Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Pulm Circ ; 8(3): 2045894018778156, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29718758

RESUMEN

Pulmonary arterial (PA) wall modifications are key pathological features of pulmonary hypertension (PH). Although such abnormalities correlate with heightened phosphorylation of c-Jun N-terminal kinases 1/2 (JNK1/2) in a rat model of PH, the contribution of specific JNK isoforms to the pathophysiology of PH is unknown. Hence, we hypothesized that activation of either one, or both JNK isoforms regulates PA remodeling in PH. We detected increased JNK1/2 phosphorylation in the thickened vessels of PH patients' lungs compared to that in lungs of healthy individuals. JNK1/2 phosphorylation paralleled a marked reduction in MAP kinase phosphatase 1 (JNK dephosphorylator) expression in patients' lungs. Association of JNK1/2 activation with vascular modification was confirmed in the calf model of severe hypoxia-induced PH. To ascertain the role of each JNK isoform in pathophysiology of PH, wild-type (WT), JNK1 null (JNK1-/-), and JNK2 null (JNK2-/-) mice were exposed to chronic hypoxia (10% O2 for six weeks) to develop PH. In hypoxic WT lungs, an increase in JNK1/2 phosphorylation was associated with PH-like pathology. Hallmarks of PH pathophysiology, i.e. excessive accumulation of extracellular matrix and vessel muscularization with medial wall thickening, was also detected in hypoxic JNK1-/- lungs, but not in hypoxia-exposed JNK2-/- lungs. However, hypoxia-induced increases in right ventricular systolic pressure (RVSP) and in right ventricular hypertrophy (RVH) were similar in all three genotypes. Our findings suggest that JNK2 participates in PA remodeling (but likely not in vasoconstriction) in murine hypoxic PH and that modulating JNK2 actions might quell vascular abnormalities and limit the course of PH.

2.
Acta Neuropathol Commun ; 3: 9, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25645462

RESUMEN

BACKGROUND: In rodent models of Parkinson's disease (PD), dopamine neuron loss is accompanied by increased expression of angiotensin II (AngII), its type 1 receptor (AT1), and NADPH oxidase (Nox) in the nigral dopamine neurons and microglia. AT1 blockers (ARBs) stymie such oxidative damage and neuron loss. Whether changes in the AngII/AT1/Nox4 axis contribute to Parkinson neuropathogenesis is unknown. Here, we studied the distribution of AT1 and Nox4 in dopamine neurons in two nigral subregions: the less affected calbindin-rich matrix and the first-affected calbindin-poor nigrosome 1 of three patients, who were clinically asymptomatic, but had nigral dopamine cell loss and Braak stages consistent with a neuropathological diagnosis of PD (prePD). For comparison, five clinically- and neuropathologically-confirmed PD patients and seven age-matched control patients (AMC) were examined. RESULTS: AT1 and Nox4 immunoreactivity was noted in dopamine neurons in both the matrix and the nigrosome 1. The total cellular levels of AT1 in surviving dopamine neurons in the matrix and nigrosome 1 declined from AMC>prePD>PD, suggesting that an AngII/AT1/Nox4 axis orders neurodegenerative progression. In this vein, the loss of dopamine neurons was paralleled by a decline in total AT1 per surviving dopamine neuron. Similarly, AT1 in the nuclei of surviving neurons in the nigral matrix declined with disease progression, i.e., AMC>prePD>PD. In contrast, in nigrosome 1, the expression of nuclear AT1 was unaffected and similar in all groups. The ratio of nuclear AT1 to total AT1 (nuclear + cytoplasmic + membrane) in dopamine neurons increased stepwise from AMC to prePD to PD. The proportional increase in nuclear AT1 in dopamine neurons in nigrosome 1 of prePD and PD patients was accompanied by elevated nuclear expression of Nox4, oxidative damage to DNA, and caspase-3-mediated cell loss. CONCLUSIONS: Our observations are consistent with the idea that AngII/AT1/Nox4 axis-mediated oxidative stress gives rise to the dopamine neuron dysfunction and loss characteristic of the neuropathological and clinical manifestations of PD and suggest that the chance for a neuron to survive increases in association with lower total as well as nuclear AT1 expression. Our results support the need for further evaluation of ARBs as disease-modifying agents in PD.


Asunto(s)
Angiotensina II/metabolismo , Caspasa 3/metabolismo , Neuronas Dopaminérgicas/metabolismo , Guanosina/metabolismo , NADPH Oxidasas/metabolismo , Enfermedad de Parkinson/patología , Receptor de Angiotensina Tipo 1/metabolismo , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Estrés Oxidativo , Enfermedad de Parkinson/metabolismo , Sustancia Negra/metabolismo
3.
Am J Physiol Cell Physiol ; 306(7): C648-58, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24500281

RESUMEN

Epithelial cells are key players in the pathobiology of numerous hypoxia-induced lung diseases. The mechanisms mediating such hypoxic responses of epithelial cells are not well characterized. Earlier studies reported that hypoxia stimulates protein kinase C (PKC)δ activation in renal cancer cells and an increase in expression of a heparin-binding growth factor, midkine (MK), in lung alveolar epithelial cells. We reasoned that hypoxia might regulate MK levels via a PKCδ-dependent pathway and hypothesized that PKCδ-driven MK expression is required for hypoxia-induced lung epithelial cell proliferation and differentiation. Replication of human lung epithelial cells (A549) was significantly increased by chronic hypoxia (1% O2) and was dependent on expression of PKCδ. Hypoxia-induced proliferation of epithelial cells was accompanied by translocation of PKCδ from Golgi into the nuclei. Marked attenuation in MK protein levels by rottlerin, a pharmacological antagonist of PKC, and by small interfering RNA-targeting PKCδ, revealed that PKCδ is required for MK expression in both normoxic and hypoxic lung epithelial cells. Sequestering MK secreted into the culture media with a neutralizing antibody reduced hypoxia-induced proliferation demonstrating that an increase in MK release from cells is linked with epithelial cell division under hypoxia. In addition, recombinant MK accelerated transition of hypoxic epithelial cells to cells of mesenchymal phenotype characterized by elongated morphology and increased expression of mesenchymal markers, α-smooth muscle actin, and vimentin. We conclude that PKCδ/MK axis mediates hypoxic proliferation and differentiation of lung epithelial cells. Manipulation of PKCδ and MK activity in epithelial cells might be beneficial for the treatment of hypoxia-mediated lung diseases.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Células Epiteliales/enzimología , Pulmón/enzimología , Factores de Crecimiento Nervioso/metabolismo , Proteína Quinasa C-delta/metabolismo , Actinas/metabolismo , Transporte Activo de Núcleo Celular , Anticuerpos Neutralizantes/farmacología , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Hipoxia de la Célula , Línea Celular Tumoral , Núcleo Celular/enzimología , Proliferación Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Transición Epitelial-Mesenquimal , Aparato de Golgi/enzimología , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Midkina , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Factores de Crecimiento Nervioso/inmunología , Fenotipo , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/genética , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , Proteínas Recombinantes/metabolismo , Transducción de Señal , Factores de Tiempo , Transfección , Vimentina/metabolismo
4.
Dev Neurosci ; 35(4): 293-305, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23751520

RESUMEN

Reactive oxygen species (ROS) have been reported to affect neural stem cell self-renewal and therefore may be important for normal development and may influence neurodegenerative processes when ROS activity is elevated. To determine if increasing production of superoxide, via activation of NADPH oxidase (Nox), increases neural stem cell proliferation, 100 nM angiotensin II (Ang II) - a strong stimulator of Nox - was applied to cultures of a murine neural stem cell line, C17.2. Twelve hours following a single treatment with Ang II, there was a doubling of the number of neural stem cells. This increase in neural stem cell numbers was preceded by a gradual elevation of superoxide levels (detected by dihydroethidium fluorescence) from the steady state at 0, 5, and 30 min and gradually increasing from 1 h to the maximum at 12 h, and returning to baseline at 24 h. Ang II-dependent proliferation was blocked by the antioxidant N-acetyl-L-cysteine. Confocal microscopy revealed the presence of two sources of intracellular ROS in C17.2 cells: (i) mitochondrial and (ii) extramitochondrial; the latter indicative of the involvement of one or more specific isoforms of Nox. Of the Nox family, mRNA expression for one member, Nox4, is abundant in neural stem cell cultures, and Ang II treatment resulted in elevation of the relative levels of Nox4 protein. SiRNA targeting of Nox4 mRNA reduced both the constitutive and Ang II-induced Nox4 protein levels and attenuated Ang II-driven increases in superoxide levels and stem cell proliferation. Our findings are consistent with our hypothesis that Ang II-induced proliferation of neural stem cells occurs via Nox4-generated superoxide, suggesting that an Ang II/Nox4 axis is an important regulator of neural stem cell self-renewal and as such may fine-tune normal, stress- or disease-modifying neurogenesis.


Asunto(s)
Angiotensina II/farmacología , Proliferación Celular/efectos de los fármacos , NADPH Oxidasas/metabolismo , Células-Madre Neurales/efectos de los fármacos , Superóxidos/metabolismo , Animales , Western Blotting , Recuento de Células , Células Cultivadas , Interpretación Estadística de Datos , Ratones , Microscopía Confocal , NADPH Oxidasa 4 , NADPH Oxidasas/genética , Células-Madre Neurales/ultraestructura , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
5.
Cardiovasc Res ; 95(3): 356-65, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22735370

RESUMEN

AIMS: Pulmonary hypertension (PH) is a devastating condition for which no disease-modifying therapies exist. PH is recognized as proliferative disease of the pulmonary artery (PA). In the experimental newborn calf model of hypoxia-induced PH, adventitial fibroblasts in the PA wall exhibit a heightened replication index. Because elevated platelet-derived growth factor ß receptor (PDGFß-R) signalling is associated with PH, we tested the hypothesis that the activation of PDGFß-R contributes to fibroblast proliferation and adventitial remodelling in PH. METHODS AND RESULTS: Newborn calves were exposed to either ambient air (P(B) = 640 mmHg) (Neo-C) or high altitude (P(B) = 445 mm Hg) (Neo-PH) for 2 weeks. PDGFß-R phosphorylation was markedly elevated in PA adventitia of Neo-PH calves as well as in cultured PA fibroblasts isolated from Neo-PH animals. PDGFß-R activation with PDGF-BB stimulated higher replication in Neo-PH cells compared with that of control fibroblasts. PDGF-BB-induced proliferation was dependent on reactive oxygen species generation and extracellular signal-regulated kinase1/2 activation in both cell populations; however, only Neo-PH cell division via PDGFß-R activation displayed a unique dependence on c-Jun N-terminal kinase1 (JNK1) stimulation as the blockade of JNK1 with SP600125, a pharmacological antagonist of the JNK pathway, and JNK1-targeted siRNA selectively blunted Neo-PH cell proliferation. CONCLUSIONS: Our data strongly suggest that hypoxia-induced modified cells engage the PDGFß-R-JNK1 axis to confer distinctively heightened proliferation and adventitial remodelling in PH.


Asunto(s)
Adventicia/enzimología , Proliferación Celular , Fibroblastos/enzimología , Hipertensión Pulmonar/enzimología , Hipoxia/enzimología , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Arteria Pulmonar/enzimología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Adventicia/patología , Altitud , Animales , Animales Recién Nacidos , Becaplermina , Bovinos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Activación Enzimática , Hipertensión Pulmonar Primaria Familiar , Fibroblastos/patología , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Hipoxia/etiología , Hipoxia/genética , Hipoxia/patología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 8 Activada por Mitógenos/genética , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-sis/metabolismo , Arteria Pulmonar/patología , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección
6.
J Neuroinflammation ; 8: 129, 2011 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-21975039

RESUMEN

BACKGROUND: Reactive oxygen species (ROS), superoxide and hydrogen peroxide (H2O2), are necessary for appropriate responses to immune challenges. In the brain, excess superoxide production predicts neuronal cell loss, suggesting that Parkinson's disease (PD) with its wholesale death of dopaminergic neurons in substantia nigra pars compacta (nigra) may be a case in point. Although microglial NADPH oxidase-produced superoxide contributes to dopaminergic neuron death in an MPTP mouse model of PD, this is secondary to an initial die off of such neurons, suggesting that the initial MPTP-induced death of neurons may be via activation of NADPH oxidase in neurons themselves, thus providing an early therapeutic target. METHODS: NADPH oxidase subunits were visualized in adult mouse nigra neurons and in N27 rat dopaminergic cells by immunofluorescence. NADPH oxidase subunits in N27 cell cultures were detected by immunoblots and RT-PCR. Superoxide was measured by flow cytometric detection of H2O2-induced carboxy-H2-DCFDA fluorescence. Cells were treated with MPP+ (MPTP metabolite) following siRNA silencing of the Nox2-stabilizing subunit p22phox, or simultaneously with NADPH oxidase pharmacological inhibitors or with losartan to antagonize angiotensin II type 1 receptor-induced NADPH oxidase activation. RESULTS: Nigral dopaminergic neurons in situ expressed three subunits necessary for NADPH oxidase activation, and these as well as several other NADPH oxidase subunits and their encoding mRNAs were detected in unstimulated N27 cells. Overnight MPP+ treatment of N27 cells induced Nox2 protein and superoxide generation, which was counteracted by NADPH oxidase inhibitors, by siRNA silencing of p22phox, or losartan. A two-wave ROS cascade was identified: 1) as a first wave, mitochondrial H2O2 production was first noted at three hours of MPP+ treatment; and 2) as a second wave, H2O2 levels were further increased by 24 hours. This second wave was eliminated by pharmacological inhibitors and a blocker of protein synthesis. CONCLUSIONS: A two-wave cascade of ROS production is active in nigral dopaminergic neurons in response to neurotoxicity-induced superoxide. Our findings allow us to conclude that superoxide generated by NADPH oxidase present in nigral neurons contributes to the loss of such neurons in PD. Losartan suppression of nigral-cell superoxide production suggests that angiotensin receptor blockers have potential as PD preventatives.


Asunto(s)
1-Metil-4-fenilpiridinio/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Línea Celular , Neuronas Dopaminérgicas/citología , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/metabolismo , Femenino , Herbicidas/farmacología , Peróxido de Hidrógeno/farmacología , Losartán/farmacología , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasas/química , NADPH Oxidasas/genética , Oxidantes/farmacología , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Sustancia Negra/citología
7.
Am J Pathol ; 178(1): 98-109, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21224048

RESUMEN

Although mitogen-activated protein kinase phosphatase-1 (MKP-1) is a key deactivator of MAP kinases, known effectors of lung vessel formation, whether it plays a role in the expression of proangiogenic vascular endothelial growth factor (VEGF) in hypoxic lung is unknown. We therefore hypothesized that MKP-1 is a crucial modulator of hypoxia-stimulated vessel development by regulating lung VEGF levels. Wild-type MKP-1(+/+), heterozygous MKP-1(+/-), and deficient MKP-1(-/-) mice were exposed to sea level (SL), Denver altitude (DA) (1609 m [5280 feet]), and severe high altitude (HYP) (∼5182 m [∼17,000 feet]) for 6 weeks. Hypoxia enhanced phosphorylation of p38 MAP kinase, a substrate of MKP-1, as well as α smooth muscle actin (αSMA) expression in vessels, respiratory epithelium, and interstitium of phosphatase-deficient lung. αSMA-positive vessel (<50 µm outside diameter) densities were markedly reduced, whereas vessel wall thickness was increased in hypoxic MKP-1(-/-) lung. Mouse embryonic fibroblasts (MEFs) of all three genotypes were isolated to pinpoint the mechanism involved in hypoxia-induced vascular abnormalities of MKP-1(-/-) lung. Sustained phosphorylation of p38 MAP kinase was observed in MKP-1-null MEFs in response to hypoxia exposure. Although hypoxia up-regulated VEGF levels in MKP-1(+/+) MEFs eightfold, only a 70% increase in VEGF expression was observed in MKP-1-deficient cells. Therefore, our data strongly suggest that MKP-1 might be the key regulator of vascular densities through the regulation of VEGF levels in hypoxic lung.


Asunto(s)
Fosfatasa 1 de Especificidad Dual/fisiología , Hipoxia/enzimología , Pulmón/irrigación sanguínea , Neovascularización Fisiológica , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Actinas/metabolismo , Animales , Fosfatasa 1 de Especificidad Dual/genética , Hipoxia/fisiopatología , Antígeno Ki-67/metabolismo , Pulmón/fisiología , Ratones , Ratones Mutantes , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Stem Cell Res Ther ; 1(5): 36, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21122109

RESUMEN

INTRODUCTION: Regulated neurotransmitter actions in the mammalian central nervous system determine brain function and control peripheral organs and behavior. Although drug-seeking behaviors, including alcohol consumption, depend on central neurotransmission, modification of neurotransmitter actions in specific brain nuclei remains challenging. Herein, we report a novel approach for neurotransmission modification in vivo by transplantation of stem cells engineered to take up the neurotransmitter dopamine (DA) efficiently through the action of the human dopamine transporter (hDAT). As a functional test in mice, we used voluntary alcohol consumption, which is known to release DA in nucleus accumbens (NAC), an event hypothesized to help maintain drug-seeking behavior. We reasoned that reducing extracellular DA levels, by engrafting into NAC DA-sequestering stem cells expressing hDAT, would alter alcohol intake. METHODS: We have generated a neural stem cell line stably expressing the hDAT. Uptake kinetics of DA were determined to select a clone for transplantation. These genetically modified stem cells (or cells transfected with a construct lacking the hDAT sequence) were transplanted bilaterally into the NAC of wild-type mice trained to consume 10% alcohol in a two-bottle free-choice test for alcohol consumption. Alcohol intake was then ascertained for 1 week after transplantation, and brain sections through the NAC were examined for surviving grafted cells. RESULTS: Modified stem cells expressed hDAT and uptaken DA selectively via hDAT. Mice accustomed to drinking 10% ethanol by free choice reduced their alcohol consumption after being transplanted with hDAT-expressing stem cells. By contrast, control stem cells lacked that effect. Histologic examination revealed surviving stem cells in the NAC of all engrafted brains. CONCLUSIONS: Our findings represent proof of principle suggesting that genetically engineered stem cells can be useful for exploring the role of neurotransmitters (or other signaling molecules) in alcohol consumption and potentially in other aspects of brain function.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Comportamiento de Búsqueda de Drogas/fisiología , Células-Madre Neurales/metabolismo , Consumo de Bebidas Alcohólicas/metabolismo , Animales , Células Cultivadas , Dopamina/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/biosíntesis , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/trasplante , Núcleo Accumbens/metabolismo , Ratas , Ratas Sprague-Dawley , Transfección , beta-Galactosidasa/metabolismo
9.
Alcohol Res Health ; 31(3): 256-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-23584871

RESUMEN

One strategy to determine the contribution of individual genes to the development of complex traits and behaviors such as alcohol consumption is to reduce or completely eliminate the activity of those genes in the cells or organism under investigation and then study the effects of this modification. But how can a single specific gene be inactivated? One strategy that has been developed in recent years is the use of small, artificially generated molecules called short-interfering RNAs (siRNAs). This article briefly describes the principles of this strategy and presents some initial results obtained with this approach.


Asunto(s)
Alcoholismo/genética , Encéfalo/fisiología , Silenciador del Gen/fisiología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Alcoholismo/diagnóstico , Alcoholismo/enzimología , Animales , Encéfalo/patología , Humanos , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
10.
Neurochem Res ; 33(5): 886-901, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-17992568

RESUMEN

Among various types of neurons affected in Parkinson's disease, dopamine (DA) neurons of the substantia nigra undergo the most pronounced degeneration. Products of DA oxidation and consequent cellular damage have been hypothesized to contribute to neuronal death. To examine whether elevated intracellular DA will selectively predispose the dopaminergic subpopulation of nigral neurons to damage by an oxidative insult, we first cultured rat primary mesencephalic cells in the presence of rotenone to elevate reactive oxygen species. Although MAP2(+) neurons were more sensitive to rotenone-induced toxicity than type 1 astrocytes, rotenone affected equally both DA (TH(+)) neurons and MAP2(+) neurons. In contrast, when intracellular DA concentration was elevated, DA neurons became selectively sensitized to rotenone. Raising intracellular DA levels in primary DA neurons resulted in dopaminergic neuron death in the presence of subtoxic concentrations of rotenone. Furthermore, mitochondrial superoxide dismutase mimetic, manganese (III) meso-tetrakis (4-benzoic acid) porphyrin, blocked activation of caspase-3, and consequent cell death. Our results demonstrate that an inhibitor of mitochondrial complex I and increased cytosolic DA may cooperatively lead to conditions of elevated oxidative stress and thereby promote selective demise of dopaminergic neurons.


Asunto(s)
Apoptosis/efectos de los fármacos , Dopamina/farmacología , Neuronas/efectos de los fármacos , Rotenona/toxicidad , Animales , Dopamina/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Inmunohistoquímica , Neuronas/metabolismo , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley
11.
Mol Neurodegener ; 2: 1, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17224059

RESUMEN

BACKGROUND: Recent attention has focused on understanding the role of the brain-renin-angiotensin-system (RAS) in stroke and neurodegenerative diseases. Direct evidence of a role for the brain-RAS in Parkinson's disease (PD) comes from studies demonstrating the neuroprotective effect of RAS inhibitors in several neurotoxin based PD models. In this study, we show that an antagonist of the angiotensin II (Ang II) type 1 (AT1) receptor, losartan, protects dopaminergic (DA) neurons against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) toxicity both in primary ventral mesencephalic (VM) cultures as well as in the substantia nigra pars compacta (SNpc) of C57BL/6 mice (Fig. 1). RESULTS: In the presence of exogenous Ang II, losartan reduced MPP+ (5 muM) induced DA neuronal loss by 72% in vitro. Mice challenged with MPTP showed a 62% reduction in the number of DA neurons in the SNpc and a 71% decrease in tyrosine hydroxylase (TH) immunostaining of the striatum, whereas daily treatment with losartan lessened MPTP-induced loss of DA neurons to 25% and reduced the decrease in striatal TH+ immunostaining to 34% of control. CONCLUSION: Our study demonstrates that the brain-RAS plays an important neuroprotective role in the MPTP model of PD and points to AT1 receptor as a potential novel target for neuroprotection.

12.
Brain Res ; 1045(1-2): 64-71, 2005 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-15910763

RESUMEN

In this study, we demonstrate that angiotensin II (Ang II) protects dopamine (DA) neurons from rotenone toxicity in vitro. Primary ventral mesencephalic (VM) cultures from E15 rats were grown for 5 days and then cultured in the presence of the mitochondrial complex I inhibitor, rotenone. Acute exposure (20 h) to 20 nM rotenone reduced the number of tyrosine hydroxylase-positive (TH+) neurons by 50 +/- 6% when compared to untreated cultures. Pre-treatment of VM cultures with 100 nM Ang II decreased TH+ neuronal loss to 25 +/- 10% at the 20-nM rotenone concentration. Ang II in the presence of the angiotensin type 1 receptor (AT1R) antagonist, losartan, was even more effective in protecting DA neurons showing a loss of only 13 +/- 4% at 20 nM rotenone. Conversely, the AT2R antagonist, PD123319, abolished the protective effects of Ang II. Furthermore, both the NMDA receptor antagonist, MK801, and the antioxidant, alpha-tocopheryl succinate (vitamin E analogue), prevented rotenone-induced toxicity. Here, we show that acute exposure of VM cultures to the pesticide rotenone leads to dopaminergic neuronal cell death and that angiotensin acting through the AT2 receptor protects dopamine neurons from rotenone toxicity.


Asunto(s)
Angiotensina II/farmacología , Dopamina/metabolismo , Neuronas/efectos de los fármacos , Rotenona/antagonistas & inhibidores , Sustancia Negra/efectos de los fármacos , Angiotensina II/uso terapéutico , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Antioxidantes/farmacología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Antagonistas de Aminoácidos Excitadores/farmacología , Insecticidas/antagonistas & inhibidores , Losartán/farmacología , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/prevención & control , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Neurotoxinas/antagonistas & inhibidores , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/prevención & control , Ratas , Ratas Sprague-Dawley , Sustancia Negra/patología , Sustancia Negra/fisiopatología , Tirosina 3-Monooxigenasa/metabolismo
13.
J Biol Chem ; 279(29): 30760-70, 2004 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-15128747

RESUMEN

The plasma membrane dopamine transporter (DAT) has an essential role in terminating dopaminergic neurotransmission by reuptake of dopamine into the presynaptic neurons. Therefore, the amount of DAT at the cell surface is a critical determinant of DAT function. In this study, we examined the role of the carboxyl terminus of DAT in trafficking of the transporter through the biosynthetic pathway to the plasma membrane. Live cell fluorescence microscopy and cell surface biotinylation were used to study the effects of systematic deletions and alanine substitutions in the carboxyl terminus on DAT localization. It was found that alanine substitutions of Lys-590 and Asp-600 significantly delayed the delivery of DAT to the plasma membrane because of retention of DAT in the endoplasmic reticulum (ER). Most surprising, mutation of Gly-585 to alanine completely blocked the exit of DAT from the ER and surface expression of the transporter. The effect of these three mutations on ER export of DAT was demonstrated in porcine aortic endothelial cells and the immortalized neuronal cell line 1RB3AN27. In primary cultures of rat embryonic midbrain neurons, DAT G585A, K590A, and D600A mutants were restricted to the cell soma and did not traffic to the dendrites or axonal processes. These data are consistent with the model whereby the local conformation and/or intramolecular interactions of the sequences of the DAT carboxyl terminus proximal to the last transmembrane domain are essential for the ER export of the transporter.


Asunto(s)
Retículo Endoplásmico/metabolismo , Glicoproteínas de Membrana , Proteínas de Transporte de Membrana/química , Proteínas del Tejido Nervioso/química , Alanina/química , Secuencia de Aminoácidos , Animales , Aorta/patología , Ácido Aspártico/química , Proteínas Bacterianas/metabolismo , Biotinilación , Membrana Celular/metabolismo , Células Cultivadas , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática , Endotelio Vascular/metabolismo , Factor de Crecimiento Epidérmico/química , Transferencia Resonante de Energía de Fluorescencia , Glicina/química , Proteínas Luminiscentes/metabolismo , Lisina/química , Microscopía Fluorescente , Modelos Biológicos , Datos de Secuencia Molecular , Mutagénesis , Mutación , Neuronas/metabolismo , Plásmidos/metabolismo , Unión Proteica , Conformación Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Ratas , Porcinos , Transfección
14.
Am J Physiol Cell Physiol ; 286(2): C416-25, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14561589

RESUMEN

Activation of the alpha-smooth muscle actin (alpha-SMA) gene during the conversion of fibroblasts into myofibroblasts is an essential feature of various fibrotic conditions. Microvascular compromise and thus local environmental hypoxia are important components of the fibrotic response. The present study was thus undertaken to test the hypothesis that hypoxia can induce transdifferentiation of vascular fibroblasts into myofibroblasts and also to evaluate potential signaling mechanisms governing this process. We found that hypoxia significantly upregulates alpha-SMA protein levels in bovine pulmonary artery adventitial fibroblasts. Increased alpha-SMA expression is controlled at the transcriptional level because the alpha-SMA gene promoter activity, assayed via a luciferase reporter, was markedly increased in transfected fibroblasts exposed to hypoxia. Hypoxic induction of the alpha-SMA gene was mimicked by overexpression of constitutively active Galphai2 (alphai2Q205L) but not Galpha16 (alpha-16Q212L). Blockade of hypoxia-induced alpha-SMA expression with pertussis toxin, a Galphai antagonist, confirmed a role for Galphai in the hypoxia-induced transdifferentiation process. c-Jun NH2-terminal kinase (JNK) inhibitor II and SB202190, but not U0126, also attenuated alpha-SMA expression in hypoxic fibroblasts, suggesting the importance of JNK in the differentiation process. Hypoxia-induced increase in bromodeoxyuridine incorporation, which occurred concomitantly with hypoxia-induced differentiation, was blocked by U0126, suggesting that DNA synthesis and alpha-SMA expression take place through simultaneously activated parallel signaling pathways. Neutralizing antibody against transforming growth factor-beta1 blocked only 30% of the hypoxia-induced alpha-SMA promoter activity. Taken together, our results suggest that hypoxia induces differentiation of vascular fibroblasts into myofibroblasts by upregulating the expression of alpha-SMA, and this increase in alpha-SMA level occurs through Galphai- and JNK-dependent signaling pathways.


Asunto(s)
Fibroblastos/patología , Hipoxia/patología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Arteria Pulmonar/patología , Actinas/genética , Actinas/metabolismo , Animales , Animales Recién Nacidos , Anticuerpos/farmacología , Bromodesoxiuridina/metabolismo , Bovinos , Diferenciación Celular , Células Cultivadas , Subunidad alfa de la Proteína de Unión al GTP Gi2 , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Liso/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta1 , Regulación hacia Arriba
15.
J Neurochem ; 87(4): 914-21, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14622122

RESUMEN

In vivo, the pesticide rotenone induces degeneration of dopamine neurons and parkinsonian-like pathology in adult rats. In the current study, we utilized primary ventral mesencephalic (VM) cultures from E15 rats as an in vitro model to examine the mechanism underlying rotenone-induced death of dopamine neurons. After 11 h of exposure to 30 nm rotenone, the number of dopamine neurons identified by tyrosine hydroxylase (TH) immunostaining declined rapidly with only 23% of the neurons surviving. By contrast, 73% of total cells survived rotenone treatment, indicating that TH+ neurons are more sensitive to rotenone. Examination of the role of apoptosis in TH+ neuron death, revealed that 10 and 30 nm rotenone significantly increased the number of apoptotic TH+ neurons from 7% under control conditions to 38 and 55%, respectively. The increase in apoptotic TH+ neurons correlated with an increase in immunoreactivity for active caspase-3 in TH+ neurons. The caspase-3 inhibitor, DEVD, rescued a significant number of TH+ neurons from rotenone-induced death. Furthermore, this protective effect lasted for at least 32 h post-rotenone and DEVD exposure, indicating lasting neuroprotection achieved with an intervention prior to the death commitment point. Our results show for the first time in primary dopamine neurons that, at low nanomolar concentrations, rotenone induces caspase-3-mediated apoptosis. Understanding the mechanism of rotenone-induced apoptosis in dopamine neurons may contribute to the development of new neuroprotective strategies against Parkinson's disease.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Mesencéfalo , Neuronas/efectos de los fármacos , Plaguicidas/toxicidad , Rotenona/toxicidad , Animales , Apoptosis/efectos de los fármacos , Caspasa 3 , Inhibidores de Caspasas , Células Cultivadas , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Mesencéfalo/citología , Mesencéfalo/embriología , Neuronas/citología , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Oligopéptidos/farmacología , Ratas , Ratas Sprague-Dawley , Tirosina 3-Monooxigenasa/biosíntesis
16.
Alcohol Clin Exp Res ; 27(2): 324-35, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12605082

RESUMEN

This article summarizes the proceedings of a symposium held at the 2002 Research Society on Alcoholism Meeting in San Francisco, California. The aim of this symposium was to review research on the effects of ethanol on neural stems cells and neurogenesis. Ethanol is known to alter neurogenesis during development; however, recent studies indicate that the brain forms new neurons from stem cells throughout life. Furthermore, stem cells can be transplanted into the brain, creating exciting new possibilities to study brain function. The symposium covered these research areas. Dr. Michael W. Miller reviewed knowledge on the effects of ethanol on stem cell proliferation and differentiation during development. Dr. Wu Ma described studies in culture indicating that (1) neural stem cells express functional muscarinic acetylcholine receptors (mAchR), (2) mAchR-mediated proliferation involves Ca signaling and mitogen-activated protein kinase phosphorylation, and (3) phosphoinositol-3 kinase is a downstream effector for mAchR-mediated cell proliferation via activation of Akt. Drs. Kim Nixon and Fulton T. Crews followed with in vivo studies on ethanol's effects on adult neural stem cell proliferation and differentiation. Dr. W. Michael Zawada described studies directed at dopamine neuron cell transplants into mammalian central nervous system. These studies clearly establish that ethanol has significant effects on stem cells.


Asunto(s)
Trasplante de Tejido Encefálico , Diferenciación Celular/efectos de los fármacos , Etanol/toxicidad , Neuroglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Trasplante de Células Madre , Células Madre/efectos de los fármacos , Adulto , Animales , División Celular/efectos de los fármacos , Niño , Femenino , Humanos , Embarazo , Receptores Muscarínicos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
17.
Cell Transplant ; 11(1): 5-16, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12095220

RESUMEN

The limited availability of human embryonic tissue for dopamine cell transplants in Parkinson's patients has led to an increased interest in using xenogeneic donor tissue. Unfortunately, without aggressive immunosuppression, such brain xenografts are rejected by the host immune system. Chronic brain xenograft rejection is largely mediated by helper T cells, which require presentation of xenoantigens by major histocompatability complex (MHC) class II for their activation. We examined survival and function of xenografts of E13 mouse mesencephalon deficient in either MHC class I, class II, or both after transplantation into adult hemiparkinsonian rats without immunosuppression. Recipients received grafts from C57BL/6 mice that were either: 1) wild-type (wt), 2) MHC class I knockout (KO), 3) MHC class II KO, 4) MHC class I and II double KO, or 5) saline sham transplants. At 6 weeks after transplantation, recipients of MHC class I KO, class II KO, and double KO xenografts significantly reduced methamphetamine-induced circling rate while rats with wt xenografts and sham-operated rats showed no improvement. MHC class II KO grafts had the greatest number of surviving dopamine neurons. All transplants, including saline sham controls, contained infiltrating host MHC class II-positive cells. Saline sham grafts and MHC class II KO xenografts contained significantly fewer infiltrating host MHC class II-positive cells than did wt grafts. Our results show that MHC class II-deficient xenografts survive transplantation for at least 6 weeks in the absence of immunosuppression, reduce rotational asymmetry, and provoke lesser immune reaction than wt grafts.


Asunto(s)
Conducta Animal/efectos de los fármacos , Complejo Mayor de Histocompatibilidad/genética , Mesencéfalo/trasplante , Enfermedad de Parkinson/terapia , Recuperación de la Función , Trasplante Heterólogo/inmunología , Animales , Conducta Animal/fisiología , Cuerpo Estriado/citología , Cuerpo Estriado/metabolismo , Cuerpo Estriado/cirugía , Dopamina/metabolismo , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Inmunohistoquímica , Complejo Mayor de Histocompatibilidad/inmunología , Masculino , Mesencéfalo/citología , Mesencéfalo/embriología , Metanfetamina/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neuronas/química , Neuronas/metabolismo , Oxidopamina/farmacología , Enfermedad de Parkinson/inmunología , Ratas , Ratas Sprague-Dawley , Simpaticolíticos/farmacología , Factores de Tiempo , Trasplante Heterotópico/inmunología , Tirosina 3-Monooxigenasa/metabolismo
18.
Brain Res ; 926(1-2): 42-50, 2002 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11814405

RESUMEN

Mutations in the alpha-synuclein gene have been linked to rare cases of familial Parkinson's disease (PD). Alpha-synuclein is a major component of Lewy bodies (LB), a pathological hallmark of PD. Transgenic mice and Drosophila expressing either wild-type or mutant human alpha-synuclein develop motor deficits, LB-like inclusions in some neurons, and neuronal degeneration. However, the relationship between abnormal aggregates of alpha-synuclein and human dopamine (DA) neuron degeneration remains unclear. In this report, we have investigated the influence of alpha-synuclein expression on DA neurons in primary culture of embryonic human mesencephalon. Two days after culture, human DA cells were transduced with wild-type or mutant human (Ala(53)Thr) alpha-synuclein adenoviruses and maintained for 5 days. Overexpression of mutant and wild-type human alpha-synuclein resulted in 49% (P<0.01) and 27% (P<0.05) loss of DA neurons, respectively, while not affecting viability of other cells in the culture. Overexpression of rat alpha-synuclein or GFP (green fluorescent protein) had no effect on DA neuron survival. Cytoplasmic inclusions of alpha-synuclein were detected immunohistochemically in DA cells transduced with mutant human alpha-synuclein, but not wild-type alpha-synuclein. These results show that overexpression of human alpha-synuclein, particularly the mutant form, can cause human DA neuron death, suggesting that alpha-synuclein may have a primary role in the pathogenesis of PD.


Asunto(s)
Apoptosis/fisiología , Dopamina/fisiología , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Adenoviridae/genética , Células Cultivadas , Feto/citología , Regulación Viral de la Expresión Génica , Proteínas Fluorescentes Verdes , Humanos , Cuerpos de Inclusión/química , Indicadores y Reactivos/metabolismo , Proteínas Luminiscentes/genética , Mesencéfalo/citología , Proteínas del Tejido Nervioso/análisis , Neuronas/fisiología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Sustancia Negra/patología , Sinucleínas , alfa-Sinucleína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...