Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Transl Neurodegener ; 13(1): 22, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38622720

RESUMO

The renin-angiotensin system (RAS) was classically considered a circulating hormonal system that regulates blood pressure. However, different tissues and organs, including the brain, have a local paracrine RAS. Mutual regulation between the dopaminergic system and RAS has been observed in several tissues. Dysregulation of these interactions leads to renal and cardiovascular diseases, as well as progression of dopaminergic neuron degeneration in a major brain center of dopamine/angiotensin interaction such as the nigrostriatal system. A decrease in the dopaminergic function induces upregulation of the angiotensin type-1 (AT1) receptor activity, leading to recovery of dopamine levels. However, AT1 receptor overactivity in dopaminergic neurons and microglial cells upregulates the cellular NADPH-oxidase-superoxide axis and Ca2+ release, which mediate several key events in oxidative stress, neuroinflammation, and α-synuclein aggregation, involved in Parkinson's disease (PD) pathogenesis. An intraneuronal antioxidative/anti-inflammatory RAS counteracts the effects of the pro-oxidative AT1 receptor overactivity. Consistent with this, an imbalance in RAS activity towards the pro-oxidative/pro-inflammatory AT1 receptor axis has been observed in the substantia nigra and striatum of several animal models of high vulnerability to dopaminergic degeneration. Interestingly, autoantibodies against angiotensin-converting enzyme 2 and AT1 receptors are increased in PD models and PD patients and contribute to blood-brain barrier (BBB) dysregulation and nigrostriatal pro-inflammatory RAS upregulation. Therapeutic strategies addressed to the modulation of brain RAS, by AT1 receptor blockers (ARBs) and/or activation of the antioxidative axis (AT2, Mas receptors), may be neuroprotective for individuals with a high risk of developing PD or in prodromal stages of PD to reduce progression of the disease.


Assuntos
Doença de Parkinson , Sistema Renina-Angiotensina , Animais , Humanos , Antagonistas de Receptores de Angiotensina/farmacologia , Angiotensinas/metabolismo , Pressão Sanguínea , Encéfalo/metabolismo , Dopamina , Doença de Parkinson/patologia , Receptor Tipo 1 de Angiotensina/metabolismo , Sistema Renina-Angiotensina/fisiologia
2.
Antioxidants (Basel) ; 12(7)2023 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-37507992

RESUMO

Statins have been proposed for L-DOPA-induced dyskinesia (LID) treatment. Statin anti-dyskinetic effects were related to the inhibition of the Ras-ERK pathway. However, the mechanisms responsible for the anti-LID effect are unclear. Changes in cholesterol homeostasis and oxidative stress- and inflammation-related mechanisms such as angiotensin II and Rho-kinase (ROCK) inhibition may be involved. The nigra and striatum of dyskinetic rats showed increased levels of cholesterol, ROCK, and the inflammatory marker IL-1ß, which were reduced by the angiotensin type-1 receptor (AT1) antagonist candesartan, simvastatin, and the ROCK inhibitor fasudil. As observed for LID, angiotensin II-induced, via AT1, increased levels of cholesterol and ROCK in the rat nigra and striatum. In cultured dopaminergic neurons, angiotensin II increased cholesterol biosynthesis and cholesterol efflux without changes in cholesterol uptake. In astrocytes, angiotensin induced an increase in cholesterol uptake, decrease in biosynthesis, and no change in cholesterol efflux, suggesting a neuronal accumulation of cholesterol that is reduced via transfer to astrocytes. Our data suggest mutual interactions between angiotensin/AT1, cholesterol, and ROCK pathways in LID, which are attenuated by the corresponding inhibitors. Interestingly, these three drugs have also been suggested as neuroprotective treatments against Parkinson's disease. Therefore, they may reduce dyskinesia and the progression of the disease using common mechanisms.

3.
J Neurochem ; 157(6): 1789-1808, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32931038

RESUMO

Pannexin-1 (Panx1) forms plasma membrane channels that allow the exchange of small molecules between the intracellular and extracellular compartments, and are involved in diverse physiological and pathological responses in the nervous system. However, the signaling mechanisms that induce their opening still remain elusive. Here, we propose a new mechanism for Panx1 channel activation through a functional crosstalk with the highly Ca2+ permeable α7 nicotinic acetylcholine receptor (nAChR). Consistent with this hypothesis, we found that activation of α7 nAChRs induces Panx1-mediated dye uptake and ATP release in the neuroblastoma cell line SH-SY5Y-α7. Using membrane permeant Ca2+ chelators, total internal reflection fluorescence microscopy in SH-SY5Y-α7 cells expressing a membrane-tethered GCAMP3, and Src kinase inhibitors, we further demonstrated that Panx1 channel opening depends on Ca2+ signals localized in submembrane areas, as well as on Src kinases. In turn, Panx1 channels amplify cytosolic Ca2+ signals induced by the activation of α7 nAChRs, by a mechanism that seems to involve ATP release and P2X7 receptor activation, as hydrolysis of extracellular ATP with apyrase or blockage of P2X7 receptors with oxidized ATP significantly reduces the α7 nAChR-Ca2+ signal. The physiological relevance of this crosstalk was also demonstrated in neuroendocrine chromaffin cells, wherein Panx1 channels and P2X7 receptors contribute to the exocytotic release of catecholamines triggered by α7 nAChRs, as measured by amperometry. Together these findings point to a functional coupling between α7 nAChRs, Panx1 channels and P2X7 receptors with physiological relevance in neurosecretion.


Assuntos
Células Cromafins/metabolismo , Conexinas/metabolismo , Exocitose/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Receptor Cross-Talk/fisiologia , Receptores Purinérgicos P2X7/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Animais , Quelantes de Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Bovinos , Linhagem Celular Tumoral , Células Cromafins/efeitos dos fármacos , Exocitose/efeitos dos fármacos , Humanos , Camundongos , Receptor Cross-Talk/efeitos dos fármacos
4.
J Gerontol A Biol Sci Med Sci ; 75(3): 416-424, 2020 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-30412227

RESUMO

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.


Assuntos
Angiotensina II/fisiologia , Doenças Neurodegenerativas/etiologia , Estresse Oxidativo , Receptor Tipo 1 de Angiotensina/fisiologia , Sirtuína 3/metabolismo , Substância Negra/química , Substância Negra/metabolismo , Fatores Etários , Animais , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley , Sirtuína 3/análise
5.
J Neurochem ; 151(6): 703-715, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31418818

RESUMO

ß-Subunits of the Ca2+ channel have been conventionally regarded as auxiliary subunits that regulate the expression and activity of the pore-forming α1 subunit. However, they comprise protein-protein interaction domains, such as a SRC homology 3 domain (SH3) domain, which make them potential signaling molecules. Here we evaluated the role of the ß2a subunit of the Ca2+ channels (CaV ß2a) and its SH3 domain (ß2a-SH3) in late stages of channel trafficking in bovine adrenal chromaffin cells. Cultured bovine adrenal chromaffin cells were injected with CaV ß2a or ß2a-SH3 under different conditions, in order to acutely interfere with endogenous associations of these proteins. As assayed by whole-cell patch clamp recordings, Ca2+ currents were reduced by CaV ß2a in the presence of exogenous α1-interaction domain. ß2a-SH3, but not its dimerization-deficient mutant, also reduced Ca2+ currents. Na+ currents were also diminished following ß2a-SH3 injection. Furthermore, ß2a-SH3 was still able to reduce Ca2+ currents when dynamin-2 function was disrupted, but not when SNARE-dependent exocytosis or actin polymerization was inhibited. Together with the additional finding that both CaV ß2a and ß2a-SH3 diminished the incorporation of new actin monomers to cortical actin filaments, ß2a-SH3 emerges as a signaling module that might down-regulate forward trafficking of ion channels by modulating actin dynamics.


Assuntos
Actinas/metabolismo , Canais de Cálcio Tipo L/metabolismo , Células Cromafins/metabolismo , Regulação para Baixo/fisiologia , Domínios de Homologia de src/fisiologia , Animais , Bovinos , Células Cultivadas , Subunidades Proteicas/metabolismo , Transporte Proteico/fisiologia , Coelhos
6.
Oncotarget ; 9(13): 10834-10846, 2018 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-29541380

RESUMO

Gastrointestinal dysfunction is a common problem in the elderly. Aging-related changes in interactions between local dopaminergic and renin-angiotensin systems (RAS) have been observed in the brain, renal and vascular tissues. However, it is not known if these interactions also occur in the gut, and are dysregulated with aging. We showed a mutual regulation between the colonic dopaminergic system and RAS using young and aged mice deficient for major angiotensin and dopamine receptors. Aged rats showed a marked decrease in colonic dopamine D2 receptor expression, together with an increase in angiotensin type 1 (AT1) receptor expression, a decrease in angiotensin type 2 (AT2) receptor expression (i.e. an increase in the RAS pro-inflammatory arm activity), and increased levels of inflammatory and oxidative markers. Aged rats also showed increased levels of colonic dopamine and noradrenalin, and a marked decrease in acetylcholine and serotonin levels. The present observations contribute to explain an aging-related pro-inflammatory state and dysregulation in gastrointestinal function, which may be counteracted by treatment of aged animals with the AT1 receptor blocker candesartan.

7.
Mol Neurobiol ; 55(9): 7297-7316, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29404956

RESUMO

The exact mechanism of gut dysfunction in Parkinson's disease and, conversely, the role of gut pathology in brain dopaminergic degeneration are controversial. We investigated the effects of nigral lesions on the colonic neurotransmission, the effect of gut inflammation on the nigrostriatal dopaminergic function, and the possible involvement of the vagus nerve and the local renin-angiotensin system (RAS). Nigrostriatal dopamine depletion was performed by bilateral injection 6-hydroxydopamine, and gut inflammation was induced by dextran sulfate sodium salt treatment in rats and mice, respectively, with or without vagal disruption. A decrease in central dopamine levels induced a decrease in colonic dopamine types 1 and 2 receptor expression together with an increase in the colonic levels of dopamine and a decrease in the levels of acetylcholine, which may explain a decrease in gut motility. Central dopaminergic depletion also induced an increase in the colonic levels of inflammatory and oxidative stress markers together with activation of the pro-inflammatory arm of the local RAS. Mice with acute (1 week) or subchronic (3 weeks) gut inflammation did not show a significant increase in colonic α-synuclein and phosphorylated α-synuclein expression during this relatively short survival period. Interestingly, we observed early changes in the nigrostriatal dopaminergic homeostasis, dopaminergic neuron death, and increased levels of nigral pro-inflammatory markers and RAS pro-inflammatory activity. The present results show that a dysregulation of the neural bidirectional gut-brain interaction may explain the early gut disturbances observed in parkinsonian patients, and also the increase in vulnerability of nigral dopaminergic neurons after gut inflammation.


Assuntos
Dopamina/metabolismo , Trato Gastrointestinal/inervação , Trato Gastrointestinal/patologia , Doença de Parkinson/patologia , Animais , Colo/inervação , Colo/patologia , Modelos Animais de Doenças , Inflamação/patologia , Masculino , Camundongos Endogâmicos C57BL , Oxidopamina , Ratos Sprague-Dawley , Substância Negra/patologia , Tirosina 3-Mono-Oxigenase/metabolismo , Vagotomia
8.
Front Aging Neurosci ; 9: 129, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28515690

RESUMO

Microglia can transform into proinflammatory/classically activated (M1) or anti-inflammatory/alternatively activated (M2) phenotypes following environmental signals related to physiological conditions or brain lesions. An adequate transition from the M1 (proinflammatory) to M2 (immunoregulatory) phenotype is necessary to counteract brain damage. Several factors involved in microglial polarization have already been identified. However, the effects of the brain renin-angiotensin system (RAS) on microglial polarization are less known. It is well known that there is a "classical" circulating RAS; however, a second RAS (local or tissue RAS) has been observed in many tissues, including brain. The locally formed angiotensin is involved in local pathological changes of these tissues and modulates immune cells, which are equipped with all the components of the RAS. There are also recent data showing that brain RAS plays a major role in microglial polarization. Level of microglial NADPH-oxidase (Nox) activation is a major regulator of the shift between M1/proinflammatory and M2/immunoregulatory microglial phenotypes so that Nox activation promotes the proinflammatory and inhibits the immunoregulatory phenotype. Angiotensin II (Ang II), via its type 1 receptor (AT1), is a major activator of the NADPH-oxidase complex, leading to pro-oxidative and pro-inflammatory effects. However, these effects are counteracted by a RAS opposite arm constituted by Angiotensin II/AT2 receptor signaling and Angiotensin 1-7/Mas receptor (MasR) signaling. In addition, activation of prorenin-renin receptors may contribute to activation of the proinflammatory phenotype. Aged brains showed upregulation of AT1 and downregulation of AT2 receptor expression, which may contribute to a pro-oxidative pro-inflammatory state and the increase in neuron vulnerability. Several recent studies have shown interactions between the brain RAS and different factors involved in microglial polarization, such as estrogens, Rho kinase (ROCK), insulin-like growth factor-1 (IGF-1), tumor necrosis factor α (TNF)-α, iron, peroxisome proliferator-activated receptor gamma, and toll-like receptors (TLRs). Metabolic reprogramming has recently been involved in the regulation of the neuroinflammatory response. Interestingly, we have recently observed a mitochondrial RAS, which is altered in aged brains. In conclusion, dysregulation of brain RAS plays a major role in aging-related changes and neurodegeneration by exacerbation of oxidative stress (OS) and neuroinflammation, which may be attenuated by pharmacological manipulation of RAS components.

9.
Front Cell Neurosci ; 11: 130, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28522963

RESUMO

Upon cell stimulation, the network of cortical actin filaments is rearranged to facilitate the neurosecretory process. This actin rearrangement includes both disruption of the preexisting actin network and de novo actin polymerization. However, the mechanism by which a Ca2+ signal elicits the formation of new actin filaments remains uncertain. Cortactin, an actin-binding protein that promotes actin polymerization in synergy with the nucleation promoting factor N-WASP, could play a key role in this mechanism. We addressed this hypothesis by analyzing de novo actin polymerization and exocytosis in bovine adrenal chromaffin cells expressing different cortactin or N-WASP domains, or cortactin mutants that fail to interact with proline-rich domain (PRD)-containing proteins, including N-WASP, or to be phosphorylated by Ca2+-dependent kinases, such as ERK1/2 and Src. Our results show that the activation of nicotinic receptors in chromaffin cells promotes cortactin translocation to the cell cortex, where it colocalizes with actin filaments. We further found that, in association with PRD-containing proteins, cortactin contributes to the Ca2+-dependent formation of F-actin, and regulates fusion pore dynamics and the number of exocytotic events induced by activation of nicotinic receptors. However, whereas the actions of cortactin on the fusion pore dynamics seems to depend on the availability of monomeric actin and its phosphorylation by ERK1/2 and Src kinases, cortactin regulates the extent of exocytosis by a mechanism independent of actin polymerization. Together our findings point out a role for cortactin as a critical modulator of actin filament formation and exocytosis in neuroendocrine cells.

10.
Brain Behav Immun ; 62: 277-290, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28232171

RESUMO

Dopamine is an immunomodulatory molecule that acts on immune effector cells both in the CNS and peripheral tissues. However, the role of changes in dopamine levels in the neuroinflammatory response is controversial. The local/paracrine renin-angiotensin system (RAS) plays a major role in inflammatory processes in peripheral tissues and brain. In the present study, we investigated the possible role of the brain RAS in the effects of dopamine on the glial inflammatory responses. Astrocytes are the major source of the precursor protein angiotensinogen and angiotensin II (AII) in the brain. Neurotoxins such as MPP+ (1-methyl-4-phenylpyridinium) can act directly on astrocytes to increase levels of angiotensinogen and AII. Conversely, dopamine, via type-2 (D2) receptors, inhibited production of angiotensinogen, decreased expression of angiotensin type-1 (AT1) receptors and increased expression of AT2 receptors. In microglia, dopamine and dopamine agonists also regulated RAS activity. First, indirectly, via downregulation of the astrocyte-derived AII. Second, via dopamine-induced regulation of microglial angiotensin receptors. Dopamine decreased the microglial AT1/AT2 ratio leading to inhibition of the pro-inflammatory AT1/NADPH-oxidase/superoxide axis. D2 receptors were particularly responsible for microglial RAS inhibition in basal culture conditions. However, both D1 and D2 agonists inhibited the AT1/NADPH-oxidase axis in lipopolysaccharide-treated (LPS; i.e. activated) microglia. The results indicate that the decrease in dopamine levels observed in early stages of Parkinson's disease and aging may promote neuroinflammation and disease progression via glial RAS exacerbation.


Assuntos
Angiotensinogênio/metabolismo , Astrócitos/efeitos dos fármacos , Dopamina/farmacologia , Microglia/efeitos dos fármacos , Sistema Renina-Angiotensina/efeitos dos fármacos , 1-Metil-4-fenilpiridínio/farmacologia , Animais , Animais Recém-Nascidos , Astrócitos/metabolismo , Benzazepinas/farmacologia , Células Cultivadas , Antagonistas de Dopamina/farmacologia , Microglia/metabolismo , Ratos , Ratos Sprague-Dawley , Salicilamidas/farmacologia
11.
Front Neuroendocrinol ; 43: 44-59, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27693730

RESUMO

The neuroprotective effects of menopausal hormonal therapy in Parkinson's disease (PD) have not yet been clarified, and it is controversial whether there is a critical period for neuroprotection. Studies in animal models and clinical and epidemiological studies indicate that estrogens induce dopaminergic neuroprotection. Recent studies suggest that inhibition of the brain renin-angiotensin system (RAS) mediates the effects of estrogens in PD models. In the substantia nigra, ovariectomy induces a decrease in levels of estrogen receptor-α (ER-α) and increases angiotensin activity, NADPH-oxidase activity and expression of neuroinflammatory markers, which are regulated by estrogen replacement therapy. There is a critical period for the neuroprotective effect of estrogen replacement therapy, and local ER-α and RAS play a major role. Astrocytes play a major role in ER-α-induced regulation of local RAS, but neurons and microglia are also involved. Interestingly, treatment with angiotensin receptor antagonists after the critical period induced neuroprotection.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Estrogênios/metabolismo , Menopausa/metabolismo , Degeneração Neural/metabolismo , Fármacos Neuroprotetores/metabolismo , Doença de Parkinson/metabolismo , Sistema Renina-Angiotensina/fisiologia , Animais , Humanos
12.
Eur J Immunol ; 45(9): 2615-27, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26115479

RESUMO

Delta-like protein 1 (DLK1) is a noncanonical ligand that inhibits NOTCH1 receptor activity and regulates multiple differentiation processes. In macrophages, NOTCH signaling increases TLR-induced expression of key pro-inflammatory mediators. We have investigated the role of DLK1 in macrophage activation and inflammation using Dlk1-deficient mice and Raw 264.7 cells overexpressing Dlk1. In the absence of Dlk1, NOTCH1 expression is increased and the activation of macrophages with TLR3 or TLR4 agonists leads to higher production of IFN-ß and other pro-inflammatory cytokines, including TNF-α, IL-12, and IL-23. The expression of key proteins involved in IFN-ß signaling, such as IRF3, IRF7, IRF1, or STAT1, as well as cRel, or RelB, which are responsible for the generation of IL-12 and IL-23, is enhanced in Dlk1 KO macrophages. Consistently, Dlk1 KO mice are more sensitive to LPS-induced endotoxic shock. These effects seem to be mediated through the modulation of NOTCH1 signaling. TLR4 activation reduces DLK1 expression, whereas increases NOTCH1 levels. In addition, DLK1 expression diminishes during differentiation of human U937 cells to macrophages. Overall, these results reveal a novel role for DLK1 as a regulator of NOTCH-mediated, pro-inflammatory macrophage activation, which could help to ensure a baseline level preventing constant tissue inflammation.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Macrófagos/imunologia , Receptor Notch1/imunologia , Receptor 3 Toll-Like/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Proteínas de Ligação ao Cálcio , Diferenciação Celular , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/imunologia , Interferon beta/genética , Interferon beta/imunologia , Interleucina-12/genética , Interleucina-12/imunologia , Interleucina-23/genética , Interleucina-23/imunologia , Ativação de Macrófagos , Macrófagos/citologia , Camundongos , Camundongos Knockout , Receptor Notch1/genética , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/imunologia , Transdução de Sinais , Receptor 3 Toll-Like/genética , Receptor 4 Toll-Like/genética , Fator de Transcrição RelB/genética , Fator de Transcrição RelB/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Células U937
13.
Neuroscientist ; 21(6): 616-29, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25323761

RESUMO

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.


Assuntos
Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/enzimologia , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/enzimologia , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo , Animais , Antiparkinsonianos/farmacologia , Antiparkinsonianos/uso terapêutico , Humanos , Degeneração Neural/tratamento farmacológico , Degeneração Neural/enzimologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico
14.
Glia ; 63(3): 466-82, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25377425

RESUMO

Previous studies have shown that the brain renin-angiotensin system may play a major role, via angiotensin type 1 (AT1) receptors, in the regulation of neuroinflammation, oxidative stress and progression of dopaminergic degeneration. Angiotensin-induced activation of the microglial nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase complex and microglial Rho-kinase are particularly important in this respect. However, it is not known whether crosstalk between Rho-kinase and NADPH-oxidase leads to microglial activation. In the present study, we found that, in the substantia nigra of rats, NADPH-oxidase activation was involved in angiotensin-induced Rho-kinase activation, which, in turn, was involved in angiotensin-induced NADPH-oxidase activation. In N9 microglial cell line and primary microglial cultures, a crosstalk signaling between NADPH-oxidase and Rho-kinase occurred in a positive feedback fashion during angiotensin-induced microglial activation. Angiotensin-induced NADPH-oxidase activation and superoxide generation led to NF-кB translocation and Rho-kinase activation. Rho-kinase activation was involved in regulation of NADPH-oxidase activation via p38 mitogen-activated protein kinase. Moreover, Rho-kinase activation, via NF-кB, upregulated AT1 receptor expression in microglial cells through a feed-forward mechanism. NADPH-oxidase and Rho-kinase pathways are known to be responsible for major components of the microglial response, such as changes involving microglial motility and phagocytosis, generation of superoxide, and release of inflammatory cytokines. The present results show that both pathways are linked by a common mechanism that may constitute a basic means of coordinating the microglial response.


Assuntos
Angiotensina II/metabolismo , Microglia/enzimologia , NADPH Oxidases/metabolismo , Quinases Associadas a rho/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Masculino , Camundongos , NF-kappa B/metabolismo , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/metabolismo , Substância Negra/imunologia , Superóxidos/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Neuropharmacology ; 85: 1-8, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24878243

RESUMO

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+.


Assuntos
Amidas/farmacologia , Morte Celular/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Intoxicação por MPTP/tratamento farmacológico , Microglia/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Piridinas/farmacologia , Animais , Axônios/efeitos dos fármacos , Axônios/patologia , Axônios/fisiologia , Técnicas de Cultura de Células , Morte Celular/fisiologia , Linhagem Celular Tumoral , Células Cultivadas , Neurônios Dopaminérgicos/patologia , Neurônios Dopaminérgicos/fisiologia , Intoxicação por MPTP/patologia , Intoxicação por MPTP/fisiopatologia , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/patologia , Mesencéfalo/fisiopatologia , Microglia/enzimologia , Neuritos/efeitos dos fármacos , Neuritos/patologia , Neuritos/fisiologia , Ratos , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo
16.
Glia ; 62(1): 145-57, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24272709

RESUMO

In vitro and in vivo models of Parkinson's disease were used to investigate whether TNF-α plays a major role in the enhancement of the microglial response and dopaminergic degeneration induced by brain angiotensin hyperactivity. Treatment of primary mesencephalic cultures with low doses of the neurotoxin MPP(+) induced a significant loss of dopaminergic neurons, which was enhanced by cotreatment with angiotensin II and inhibited by TNF-α inhibitors. Treatment of primary cultures with angiotensin induced a marked increase in levels of TNF-α, which was inhibited by treatment with angiotensin type-1-receptor antagonists, NADPH-oxidase inhibitors and NFK-ß inhibitors. However, TNF-α levels were not significantly affected by treatment with angiotensin in the absence of microglia. The microglial origin of the angiotensin-induced increase in TNF-α levels was confirmed using dopaminergic (MES 23.5) and microglial (N9) cell lines. Inhibition of the microglial Rho-kinase activity also blocked the AII-induced increase in TNF-α levels. Treatment of the dopaminergic cell line with TNF-α revealed that NFK-ß activation mediates the deleterious effect of microglial TNF-α on dopaminergic neurons. Treatment of mice with MPTP also induced significant increases in striatal and nigral TNF-α levels, which were inhibited by angiotensin type-1-receptor antagonists or NFK-ß inhibitors. The present results show that microglial TNF-α plays a major role in angiotensin-induced dopaminergic cell death and that the microglial release of TNF-α is mediated by activation of angiotensin type-1 receptors, NADPH-oxidase, Rho-kinase and NFK-ß.


Assuntos
Neurônios Dopaminérgicos/patologia , Calicreínas/metabolismo , Microglia/metabolismo , Degeneração Neural/fisiopatologia , Extratos Pancreáticos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Angiotensina II/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Benzimidazóis/farmacologia , Compostos de Bifenilo , Células Cultivadas , CMP Cíclico/análogos & derivados , CMP Cíclico/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Masculino , Mesencéfalo/citologia , Camundongos , Camundongos Endogâmicos C57BL , Degeneração Neural/induzido quimicamente , Degeneração Neural/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Neurotoxinas/farmacologia , Tetrazóis/farmacologia
17.
Neuropharmacology ; 76 Pt A: 156-68, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23973568

RESUMO

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'.


Assuntos
Antagonistas de Receptores de Angiotensina/farmacologia , Antagonistas de Receptores de Angiotensina/uso terapêutico , Corpo Estriado/efeitos dos fármacos , Dopamina/metabolismo , Levodopa/farmacologia , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson Secundária/tratamento farmacológico , Animais , Benzimidazóis/farmacologia , Benzimidazóis/uso terapêutico , Compostos de Bifenilo , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Levodopa/uso terapêutico , Masculino , Atividade Motora/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Oxidopamina/toxicidade , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/metabolismo , Ratos , Receptor 5-HT1B de Serotonina/metabolismo , Receptores Dopaminérgicos/metabolismo , Serotonina/metabolismo , Tetrazóis/farmacologia , Tetrazóis/uso terapêutico
18.
Exp Neurol ; 250: 384-96, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24184051

RESUMO

Dysfunction of iron homeostasis has been shown to be involved in ageing, Parkinson's disease and other neurodegenerative diseases. Increased levels of labile iron result in increased reactive oxygen species and oxidative stress. Angiotensin II, via type-1 receptors, exacerbates oxidative stress, the microglial inflammatory response and progression of dopaminergic degeneration. Angiotensin activates the NADPH-oxidase complex, which produces superoxide. However, it is not known whether angiotensin affects iron homeostasis. In the present study, administration of angiotensin to primary mesencephalic cultures, the dopaminergic cell line MES23.5 and to young adult rats, significantly increased levels of transferrin receptors, divalent metal transporter-1 and ferroportin, which suggests an increase in iron uptake and export. In primary neuron-glia cultures and young rats, angiotensin did not induce significant changes in levels of ferritin or labile iron, both of which increased in neurons in the absence of glia (neuron-enriched cultures, dopaminergic cell line) and in the N9 microglial cell line. In aged rats, which are known to display high levels of angiotensin activity, ferritin levels and iron deposits in microglial cells were enhanced. Angiotensin-induced changes were inhibited by angiotensin type-1 receptor antagonists, NADPH-oxidase inhibitors, antioxidants and NF-kB inhibitors. The results demonstrate that angiotensin, via type-1 receptors, modulates iron homeostasis in dopaminergic neurons and microglial cells, and that glial cells play a major role in efficient regulation of iron homeostasis in dopaminergic neurons.


Assuntos
Angiotensina II/metabolismo , Neurônios Dopaminérgicos/metabolismo , Homeostase/fisiologia , Ferro/metabolismo , Microglia/metabolismo , Angiotensina II/farmacologia , Animais , Western Blotting , Encéfalo/metabolismo , Células Cultivadas , Imunofluorescência , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Angiotensina/metabolismo
19.
Neurobiol Dis ; 58: 209-19, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23774254

RESUMO

The mechanism by which estrogen protects dopaminergic neurons has not yet been clarified. It is not known if changes in RhoA/Rho kinase activity are involved in the enhanced vulnerability of dopaminergic neurons observed after estrogen depletion. The present study shows that the MPTP-induced loss of dopaminergic neurons is increased by estrogen depletion and inhibited by estrogen replacement, the Rho kinase inhibitor Y27632 and deletion of the angiotensin type-1 receptor. In ovariectomized mice, treatment with MPTP induced a marked increase in Rho kinase activity, and RhoA and RhocK II mRNA and protein expression, which were significantly higher than in ovariectomized mice treated with MPTP and estrogen replacement or type-1 receptor deletion. Estrogen depletion increased Rho kinase activity, via enhancement of the angiotensin type-1 receptor pathway, and Rho kinase activation increased type-1 receptor expression suggesting a vicious cycle in which Rho kinase and type-1 receptor activate each other and promote the degenerative process. The results suggest that type-1 receptor antagonists and Rho kinase inhibitors may provide a new neuroprotective strategy, which may circumvent the potential risks of estrogen replacement therapy and be particularly useful in elderly women or women affected by long-term lack of estrogen.


Assuntos
Estradiol/uso terapêutico , Intoxicação por MPTP/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Quinases Associadas a rho/metabolismo , Amidas/administração & dosagem , Bloqueadores do Receptor Tipo 1 de Angiotensina II/administração & dosagem , Animais , Benzimidazóis/administração & dosagem , Compostos de Bifenilo , Modelos Animais de Doenças , Inibidores Enzimáticos/administração & dosagem , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Ovariectomia , Piridinas/administração & dosagem , Receptor Tipo 1 de Angiotensina/deficiência , Tetrazóis/administração & dosagem , Tirosina 3-Mono-Oxigenase/metabolismo , Quinases Associadas a rho/antagonistas & inibidores
20.
Rev. cuba. plantas med ; 18(1): 71-83, ene.-mar. 2013.
Artigo em Espanhol | LILACS | ID: lil-667492

RESUMO

Introducción: la decocción de partes aéreas frescas de Phania matricarioides (Spreng. ) Griseb (manzanilla), se emplea tradicionalmente en Cuba para afecciones digestivas como malas digestiones y diarrea aguda simple; no se encontraron estudios de validación preclínica del efecto antidiarreico y su seguridad. Objetivos: evaluar la acción sobre el tránsito intestinal y la toxicología aguda oral y tópica en modelos preclínicos de la decocción de partes aéreas frescas de Phania matricarioides. Métodos: se colectaron las partes aéreas frescas de Phania matricarioides y se realizó decocción (30 y 50 por ciento). Se aplicó el modelo experimental: tránsito intestinal en ratones con una sola administración de la decocción al 30 por ciento en dosis de 1,0, 5,0 y 10,0 g de material vegetal/kg de peso corporal por 1 día; y en dosis de 1,0 y 5,0 g de material vegetal/kg de peso corporal por 4 días. El estudio toxicológico oral y tópico (decocción 50 por ciento) se efectuó en los modelos: clases tóxicas agudas y toxicidad dérmica aguda en ratas con dosis de 2 000 mg/kg de peso corporal e irritabilidad dérmica primaria en conejos. Resultados: la decocción administrada por 1 día no modificó de forma significativa el tránsito intestinal, la administración por 4 días disminuyó de forma significativa y dosis dependiente el tránsito intestinal (5,0 g/kg). en el estudio toxicológico no se produjo ninguna muerte, no se evidenciaron signos de toxicidad ni lesiones macroscópicas en los órganos de las ratas, el aumento de peso fue el esperado. El índice de irritación primaria reflejó 0. Conclusiones: los resultados permiten validar el efecto antidiarreico de la decocción de partes aéreas frescas de Phania matricarioides para afecciones digestivas y no clasifica como tóxico


Introduction: Phania matricarioides (Spreng. ) Griseb (chamomile) fresh aerial part decoction is traditionally used in Cuba to treat digestive disorders as upset stomach and simple acute diarrheas. However, there were no previous preclinical validation studies on the antidiarrheal effect and safety of this species. Objectives: to evaluate the action of decoction from Phania matricarioides fresh aerial parts on the intestinal transit and the oral and topical acute toxicology in preclinical models. Methods: the fresh aerial parts of this plant were harvested and decoction was obtained (30 and 50 percent). The experimental model of intestinal transit in mice, with 30 percent decoction being administrated once at doses of 1.0, 5.0 and 10.0 g of vegetal material/kg of bodyweight for one day, and at doses of 1.0 and 5.0 g/kg for 4 days, was applied. The oral and topical toxicological study (50 percent decoction) was conducted in the models acute-toxic and acute dermal toxic classes in rats at a dose of 2000 mg/kg of bodyweight and primary dermal irritability in rabbits. Results: the decoction administered for one day did not significantly change the intestinal transit, but the administration for 4 days did significantly change, depending on dose, the intestinal transit (5.0 g/kg). There was no death in the study, there were neither signs of toxicity nor macroscopic lesions in the rats' organs, whereas the weight gain behaved as expected. The index of primary irritation was null. Conclusions: the results allow validating the antidiarrheal effect of Phania matricarioides fresh aerial parts decoction on digestive disorders and it is not toxic


Assuntos
Antidiarreicos , Camomila/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...