Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
NPJ Parkinsons Dis ; 10(1): 37, 2024 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-38368444

RESUMEN

The brain renin-angiotensin system (RAS) has been related to dopaminergic degeneration, and high expression of the angiotensin II (AngII) type 1 receptor (AT1) gene is a marker of the most vulnerable neurons in humans. However, it is unknown whether AngII/AT1 overactivation affects α-synuclein aggregation and transmission. In vitro, AngII/AT1 activation increased α-synuclein aggregation in dopaminergic neurons and microglial cells, which was related to AngII-induced NADPH-oxidase activation and intracellular calcium raising. In mice, AngII/AT1 activation was involved in MPTP-induced increase in α-synuclein expression and aggregation, as they significantly decreased in mice treated with the AT1 blocker telmisartan and AT1 knockout mice. Cell co-cultures (transwells) revealed strong transmission of α-synuclein from dopaminergic neurons to astrocytes and microglia. AngII induced a higher α-synuclein uptake by microglial cells and an increase in the transfer of α-synuclein among astroglial cells. However, AngII did not increase the release of α-synuclein by neurons. The results further support brain RAS dysregulation as a major mechanism for the progression of Parkinson's disease, and AT1 inhibition and RAS modulation as therapeutic targets.

2.
Antioxidants (Basel) ; 11(2)2022 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-35204211

RESUMEN

The tissue renin-angiotensin system (RAS) has been shown to be involved in prooxidative and proinflammatory changes observed in aging and aging-related diseases such as dopaminergic degeneration in Parkinson's disease (PD). We studied the activation of the NLRP3 inflammasome in the substantia nigra with aging and early stages of dopaminergic degeneration in PD models and, particularly, if the brain RAS, via its prooxidative proinflammatory angiotensin II (AngII) type 1 (AT1) receptors, mediates the inflammasome activation. Nigras from aged rats and mice and 6-hydroxydopamine PD models showed upregulation in transcription of inflammasome-related components (NLRP3, pro-IL1ß and pro-IL18) and IL1ß and IL18 protein levels, which was inhibited by the AT1 receptor antagonist candesartan. The role of the AngII/AT1 axis in inflammasome activation was further confirmed in rats intraventricularly injected with AngII, and in primary mesencephalic cultures treated with 6-hydroxydopamine, which showed inflammasome activation that was blocked by candesartan. Observations in the nigra of young and aged AT1 and AT2 knockout mice confirmed the major role of AT1 receptors in nigral inflammasome activation. In conclusion, the inflammasome is upregulated by aging and dopaminergic degeneration in the substantia nigra, possibly related with a decrease in dopamine levels, and it is mediated by the AngII/AT1 axis.

3.
Antioxidants (Basel) ; 10(9)2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-34572972

RESUMEN

Dysregulation of the tissue renin-angiotensin system (RAS) is involved in tissue oxidative and inflammatory responses. Among RAS components, renin, its precursor (pro)renin and its specific receptor (PRR) have been less investigated, particularly in the brain. We previously showed the presence of PRR in neurons and glial cells in the nigrostriatal system of rodents and primates, including humans. Now, we used rat and mouse models and cultures of BV2 and primary microglial cells to study the role of PRR in microglial pro-inflammatory responses. PRR was upregulated in the nigral region, particularly in microglia during the neuroinflammatory response. In the presence of the angiotensin type-1 receptor blocker losartan, to exclude angiotensin-related effects, treatment of microglial cells with (pro)renin induces the expression of microglial pro-inflammatory markers, which is mediated by upregulation of NADPH-oxidase and Rho-kinase activities, downregulation of autophagy and upregulation of inflammasome activity. Conditioned medium from (pro)renin-treated microglia increased dopaminergic cell death relative to medium from non-treated microglia. However, these effects were blocked by pre-treatment of microglia with the Rho-kinase inhibitor fasudil. Activation of microglial PRR enhances the microglial pro-inflammatory response and deleterious effects of microglia on dopaminergic cells, and microglial NADPH-oxidase, Rho-Kinase and autophagy are involved in this process.

4.
Prog Neurobiol ; 199: 101919, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33039415

RESUMEN

The renin-angiotensin system (RAS) is one of the oldest hormone systems in vertebrate phylogeny. RAS was initially related to regulation of blood pressure and sodium and water homeostasis. However, local or paracrine RAS were later identified in many tissues, including brain, and play a major role in their physiology and pathophysiology. In addition, a major component, ACE2, is the entry receptor for SARS-CoV-2. Overactivation of tissue RAS leads several oxidative stress and inflammatory processes involved in aging-related degenerative changes. In addition, a third level of RAS, the intracellular or intracrine RAS (iRAS), with still unclear functions, has been observed. The possible interaction between the intracellular and extracellular RAS, and particularly the possible deleterious or beneficial effects of the iRAS activation are controversial. The dopaminergic system is particularly interesting to investigate the RAS as important functional interactions between dopamine and RAS have been observed in the brain and several peripheral tissues. Our recent observations in mitochondria and nucleus of dopaminergic neurons may clarify the role of the iRAS. This may be important for the developing of new therapeutic strategies, since the effects on both extracellular and intracellular RAS must be taken into account, and perhaps better understanding of COVID-19 cell mechanisms.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Sistema Renina-Angiotensina , Animales , COVID-19 , Humanos , Espacio Intracelular/metabolismo , Estrés Oxidativo , SARS-CoV-2
5.
J Gerontol A Biol Sci Med Sci ; 75(3): 416-424, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-30412227

RESUMEN

Sirtuin 3 (SIRT3) and angiotensin play a major role in aging-related disorders. Both modulate oxidative stress and neurodegeneration. We investigated the interaction between SIRT3 and angiotensin II (AngII) in the dopaminergic system. Both in vivo and in vitro, treatment with AngII decreased SIRT3 expression, which was reversed by angiotensin type 1 receptor (AT1) antagonists. Aged animals showed enhanced pro-oxidative RAS activity and low nigral SIRT3 levels, which significantly increased with treatment with the AT1 antagonist candesartan or AT1 deletion. Consistent with this, AT2 knockout mice and cells treated with AT2 blockers showed downregulation of SIRT3. Treatment with the specific SIRT3 inhibitor AGK7 induced overexpression of AT1 and AT2 in substantia nigra (SN) of rats, and in dopaminergic neuronal MES23.5 and microglial N9 cell lines. The results suggest that SIRT3 may initially counteract low levels of oxidative stress as part of the antioxidant response. However, high or persistent oxidative stress induced by overactivation of the angiotensin/AT1 pro-oxidative axis induces a decrease in nigral SIRT3 levels. Furthermore, a decrease in SIRT3 levels further increases AT1 activity, which may lead to a feed-forward mechanism. This is observed in aged rats and can be counteracted by treatment with AT1 antagonists such as candesartan.


Asunto(s)
Angiotensina II/fisiología , Enfermedades Neurodegenerativas/etiología , Estrés Oxidativo , Receptor de Angiotensina Tipo 1/fisiología , Sirtuina 3/metabolismo , Sustancia Negra/química , Sustancia Negra/metabolismo , Factores de Edad , Animales , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Sirtuina 3/análisis
6.
Mol Neurobiol ; 55(7): 5847-5867, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29086247

RESUMEN

In addition to the classical hormonal (tissue-to-tissue) renin-angiotensin system (RAS), there are a paracrine (cell-to-cell) and an intracrine (intracellular/nuclear) RAS. A local paracrine brain RAS has been associated with several brain disorders, including Parkinson's disease (PD). Classically, angiotensin II (Ang II) is the main RAS effector peptide and acts through two major receptors: Ang II type 1 and 2 (AT1 and AT2) receptors. It has been shown that enhanced activation of the Ang II/AT1 axis exacerbates dopaminergic cell death. Several new components of the RAS have more recently been discovered. However, the role of new Ang 1-7/Mas receptor RAS component was not investigated in the brain and particularly in the dopaminergic system. In the present study, we observed Mas receptor labeling in dopaminergic neurons and glial cells in rat mesencephalic primary cultures; substantia nigra of rats, monkeys, and humans; and human induced pluripotent stem (iPS) cells derived from healthy controls and sporadic PD patients. The present data support a neuroprotective role of the Ang 1-7/Mas receptor axis in the dopaminergic system. We observed that this axis is downregulated with aging, which may contribute to the aging-related vulnerability to neurodegeneration. We have also identified an intracellular Ang 1-7/Mas axis that modulates mitochondrial and nuclear levels of superoxide. The present data suggest that nuclear RAS receptors regulate the adequate balance between the detrimental and the protective arms of the cell RAS. The results further support that the brain RAS should be taken into account for the design of new therapeutic strategies for PD.


Asunto(s)
Angiotensina I/metabolismo , Comunicación Paracrina , Fragmentos de Péptidos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Sustancia Negra/metabolismo , Envejecimiento/metabolismo , Angiotensina II , Animales , Astrocitos/metabolismo , Estudios de Casos y Controles , Núcleo Celular/metabolismo , Células Cultivadas , Neuronas Dopaminérgicas/metabolismo , Eliminación de Gen , Haplorrinos , Masculino , Mesencéfalo/citología , Microglía/metabolismo , Mitocondrias/metabolismo , Modelos Biológicos , Neuroprotección , Estrés Oxidativo , Peptidil-Dipeptidasa A/metabolismo , Proto-Oncogenes Mas , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo , Roedores
7.
Front Aging Neurosci ; 9: 365, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29163145

RESUMEN

Insulin-like growth factor-1 (IGF-1) effects on aging and neurodegeneration is still controversial. However, it is widely admitted that IGF-1 is involved in the neuroinflammatory response. In peripheral tissues, several studies showed that IGF-1 inhibited the expression of inflammatory markers, although other studies concluded that IGF-1 has proinflammatory functions. Furthermore, proinflammatory cytokines such as TNF-α impaired IGF-1 signaling. In the brain, there are controversial results on effects of IGF-1 in neuroinflammation. In addition to direct protective effects on neurons, several studies revealed anti-inflammatory effects of IGF-1 acting on astrocytes and microglia, and that IGF-1 may also inhibit blood brain barrier permeability. Altogether suggests that the aging-related decrease in IGF-1 levels may contribute to the aging-related pro-inflammatory state. IGF-1 inhibits the astrocytic response to inflammatory stimuli, and modulates microglial phenotype (IGF-1 promotes the microglial M2 and inhibits of M1 phenotype). Furthermore, IGF-1 is mitogenic for microglia. IGF-1 and estrogen interact to modulate the neuroinflammatory response and microglial and astrocytic phenotypes. Brain renin-angiotensin and IGF-1 systems also interact to modulate neuroinflammation. Induction of microglial IGF-1 by angiotensin, and possibly by other pro-inflammatory inducers, plays a major role in the repression of the M1 microglial neurotoxic phenotype and the enhancement of the transition to an M2 microglial repair/regenerative phenotype. This mechanism is impaired in aged brains. Aging-related decrease in IGF-1 may contribute to the loss of capacity of microglia to undergo M2 activation. Fine tuning of IGF-1 levels may be critical for regulating the neuroinflammatory response, and IGF-1 may be involved in inflammation in a context-dependent mode.

8.
Cell Death Dis ; 8(9): e3044, 2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28880266

RESUMEN

The 'classical' renin-angiotensin system (RAS) is a circulating system that controls blood pressure. Local/paracrine RAS, identified in a variety of tissues, including the brain, is involved in different functions and diseases, and RAS blockers are commonly used in clinical practice. A third type of RAS (intracellular/intracrine RAS) has been observed in some types of cells, including neurons. However, its role is still unknown. The present results indicate that in brain cells the intracellular RAS counteracts the intracellular superoxide/H2O2 and oxidative stress induced by the extracellular/paracrine angiotensin II acting on plasma membrane receptors. Activation of nuclear receptors by intracellular or internalized angiotensin triggers a number of mechanisms that protect the cell, such as an increase in the levels of protective angiotensin type 2 receptors, intracellular angiotensin, PGC-1α and IGF-1/SIRT1. Interestingly, this protective mechanism is altered in isolated nuclei from brains of aged animals. The present results indicate that at least in the brain, AT1 receptor blockers acting only on the extracellular or paracrine RAS may offer better protection of cells.


Asunto(s)
Envejecimiento/metabolismo , Angiotensina II/metabolismo , Neuronas Dopaminérgicas/metabolismo , Comunicación Paracrina/fisiología , Receptor de Angiotensina Tipo 2/genética , Sistema Renina-Angiotensina/genética , Envejecimiento/genética , Angiotensina II/farmacología , Animales , Presión Sanguínea/fisiología , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/ultraestructura , Regulación de la Expresión Génica , Humanos , Peróxido de Hidrógeno/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Cultivo Primario de Células , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 2/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo , Sustancia Negra/citología , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Superóxidos/metabolismo
9.
Cell Death Dis ; 7(10): e2427, 2016 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-27763643

RESUMEN

The renin-angiotensin system (RAS) was initially considered as a circulating humoral system controlling blood pressure, being kidney the key control organ. In addition to the 'classical' humoral RAS, a second level in RAS, local or tissular RAS, has been identified in a variety of tissues, in which local RAS play a key role in degenerative and aging-related diseases. The local brain RAS plays a major role in brain function and neurodegeneration. It is normally assumed that the effects are mediated by the cell-surface-specific G-protein-coupled angiotensin type 1 and 2 receptors (AT1 and AT2). A combination of in vivo (rats, wild-type mice and knockout mice) and in vitro (primary mesencephalic cultures, dopaminergic neuron cell line cultures) experimental approaches (confocal microscopy, electron microscopy, laser capture microdissection, transfection of fluorescent-tagged receptors, treatments with fluorescent angiotensin, western blot, polymerase chain reaction, HPLC, mitochondrial respirometry and other functional assays) were used in the present study. We report the discovery of AT1 and AT2 receptors in brain mitochondria, particularly mitochondria of dopaminergic neurons. Activation of AT1 receptors in mitochondria regulates superoxide production, via Nox4, and increases respiration. Mitochondrial AT2 receptors are much more abundant and increase after treatment of cells with oxidative stress inducers, and produce, via nitric oxide, a decrease in mitochondrial respiration. Mitochondria from the nigral region of aged rats displayed altered expression of AT1 and AT2 receptors. AT2-mediated regulation of mitochondrial respiration represents an unrecognized primary line of defence against oxidative stress, which may be particularly important in neurons with increased levels of oxidative stress such as dopaminergic neurons. Altered expression of AT1 and AT2 receptors with aging may induce mitochondrial dysfunction, the main risk factor for neurodegeneration.


Asunto(s)
Envejecimiento/patología , Citoprotección , Neuronas Dopaminérgicas/metabolismo , Mitocondrias/metabolismo , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Respiración de la Célula , Células Cultivadas , Fluorescencia , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Luminiscentes/metabolismo , Masculino , Potencial de la Membrana Mitocondrial , Ratones Endogámicos C57BL , Modelos Biológicos , NADPH Oxidasa 4 , NADPH Oxidasas/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Fosforilación Oxidativa , Estrés Oxidativo , Ratas Sprague-Dawley , Sustancia Negra/metabolismo , Sustancia Negra/patología , Superóxidos/metabolismo
10.
J Gerontol A Biol Sci Med Sci ; 71(10): 1254-7, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26503374

RESUMEN

Abnormal activation of the RhoA/Rho kinase (ROCK) pathway plays a pivotal role in neuroinflammatory and pro-oxidative responses, axonal retraction, and apoptosis. We observed increased expression of RhoA, ROCK II, and ROCK activity in the brain of aged rats, particularly in the substantia nigra. Increased ROCK activity may enhance major mechanisms responsible for aging-related neurodegeneration, thus representing a major factor in the vulnerability of dopaminergic neurons to damage. We also observed that physical exercise decreased ROCK activation in aged rats. This suggests that decreased ROCK activation plays an important role in the neuroprotective effects of exercise observed in several previous studies. Furthermore, the present results suggest that ROCK inhibitors may constitute an effective neuroprotective strategy against aging-related risk of dopaminergic degeneration and possibly against other aging-related neurodegenerative processes.


Asunto(s)
Apoptosis/fisiología , Neuroprotección/fisiología , Condicionamiento Físico Animal , Sustancia Negra/enzimología , Quinasas Asociadas a rho/metabolismo , Animales , Western Blotting , Neuronas Dopaminérgicas/enzimología , Masculino , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
11.
Neuroscientist ; 21(6): 616-29, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25323761

RESUMEN

The small GTP-binding protein Rho plays an important role in several cellular functions. RhoA, which is a member of the Rho family, initiates cellular processes that act on its direct downstream effector Rho-associated kinase (ROCK). ROCK inhibition protects against dopaminergic cell death induced by dopaminergic neurotoxins. It has been suggested that ROCK inhibition activates neuroprotective survival cascades in dopaminergic neurons. Axon-stabilizing effects in damaged neurons may represent another mechanism of neuroprotection of dopaminergic neurons by ROCK inhibition. However, it has been shown that microglial cells play a crucial role in neuroprotection by ROCK inhibition and that activation of microglial ROCK mediates major components of the microglial inflammatory response. Additional mechanisms such as interaction with autophagy may also contribute to the neuroprotective effects of ROCK inhibition. Interestingly, ROCK interacts with several brain factors that play a major role in dopaminergic neuron vulnerability such as NADPH-oxidase, angiotensin, and estrogen. ROCK inhibition may provide a new neuroprotective strategy for Parkinson's disease. This is of particular interest because ROCK inhibitors are currently used against vascular diseases in clinical practice. However, it is necessary to develop more potent and selective ROCK inhibitors to reduce side effects and enhance the efficacy.


Asunto(s)
Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/enzimología , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/enzimología , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo , Animales , Antiparkinsonianos/farmacología , Antiparkinsonianos/uso terapéutico , Humanos , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/enzimología , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico
12.
Neuropharmacology ; 85: 1-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24878243

RESUMEN

Several recent studies have shown that activation of the RhoA/Rho-associated kinase (ROCK) pathway is involved in the MPTP-induced dopaminergic cell degeneration and possibly in Parkinson's disease. ROCK inhibitors have been suggested as candidate neuroprotective drugs for Parkinson's disease. However, the mechanism responsible for the increased survival of dopaminergic neurons after treatment with ROCK inhibitors is not clear. We exposed primary (neuron-glia) mesencephalic cultures, cultures of the MES 23.5 dopaminergic neuron cell line and primary mesencephalic cultures lacking microglial cells to the dopaminergic neurotoxin MPP+ and the ROCK inhibitor Y-27632 in order to study the effects of ROCK inhibition on dopaminergic cell loss and the length of neurites of surviving dopaminergic neurons. In primary (neuron-glia) cultures, simultaneous treatment with MPP+ and the ROCK inhibitor significantly reduced the loss of dopaminergic neurons. In the absence of microglia, treatment with the ROCK inhibitor did not induce a significant reduction in the dopaminergic cell loss. Treatment with the ROCK inhibitor induced a significant decrease in axonal retraction in primary cultures with and without microglia and in cultures of the MES 23.5 neuron cell line. In conclusion, inhibition of microglial ROCK is essential for the neuroprotective effects of ROCK inhibitors against cell death induced by the dopaminergic neurotoxin MPP+. In addition, ROCK inhibition induced a direct effect against axonal retraction in surviving neurons. However, the latter effect was not sufficient to cause a significant increase in the survival of dopaminergic neurons after treatment with MPP+.


Asunto(s)
Amidas/farmacología , Muerte Celular/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Intoxicación por MPTP/tratamiento farmacológico , Microglía/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Piridinas/farmacología , Animales , Axones/efectos de los fármacos , Axones/patología , Axones/fisiología , Técnicas de Cultivo de Célula , Muerte Celular/fisiología , Línea Celular Tumoral , Células Cultivadas , Neuronas Dopaminérgicas/patología , Neuronas Dopaminérgicas/fisiología , Intoxicación por MPTP/patología , Intoxicación por MPTP/fisiopatología , Mesencéfalo/efectos de los fármacos , Mesencéfalo/patología , Mesencéfalo/fisiopatología , Microglía/enzimología , Neuritas/efectos de los fármacos , Neuritas/patología , Neuritas/fisiología , Ratas , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
13.
Neurobiol Aging ; 35(7): 1726-38, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24529758

RESUMEN

It is not known whether the aging-related decrease in dopaminergic function leads to the aging-related higher vulnerability of dopaminergic neurons and risk for Parkinson's disease. The renin-angiotensin system (RAS) plays a major role in the inflammatory response, neuronal oxidative stress, and dopaminergic vulnerability via type 1 (AT1) receptors. In the present study, we observed a counterregulatory interaction between dopamine and angiotensin receptors. We observed overexpression of AT1 receptors in the striatum and substantia nigra of young adult dopamine D1 and D2 receptor-deficient mice and young dopamine-depleted rats, together with compensatory overexpression of AT2 receptors or compensatory downregulation of angiotensinogen and/or angiotensin. In aged rats, we observed downregulation of dopamine and dopamine receptors and overexpression of AT1 receptors in aged rats, without compensatory changes observed in young animals. L-Dopa therapy inhibited RAS overactivity in young dopamine-depleted rats, but was ineffective in aged rats. The results suggest that dopamine may play an important role in modulating oxidative stress and inflammation in the substantia nigra and striatum via the RAS, which is impaired by aging.


Asunto(s)
Envejecimiento/genética , Dopamina/fisiología , Receptor de Angiotensina Tipo 1/fisiología , Envejecimiento/metabolismo , Animales , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Dopamina/genética , Dopamina/metabolismo , Neuronas Dopaminérgicas/fisiología , Regulación hacia Abajo , Expresión Génica , Inflamación/genética , Masculino , Ratones , Estrés Oxidativo/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Receptores Dopaminérgicos/genética , Receptores Dopaminérgicos/metabolismo , Sistema Renina-Angiotensina/fisiología , Riesgo , Sustancia Negra/metabolismo , Sustancia Negra/patología
14.
Neuropharmacology ; 76 Pt A: 156-68, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23973568

RESUMEN

Beneficial effects of angiotensin type-1 receptor (AT1) inhibition have been observed in a number of brain processes mediated by oxidative stress and neuroinflammation, including Parkinson's disease. However, important counterregulatory interactions between dopamine and angiotensin systems have recently been demonstrated in several peripheral tissues, and it is possible that a decrease in dopamine levels due to AT1 inhibition may interfere with neuroprotective strategies. The present experiments involving rats with normal dopaminergic innervation indicate that chronic treatment with the AT1 antagonist candesartan does not significantly affect striatal levels of dopamine, serotonin or metabolites, as does not significantly affect motor behavior, as evaluated by the rotarod test. Interestingly, chronic administration of candesartan to normal rats induced a marked increase in dopamine D1 and a decrease in dopamine D2 receptor expression. In a rat model of Parkinson's disease treated with L-DOPA, no differences in striatal dopamine and serotonin levels were observed between candesartan-treated rats and untreated, which suggests that chronic treatment with candesartan does not significantly affect the process of L-DOPA decarboxylation and dopamine release in Parkinson's disease patients. Candesartan did not induce any differences in the striatal expression of dopamine D1 and D2 and serotonin 5-HT1B receptors in 6ydroxydopamine-lesioned rats treated with L-DOPA. The results suggest that chronic treatment with AT1 antagonists as a neuroprotective strategy does not significantly affect striatal dopamine release or motor behavior. This article is part of the Special Issue entitled 'The Synaptic Basis of Neurodegenerative Disorders'.


Asunto(s)
Antagonistas de Receptores de Angiotensina/farmacología , Antagonistas de Receptores de Angiotensina/uso terapéutico , Cuerpo Estriado/efectos de los fármacos , Dopamina/metabolismo , Levodopa/farmacología , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson Secundaria/tratamiento farmacológico , Animales , Bencimidazoles/farmacología , Bencimidazoles/uso terapéutico , Compuestos de Bifenilo , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Levodopa/uso terapéutico , Masculino , Actividad Motora/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Oxidopamina/toxicidad , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/metabolismo , Ratas , Receptor de Serotonina 5-HT1B/metabolismo , Receptores Dopaminérgicos/metabolismo , Serotonina/metabolismo , Tetrazoles/farmacología , Tetrazoles/uso terapéutico
15.
Mov Disord ; 28(10): 1337-42, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23925977

RESUMEN

Renin-angiotensin systems are known to act in many tissues, for example, the blood vessel wall or kidney, where a close interaction between angiotensin and dopamine has been demonstrated. Regulatory interactions between the dopaminergic and renin-angiotensin systems have recently been described in the substantia nigra and striatum. In animal models, dopamine depletion induces compensatory overactivation of the local renin-angiotensin system, which primes microglial responses and neuron vulnerability by activating NADPH-oxidase. Hyperactivation of the local renin-angiotensin system exacerbates the inflammatory microglial response, oxidative stress, and dopaminergic degeneration, all of which are inhibited by angiotensin receptor blockers and inhibitors of angiotensin-converting enzymes. In this review we provide evidence suggesting that the renin-angiotensin system may play an important role in dopamine's mediated neuroinflammation and oxidative stress changes in Parkinson's disease. We suggest that manipulating brain angiotensin may constitute an effective neuroprotective strategy for Parkinson's disease.


Asunto(s)
Angiotensinas/fisiología , Ganglios Basales/fisiología , Dopamina/fisiología , Enfermedad de Parkinson/fisiopatología , Sistema Renina-Angiotensina/fisiología , Encéfalo/fisiopatología , Interpretación Estadística de Datos , Humanos , Neostriado/fisiopatología , Comunicación Paracrina/fisiología , Sustancia Negra/fisiopatología
16.
Brain Struct Funct ; 218(2): 373-88, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22407459

RESUMEN

We have previously obtained in rodents a considerable amount of data suggesting a major role for the brain renin-angiotensin system (RAS) in dopaminergic neuron degeneration and potentially in Parkinson's disease. However, the presence of a local RAS has not been demonstrated in the monkey or the human substantia nigra compacta (SNc). The present study demonstrates the presence of major RAS components in dopaminergic neurons, astrocytes and microglia in both the monkey and the human SNc. Angiotensin type 1 and 2 and renin-prorenin receptors were located at the surface of dopaminergic neurons and glial cells, as expected for a tissular RAS. However, angiotensinogen and receptors for angiotensin and renin-prorenin were also observed at the cytoplasm and nuclear level, which suggests the presence of an intracrine or intracellular RAS in monkey and human SNc. Although astrocytes and microglia were labeled for angiotensin and prorenin receptors in the normal SNc, most glial cells appeared less immunoreactive than the dopaminergic neurons. However, our previous studies in rodent models of PD and studies in other animal models of brain diseases suggest that the RAS activity is significantly upregulated in glial cells in pathological conditions. The present results together with our previous findings in rodents suggest a major role for the nigral RAS in the normal functioning of the dopaminergic neurons, and in the progression of the dopaminergic degeneration.


Asunto(s)
Angiotensinógeno/análisis , Receptor de Angiotensina Tipo 1/análisis , Receptor de Angiotensina Tipo 2/análisis , Receptores de Superficie Celular/análisis , Sistema Renina-Angiotensina , Sustancia Negra/química , ATPasas de Translocación de Protón Vacuolares/análisis , Adulto , Animales , Astrocitos/química , Autopsia , Neuronas Dopaminérgicas/química , Técnica del Anticuerpo Fluorescente , Humanos , Macaca fascicularis , Masculino , Microglía/química , Microscopía Confocal , Sustancia Negra/citología , Receptor de Prorenina
17.
Neurobiol Dis ; 47(2): 268-79, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22542954

RESUMEN

It has recently been shown that the dopaminergic cell loss induced by neurotoxins is enhanced by brain angiotensin II (AII) via type 1 receptors (AT1). However, the mechanisms involved in the dopaminergic degeneration and the brain inflammatory effects of AII have not been clarified. The RhoA-Rho-Kinase (ROCK) pathway may play a critical role in the inflammatory and oxidative effects of AII. In the substantia nigra of mice, administration of the dopaminergic neurotoxin MPTP induced an increase in the expression of RhoA and ROCK II mRNA levels and ROCK activity, which were inhibited by AT1 receptor deletion (i.e., in AT1a null mice treated with MPTP). Administration of the ROCK inhibitor Y-27632 or AT1 deletion induced a significant decrease in MPTP-induced microglial activation and dopaminergic cell death. In rat primary mesencephalic cultures treated with MPP(+), the increase in dopaminergic cell loss induced by AII administration was also inhibited by treatment with Y27632. Intense expression of ROCK II was observed in the microglial cells in the substantia nigra of mice treated with MPTP, and the major role of the microglial ROCK was confirmed by comparing mesencephalic cultures with and without microglia. Activation of the RhoA/ROCK pathway is involved in the MPTP-induced dopaminergic degeneration, and in the enhancing effect of AII/AT1 activation on the microglial response and dopaminergic degeneration. ROCK inhibitors and AT1 receptor antagonists may provide new neuroprotective strategies against the progression of Parkinson's disease.


Asunto(s)
Angiotensinas/fisiología , Neuronas Dopaminérgicas/metabolismo , Microglía/metabolismo , Receptor de Angiotensina Tipo 1/fisiología , Transducción de Señal/fisiología , Proteínas de Unión al GTP rho/fisiología , Amidas/farmacología , Amidas/uso terapéutico , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Células Cultivadas , Neuronas Dopaminérgicas/enzimología , Neuronas Dopaminérgicas/patología , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/enzimología , Microglía/patología , Enfermedad de Parkinson/fisiopatología , Enfermedad de Parkinson/prevención & control , Piridinas/farmacología , Piridinas/uso terapéutico , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/fisiología , Proteína de Unión al GTP rhoA
18.
Am J Neurodegener Dis ; 1(3): 226-44, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23383395

RESUMEN

The pathogenic mechanism of Parkinson's disease (PD) appears to be multifactorial. However, oxidative stress and neuroinflammation, including activation of NADPH-dependent oxidases, play a major role in the progression of dopaminergic cell death. The renin-angiotensin system (RAS) was described as a circulating humoral system that regulates blood pressure and water homeostasis. However, there exist local RAS in many tissues, and locally formed angiotensin activates NADPH-dependent oxidases, which are a major source of superoxide and are upregulated in major aging-related diseases such as hypertension, diabetes and atherosclerosis. Furthermore, an intracellular or intracrine RAS, with still unknown functions, has been identified in several cell types. The brain has an independent local RAS, which has been involved in several brain disorders, including neurodegenerative diseases. It is particularly interesting for PD the important interaction observed between angiotensin and dopamine, which counterregulate each other in renal cells and also in the striatum and substantia nigra. In recent studies, we have observed both a local and an intracellular RAS in the rodent, monkey and human substantia nigra, and that dopamine depletion induced RAS upregulation possibly as a compensatory mechanism. However, RAS hyperactivation also exacerbated oxidative stress and neuroinflammation, which contributed to progression of dopaminergic degeneration. In addition, we observed increased RAS activity in the nigra of animals with higher vulnerability of dopaminergic neurons to degeneration, such as aged males, menopausal females and rats subjected to chronic brain hypoperfusion. RAS activity and dopaminergic vulnerability were significantly reduced by treatment with angiotensin type I receptor antagonists. Manipulation of the brain RAS may constitute an effective neuroprotective strategy against dopaminergic degeneration in PD.

19.
Neurobiol Aging ; 33(1): 204.e1-11, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20888078

RESUMEN

An age-related proinflammatory, pro-oxidant state in the nigra may increase the vulnerability of dopaminergic neurons to additional damage. Angiotensin II, via type 1 (AT1) receptors, is one of the most important known inflammation and oxidative stress inducers. However, it is not known if there are age-related changes in the nigral angiotensin system. In aged rats, we observed increased activation of the nicotinamide adenine dinucleotide phosphate-oxidase (NADPH oxidase) complex and increased levels of the proinflammatory cytokines interleukin (IL)-1ß and tumor necrosis factor (TNF)-α, which indicate pro-oxidative, proinflammatory state in the nigra. We also observed enhanced 6-hydroxydopamine (6-OHDA)-induced dopaminergic cell death in aged rats. This is associated with increased expression of AT1 receptors and decreased expression of AT2 receptors in aged rats, and is reduced by treatment with the AT1 antagonist candesartan. The present results indicate that brain angiotensin is involved in changes that may increase the risk of Parkinson's disease with aging. Furthermore, the results suggest that manipulation of the brain angiotensin system may constitute an effective neuroprotective strategy against aging-related risk of dopaminergic degeneration.


Asunto(s)
Envejecimiento/patología , Envejecimiento/fisiología , Angiotensina II/fisiología , Neuronas Dopaminérgicas/patología , Mediadores de Inflamación/metabolismo , Interleucina-1beta/metabolismo , NADPH Oxidasas/metabolismo , Estrés Oxidativo , Oxidopamina/farmacología , Especies Reactivas de Oxígeno/metabolismo , Sustancia Negra/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Envejecimiento/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Bencimidazoles/farmacología , Compuestos de Bifenilo , Neuronas Dopaminérgicas/metabolismo , Masculino , Terapia Molecular Dirigida , Degeneración Nerviosa , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/terapia , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/fisiología , Sustancia Negra/patología , Tetrazoles/farmacología
20.
Brain ; 135(Pt 1): 124-38, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22189567

RESUMEN

There is a lack of consensus about the effects of the type of menopause (surgical or natural) and of oestrogen replacement therapy on Parkinson's disease. The effects of the timing of replacement therapy and the female's age may explain the observed differences in such effects. However, the mechanisms involved are poorly understood. The renin-angiotensin system mediates the beneficial effects of oestrogen in several tissues, and we have previously shown that dopaminergic cell loss is enhanced by angiotensin via type 1 receptors, which is activated by ageing. In rats, we compared the effects of oestrogen replacement therapy on 6-hydroxydopamine-induced dopaminergic degeneration, nigral renin-angiotensin system activity, activation of the nicotinamide adenine dinucleotide phosphate oxidase complex and levels of the proinflammatory cytokine interleukin-1ß in young (surgical) menopausal rats and aged menopausal rats. In young surgically menopausal rats, the renin-angiotensin system activity was higher (i.e. higher angiotensin converting enzyme activity, higher angiotensin type-1 receptor expression and lower angiotensin type-2 receptor expression) than in surgically menopausal rats treated with oestrogen; the nicotinamide adenine dinucleotide phosphate oxidase activity and interleukin-1ß expression were also higher in the first group than in the second group. In aged menopausal rats, the levels of nigral renin-angiotensin and nicotinamide adenine dinucleotide phosphate oxidase activity were similar to those observed in surgically menopausal rats. However, oestrogen replacement therapy significantly reduced 6-hydroxydopamine-induced dopaminergic cell loss in young menopausal rats but not in aged rats. Treatment with oestrogen also led to a more marked reduction in nigral renin-angiotensin and nicotinamide adenine dinucleotide phosphate oxidase activity in young surgically menopausal rats (treated either immediately or after a period of hypo-oestrogenicity) than in aged menopausal rats. Interestingly, treatment with the angiotensin type-1 receptor antagonist candesartan led to remarkable reduction in renin-angiotensin system activity and dopaminergic neuron loss in both groups of menopausal rats. This suggests that manipulation of the brain renin-angiotensin system may be an efficient approach for the prevention or treatment of Parkinson's disease in oestrogen-deficient females, together with or instead of oestrogen replacement therapy.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Estradiol/análogos & derivados , Degeneración Nerviosa/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Sistema Renina-Angiotensina/efectos de los fármacos , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Animales , Bencimidazoles/farmacología , Bencimidazoles/uso terapéutico , Compuestos de Bifenilo , Cuerpo Estriado/metabolismo , Neuronas Dopaminérgicas/metabolismo , Estradiol/farmacología , Estradiol/uso terapéutico , Terapia de Reemplazo de Estrógeno , Femenino , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Ovariectomía , Oxidopamina/farmacología , Ratas , Ratas Sprague-Dawley , Sistema Renina-Angiotensina/fisiología , Tetrazoles/farmacología , Tetrazoles/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA