Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 291(44): 22970-22976, 2016 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-27624933

RESUMEN

When treated with nerve growth factor, PC12 cells will differentiate over the course of several days. Here, we have followed changes during differentiation in the cellular levels of phosphoinositide-specific phospholipase Cß (PLCß) and its activator, Gαq, which together mediate Ca2+ release. We also followed changes in the level of the novel PLCß binding partner TRAX (translin-associated factor X), which promotes RNA-induced gene silencing. We find that the level of PLCß increases 4-fold within 24 h, whereas Gαq increases only 1.4-fold, and this increase occurs ∼24 h later than PLCß. Alternately, the level of TRAX remains constant over the 72 h tested. When PLCß1 or TRAX is down-regulated, differentiation does not occur. The impact of PLCß on differentiation appears independent of Gαq as down-regulating Gαq at constant PLCß does not affect differentiation. Förster resonance energy transfer studies after PLCß association with its partners indicate that PLCß induced soon after nerve growth factor treatment associates with TRAX rather than Gαq Functional measurements of Ca2+ signals to assess the activity of PLCß-Gαq complexes and measurements of the reversal of siRNA(GAPDH) to assess the activity of PLCß-TRAX complexes additionally suggest that the newly synthesized PLCß associates with TRAX to impact RNA-induced silencing. Taken together, our studies show that PLCß, through its ability to bind TRAX and reverse RNA silencing of specific genes, plays a key role in switching PC12 cells to their differentiated state.


Asunto(s)
Diferenciación Celular , Células PC12/enzimología , Fosfolipasa C beta/metabolismo , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Células PC12/citología , Fosfolipasa C beta/genética , Unión Proteica , Ratas
2.
J Neurosci Res ; 91(2): 273-84, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23161662

RESUMEN

During the neurodegenerative process in several brain diseases, oxidative stress is known to play important roles in disease severity and evolution. Although early events of stress, such as increased lipid peroxidation and decreased superoxide dismutase, are known to characterize early onsets of these diseases, little is known about the events that participate in maintaining the chronic evolving phase influencing the disease progression in neurons. Here, we used differentiated PC12 cells to identify premitochondrial and postmitochondrial events occurring during the oxidative stress cascade leading to apoptosis. Our data indicate that an acute and strong oxidative impulse (500 µM H(2)O(2), 30 min) can induce, in this model, a 24-hr self-evolving stress, which advances from a premitochondrial phase characterized by lysosomes and cathepsin B and D translocations to cytosol and early mitochondrial membrane hyperpolarization. This phase lasts for about 5 hr and is followed by a postmitochondrial phase distinguished by mitochondrial membrane depolarization, reactive oxygen species increase, caspase-9 and caspase-3 activations, and apoptosis. Inhibition of cathepsins B and D suggests that cells can be protected at the premitochondrial phase of stress evolution and that new cathepsins regulators, such as glycosaminoglycans mimetics, can be considered as new therapeutic prototypes for neurodegeneration. Insofar as early oxidative stress markers have been related to the early onset of neurodegeneration, strategies protecting cells at the premitochondrial phase of oxidative stress may have important therapeutic applications.


Asunto(s)
Mitocondrias/metabolismo , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo , Aconitato Hidratasa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Catepsina D/metabolismo , Catepsina E/metabolismo , Supervivencia Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Peróxido de Hidrógeno/farmacología , Peroxidación de Lípido/efectos de los fármacos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Malondialdehído/metabolismo , Mitocondrias/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Células PC12/efectos de los fármacos , Células PC12/enzimología , Ratas , Superóxido Dismutasa/metabolismo , Factores de Tiempo
3.
J Neural Transm (Vienna) ; 119(11): 1327-42, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22392058

RESUMEN

Aripiprazole is the only atypical antipsychotic drug known to cause the phosphorylation of AMP-activated protein kinase (AMPK) in PC12 cells. However, the molecular mechanisms underlying this phosphorylation in aripiprazole-treated PC12 cells have not yet been clarified. Here, using PC12 cells, we show that these cells incubated for 24 h with aripiprazole at 50 µM and 25 mM glucose underwent a decrease in their NAD⁺/NADH ratio. Aripiprazole suppressed cytochrome c oxidase (COX) activity but enhanced the activities of pyruvate dehydrogenase (PDH), citrate synthase and Complex I. The changes in enzyme activities coincided well with those in NADH, NAD⁺, and NAD⁺/NADH ratio. However, the bioenergetic peril judged by the lowered COX activity might not be accompanied by excessive occurrence of apoptotic cell death in aripiprazole-treated cells, because the mitochondrial membrane potential was not decreased, but rather increased. On the other hand, when PC12 cells were incubated for 24 h with clozapine at 50 µM and 25 mM glucose, the NAD⁺/NADH ratio did not change. Also, the COX activity was decreased; and the PDH activity was enhanced. These results suggest that aripiprazole-treated PC12 cells responded to the bioenergetic peril more effectively than the clozapine-treated ones to return the ATP biosynthesis back toward its ordinary level. This finding might be related to the fact that aripiprazole alone causes phosphorylation of AMPK in PC12 cells.


Asunto(s)
Antipsicóticos/farmacología , Carbono/metabolismo , Clozapina/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucólisis/efectos de los fármacos , Piperazinas/farmacología , Quinolonas/farmacología , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/efectos de los fármacos , Aripiprazol , Supervivencia Celular/efectos de los fármacos , Dihidrolipoamida Deshidrogenasa/genética , Dihidrolipoamida Deshidrogenasa/metabolismo , Relación Dosis-Respuesta a Droga , Complejo IV de Transporte de Electrones/metabolismo , Líquido Extracelular/efectos de los fármacos , Glucosa/farmacología , Hipoxantina Fosforribosiltransferasa/genética , Hipoxantina Fosforribosiltransferasa/metabolismo , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Cetona Oxidorreductasas/genética , Cetona Oxidorreductasas/metabolismo , Ácido Láctico/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , NAD/metabolismo , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Células PC12/efectos de los fármacos , Células PC12/enzimología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Ácido Pirúvico/metabolismo , ARN Mensajero/metabolismo , Ratas , Factores de Tiempo
4.
Neuroscience ; 158(4): 1436-45, 2009 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-19095046

RESUMEN

Intermittent hypoxia (IH) is a major pathological factor in the development of neural deficits associated with sleep-disordered breathing. Here we demonstrate that IH lasting 2 or 30 days, but not sustained hypoxia (SH) of the same duration, was accompanied by several posttranslational modifications of the large subunit of RNA polymerase II, Rpb1, including hydroxylation of proline 1465, phosphorylation of serine 5 residues within the C-terminal domain, and nondegradative ubiquitylation. These modifications were found to occur in two regions of the brain, hippocampal region CA1 and the prefrontal cortex, but not in neocortex, brainstem and CA3 region of hippocampus. We also found that mice exposed to 14 or 30 days of IH, but not SH, demonstrated cognitive deficits in behavioral assays. Furthermore, by using the pheochromocytoma-derived PC12 cell line, we showed that, under in vitro IH conditions, induction of Rpb1 hydroxylation, phosphorylation, and ubiquitylation required that the von Hippel-Lindau protein be present. We hypothesize that the observed modifications of Rpb1 participate in regulating the expression of genes involved in mediating cognitive deficits evoked by chronic IH.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Hipocampo/enzimología , Hipoxia/patología , Corteza Prefrontal/enzimología , ARN Polimerasa II/metabolismo , Animales , Proteínas Portadoras/metabolismo , Proteínas Cullin/metabolismo , Hipoxia/fisiopatología , Discapacidades para el Aprendizaje/etiología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Células PC12/enzimología , Células PC12/patología , ARN Polimerasa II/genética , Ratas , Proteínas Celulares de Unión al Retinol/metabolismo , Serina/metabolismo , Factores de Tiempo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
5.
J Cell Biol ; 134(6): 1519-29, 1996 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8830779

RESUMEN

Cadherins are calcium-dependent cell adhesion molecules that play fundamental roles in embryonic development, tissue morphogenesis, and cancer. A prerequisite for their function is association with the actin cytoskeleton via the catenins. Tyrosine phosphorylation of beta-catenin, which correlates with a reduction in cadherin-dependent cell adhesion, may provide cells with a mechanism to regulate cadherin activity. Here we report that beta-catenin immune precipitates from PC12 cells contain tyrosine phosphatase activity which dephosphorylates beta-catenin in vitro. In addition, we show that a member of the leukocyte antigen-related protein (LAR)-related transmembrane tyrosine phosphatase family (LAR-PTP) associates with the cadherin-catenin complex. This association required the amino-terminal domain of beta-catenin but does not require the armadillo repeats, which mediate association with cadherins. The interaction also is detected in PC9 cells, which lack alpha-catenin. Thus, the association is not mediated by alpha-catenin or by cadherins. Interestingly, LAR-PTPs are phosphorylated on tyrosine in a TrkA-dependent manner, and their association with the cadherin-catenin complex is reduced in cells treated with NGF. We propose that changes in tyrosine phosphorylation of beta-catenin mediated by TrkA and LAR-PTPs control cadherin adhesive function during processes such as neurite outgrowth.


Asunto(s)
Cadherinas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Receptor trkA , Transactivadores , Secuencia de Aminoácidos , Animales , Células COS/química , Células COS/enzimología , Proteínas Portadoras/metabolismo , Proteínas del Citoesqueleto/química , Regulación hacia Abajo/efectos de los fármacos , Fibronectinas/química , Inmunoglobulinas/química , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Factores de Crecimiento Nervioso/farmacología , Células PC12/química , Células PC12/enzimología , Fosforilación , Pruebas de Precipitina , Estructura Terciaria de Proteína , Proteínas Tirosina Fosfatasas/química , Ratas , Tirosina/metabolismo , beta Catenina
6.
J Neurochem ; 104(2): 409-19, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17949411

RESUMEN

Besides its role in terminating acetylcholine-mediated neurotransmission, acetylcholinesterase (AChE) is found to be expressed and participate in the process of apoptosis in various cell types. However, the mechanisms underlying AChE up-regulation in neuronal cells remain elusive. Herein we demonstrated that glycogen synthase kinase-3beta (GSK3beta) mediates induced AChE-S expression during apoptosis. In this study, A23187 and thapsigargin (TG) were employed to induce apoptosis in neuroendocrine PC12 cells. The results showed that exposure of PC12 cells to A23187 and TG up-regulated AChE activity significantly. The same treatment also led to activation of GSK3beta. Two different inhibitors of GSK3beta (lithium and GSK3beta-specific inhibitor VIII) could block A23187- or TG-induced up-regulation of AChE activity, AChE-S mRNA level and protein expression. However, lithium could not inhibit the induction of AChE-R mRNA and protein under similar conditions. Taken together, our results show that GSK3beta is specifically involved in the induction of AChE-S expression in PC12 cells during apoptosis.


Asunto(s)
Acetilcolinesterasa/metabolismo , Apoptosis/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Sinapsis/enzimología , Acetilcolinesterasa/genética , Animales , Apoptosis/efectos de los fármacos , Calcimicina/farmacología , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta , Yoduros , Ionóforos/farmacología , Cloruro de Litio/farmacología , Células PC12/citología , Células PC12/efectos de los fármacos , Células PC12/enzimología , ARN Mensajero/metabolismo , Ratas , Sinapsis/efectos de los fármacos , Tapsigargina/farmacología , Regulación hacia Arriba/efectos de los fármacos
7.
J Vet Sci ; 8(4): 377-82, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17993752

RESUMEN

Mesenchymal stem cells (MSCs) secrete a variety of neuroregulatory molecules, such as nerve growth factor, brain-derived neurotrophic factor, and glial cell-derived neurotrophic factor, which upregulate tyrosine hydroxylase (TH) gene expression in PC12 cells. Enhancing TH gene expression is a critical step for treatment of Parkinson's disease (PD). The objective of this study was to assess the effects of co-culturing PC12 cells with MSCs from feline bone marrow on TH protein expression. We divided the study into three groups: an MSC group, a PC12 cell group, and the combined MSC + PC12 cell group (the co-culture group). All cells were cultured in DMEM-HG medium supplemented with 10% fetal bovine serum for three days. Thereafter, the cells were examined using western blot analysis and immunocytochemistry. In western blots, the co-culture group demonstrated a stronger signal at 60 kDa than the PC12 cell group (p < 0.001). TH was not expressed in the MSC group, either in western blot or immunocytochemistry. Thus, the MSCs of feline bone marrow can up-regulate TH expression in PC12 cells. This implies a new role for MSCs in the neurodegenerative disease process.


Asunto(s)
Gatos/fisiología , Regulación Enzimológica de la Expresión Génica , Células Madre Mesenquimatosas/citología , Células PC12/enzimología , Tirosina 3-Monooxigenasa/metabolismo , Animales , Antígenos de Superficie/metabolismo , Western Blotting , Técnicas de Cultivo de Célula , Células Cultivadas , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Inmunohistoquímica , Células Madre Mesenquimatosas/metabolismo , Microscopía de Contraste de Fase , Células PC12/citología , Ratas
8.
Oncogene ; 11(2): 239-44, 1995 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-7624141

RESUMEN

Treatment of PC12 cells with nerve growth factor (NGF) results in neural differentiation of the cells, inducing neurite outgrowth. Ras protein has been shown to play an essential role in this process. To examine whether or not the MAP kinase (MAPK) cascade mediates the NGF- and Ras-induced neural differentiation process, we injected PC12 cells with constitutive active forms of each components of the MAPK cascade. When a moderately active mutant of Xenopus MAPK kinase (S222E-MAPKK) in which Ser 222 was changed into glutamic acid was injected, the neurite outgrowth of PC12 cells occurred to some extent. Injection of an N-terminal truncated STE11 protein (delta N-STE11), a constitutively active form of STE11 which is a yeast MAPKK kinase, induced neurite outgrowth in PC12 cells. Furthermore, injection of thiophosphorylated MAPK, but not purified active MAPK, into PC12 cells resulted in neurite outgrowth. Thiophosphorylated MAPK was resistant to protein phosphatase 2A treatment, while purified active MAPK was inactivated by this treatment. All these results have suggested that sustained activation of MAPK is sufficient for PC12 cell differentiation. In accord with this, the delta N-STE11- or S222E- MAPKK-induced neurite outgrowth was inhibited by coinjection of CL-100 protein, a dual-specificity phosphatase that is capable of inactivating MAPK.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Neuritas/fisiología , Células PC12/fisiología , Animales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/farmacología , Diferenciación Celular/efectos de los fármacos , Activación Enzimática , Neuritas/efectos de los fármacos , Neuritas/enzimología , Células PC12/efectos de los fármacos , Células PC12/enzimología , Monoéster Fosfórico Hidrolasas/farmacología , Fosforilación , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Xenopus , Levaduras/enzimología
9.
J Neurosci ; 24(30): 6650-8, 2004 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-15282267

RESUMEN

Neurotrophins, such as NGF and BDNF, activate Trk receptor tyrosine kinases through receptor dimerization at the cell surface followed by autophosphorylation and intracellular signaling. It has been shown that activation of Trk receptor tyrosine kinases can also occur via a G-protein-coupled receptor (GPCR) mechanism, without involvement of neurotrophins. Two GPCR ligands, adenosine and pituitary adenylate cyclase-activating polypeptide (PACAP), can activate Trk receptor activity to increase the survival of neural cells through stimulation of Akt activity. To investigate the mechanism of Trk receptor transactivation, we have examined the localization of Trk receptors in PC12 cells and primary neurons after treatment with adenosine agonists and PACAP. In contrast to neurotrophin treatment, Trk receptors were sensitive to transcriptional and translational inhibitors, and they were found predominantly in intracellular locations particularly associated with Golgi membranes. Biotinylation and immunostaining experiments confirm that most of the transactivated Trk receptors are found in intracellular membranes. These results indicate that there are alternative modes of activating Trk receptor tyrosine kinases in the absence of neurotrophin binding at the cell surface and that receptor signaling may occur and persist inside of neuronal cells.


Asunto(s)
Adenosina/análogos & derivados , Adenosina/farmacología , Aparato de Golgi/enzimología , Membranas Intracelulares/enzimología , Factores de Crecimiento Nervioso/farmacología , Neuropéptidos/farmacología , Neurotransmisores/farmacología , Fenetilaminas/farmacología , Receptor trkA/efectos de los fármacos , Receptor trkB/efectos de los fármacos , Receptores Acoplados a Proteínas G/efectos de los fármacos , Animales , Brefeldino A/farmacología , Cromonas/farmacología , Cicloheximida/farmacología , Dactinomicina/farmacología , Activación Enzimática/efectos de los fármacos , Morfolinas/farmacología , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Células PC12/efectos de los fármacos , Células PC12/enzimología , Fosfatidilinositol 3-Quinasas/fisiología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa , Prosencéfalo/enzimología , Biosíntesis de Proteínas/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/fisiología , Inhibidores de la Síntesis de la Proteína/farmacología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-akt , Ratas , Ratas Sprague-Dawley , Receptor trkA/metabolismo , Receptor trkB/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
10.
Biochim Biophys Acta ; 1440(2-3): 235-43, 1999 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-10521707

RESUMEN

Ultraviolet-C (UVC) irradiation induces DNA damage and UVC-irradiated cells undergo cell growth arrest to repair the damaged DNA or the induction of apoptosis to prevent the risk of neoplastic transformation. Phospholipase C-gamma1 (PLC-gamma1) is a mediator of growth factor induced-signal cascade, catalyzing the hydrolysis of phosphatidyl 4,5-bisphosphate to generate second messengers, diacylglycerol and inositol 1,4,5-trisphosphate (IP(3)). PLC-gamma1 is activated by phosphorylation of tyrosine residues upon occupation of cell surface receptors by growth factors and plays an important role in controlling cellular proliferation and differentiation. In this study, we found that PLC-gamma1 was tyrosine phosphorylated within 2.5 min after UVC irradiation. To investigate the role of UVC-induced tyrosine phosphorylation of PLC-gamma1, we compared the effect of UVC between PLC-gamma1 overexpressing cells and empty vector transfected cells. Overexpression of PLC-gamma1 inhibited UVC-induced sub-diploid peak and DNA fragmentation. Northern blot analysis revealed that UVC-induced c-fos mRNA accumulation was inhibited in PLC-gamma1 overexpressing cells, while c-jun expression was not affected. In addition, UVC-induced activation of c-Jun N-terminal kinase (JNK) was significantly suppressed in PLC-gamma1 overexpressing cells. These results suggest that PLC-gamma1 may associate with the protective function against the UVC-induced cell death progression via the inhibition of accumulation of c-fos mRNA and the inhibition of JNK kinase activity.


Asunto(s)
Apoptosis , Isoenzimas/biosíntesis , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Células PC12/efectos de la radiación , Fosfolipasas de Tipo C/biosíntesis , Animales , Regulación hacia Abajo , Activación Enzimática , Genes fos , Genes jun , Proteínas Quinasas JNK Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/análisis , Células PC12/enzimología , Fosfolipasa C gamma , Ratas , Transcripción Genética/efectos de la radiación , Transfección , Rayos Ultravioleta
11.
Mol Endocrinol ; 11(11): 1618-25, 1997 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9328344

RESUMEN

Mitogen-activated protein (MAP)/ERK kinase (MEK)1 and MEK2 are the upstream activators of the MAP kinases, ERK1 and ERK2. MEK1 and MEK2 are approximately 85% identical in sequence but have unique inserts in their C-terminal domains. MEK isoform-specific antibodies were used to examine expression and regulation of each enzyme. MEK1 and MEK2 were expressed in approximately equal amounts in several cell lines; in some, MEK1 was present in slight excess. Activation of tyrosine kinase-containing receptors, heterotrimeric G proteins, and protein kinase C enhanced the activities of both MEK isoforms in 293 and PC12 cells. AIF4-stimulated both MEK1 and MEK2 in PC12 cells expressing a dominant interfering Ras mutant that prevents nerve growth factor-dependent activation of the cascade. Carbachol also stimulated the pathway in these cells. Thus, in addition to their ability to activate Ras/Raf and the downstream ERK pathway, heterotrimeric G proteins also appear to trigger a Ras-independent mechanism to regulate this kinase cascade. In U373, Chinese hamster ovary (CHO), and INS-1 cells, MEK1 was activated by regulators of ERKs, while MEK2 was not. These data suggest that, like the MAP kinases ERK1 and ERK2, in some cell settings the two similar MEK isoforms are differentially regulated.


Asunto(s)
Proteínas de Unión al GTP/fisiología , Regulación Enzimológica de la Expresión Génica , Quinasas de Proteína Quinasa Activadas por Mitógenos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptores de Superficie Celular/efectos de los fármacos , Transducción de Señal/fisiología , Células 3T3/efectos de los fármacos , Células 3T3/metabolismo , Compuestos de Aluminio/farmacología , Animales , Anticuerpos Monoclonales/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Células CHO , Carbacol/farmacología , Línea Celular , Colforsina/farmacología , Cricetinae , Cricetulus , Embrión de Mamíferos , Activación Enzimática , Inducción Enzimática , Fluoruros/farmacología , Glucosa/farmacología , Insulina/metabolismo , Secreción de Insulina , Insulinoma/metabolismo , Insulinoma/patología , MAP Quinasa Quinasa 1 , MAP Quinasa Quinasa 2 , Ratones , Células PC12/enzimología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Feocromocitoma/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Ratas , Receptores de Superficie Celular/fisiología , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Transfección , Células Tumorales Cultivadas
12.
Endocrinology ; 144(1): 94-100, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12488334

RESUMEN

The observation that natriuretic peptides and protein kinase C activators influence evoked neurotransmitter efflux by diametrically opposed mechanisms prompted an investigation of the influence of natriuretic peptides on protein kinase C activity and the potential involvement of this pathway in neuromodulatory responses to natriuretic peptides. C-Type natriuretic peptide attenuated both evoked dopamine efflux and protein kinase C activity in a concentration-dependent manner consistent with a 10% diminution in protein kinase C activity producing a 4.6-6.2% reduction in evoked dopamine efflux. The ability of C-type natriuretic peptide to suppress evoked dopamine efflux was abolished by treatment with the protein kinase C inhibitors chelerythrine (10 micro M) and staurosporine (10 nM). Both chelerythrine and staurosporine attenuated protein kinase C activity at the concentrations used. The natriuretic peptide C receptor (NPR-C) appeared to mediate the attenuation of protein kinase C activity, because the effect was mimicked by a pentadecapeptide fragment of the NPR-C, and the effect of C-type natriuretic peptide was attenuated by an antibody generated against the same region of the receptor. These data suggest that C-type natriuretic peptide attenuates neurotransmitter efflux by a mechanism involving suppression of neuronal protein kinase C activity via an interaction with the NPR-C.


Asunto(s)
Dopamina/metabolismo , Péptido Natriurético Tipo-C/farmacología , Células PC12/metabolismo , Proteína Quinasa C/metabolismo , Alcaloides , Animales , Benzofenantridinas , Guanilato Ciclasa/fisiología , Células PC12/enzimología , Fenantridinas/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Ratas , Receptores del Factor Natriurético Atrial/fisiología , Estaurosporina/farmacología
13.
Mol Neurobiol ; 22(1-3): 99-113, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11414283

RESUMEN

Neuromodulin (GAP-43), neurogranin (RC3), and PEP-19 are small acid-stable proteins that bind calcium-poor calmodulin through a loosely conserved IQ-motif. Even though these proteins have been known for many years, much about their function in cells is not understood. It has recently become appreciated that calmodulin activity in cells is tightly controlled and that pools of otherwise free calmodulin are sequestered so as to restrict its availability for activating calcium/calmodulin-dependent enzymes. Neuromodulin, neurogranin, and PEP-19 appear to be major participants in this type of regulation. One way in which they do this is by providing localized increases in the concentration of calmodulin in cells so that the maximal level of target activation is increased. Additionally, they can function as calmodulin antagonists by directly inhibiting the association of calcium/calmodulin with enzymes and other proteins. Although neuromodulin, neurogranin, and PEP-19 were early representatives of the small IQ-motif-containing protein family, newer examples have come to light that expand the number of cellular systems through which the IQ-peptide/calmodulin interaction could regulate biological processes including gene transcription. It is the purpose of this review to examine the behavior of neuromodulin, neurogranin, and PEP-19 in paradigms that include both in vitro and in situ systems in order to summarize possible biological consequences that are linked to the expression of this type of protein. The use of protein:protein interaction chromatography is also examined in the recovery of a new calmodulin-binding peptide, CAP-19 (ratMBF1). Consistent with earlier predictions, at least one function of small IQ-motif proteins appears to be that they lessen the extent to which calcium-calmodulin-dependent enzymes become or stay activated. It also appears that these polypeptides can function to selectively inhibit activation of intracellular targets by some agonists while simultaneously permitting activation of these same targets by other agonists. Much of the mechanism for how this occurs is unknown, and possible explanations are examined. One of the biological consequences for a cell that expresses a calmodulin-regulatory protein could be an increased resistance to calcium-mediated toxicity. This possibility is examined for cells expressing PEP-19 and both anatomical and cell-biological data is described. The study of IQ-motif-containing small proteins has stimulated considerable thought as to how calcium signaling is refined in neurons. Current evidence suggests that signaling through calmodulin is not a fulminating and homogenous process but a spatially limited and highly regulated one. Data from studies on neuromodulin, neurogranin, and PEP-19 suggest that they play an important role in establishing some of the processes by which this regulation is accomplished.


Asunto(s)
Señalización del Calcio/fisiología , Proteínas de Unión a Calmodulina/fisiología , Calmodulina/fisiología , Proteína GAP-43/fisiología , Proteínas del Tejido Nervioso/fisiología , Enfermedad de Alzheimer/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Química Encefálica , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Calmodulina/química , Proteínas de Unión a Calmodulina/química , Proteínas de Unión a Calmodulina/farmacología , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Activación Enzimática/efectos de los fármacos , Proteína GAP-43/química , Proteína GAP-43/farmacología , Homeostasis , Humanos , Enfermedad de Huntington/metabolismo , Datos de Secuencia Molecular , Proteínas de Neoplasias/metabolismo , Degeneración Nerviosa , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/farmacología , Neurogranina , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo I , Células PC12/efectos de los fármacos , Células PC12/enzimología , Fosforilación , Procesamiento Proteico-Postraduccional , Ratas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
14.
Mol Neurobiol ; 23(1): 1-19, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11642541

RESUMEN

Mitogen-activated protein kinases (MAPKs) have crucial roles in signal transduction from the cell surface to the nucleus and regulate cell death and survival. Recent papers support the hypothesis that neuronal apoptosis and cerebral ischemia induce the robust activation of MAPK cascades. Although extracellular signal-regulated kinases pathways promote cell survival and proliferation, and c-Jun N-terminal protein kinases/p38 pathways induce apoptosis in general, the roles of MAPK cascades in neuronal death and survival seem to be complicated and altered by the type of cells and the magnitude and timing of insults. Some specific inhibitors of MAPK cascades provide important information in clarifying the roles of each molecule in neuronal death and survival, but the results are still controversial. Further studies are necessary to elucidate the activated signal transduction upstream and downstream of the cascades in cerebral ischemia, and to define the crosstalk between the cascades and other signaling pathways, before MAPK cascades can be candidate molecules in the treatment of cerebral ischemia.


Asunto(s)
Isquemia Encefálica/enzimología , Sistema de Señalización de MAP Quinasas , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Señalización del Calcio , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Regulación de la Expresión Génica , Sustancias de Crecimiento/fisiología , Hipocampo/irrigación sanguínea , Hipocampo/enzimología , Hipocampo/patología , Humanos , Precondicionamiento Isquémico , Proteínas Quinasas JNK Activadas por Mitógenos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/fisiología , Modelos Biológicos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/fisiología , Neuronas/efectos de los fármacos , Neuronas/enzimología , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Células PC12/enzimología , Células PC12/metabolismo , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas , Receptores de Glutamato/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos
15.
Gene ; 162(2): 279-84, 1995 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-7557444

RESUMEN

A novel cDNA encoding PTP (protein tyrosine phosphatase) was cloned from PC12h cells and designated as PCPTP1 (gene encoding PC12 protein Tyr phosphatase). The longest open reading frame (ORF) of this clone encodes a 656-amino-acid (aa) protein with a single PTP catalytic domain. Western blot analysis using a polyclonal Ab (antibody) raised against the cytoplasmic region of PCPTP1 detected two products, a major 65-kDa and minor 42-kDa protein, designated PCPTP1-MFI and PCPTP1-MVQ, respectively, in PC12h cells. These two proteins correspond to the products translated from the second and fifth methionine of PCPTP1, respectively. The bacterially expressed GST::PCPTP1-MVQ fusion protein had phosphatase activity with pNPP (p-nitrophenyl phosphate) as a substrate. Alignment of the aa sequence of PCPTP1-MVQ with those of other PTP showed the highest similarity to STEP and LC-PTP/HePTP, with 54 and 51% identity, respectively. Northern blot analysis showed only one 3.9-kb transcript in PC12h cells, indicating that PCPTP1 corresponds to this 3.9-kb transcript. The 3.9-kb PCPTP1 mRNA was detected in the brain and adrenal gland, but not in other non-neuronal tissues in adult rats. Two other transcripts of 3.3 and 1.7 kb were also detected in brain. NGF (nerve growth factor) and glucocorticoid are known to bimodally regulate the cell fate decision of sympathoadrenal precursors like PC12 cells, with NGF promoting the neuronal phenotype and glucocorticoid promoting the chromaffin phenotype. Still, both agents decreased the level of PCPTP1 mRNA in PC12h cells. Therefore, it is likely that the decrease in the level of PCPTP1 mRNA might be associated or correlated with cell differentiation in PC12h cells.


Asunto(s)
Proteínas del Tejido Nervioso , Proteínas Tirosina Fosfatasas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Encéfalo/enzimología , Diferenciación Celular , Clonación Molecular , Cartilla de ADN/química , ADN Complementario/genética , Expresión Génica , Genes , Péptidos y Proteínas de Señalización Intracelular , Datos de Secuencia Molecular , Células PC12/enzimología , ARN Mensajero/genética , Ratas , Ratas Wistar , Proteínas Tirosina Fosfatasas Clase 7 Similares a Receptores , Distribución Tisular
16.
FEBS Lett ; 319(1-2): 31-4, 1993 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-8454058

RESUMEN

Phorbol esters such as phorbol 12-myristate,13-acetate (PMA) are potent activators of protein kinase C (PKC), and activate all PKC isozymes except zeta and lambda. Recently, 12-deoxyphorbol-13-O-phenylacetate-20-acetate (dPPA) and thymeleatoxin (Tx) were reported to selectively activate PKC beta 1 (dPPA) and PKC alpha, -beta, and -gamma (Tx), but not PKC delta or PKC epsilon in vitro. We examined the ability of these phorbol derivatives to translocate and down-regulate PKC isozymes in intact cells. Our findings demonstrate that dPPA and Tx cause translocation and down-regulation of multiple PKC isozymes, including delta and epsilon.


Asunto(s)
Isoenzimas/metabolismo , Ésteres del Forbol/farmacología , Proteína Quinasa C/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Animales , Transporte Biológico/efectos de los fármacos , Citosol/enzimología , Electroforesis en Gel de Poliacrilamida , Activación Enzimática/efectos de los fármacos , Células PC12/enzimología
17.
Neurology ; 54(8): 1665-70, 2000 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-10762511

RESUMEN

OBJECTIVE: To examine the origin of hyperphosphorylated proteins within the vacuolated myofibers in sporadic inclusion body myositis (s-IBM) and search for dysregulated intracellular protein phosphorylation. BACKGROUND: s-IBM is morphologically characterized by primary endomysial inflammation and vacuolated myofibers containing tubulofilaments that originate from cytoskeletal proteins. Mitogen-activated protein kinases (MAPKs) play a role in regulating phosphorylation and maintaining the stability of the cytoskeletal architecture. METHODS: Muscle biopsies from seven patients with s-IBM and 15 controls were examined for the expression of the active components of the various MAPKs, including p44/42MAPK, p38MAPK, p46JNK1, p54JNK2, and p54JNK3, using immunocytochemistry and Western blot analysis. The expression of selected phosphorylated components was also examined in the same specimens. RESULTS: In s-IBM, but not the disease controls, the vacuolated muscle fibers express active p42MAPK but not JNK or p38MAPK. Western blots of cell lysates confirmed the hyperexpression of p42MAPK and demonstrated a novel 35 kD phosphoprotein. Antibodies against phosphoepitopes of the 35 kD protein preferentially immunostained antigens within the vacuolated muscle fibers of s-IBM but not disease controls. CONCLUSION: In s-IBM, there is increased p42MAPK activation and abnormal intracellular protein phosphorylation with selective accumulation of a 35 kD phosphoprotein within the vacuolated fibers. Although the hyperexpression of 35kD protein may represent cytoskeletal by-products due to heightened p42MAPK activation, its abundant expression only in s-IBM implies that hyperphosphorylated myofibrillar proteins may be involved in the primary disease process.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Esquelético/enzimología , Miositis por Cuerpos de Inclusión/enzimología , Anciano , Animales , Biopsia , Western Blotting , Proteínas del Citoesqueleto/biosíntesis , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Fibras Musculares Esqueléticas/enzimología , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/patología , Miositis por Cuerpos de Inclusión/patología , Células PC12/efectos de los fármacos , Células PC12/enzimología , Fosforilación , Fosfoserina/metabolismo , Ratas , Vacuolas/metabolismo
18.
Neuroscience ; 104(2): 593-8, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11377858

RESUMEN

PC12-GFRalpha1 cells, a clonal cell line engineered to express glial cell line-derived neurotrophic factor receptor alpha1 were constructed. Given glial cell line-derived neurotrophic factor could induce the differentiation and promote the survival of PC12-GFRalpha1 cells at low concentrations, the cells provide an unlimited source of monoclonal cells for studies on the signal transduction pathway of glial cell line-derived neurotrophic factor. To characterize the involvement of the mitogen-activated protein kinase and phosphatidylinositol 3-kinase pathways in the biological effect of glial cell line-derived neurotrophic factor, we used the mitogen-activated protein kinase kinase inhibitor PD98059 and the phosphatidylinositol 3-kinase inhibitor LY294002. PD98059 blocked glial cell line-derived neurotrophic factor-induced PC12-GFRalpha1 cells neurite formation in a dose-dependent manner, without significantly altering cell viability. LY294002 reversed the survival-promoting effect of glial cell line-derived neurotrophic factor on the PC12-GFRalpha1 cells in serum-deprived medium. The present study demonstrates that phosphatidylinositol 3-kinase pathway seems to mediate the survival-promoting effect of glial cell line-derived neurotrophic factor on PC12-GFRalpha1 cells, while the activation of mitogen-activated protein kinase pathway could be an important step in mediating PC12-GFRalpha1 cells differentiation induced by glial cell line-derived neurotrophic factor. Therefore, it is inferred that similar intracellular signaling components are used by distinct growth factors toward a common biological effect.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Proteínas de Drosophila , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Factores de Crecimiento Nervioso , Proteínas del Tejido Nervioso/farmacología , Células PC12/enzimología , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Animales , Diferenciación Celular/fisiología , Supervivencia Celular/fisiología , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Factor Neurotrófico Derivado de la Línea Celular Glial , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial , Sistema de Señalización de MAP Quinasas/fisiología , Morfolinas/farmacología , Proteínas del Tejido Nervioso/metabolismo , Células PC12/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-ret , Ratas , Proteínas Tirosina Quinasas Receptoras/genética , Transfección
19.
J Mol Neurosci ; 4(2): 125-39, 1993.
Artículo en Inglés | MEDLINE | ID: mdl-8105857

RESUMEN

The functional organization of rat tyrosine hydroxylase was investigated by deletion mutagenesis of the regulatory and catalytic domains. A series of tyrosine hydroxylase cDNA deletion mutants were amplified by PCR, cloned into the pET3C prokaryotic expression vector, and the mutant proteins were partially purified from E. coli. The results show that the deletion of up to 157 N-terminal amino acids activated the enzyme, but further deletion to position 184 completely destroyed catalytic activity. On the carboxyl end, the removal of 43 amino acids decreased but did not eliminate activity, suggesting that this region may play a different role in the regulation of the enzyme. These findings place the amino end of the catalytic domain between residues 158 and 184 and the carboxyl end at or prior to position 455. Deletions within the first 157 amino acids in the N-terminus caused an increase in hydroxylating activity, a decrease in the apparent Km for tyrosine and phenylalanine substrates, and a substantial increase in the Ki for dopamine inhibition. The results define this region of the N-terminus as the regulatory domain of tyrosine hydroxylase, whose primary functions are to restrict the binding of amino acid substrates and to facilitate catecholamine inhibition. The results also suggest that the well-established role of the regulatory domain in restricting cofactor binding may be secondary to an increase in catecholamine binding, which in turn lowers the affinity for the cofactor. These findings provide new insight into the functional organization and mechanisms of regulation of tyrosine hydroxylase.


Asunto(s)
Mutagénesis , Proteínas de Neoplasias/genética , Células PC12/enzimología , Tirosina 3-Monooxigenasa/genética , Animales , Secuencia de Bases , Sitios de Unión , ADN Complementario/genética , ADN de Neoplasias/genética , Dopamina/metabolismo , Dopamina/farmacología , Retroalimentación , Datos de Secuencia Molecular , Proteínas de Neoplasias/química , Fosforilación , Reacción en Cadena de la Polimerasa , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Ratas , Proteínas Recombinantes de Fusión/química , Eliminación de Secuencia , Tirosina 3-Monooxigenasa/química
20.
J Mol Neurosci ; 4(2): 97-105, 1993.
Artículo en Inglés | MEDLINE | ID: mdl-8217523

RESUMEN

Pheochromocytoma PC12 cells grown in the presence of nerve growth factor (NGF) undergo marked neuronal differentiation. During this process gene expression is altered, resulting in the activation of genes specific for neuronal properties, including the gene encoding neuropeptide Y (NPY). Here we sought to determine whether NGF also induces the activity of peptidylglycine alpha-hydroxylating monooxygenase (PHM) (EC1.4.17.3). PHM catalyzes the rate limiting step in the formation of alpha-amidated NPY from its glycine extended precursor, a posttranslational modification essential for biologic activity. PC12 cells were grown with or without NGF and assayed for PHM activity under optimal conditions. Whole cell extracts, medium and soluble and membrane bound fractions were assayed; total cellular PHM activity was found to be primarily membrane bound (fivefold greater than in soluble) and very little activity was released into the medium. Compared to control cells, PHM activity was increased significantly by NGF by 24 h but not before 4 h exposure. Through kinetic analysis, it was determined that the NGF-induction of PHM was a result of an increase in Vmax with no change in Km. It was found that the glucocorticoid, dexamethasone (DEX), decreased basal PHM activity and prevented its induction by NGF. Cotreatment with DEX for up to 7 d, however, did not dramatically alter the pronounced changes in cell morphology that occurred in response to NGF. These findings indicate that NGF and glucocorticoids exert reciprocal control over the activity of PHM in PC12 cells. As such, the process of differentiation in PC12 cells is a model for studying the mechanisms that coordinate the expression and activity of peptide processing enzymes with the regulation of their substrates and products.


Asunto(s)
Oxigenasas de Función Mixta/biosíntesis , Complejos Multienzimáticos , Proteínas de Neoplasias/biosíntesis , Factores de Crecimiento Nervioso/farmacología , Células PC12/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Diferenciación Celular/efectos de los fármacos , Dexametasona/farmacología , Inducción Enzimática/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Cinética , Datos de Secuencia Molecular , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Células PC12/enzimología , Células PC12/ultraestructura , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA