profiles of the three transcripts suggest that regulation of both the annexin IV gene expression and the cellular role of the protein are complex. The AIVa-/- mouse may become a valuable model to further study transcription and the physiological role of annexin IV.


Assuntos
Anexina A4/genética , Íntrons , RNA Mensageiro/metabolismo , Animais , Anexina A4/metabolismo , Northern Blotting , Cálcio/metabolismo , DNA Complementar/metabolismo , Immunoblotting , Imuno-Histoquímica , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Rim/metabolismo , Bicamadas Lipídicas/metabolismo , Camundongos , Modelos Genéticos , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Distribuição Tecidual
18.
Am J Physiol Cell Physiol ; 282(6): C1313-21, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11997246

RESUMO

The exposure of anionic phospholipids on the external surface of injured endothelial cells and activated platelets is a primary biological signal to initiate blood coagulation. Disease conditions that promote the formation of ectopic thrombi result in tissue ischemia. Annexins, Ca2+-dependent anionic phospholipid binding proteins, are potential therapeutic agents for the inhibition of coagulation. We have designed a transgene that targets secretion of annexin V from cultured thyroid cells under the control of doxycycline. Our results indicate that annexin V in the endoplasmic reticulum (ER)/Golgi lumen does not affect the synthesis, processing, and secretion of thyroglobulin. ER luminal Ca2+ was moderately increased and can be released by inositol 1,4,5-trisphosphate. Our study demonstrates that targeting and secretion of annexin V through the secretory pathway of mammalian cells does not adversely affect cellular function. Regulated synthesis and release of annexin V may exert anticoagulatory and anti-inflammatory effects systemically and may prove useful in further developing therapeutic strategies for conditions including antiphospholipid syndrome.


Assuntos
Anexina A5/metabolismo , Células Epiteliais/metabolismo , Glândula Tireoide/metabolismo , Sequência de Aminoácidos , Animais , Anexina A5/genética , Cálcio/metabolismo , Divisão Celular , Células Cultivadas , Doxiciclina/farmacologia , Retículo Endoplasmático/metabolismo , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Epitopos/fisiologia , Marcação de Genes , Complexo de Golgi/metabolismo , Inositol 1,4,5-Trifosfato/farmacologia , Ligantes , Dados de Sequência Molecular , Processamento de Proteína Pós-Traducional/fisiologia , Sinais Direcionadores de Proteínas/fisiologia , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tireoglobulina/metabolismo , Glândula Tireoide/citologia , Glândula Tireoide/efeitos dos fármacos , Transgenes
19.
J Am Chem Soc ; 125(27): 8290-301, 2003 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-12837101

RESUMO

Host-[2]rotaxanes, containing a diarginine-derivatized dibenzo-24-crown-8 (DB24C8) ether as the ring and a cyclophane pocket or an aromatic cleft as one blocking group, are cell transport agents. These hosts strongly associate with a variety of amino acids, dipeptides, and fluorophores in water (1 mM phosphate buffer, pH 7.0), DMSO, and a 75/25 (v/v) buffer to DMSO solution. All peptidic guests in all solvent systems have association constants (K(A)'s) in the range of 1 x 10(4) to 5 x 10(4) M(-)(1), whereas the K(A) range for the fluorophores is 1 x 10(4) to 9 x 10(5) M(-)(1). Association constants for the cyclophane itself, cyclophane 3, are smaller. These values are in the 1 x 10(3) to 5 x 10(3) M(-)(1) range, which shows that the rotaxane architecture is advantageous for guest binding. Cyclophane-[2]rotaxane 1 efficiently transports fluorescein and a fluorescein-protein kinase C (PKC) inhibitor into eukaryotic COS-7 cells, including the nucleus. Interestingly, cleft-[2]rotaxane 2 does not transport fluorescein as efficiently, even though the results from the fluorescence assays show that both [2]rotaxanes bind fluorescein with the same ability.


Assuntos
Portadores de Fármacos/química , Animais , Arginina/análogos & derivados , Arginina/química , Arginina/farmacocinética , Materiais Biomiméticos/química , Materiais Biomiméticos/metabolismo , Células COS , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Chlorocebus aethiops , Portadores de Fármacos/síntese química , Portadores de Fármacos/farmacocinética , Éteres Cíclicos/química , Éteres Cíclicos/farmacocinética , Fluoresceína/química , Fluoresceína/farmacocinética , Cinética , Modelos Moleculares , Rotaxanos
20.
Am J Physiol Cell Physiol ; 284(6): C1334-45, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12570987

RESUMO

Ca(2+)/calmodulin-regulated protein kinase II (CaMKII) mediates many cellular events. The four CaMKII isoforms have numerous splice variants, three of which contain nuclear localization signals. Little is known about the role of nuclear localized CaMKII in neuronal development. To study this process, PC12 cells were transfected to produce CaMKII targeted to either the cytoplasm or the nucleus and then treated with nerve growth factor (NGF). NGF triggers a signaling cascade (MAPK) that results in the differentiation of PC12 cells into a neuronal phenotype, marked by neurite outgrowth. The present study found that cells expressing nuclear targeted CaMKII failed to grow neurites, whereas cells expressing cytoplasmic CaMKII readily produced neurites. Inhibition of neuronal differentiation by nuclear CaMKII was independent of MAPK signaling, as sustained Erk phosphorylation was not affected. Phosphorylation of CREB was also unaffected. Thus nuclear CaMKII modifies neuronal differentiation by a mechanism independent of MAPK and CREB activation.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Diferenciação Celular/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neurônios/fisiologia , Proteínas Nucleares/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Citoplasma/química , Ativação Enzimática , Isoenzimas/genética , Isoenzimas/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Fator de Crescimento Neural/farmacologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Proteínas Nucleares/genética , Células PC12 , Fosforilação , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA