Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Intervalo de año de publicación
1.
Behav Brain Res ; 253: 240-52, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23896051

RESUMEN

Early life stress is shown to have a life-span outcome on human and animal behavior, increasing the risk for psychopathology. The gene methylenetetrahydrofolate reductase (MTHFR), which encodes for a key enzyme in one carbon metabolism, shows a high prevalence of polymorphism in patients with developmental disorders. Here we examined the hypothesis that MTHFR deficiency results in an increased susceptibility of the developing brain to mild neonatal stress (NS). Mild NS failed to alter corticosterone levels in young and adult Wt mice. However, an elevated level of corticosterone was found in the MTHFR deficient-NS female, exemplifying enhanced sensitivity to NS. Behavioral phenotyping of Wt and MTHFR deficient mice provides evidence that the effect of mild NS may be amplified by the MTHFR deficient genotype. Distinct behavioral characteristics were altered in male and female mice. In general, three patterns of influence on mice behavior were observed: (1) an additive suppressive effect of NS and MTHFR deficiency on exploration and activity was evident in females; (2) stress related parameters were significantly sensitive to genotype in females, presenting an interaction between genotype and sex; (3) various aspects of behavior in a social setting were modified preferably in males by genotype, NS and the interaction between the two, while females exhibited a smaller effect that was restricted to NS with no genotype effect. Overall, our results support an interaction between mild NS, the MTHFR genotype and sex. We suggest using this animal model to study the molecular mechanism linking these two risk factors and their involvement in neurodevelopmental disorders such as schizophrenia and autism.


Asunto(s)
Homocistinuria/fisiopatología , Metilenotetrahidrofolato Reductasa (NADPH2)/deficiencia , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Espasticidad Muscular/fisiopatología , Estrés Psicológico/fisiopatología , Animales , Animales Recién Nacidos , Ansiedad/psicología , Conducta Animal/efectos de los fármacos , Corticosterona/metabolismo , Conducta Exploratoria/fisiología , Femenino , Genotipo , Homocistinuria/psicología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Espasticidad Muscular/psicología , Trastornos Psicóticos/fisiopatología , Trastornos Psicóticos/psicología , Reflejo/fisiología , Caracteres Sexuales , Conducta Social
2.
Mol Cell Biol ; 18(2): 880-6, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9447984

RESUMEN

The guanine nucleotide exchange factor Sos mediates the coupling of receptor tyrosine kinases to Ras activation. To investigate the mechanisms that control Sos activity, we have analyzed the contribution of various domains to its catalytic activity. Using human Sos1 (hSos1) truncation mutants, we show that Sos proteins lacking either the amino or the carboxyl terminus domain, or both, display a guanine nucleotide exchange activity that is significantly higher compared with that of the full-length protein. These results demonstrate that both the amino and the carboxyl terminus domains of Sos are involved in the negative regulation of its catalytic activity. Furthermore, in vitro Ras binding experiments suggest that the amino and carboxyl terminus domains exert negative allosteric control on the interaction of the Sos catalytic domain with Ras. The guanine nucleotide exchange activity of hSos1 was not augmented by growth factor stimulation, indicating that Sos activity is constitutively maintained in a downregulated state. Deletion of both the amino and the carboxyl terminus domains was sufficient to activate the transforming potential of Sos. These findings suggest a novel negative regulatory role for the amino terminus domain of Sos and indicate a cooperation between the amino and the carboxyl terminus domains in the regulation of Sos activity.


Asunto(s)
Factor 2 Eucariótico de Iniciación/fisiología , Proteínas/fisiología , Animales , Células COS , Catálisis , Receptores ErbB/metabolismo , Factor 2 Eucariótico de Iniciación/genética , Sustancias de Crecimiento/metabolismo , Factores de Intercambio de Guanina Nucleótido , Humanos , Mutagénesis Sitio-Dirigida , Proteínas/genética , Transducción de Señal , Relación Estructura-Actividad , Factores de Intercambio de Guanina Nucleótido ras , Proteínas ras/metabolismo
3.
Virology ; 232(2): 337-44, 1997 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-9191847

RESUMEN

The LTRs of HIV-1 and HTLV-I have been shown by several laboratories to be activated by 12-O-tetradecanoylphorbol-13-acetate (TPA). This agent is a potent activator of protein kinase C (PKC). However, long exposure to TPA downregulates PKC in many cell types. We demonstrated that TPA treatment of Jurkat cells for more than 24 hr resulted in a sever depletion of this enzyme. Therefore, to explore the role of PKC in the effect of TPA on these LTRs, we transfected Jurkat cells with HIV-1 LTR-CAT or HTLV-I LTR-CAT construct after 72 hr of TPA pretreatment. While this TPA pretreatment considerably reduced the HIV-1 LTR basal expression, it strongly stimulated the expression of HTLV-I LTR. Furthermore, when TPA was added after transfection, a strong stimulation of HIV-1 LTR was observed, which could be abrogated by PKC inhibitors like H7 and chelerythryn. However, under these conditions TPA stimulated HTLV-I LTR to a lesser extent than did the long-term TPA pretreatment. Moreover, this stimulation was enhanced by the PKC inhibitors. Thus our data indicate that while the effect of TPA on HIV-1 LTR is strictly dependent on PKC activity, its effect on HTLV-I LTR is exerted via a different pathway that not only does not require PKC activation but rather seems to be antagonized by the activated PKC. Using a deletion mutant of HTLV-I LTR we mapped the PKC-independent effect of TPA to the c-ets responsive region 1 (ERR-1) located in U3 of this LTR.


Asunto(s)
Regulación Viral de la Expresión Génica/efectos de los fármacos , Duplicado del Terminal Largo de VIH , VIH-1/genética , Virus Linfotrópico T Tipo 1 Humano/genética , Proteína Quinasa C/metabolismo , Secuencias Repetitivas de Ácidos Nucleicos , Acetato de Tetradecanoilforbol/farmacología , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Regulación hacia Abajo , Activación Enzimática , Humanos , Células Jurkat , Mutación
4.
Cell Immunol ; 178(2): 141-51, 1997 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-9225005

RESUMEN

T cell activation via the antigen receptor or by PKC-activating drugs results in phosphorylation of Lck and alteration of its electrophoretic mobility. Although tyrosine phosphorylation appears to regulate Lck enzymatic activity, the significance of phosphorylation of serine residues and its relevance to the cell proliferation process are yet unclear. We found that the PKC activator, bryostatin, like PMA, induced the conversion of p56lck to a slower migrating form with an apparent molecular mass of 60 kDa. The effect of PMA lasted over 48 hr but that of bryostatin was transient and correlated in time kinetics with that of the bryostatin-induced degradation of PKC. The effects of bryostatin were dominant over those of PMA. In addition, PKC was found to affect both serine and tyrosine phosphorylation of Lck but had no significant effect on the in vitro catalytic activity of Lck. To test whether serine phosphorylation of Lck may affect its ability to bind tyrosine phosphoproteins, we compared Lck immunoprecipitates from PMA- and bryostatin-treated T cells. We found that a 36- to 38-kDa tyrosine phosphoprotein co-immunoprecipitated with Lck from cells that were treated for 24 hr with PMA, but not bryostatin. A p36-38 from PMA- but not bryostatin-treated cells also interacted with an Lck-SH2 fusion protein, suggesting differential regulation of p36-38 by PMA and bryostatin. Furthermore, in vitro phosphorylation of p36-38 occurred in lysates of cells that were treated for 24 hr with PMA, but not in lysates of bryostatin-treated cells. The results show that tyrosine phosphorylation and the association of p36-38 with Lck are differentially affected by bryostatin and PMA and suggest that PKC regulates the interaction of potential signaling molecules with Lck, thereby regulating biochemical events that are relevant to T cell mitogenesis and/or transformation.


Asunto(s)
Carcinógenos/farmacología , Lactonas/farmacología , Proteína Quinasa C/fisiología , Proteínas Proto-Oncogénicas/fisiología , Acetato de Tetradecanoilforbol/farmacología , Familia-src Quinasas/fisiología , Adyuvantes Inmunológicos/farmacología , Brioestatinas , Electroforesis en Gel de Poliacrilamida , Activación Enzimática , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito , Macrólidos , Peso Molecular , Fosfoproteínas/metabolismo , Fosfoserina/metabolismo , Fosfotreonina/metabolismo , Fosfotirosina/metabolismo , Procesamiento Proteico-Postraduccional , Factores de Tiempo
5.
Virus Genes ; 15(3): 227-34, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9482588

RESUMEN

Measles virus (MV) is among the infectious agents displaying a propensity for establishing persistent infections of the CNS. It is assumed that continuous presence of MV defective particles or viral genome in persistently infected cells may influence host cellular processes and perturb biochemical signal transduction pathways operating in linkage to various cell surface receptors. PKC expression in a MV persistently infected neuroblastoma cell line (NS20Y/MS) was investigated. The relative levels of PKC isoenzymes were determined by Western blot analysis. We found that protein levels of PKCalpha, epsilon and zeta, but not PKCdelta, were increased in NS20Y/MS cells. PKCbeta, gamma and eta were undetectable. Treatment of NS20Y/MS cells with anti-MV Abs, which downregulated MV protein synthesis, also reduced PKCalpha expression to the basal level observed in the uninfected NS20Y cells. Our results suggest that a persistent MV infection has specific effects on the expression of certain PKC isoenzymes. We postulate that the MV-associated neurologic changes may reflect virus induced changes in biochemical signaling pathways and that these effects are likely to be regulated by the host's anti-viral humoral immune response.


Asunto(s)
Isoenzimas/metabolismo , Virus del Sarampión/fisiología , Fosfoproteínas , Proteína Quinasa C/metabolismo , Latencia del Virus , Animales , Anticuerpos Monoclonales , Anticuerpos Antivirales/metabolismo , Regulación hacia Abajo , Hemaglutininas Virales/inmunología , Isoenzimas/biosíntesis , Ratones , Neuroblastoma , Nucleocápside/biosíntesis , Proteína Quinasa C/biosíntesis , Proteína Quinasa C-alfa , Células Tumorales Cultivadas , Proteínas Virales/biosíntesis
6.
Cell Immunol ; 158(1): 195-207, 1994 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-8087865

RESUMEN

Protein kinase C (PKC)-activating tumor promoters, such as PMA, are mitogenic to human T lymphocytes but, in contrast to many physiological mitogenic agonists, PMA does not operate via a receptor-coupled, IL-2-dependent signaling pathway. Bryostatin, another PKC activator, is not mitogenic to human T cells and, moreover, it inhibits the PMA-induced mitogenic response. Inhibition of the PMA-induced human T cell proliferation by bryostatin correlates with the bryostatin-induced increased degradation rate of conventional PKC (cPKC) isoenzymes. Nevertheless, bryostatin synergizes with suboptimal doses of various T cell mitogens operating via a Ca(2+)-dependent signaling pathway to induce a strong IL-2-dependent proliferative response. Our findings indicate that Ca2+ signals do not abolish the bryostatin-induced enhanced degradation rate of cPKC observed in human T cells. The results suggest that transient activation of cPKC by bryostatin induces some irreversible biochemical events that, by themselves, are not sufficient for the induction of cell proliferation, but that can be complemented by a Ca(2+)-mediated signal(s) leading to IL-2-dependent T cell proliferation. In contrast, PMA-induced IL-2-independent proliferation of human T cells appears to be dependent on the continuous presence of cPKC (that may mediate some critical events at late stages of the response) and, therefore, is inhibited by bryostatin, that induces a rapid degradation of potentially active cPKC.


Asunto(s)
Calcio/metabolismo , Lactonas/farmacología , Activación de Linfocitos/efectos de los fármacos , Mitógenos/farmacología , Proteína Quinasa C/metabolismo , Linfocitos T/efectos de los fármacos , Transporte Biológico/efectos de los fármacos , Brioestatinas , Membrana Celular/efectos de los fármacos , Membrana Celular/enzimología , Células Cultivadas , Sinergismo Farmacológico , Humanos , Interleucina-2/biosíntesis , Ionomicina/farmacología , Macrólidos , Transducción de Señal , Linfocitos T/enzimología , Linfocitos T/inmunología , Acetato de Tetradecanoilforbol/farmacología
7.
Immunol Lett ; 39(1): 17-22, 1993 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8144186

RESUMEN

The bryostatin (Bryo) is a macrocyclic lactone that binds specifically to protein kinase C (PKC) thereby affecting cell growth and differentiation and inhibits phorbol ester-induced tumor promotion. We used human peripheral blood lymphocytes (PBL) and epidermal cells in order to analyze the action mechanism of Bryo and compare it with that of the phorbol ester PMA. Bryo and PMA activated PBL- or T cell-derived PKC in a similar dose-response and induced a similar time kinetic of cytosol-to-membrane translocation of enzymatically active and immunoreactive PKC. In addition, the 2 drugs induced similar patterns of protein phosphorylation and activated the c-fos and c-jun genes that their protein products regulate transcription of TRE-containing genes. In contrast, long-term (20 h) treatment of cells with Bryo resulted in a marked loss of both cytosolic- and membrane-bound PKC while PMA induced only a slight reduction in the amount of cellular PKC. Inhibition of PMA-induced human T-cell proliferation by Bryo correlated with a reduction in the amount of cellular PKC. An opposite effect was observed in human epidermal cells where Bryo augmented growth and proliferation while PMA induced terminal differentiation and cell death. We propose that at least some of the differences in the biological effects induced by Bryo and PMA are due to distinct regulations of PKC. Thus, although both agents can initially bind to and activate PKC at a later time (approximately 16 h), Bryo, but not PMA, induces rapid PKC degradation and inhibition of PKC-regulated biological responses that are dependent on the continuous presence and/or activation of the enzyme.


Asunto(s)
Antineoplásicos/farmacología , Queratinocitos/efectos de los fármacos , Lactonas/farmacología , Proteína Quinasa C/metabolismo , Linfocitos T/efectos de los fármacos , Adyuvantes Inmunológicos , Secuencia de Aminoácidos , Brioestatinas , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Humanos , Queratinocitos/enzimología , Activación de Linfocitos/efectos de los fármacos , Macrólidos , Mitógenos , Datos de Secuencia Molecular , Piel/citología , Piel/efectos de los fármacos , Linfocitos T/enzimología , Acetato de Tetradecanoilforbol/farmacología
8.
J Immunol ; 150(4): 1195-204, 1993 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-8432975

RESUMEN

The bryostatins (Bryo) represent a group of immunomodulators that counteract the tumor-promoting effects of PMA. In contrast to the mitogenic effect of PMA on human peripheral blood T lymphocytes, Bryo was nonmitogenic and, furthermore, it inhibited PMA-induced T cell proliferation in a dose-dependent manner when added up to 2 days after PMA. Because both Bryo and PMA bind to, and activate, protein kinase C (PKC), we compared their effects on PKC expression and activity in human PBL or leukemic T cells (Jurkat). After treatment for 5 to 60 min, both Bryo and PMA were found to: a) activate PKC in vitro with similar dose-response curves; b) induce a nearly complete cytosol-to-membrane translocation of enzymatically active, Ca(2+)-dependent PKC and of distinct immunoreactive PKC isoforms in intact PBL; and c) stimulate similar patterns of protein phosphorylation. After a longer, 20-h treatment with PMA (20 nM), a considerable portion of PKC was still membrane-associated, and the total amount of immunoreactive PKC was not reduced considerably. In contrast, Bryo induced a marked loss of cellular immunoreactive PKC, including PKC-alpha and -beta. These results were paralleled by measurements of total cytosol- or membrane-associated PKC enzymatic activity. Thus, substantial PKC activity was associated with the particulate fraction of PMA-, but not Bryo-stimulated PBL. Furthermore, inhibition of PMA-induced T cell proliferation by Bryo also correlated with a reduction in the amount of cytosolic and membrane-bound immunoreactive PKC and enzymatic activity, demonstrating the dominance of Bryo over PMA. We propose that Bryo inhibits PMA-induced T cell proliferation by causing rapid degradation of PKC, reflecting a requirement of persistent PKC stimulation (lasting approximately 48 h) for the activation of human T cells and progression through the cell cycle.


Asunto(s)
Lactonas/farmacología , Activación de Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Ésteres del Forbol/farmacología , Proteína Quinasa C/metabolismo , Brioestatinas , Compartimento Celular/efectos de los fármacos , Células Cultivadas , Humanos , Técnicas In Vitro , Macrólidos , Fosfoproteínas/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA