Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Death Discov ; 4: 44, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29581895

RESUMEN

We and others have demonstrated that stimulants such as methamphetamine (METH) exerts immunosuppressive effects on the host's innate and adaptive immune systems and has profound immunological implications. Evaluation of the mechanisms responsible for T-cell immune dysregulation may lead to ways of regulating immune homeostasis during stimulant use. Here we evaluated the effects of METH on T cell cycle entry and progression following activation. Kinetic analyses of cell cycle progression of T-cell subsets exposed to METH demonstrated protracted G1/S phase transition and differentially regulated genes responsible for cell cycle regulation. This result was supported by in vivo studies where mice exposed to METH had altered G1 cell cycle phase and impaired T-cell proliferation. In addition, T cells subsets exposed to METH had significant decreased expression of cyclin E, CDK2 and transcription factor E2F1 expression. Overall, our results indicate that METH exposure results in altered T cell cycle entry and progression. Our findings suggest that disruption of cell cycle machinery due to METH may limit T-cell proliferation essential for mounting an effective adaptive immune response and thus may strongly contribute to deleterious effect on immune system.

2.
J Leukoc Biol ; 99(1): 213-23, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26302754

RESUMEN

The novel transmembrane G protein-coupled receptor, trace amine-associated receptor 1 (TAAR1), represents a potential, direct target for drugs of abuse and monoaminergic compounds, including amphetamines. For the first time, our studies have illustrated that there is an induction of TAAR1 mRNA expression in resting T lymphocytes in response to methamphetamine. Methamphetamine treatment for 6 h significantly increased TAAR1 mRNA expression (P < 0.001) and protein expression (P < 0.01) at 24 h. With the use of TAAR1 gene silencing, we demonstrate that methamphetamine-induced cAMP, a classic response to methamphetamine stimulation, is regulated via TAAR1. We also show by TAAR1 knockdown that the down-regulation of IL-2 in T cells by methamphetamine, which we reported earlier, is indeed regulated by TAAR1. Our results also show the presence of TAAR1 in human lymph nodes from HIV-1-infected patients, with or without a history of methamphetamine abuse. TAAR1 expression on lymphocytes was largely in the paracortical lymphoid area of the lymph nodes with enhanced expression in lymph nodes of HIV-1-infected methamphetamine abusers rather than infected-only subjects. In vitro analysis of HIV-1 infection of human PBMCs revealed increased TAAR1 expression in the presence of methamphetamine. In summary, the ability of methamphetamine to activate trace TAAR1 in vitro and to regulate important T cell functions, such as cAMP activation and IL-2 production; the expression of TAAR1 in T lymphocytes in peripheral lymphoid organs, such as lymph nodes; and our in vitro HIV-1 infection model in PBMCs suggests that TAAR1 may play an important role in methamphetamine -mediated immune-modulatory responses.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Metanfetamina/farmacología , Receptores Acoplados a Proteínas G/genética , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Ciclo Celular/genética , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación hacia Abajo , Infecciones por VIH/genética , Infecciones por VIH/inmunología , Humanos , Inmunomodulación/efectos de los fármacos , Interleucina-2/metabolismo , Metanfetamina/efectos adversos , Receptores Acoplados a Proteínas G/metabolismo , Trastornos Relacionados con Sustancias/genética , Trastornos Relacionados con Sustancias/inmunología , Trastornos Relacionados con Sustancias/metabolismo , Linfocitos T/inmunología
3.
J Neuroimmune Pharmacol ; 10(2): 302-8, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25666933

RESUMEN

Chronic neuroinflammatory disorders (such as HIV associated neurodegeneration) require treatment that decreases production of inflammatory factors by activated microglia and macrophages and protection of blood brain barrier (BBB) injury secondary to activation of brain endothelium. Cannabioid type 2 receptor (CB2) is highly expressed on macrophages and brain microvasular enndothelial cells (BMVEC) and is upregulated in inflammation and HIV infection. It has been shown that CB2 activation dampened inflammatory responses in macrophages and BMVEC. In this study, we assessed by PCR array the expression of a wide range of genes increased in macrophages and BMVEC in inflammation. TNFα treatment upregulated 33 genes in primary human BMVEC, and two highly selective CB2 agonists diminished expression of 31 and 32 genes. These results were confirmed by functional assays (BBB protection after inflammatory insult and decreased migration of monocytes across BMVEC monolayers after CB2 stimulation). Similarly, CB2 stimulation in primary human macrophages led to the suppression of 35 genes out of the 50 genes upregulated by LPS. Such changes in gene expression paralleled diminished secretion of proinflammatory factors. These results indicate the potential utility of CB2 agonists for the treatment of neuroinflammation.


Asunto(s)
Encéfalo/metabolismo , Endotelio/metabolismo , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Receptor Cannabinoide CB2/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Células Cultivadas , Endotelio/efectos de los fármacos , Endotelio/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , Humanos , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Receptor Cannabinoide CB2/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/farmacología
4.
Cell Cycle ; 12(10): 1569-77, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23603988

RESUMEN

Several reports have pointed to the negative involvement of p53 in transcriptional regulation of the human immunodeficiency virus type 1 long-terminal repeat (HIV-1 LTR). We recently demonstrated that through their physical interaction, cdk9 phosphorylates p53 on Ser-392, leading to p53 stability and accumulation. As a result, p53 stalled transcriptional elongation of the HIV-1 LTR and significantly reduced HIV-1 replication in primary microglia and astrocytes. Therefore, we sought to identify the mechanisms used by cdk9 to allow this p53 function. Using western blot analysis, we found that cdk9 promotes inhibition and phosphorylation of Mdm2 on Ser-395, thus preventing degradation of p53, a protein that is directly involved in promoting p53 ubiquitination. On the other hand, we showed that cdk9 phosphorylates Pirh2 on Ser-211 and Thr-217 residues through their physical interaction. Phosphorylation of Pirh2 renders it inactive and may contribute to p53-inhibition of transcriptional elongation of the HIV-1 LTR. Hence, we suggest that phosphorylation of Pirh2 may be a novel target for the inhibition of HIV-1 gene expression.


Asunto(s)
Quinasa 9 Dependiente de la Ciclina/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Línea Celular , Quinasa 9 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 9 Dependiente de la Ciclina/genética , Duplicado del Terminal Largo de VIH/genética , VIH-1/metabolismo , Humanos , Datos de Secuencia Molecular , Fosforilación , Mapeo de Interacción de Proteínas , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transfección , Proteína p53 Supresora de Tumor/genética , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
5.
J Leukoc Biol ; 93(5): 801-10, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23463725

RESUMEN

Infiltrating monocytes and macrophages play a crucial role in the progression of HIV-1 infection in the CNS. Previous studies showed that activation of the CB2 can attenuate inflammatory responses and affect HIV-1 infectivity in T cells and microglia. Here, we report that CB2 agonists can also act as immunomodulators on HIV-1-infected macrophages. First, our findings indicated the presence of elevated levels of CB2 expression on monocytes/macrophages in perivascular cuffs of postmortem HIV-1 encephalitic cases. In vitro analysis by FACS of primary human monocytes revealed a step-wise increase in CB2 surface expression in monocytes, MDMs, and HIV-1-infected MDMs. We next tested the notion that up-regulation of CB2 may allow for the use of synthetic CB2 agonist to limit HIV-1 infection. Two commercially available CB2 agonists, JWH133 and GP1a, and a resorcinol-based CB2 agonist, O-1966, were evaluated. Results from measurements of HIV-1 RT activity in the culture media of 7 day-infected cells showed a significant decrease in RT activity when the CB2 agonist was present. Furthermore, CB2 activation also partially inhibited the expression of HIV-1 pol. CB2 agonists did not modulate surface expression of CXCR4 or CCR5 detected by FACS. We speculate that these findings indicate that prevention of viral entry is not a central mechanism for CB2-mediated suppression in viral replication. However, CB2 may affect the HIV-1 replication machinery. Results from a single-round infection with the pseudotyped virus revealed a marked decrease in HIV-1 LTR activation by the CB2 ligands. Together, these results indicate that CB2 may offer a means to limit HIV-1 infection in macrophages.


Asunto(s)
Agonistas de Receptores de Cannabinoides/farmacología , VIH-1/efectos de los fármacos , Macrófagos/virología , Receptor Cannabinoide CB2/agonistas , Replicación Viral/efectos de los fármacos , Anisoles/farmacología , Cannabinoides/farmacología , Células Cultivadas , Ciclohexanoles , Duplicado del Terminal Largo de VIH/efectos de los fármacos , VIH-1/fisiología , Humanos , Indenos/farmacología , Pirazoles/farmacología , Receptor Cannabinoide CB2/análisis , Receptor Cannabinoide CB2/fisiología , Receptores CCR5/análisis , Receptores CXCR4/análisis
6.
PLoS One ; 8(2): e55972, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23418486

RESUMEN

Neuroinflammatory conditions often involve dysfunction of the Blood-Brain Barrier (BBB). Therefore, identifying molecular targets that can maintain barrier fidelity is of clinical importance. We have previously reported on the anti-inflammatory effects that glycogen synthase kinase 3ß (GSK3ß) inhibition has on primary human brain endothelial cells. Here we show that GSK3ß inhibitors also promote barrier tightness by affecting tight junction (TJ) protein stability. Transendothelial electrical resistance (TEER) was used to evaluate barrier integrity with both pharmacological inhibitors and mutants of GSK3ß. Inhibition of GSK3ß produced a gradual and sustained increase in TEER (as much as 22% over baseline). Analysis of subcellular membrane fractions revealed an increase in the amount of essential tight junction proteins, occludin and claudin-5, but not claudin-3. This phenomenon was attributed to a decrease in TJ protein turnover and not transcriptional regulation. Using a novel cell-based assay, inactivation of GSK3ß significantly increased the half-life of occludin and claudin-5 by 32% and 43%, respectively. A correlation was also established between the enhanced association of ß-catenin with ZO-1 as a function of GSK3ß inhibition. Collectively, our findings suggest the possibility of using GSK3ß inhibitors as a means to extend the half-life of key tight junction proteins to promote re-sealing of the BBB during neuroinflammation.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Claudina-5/metabolismo , Células Endoteliales/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Ocludina/metabolismo , Uniones Estrechas/metabolismo , Benzazepinas/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Células Cultivadas , Impedancia Eléctrica , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Inhibidores Enzimáticos/farmacología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Humanos , Indoles/farmacología , Cloruro de Litio/farmacología , Uniones Estrechas/efectos de los fármacos , Proteína de la Zonula Occludens-1/metabolismo
7.
Am J Pathol ; 181(4): 1414-25, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22863953

RESUMEN

Glycogen synthase kinase (GSK) 3ß has been identified as a regulator of immune responses. We demonstrated previously that GSK3ß inhibition in human brain microvascular endothelial cells (BMVECs) reduced monocyte adhesion/migration across BMVEC monolayers. Herein, we tested the idea that GSK3ß inhibition in monocytes can diminish their ability to engage the brain endothelium and migrate across the blood-brain barrier. Pretreatment of primary monocytes with GSK3ß inhibitors resulted in a decrease in adhesion (60%) and migration (85%), with similar results in U937 monocytic cells. Monocyte-BMVEC interactions resulted in diminished barrier integrity that was reversed by GSK3ß suppression in monocytic cells. Because integrins mediate monocyte rolling/adhesion, we detected the active conformational form of very late antigen 4 after stimulation with a peptide mimicking monocyte engagement by vascular cell adhesion molecule-1. Peptide stimulation resulted in a 14- to 20-fold up-regulation of the active form of integrin in monocytes that was suppressed by GSK3ß inhibitors (40% to 60%). Because small GTPases, such as Rac1, control leukocyte movement, we measured active Rac1 after monocyte activation with relevant stimuli. Stimulation enhanced the level of active Rac1 that was diminished by GSK3ß inhibitors. Monocytes treated with GSK3ß inhibitors showed increased levels of inhibitory sites of the actin-binding protein, cofilin, and vasodilator-stimulated phosphoprotein-regulating conformational changes of integrins. These results indicate that GSK3ß inhibition in monocytes affects active integrin expression, cytoskeleton rearrangement, and adhesion via suppression of Rac1-diminishing inflammatory leukocyte responses.


Asunto(s)
Movimiento Celular , Regulación hacia Abajo , Células Endoteliales/patología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Integrina alfa4beta1/química , Monocitos/patología , Proteína de Unión al GTP rac1/metabolismo , Complejo SIDA Demencia/patología , Factores Despolimerizantes de la Actina/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Encéfalo/patología , Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Impedancia Eléctrica , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Activación Enzimática/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Integrina alfa4beta1/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Proteínas de Microfilamentos/metabolismo , Monocitos/efectos de los fármacos , Monocitos/enzimología , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Migración Transendotelial y Transepitelial/efectos de los fármacos
8.
J Neurosci ; 32(12): 4004-16, 2012 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-22442067

RESUMEN

Previous studies have shown that modulation of the receptor-mediated cannabinoid system during neuroinflammation can produce potent neuroprotective and anti-inflammatory effects. However, in this context, little is known about how selective activation of the cannabinoid type-2 receptor (CB2R) affects the activated state of the brain endothelium and blood-brain barrier (BBB) function. Using human brain tissues and primary human brain microvascular endothelial cells (BMVECs), we demonstrate that the CB2R is highly upregulated during inflammatory insult. We then examined whether the CB2R agonists could attenuate inflammatory responses at the BBB using a mouse model of LPS-induced encephalitis and highly selective CB2R agonists. Visualization by intravital microscopy revealed that administration of JWH133 [(6aR,10aR)-3-(1,1-dimethylbutyl)-6a,7,10,10a-tetrahydro-6,6,9-trimethyl-6H-dibenzo[b,d]pyran] or a novel resorcinol-based compound, O-1966 (1-[4-(1,1-dimethyl-heptyl)-2,6-dimethoxy-phenyl]-3-methyl-cyclohexanol), greatly attenuated leukocyte adhesion in surface pial vessels and in deep ascending cortical postcapillary venules. BBB permeability assessments with small and large fluorescent tracers showed that CB2R agonists were effective at preventing barrier leakiness after LPS administration. To determine whether the effects by CB2R agonists on barrier protection are not only due to the CB2R modulation of immune cell function, we tested the agonists in vitro with barrier-forming primary BMVECs. Remarkably, the addition of CB2R agonist increased transendothelial electrical resistance and increased the amount of tight junction protein present in membrane fractions. Furthermore, CB2R agonists decreased the induction of intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 surface expression in BMVECs exposed to various proinflammatory mediators. Together, these results suggest that pharmacological CB2R ligands offer a new strategy for BBB protection during neuroinflammation.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Encefalitis/patología , Células Endoteliales/fisiología , Leucocitos/fisiología , Receptor Cannabinoide CB2/metabolismo , Animales , Anisoles/farmacología , Anisoles/uso terapéutico , Barrera Hematoencefálica/efectos de los fármacos , Canfanos/farmacología , Cannabinoides/farmacología , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Células Cultivadas , Ciclohexanoles , Dextranos/metabolismo , Modelos Animales de Enfermedad , Impedancia Eléctrica , Encefalitis/inducido químicamente , Endotelio/metabolismo , Citometría de Flujo , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Lipopolisacáridos/efectos adversos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfoproteínas/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Pirazoles/farmacología , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/antagonistas & inhibidores , Receptor Cannabinoide CB2/deficiencia , Estadísticas no Paramétricas , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/metabolismo , Proteína de la Zonula Occludens-1
9.
PLoS One ; 6(1): e16148, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21249135

RESUMEN

The HIV-1 protein, Tat has been implicated in AIDS pathogenesis however, the amount of circulating Tat is believed to be very low and its quantification has been difficult. We performed the quantification of Tat released from infected cells and taken up by neurons using high performance capillary electrophoresis. This is the first report to successfully measure the amount of Tat in neurons and places Tat as a key player involved in HIV-associated neurocognitive disorders.


Asunto(s)
Electroforesis Capilar/métodos , Productos del Gen tat/análisis , VIH-1/química , Neuronas/virología , Complejo SIDA Demencia/etiología , Comunicación Celular , Línea Celular , Productos del Gen tat/sangre , Humanos , Neuronas/química
10.
J Immunol ; 185(5): 2867-76, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20668216

RESUMEN

Methamphetamine (METH) abuse is known to be associated with an inordinate rate of infections. Although many studies have described the association of METH exposure and immunosuppression, so far the underlying mechanism still remains elusive. In this study, we present evidence that METH exposure resulted in mitochondrial oxidative damage and caused dysfunction of primary human T cells. METH treatment of T lymphocytes led to a rise in intracellular calcium levels that enhanced the generation of reactive oxygen species. TCR-CD28 linked calcium mobilization and subsequent uptake by mitochondria in METH-treated T cells correlated with an increase in mitochondrion-derived superoxide. Exposure to METH-induced mitochondrial dysfunction in the form of marked decrease in mitochondrial membrane potential, increased mitochondrial mass, enhanced protein nitrosylation and diminished protein levels of complexes I, III, and IV of the electron transport chain. These changes paralleled reduced IL-2 secretion and T cell proliferative responses after TCR-CD28 stimulation indicating impaired T cell function. Furthermore, antioxidants attenuated METH-induced mitochondrial damage by preserving the protein levels of mitochondrial complexes I, III, and IV. Altogether, our data indicate that METH can cause T cell dysfunction via induction of oxidative stress and mitochondrial injury as underlying mechanism of immune impairment secondary to METH abuse.


Asunto(s)
Inmunosupresores/toxicidad , Metanfetamina/toxicidad , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/inmunología , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/inmunología , Células Cultivadas , Estimulantes del Sistema Nervioso Central/toxicidad , Citosol/efectos de los fármacos , Citosol/inmunología , Citosol/metabolismo , Relación Dosis-Respuesta Inmunológica , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/inmunología , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/inmunología , Microscopía Fluorescente , Mitocondrias/patología , Especies Reactivas de Oxígeno/metabolismo , Subgrupos de Linfocitos T/patología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
11.
J Neurosci ; 30(28): 9454-64, 2010 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-20631174

RESUMEN

Human immunodeficiency virus 1 (HIV-1) infection may result in activation of peripheral monocytes followed by their infiltration into the CNS, where the release of proinflammatory mediators causes neurologic disease. Previously, we detected high levels of soluble CD40 ligand (CD40L) in CSF and plasma of HIV-infected patients with cognitive impairment. We now show that CD40, a receptor for CD40L, is highly expressed in brain endothelial cells of patients affected by HIV-1 encephalitis (HIVE), suggesting an important role for the CD40/CD40L dyad in regulating blood-brain barrier (BBB) functions. This concept was further supported by in vitro experiments. Exposure of primary human brain microvascular endothelial cells (BMVECs) to CD40L upregulated the expression of adhesion molecules intracellular adhesion molecule-1 and vascular cell adhesion molecule-1, which caused a fourfold increase in monocyte adhesion to BMVECs and stimulated migration across an in vitro BBB model. Investigations into the intracellular signaling pathways that govern these events revealed that cJUN-N-terminal kinase (JNK) is critical to CD40 activation in the BMVECs. CD40L induced activation of mixed-lineage-kinase-3 and JNK, leading to the subsequent activation of cJUN/AP-1 (activating-protein-1). JNK inhibition in the BMVECs prevented CD40L-mediated induction of adhesion molecules, monocyte adhesion, and transendothelial migration. These new findings support the concept that the CD40/CD40L dyad plays an important role in HIVE neuroinflammation.


Asunto(s)
Complejo SIDA Demencia/inmunología , Barrera Hematoencefálica/inmunología , Antígenos CD40/inmunología , Inflamación/inmunología , Proteínas Quinasas JNK Activadas por Mitógenos/inmunología , Complejo SIDA Demencia/metabolismo , Adulto , Análisis de Varianza , Barrera Hematoencefálica/metabolismo , Western Blotting , Antígenos CD40/metabolismo , Ligando de CD40/inmunología , Ligando de CD40/metabolismo , Ligando de CD40/farmacología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/inmunología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , VIH-1/inmunología , VIH-1/metabolismo , Humanos , Inflamación/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Molécula 1 de Adhesión Celular Vascular/inmunología , Molécula 1 de Adhesión Celular Vascular/metabolismo
12.
Am J Pathol ; 176(2): 881-92, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20056834

RESUMEN

Immune mediators and leukocyte engagement of brain microvascular endothelial cells (BMVECs) contribute to blood-brain barrier impairment during neuroinflammation. Glycogen synthase kinase 3beta (GSK3beta) was recently identified as a potent regulator of immune responses in in vitro systems and animal models. However, the role of GSK3beta in regulation of immune endothelial functions remains undetermined. Here we evaluated the effect of GSK3beta inhibition on the regulation of inflammatory responses in BMVECs. A focused PCR gene array of 84 genes was performed to identify the cytokine and chemokine gene expression profile in tumor necrosis factor (TNF) alpha-stimulated BMVECs after GSK3beta inactivation by specific inhibitors. Fifteen of 39 genes induced by TNFalpha stimulation were down-regulated after GSK3beta inhibition. Genes known to contribute to neuroinflammation that were most negatively affected by GSK3beta inactivation included IP-10/CXCL10, MCP-1/CCL2, IL-8/CXCL8, RANTES/CCL5, and Groalpha/CXCL1. GSK3beta suppression resulted in diminished secretion of these proinflammatory mediators by inflamed BMVECs detected by ELISA. GSK3beta inhibition in BMVECs reduced adhesion molecule expression as well as monocyte adhesion to and migration across cytokine stimulated BMVEC monolayers. Interactions of monocytes with TNFalpha-activated BMVECs led to barrier disruption, and GSK3beta suppression in the endothelium restored barrier integrity. GSK3beta inhibition in vivo substantially decreased leukocyte adhesion to brain endothelium under inflammatory conditions. In summary, inhibition of GSK3beta emerges as an important target for stabilization of the blood-brain barrier in neuroinflammation.


Asunto(s)
Encéfalo/efectos de los fármacos , Encefalitis/patología , Células Endoteliales/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Complejo SIDA Demencia/tratamiento farmacológico , Complejo SIDA Demencia/metabolismo , Complejo SIDA Demencia/patología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Encéfalo/irrigación sanguínea , Encéfalo/patología , Estudios de Casos y Controles , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Evaluación Preclínica de Medicamentos , Encefalitis/prevención & control , Células Endoteliales/inmunología , Glucógeno Sintasa Quinasa 3 beta , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Monocitos/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Tiadiazoles/farmacología , Tiadiazoles/uso terapéutico , Tiazoles/farmacología , Tiazoles/uso terapéutico , Urea/análogos & derivados , Urea/farmacología , Urea/uso terapéutico
13.
J Cereb Blood Flow Metab ; 29(12): 1933-45, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19654589

RESUMEN

Methamphetamine (METH), a potent stimulant with strong euphoric properties, has a high abuse liability and long-lasting neurotoxic effects. Recent studies in animal models have indicated that METH can induce impairment of the blood-brain barrier (BBB), thus suggesting that some of the neurotoxic effects resulting from METH abuse could be the outcome of barrier disruption. In this study, we provide evidence that METH alters BBB function through direct effects on endothelial cells and explore possible underlying mechanisms leading to endothelial injury. We report that METH increases BBB permeability in vivo, and exposure of primary human brain microvascular endothelial cells (BMVEC) to METH diminishes the tightness of BMVEC monolayers in a dose- and time-dependent manner by decreasing the expression of cell membrane-associated tight junction (TJ) proteins. These changes were accompanied by the enhanced production of reactive oxygen species, increased monocyte migration across METH-treated endothelial monolayers, and activation of myosin light chain kinase (MLCK) in BMVEC. Antioxidant treatment attenuated or completely reversed all tested aspects of METH-induced BBB dysfunction. Our data suggest that BBB injury is caused by METH-mediated oxidative stress, which activates MLCK and negatively affects the TJ complex. These observations provide a basis for antioxidant protection against brain endothelial injury caused by METH exposure.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/efectos adversos , Células Endoteliales/efectos de los fármacos , Metanfetamina/metabolismo , Estrés Oxidativo , Animales , Antioxidantes/uso terapéutico , Barrera Hematoencefálica/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Estimulantes del Sistema Nervioso Central/metabolismo , Cromanos/uso terapéutico , Células Endoteliales/metabolismo , Células Endoteliales/patología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos NOD , Monocitos/citología , Monocitos/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Uniones Estrechas/patología
14.
J Biol Chem ; 284(17): 11364-73, 2009 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-19204000

RESUMEN

The detection of biomarkers of oxidative stress in brain tissue and cerebrospinal fluid of patients with human immunodeficiency virus, type 1 (HIV)-associated dementia indicates the involvement of stress pathways in the neuropathogenesis of AIDS. Although the biological importance of oxidative stress on events involved in AIDS neuropathogenesis and the HIV-1 proteins responsible for oxidative stress remain to be elucidated, our results point to the activation of hypoxia-inducible factor 1 (HIF-1) upon HIV-1 infection and its elevation in brain cells of AIDS patients with dementia. HIF-1 is a transcription factor that is responsive to oxygen. Under hypoxic conditions, HIF-1alpha becomes stable and translocates to the nucleus where it dimerizes with aryl hydrocarbon receptor nuclear translocator and modulates gene transcription. Activation of HIF-1 can also be mediated by the HIV-1 accessory protein Vpr. In addition, cellular components, including reactive oxygen species, contribute to the induction of HIF-1alpha. Our results show that Vpr induces reactive oxygen species by increasing H(2)O(2) production, which can contribute to HIF-1alpha accumulation. Interestingly, increased levels of HIF-1alpha stimulated HIV-1 gene transcription through HIF-1 association with HIV-1 long terminal repeat. These observations point to the existence of a positive feedback interplay between HIF-1alpha and Vpr and that, by inducing oxidative stress via activation of HIF-1, Vpr can induce HIV-1 gene expression and dysregulate multiple host cellular pathways.


Asunto(s)
Regulación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo , Encéfalo/metabolismo , Línea Celular , Dimerización , Células HeLa , Humanos , Peróxido de Hidrógeno/metabolismo , Microglía/metabolismo , Modelos Biológicos , Estrés Oxidativo , Regiones Promotoras Genéticas , Interferencia de ARN , Especies Reactivas de Oxígeno
15.
J Neuroimmune Pharmacol ; 4(1): 140-9, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19023660

RESUMEN

The cytokine tumor necrosis factor alpha (TNFalpha) is a key factor in several inflammatory diseases and its levels increase in response to a variety of internal or external stimuli. The regulation of the TNFalpha promoter is mediated by several transcription factors including the nuclear factor kappa B protein (NF-kappaB). This study examines the role of NF-kappaB in the regulation of TNFalpha production by morphine in microglia. Using reverse transcriptase polymerase chain reaction, we demonstrated the presence of morphine receptors in these cells. We next demonstrated the ability of morphine to promote TNFalpha production and secretion by these cells using a cytokine array assay. Transient transfection experiments led to the identification of the region located between nucleotides -751 and -615 within the TNFalpha promoter as being responsive to morphine treatment. The DNA sequence of this region contains a motif indicative of a potential NF-kappaB binding site. The use of a small interfering RNA directed against p65, a subunit of NF-kappaB, demonstrated that TNFalpha induction by morphine is NF-kappaB-dependent. All of the effects of morphine were reversed by the morphine inhibitor, naloxone. These data provide important insights into the effects of morphine on microglia.


Asunto(s)
Analgésicos Opioides/farmacología , Morfina/farmacología , FN-kappa B/fisiología , Neuroglía/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Analgésicos Opioides/antagonistas & inhibidores , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Western Blotting , Células Cultivadas , Infecciones por VIH/virología , VIH-1 , Humanos , Microglía/efectos de los fármacos , Microglía/metabolismo , Morfina/antagonistas & inhibidores , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Neuroglía/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Células U937
16.
Cell Cycle ; 7(17): 2682-90, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18719392

RESUMEN

HIV-associated dementia (HAD) is the most common AIDS-associated neurological disorder and is characterized by the development of synaptodendritic injury to neurons. To advance HAD therapy, it is crucial to identify the mechanisms and factors involved. The viral protein HIV-1 Tat is among those factors and is released by HIV-1-infected cells and can be taken up by adjacent neuronal cells leading to neurotoxic effects. Multiple cellular host proteins have been identified as Tat cofactors in causing neuronal injury. Interestingly, most of these factors function through activation of the p53 pathway. We have now examined the ability of Tat to activate the p53 pathway leading to the induction of endogenous p53 and p73 in neuronal cells. We found that Tat induced p53 and p73 levels in SH-SY5Y cells and that this induction caused retraction of neurites. In the absence of either p53 or p73, Tat failed to induce dendritic retraction or to activate the proapoptotic proteins, such as Bax. Further, we found that p53-accumulation in Tat-treated cells depends on the presence of p73. Therefore, we conclude that Tat contributes to neuronal degeneration through activation of a pathway involving p53 and p73. This information will be valuable for the development of therapeutic agents that affect these pathways to protect CNS neurons and prevent HAD.


Asunto(s)
Complejo SIDA Demencia/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Complejo SIDA Demencia/patología , Apoptosis , Proteínas Reguladoras de la Apoptosis , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/virología , Línea Celular Tumoral , VIH-1/fisiología , Humanos , Modelos Biológicos , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Neuronas/metabolismo , Neuronas/patología , Fosforilación , Fosfoserina/metabolismo , Transporte de Proteínas , Fracciones Subcelulares/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Regulación hacia Arriba , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo
17.
Lab Invest ; 88(2): 98-111, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18059365

RESUMEN

The growth factor hepatocyte growth factor (HGF), also known as scatter factor, and its tyrosine kinase receptor c-Met play important roles in medulloblastoma malignancy. The transcription factor c-Myc is another contributor to the malignancy of these most common pediatric brain tumors. In the present study, we observed strong morphological similarities between medulloblastoma xenografts overexpressing HGF and medulloblastoma xenografts overexpressing c-Myc. We therefore hypothesized a biologically significant link between HGF/c-Met and c-Myc in medulloblastoma malignancy and studied the molecular and functional interactions between them. We found that HGF induces c-Myc mRNA and protein in established and primary medulloblastoma cells. HGF regulated c-Myc levels via transcriptional and post-transcriptional mechanisms as evidenced by HGF induction of c-Myc promoter activity and induction of c-Myc protein levels in the setting of inhibited transcription and translation. We also found that HGF induces cell cycle progression, cell proliferation, apoptosis and increase in cell size in a c-Myc-dependent manner. Activation of MAPK and PI3K, inhibition of GSK-3beta and translocation of beta-catenin to the nucleus as well as Tcf/Lef transcriptional activity were involved in mediating c-Myc induction by HGF. Induction of Cdk2 kinase activity was involved in mediating the cell cycle progression effects, and downregulation of Bcl-XL was involved in mediating the proapoptotic effects of HGF downstream of c-Myc. All molecules that mediated the effects of HGF on c-Myc expression, cell proliferation and apoptosis were expressed in human large-cell medulloblastoma tissues. We therefore established for the first time a functional cooperation between HGF/c-Met and c-Myc in human medulloblastoma and elucidated the molecular mechanisms of this cooperation. The findings provide a potential explanation for the high frequency of c-Myc overexpression in medulloblastoma and suggest a cooperative role for c-Met and c-Myc in large-cell anaplastic medulloblastoma formation.


Asunto(s)
Neoplasias Cerebelosas/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Meduloblastoma/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Apoptosis/fisiología , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Ciclo Celular/fisiología , Aumento de la Célula , Línea Celular Tumoral , Proliferación Celular , Neoplasias Cerebelosas/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Meduloblastoma/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Trasplante Heterólogo , Proteína bcl-X/metabolismo , beta Catenina/metabolismo
18.
J Mol Biol ; 366(1): 67-81, 2007 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-17157319

RESUMEN

The interaction of human immunodeficiency virus type 1 (HIV-1) with CD4+ T lymphocytes is well studied and typically results in virally induced cytolysis. In contrast, relatively little is known concerning the interplay between HIV-1 and microglia. Recent findings suggest that, counter-intuitively, HIV-1 infection may extend the lifespan of microglia. We developed a novel cell line model system to confirm and mechanistically study this phenomenon. We found that transduction of a human microglial cell line with an HIV-1 vector results in a powerful cytoprotective effect following apoptotic challenge. This effect was reproduced by ectopic expression of a single virus-encoded protein, Tat. Subsequent studies showed that the pro-survival effects of intracellular Tat could be attributed to activation of the PI-3-kinase (PI3K)/Akt pathway in the microglial cell line. Furthermore, we found that expression of Tat led to decreased expression of PTEN, a negative regulator of the PI-3-K pathway. Consistent with this, decreased p53 activity and increased E2F activity were observed. Based on these findings, a model of possible regulatory circuits that intracellular Tat and HIV-1 infection engage during the cytoprotective event in microglia has been suggested. We propose that the expression of Tat may enable HIV-1 infected microglia to survive throughout the course of infection, leading to persistent HIV-1 production and infection in the central nervous system.


Asunto(s)
Productos del Gen tat/fisiología , VIH-1/fisiología , Microglía/virología , Línea Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Productos del Gen tat/metabolismo , VIH-1/genética , Humanos , Macrófagos/fisiología , Macrófagos/virología , Microglía/citología , Microglía/fisiología , Modelos Biológicos , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción Genética , Transfección , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
19.
Vaccine ; 25(9): 1634-46, 2007 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-17145123

RESUMEN

Helper-free herpes simplex virus type-1 (HSV-1) amplicon vectors elicit robust immune responses to encoded proteins, including human immunodeficiency virus type-1 (HIV-1) antigens. To improve this vaccine delivery system, seven amplicon vectors were constructed, each encoding HIV-1 Gag under the control of a different promoter. Gag expression levels were analyzed in murine and human cell lines, as well as in biopsied tissue samples from injected mice; these data were then compared with Gag-specific T cell responses in BALB/c mice. The magnitude of the amplicon-induced immune response was found to correlate strongly with the level of Gag production both in vitro and in vivo. Interestingly, the best correlation of the strength of the amplicon-induced immune response was with antigen expression in cultured DC rather than expression at the tissue site of injection or in cultured cell lines. These findings may have implications for the generation of improved HSV-1 amplicon vectors for HIV-1 vaccine delivery.


Asunto(s)
Regulación Viral de la Expresión Génica , Productos del Gen gag/inmunología , Productos del Gen gag/metabolismo , Vectores Genéticos , VIH-1/genética , Herpesvirus Humano 1/genética , Regiones Promotoras Genéticas , Células 3T3 , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/inmunología , Animales , Línea Celular , Células Cultivadas , Células Dendríticas/metabolismo , Femenino , Productos del Gen gag/genética , Genes gag , VIH-1/metabolismo , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Linfocitos T/inmunología , Transcripción Genética
20.
Eur J Neurosci ; 23(10): 2623-34, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16817865

RESUMEN

The pathogenesis of human immunodeficiency virus type 1 (HIV-1)-associated dementia is mediated by neuronal dysfunction and death, brought about by the action of soluble neurotoxic factors that are released by virally infected macrophages and microglia. Paradoxically, many candidate HIV-1 neurotoxins also possess the ability to activate nuclear factor-kappa B (NF-kappaB), which has a potent pro-survival effect in primary neurons. The present study explored this conundrum and investigated why NF-kappaB might fail to protect neurons that are exposed to candidate HIV-1 neurotoxins. Here, we evaluated the ability of virus-depleted conditioned medium produced by HIV-1-infected human macrophages (HIV-MCMs) to modulate NF-kappaB activity in neurons. We demonstrated that HIV-MCMs inhibit the normal signaling pathways that lead to NF-kappaB activation in neurons. This inhibitory effect of HIV-MCM is dependent upon the presence of HIV-1 Tat, which activates glycogen synthase kinase (GSK)-3beta in neurons. Activation of GSK-3beta, in turn, results in modification of the NF-kappaB subunit RelA at serine 468, thereby regulating the physical interaction of RelA with histone deacetylase-3 corepressor molecules. Furthermore, neutralization of Tat or inhibition of GSK-3beta activity prevents neuronal apoptosis induced by HIV-MCM. We conclude that HIV-1 Tat may compromise neuronal function and fate by interfering with normal survival pathways subserved by NF-kappaB. These findings may have important therapeutic implications for the management of HIV-1-associated dementia.


Asunto(s)
Productos del Gen tat/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , VIH-1/metabolismo , FN-kappa B/antagonistas & inhibidores , Neuronas/metabolismo , Transducción de Señal/fisiología , Complejo SIDA Demencia/metabolismo , Complejo SIDA Demencia/virología , Animales , Apoptosis/fisiología , Células Cultivadas , Medios de Cultivo Condicionados , Activación Enzimática/fisiología , Glucógeno Sintasa Quinasa 3 beta , Histona Desacetilasas/metabolismo , Humanos , Immunoblotting , Inmunoprecipitación , Etiquetado Corte-Fin in Situ , Macrófagos/metabolismo , Macrófagos/virología , Neuronas/patología , Ratas , Factor de Transcripción ReIA/metabolismo , Transfección , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...