Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
J Neuroinflammation ; 21(1): 54, 2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38383421

RESUMO

Parkinson's disease (PD) is a common age-related neurodegenerative disorder characterized by the aggregation of α-Synuclein (αSYN) building up intraneuronal inclusions termed Lewy pathology. Mounting evidence suggests that neuron-released αSYN aggregates could be central to microglial activation, which in turn mounts and orchestrates neuroinflammatory processes potentially harmful to neurons. Therefore, understanding the mechanisms that drive microglial cell activation, polarization and function in PD might have important therapeutic implications. Here, using primary microglia, we investigated the inflammatory potential of pure αSYN fibrils derived from PD patients. We further explored and characterized microglial cell responses to a chronic-type inflammatory stimulation combining PD patient-derived αSYN fibrils (FPD), Tumor necrosis factor-α (TNFα) and prostaglandin E2 (PGE2) (TPFPD). We showed that FPD hold stronger inflammatory potency than pure αSYN fibrils generated de novo. When combined with TNFα and PGE2, FPD polarizes microglia toward a particular functional phenotype departing from FPD-treated cells and featuring lower inflammatory cytokine and higher glutamate release. Whereas metabolomic studies showed that TPFPD-exposed microglia were closely related to classically activated M1 proinflammatory cells, notably with similar tricarboxylic acid cycle disruption, transcriptomic analysis revealed that TPFPD-activated microglia assume a unique molecular signature highlighting upregulation of genes involved in glutathione and iron metabolisms. In particular, TPFPD-specific upregulation of Slc7a11 (which encodes the cystine-glutamate antiporter xCT) was consistent with the increased glutamate response and cytotoxic activity of these cells toward midbrain dopaminergic neurons in vitro. Together, these data further extend the structure-pathological relationship of αSYN fibrillar polymorphs to their innate immune properties and demonstrate that PD-derived αSYN fibrils, TNFα and PGE2 act in concert to drive microglial cell activation toward a specific and highly neurotoxic chronic-type inflammatory phenotype characterized by robust glutamate release and iron retention.


Assuntos
Síndromes Neurotóxicas , Doença de Parkinson , Humanos , Doença de Parkinson/patologia , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Microglia/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Sinais (Psicologia) , Inflamação/metabolismo , Neurônios Dopaminérgicos/patologia , Síndromes Neurotóxicas/metabolismo , Glutamatos/metabolismo , Ferro/metabolismo
2.
J Neural Transm (Vienna) ; 131(1): 1-11, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37851107

RESUMO

Over the years, evidence has accumulated on a possible contributive role of the cytosolic quinone reductase NQO2 in models of dopamine neuron degeneration induced by parkinsonian toxin, but most of the data have been obtained in vitro. For this reason, we asked the question whether NQO2 is involved in the in vivo toxicity of MPTP, a neurotoxin classically used to model Parkinson disease-induced neurodegeneration. First, we show that NQO2 is expressed in mouse substantia nigra dopaminergic cell bodies and in human dopaminergic SH-SY5Y cells as well. A highly specific NQO2 inhibitor, S29434, was able to reduce MPTP-induced cell death in a co-culture system of SH-SY5Y cells with astrocytoma U373 cells but was inactive in SH-SY5Y monocultures. We found that S29434 only marginally prevents substantia nigra tyrosine hydroxylase+ cell loss after MPTP intoxication in vivo. The compound produced a slight increase of dopaminergic cell survival at day 7 and 21 following MPTP treatment, especially with 1.5 and 3 mg/kg dosage regimen. The rescue effect did not reach statistical significance (except for one experiment at day 7) and tended to decrease with the 4.5 mg/kg dose, at the latest time point. Despite the lack of robust protective activity of the inhibitor of NQO2 in the mouse MPTP model, we cannot rule out a possible role of the enzyme in parkinsonian degeneration, particularly because it is substantially expressed in dopaminergic neurons.


Assuntos
Intoxicação por MPTP , Neuroblastoma , Camundongos , Humanos , Animais , Neurônios Dopaminérgicos/metabolismo , Substância Negra/metabolismo , Dopamina/metabolismo , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças
3.
Neurobiol Dis ; 151: 105256, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33429042

RESUMO

Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are neurodegenerative disorders characterized by the misfolding and aggregation of alpha-synuclein (aSyn). Doxycycline, a tetracyclic antibiotic shows neuroprotective effects, initially proposed to be due to its anti-inflammatory properties. More recently, an additional mechanism by which doxycycline may exert its neuroprotective effects has been proposed as it has been shown that it inhibits amyloid aggregation. Here, we studied the effects of doxycycline on aSyn aggregation in vivo, in vitro and in a cell free system using real-time quaking induced conversion (RT-QuiC). Using H4, SH-SY5Y and HEK293 cells, we found that doxycycline decreases the number and size of aSyn aggregates in cells. In addition, doxycycline inhibits the aggregation and seeding of recombinant aSyn, and attenuates the production of mitochondrial-derived reactive oxygen species. Finally, we found that doxycycline induces a cellular redistribution of aggregates in a C.elegans animal model of PD, an effect that is associated with a recovery of dopaminergic function. In summary, we provide strong evidence that doxycycline treatment may be an effective strategy against synucleinopathies.


Assuntos
Doxiciclina/farmacologia , Fármacos Neuroprotetores/farmacologia , Agregação Patológica de Proteínas/patologia , Sinucleinopatias/patologia , alfa-Sinucleína/efeitos dos fármacos , Animais , Caenorhabditis elegans , Linhagem Celular , Humanos , Corpos de Inclusão/efeitos dos fármacos , Corpos de Inclusão/metabolismo
4.
Glia ; 68(3): 561-573, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31647138

RESUMO

We used mouse microglial cells in culture activated by lipopolysaccharide (LPS, 10 ng/ml) to study the anti-inflammatory potential of cannabidiol (CBD), the major nonpsychoactive component of cannabis. Under LPS stimulation, CBD (1-10 µM) potently inhibited the release of prototypical proinflammatory cytokines (TNF-α and IL-1ß) and that of glutamate, a noncytokine mediator of inflammation. The effects of CBD were predominantly receptor-independent and only marginally blunted by blockade of CB2 receptors. We established that CBD inhibited a mechanism involving, sequentially, NADPH oxidase-mediated ROS production and NF-κB-dependent signaling events. In line with these observations, active concentrations of CBD demonstrated an intrinsic free-radical scavenging capacity in the cell-free DPPH assay. Of interest, CBD also prevented the rise in glucose uptake observed in microglial cells challenged with LPS, as did the inhibitor of NADPH oxidase apocynin and the inhibitor of IκB kinase-2, TPCA-1. This indicated that the capacity of CBD to prevent glucose uptake also contributed to its anti-inflammatory activity. Supporting this view, the glycolytic inhibitor 2-deoxy-d-glucose (2-DG) mimicked the antioxidant/immunosuppressive effects of CBD. Interestingly, CBD and 2-DG, as well as apocynin and TPCA-1 caused a reduction in glucose-derived NADPH, a cofactor required for NADPH oxidase activation and ROS generation. These different observations suggest that CBD exerts its anti-inflammatory effects towards microglia through an intrinsic antioxidant effect, which is amplified through inhibition of glucose-dependent NADPH synthesis. These results also further confirm that CBD may have therapeutic utility in conditions where neuroinflammatory processes are prominent.


Assuntos
Canabidiol/farmacologia , Glucose/metabolismo , Inflamação/prevenção & controle , Microglia/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Citocinas/farmacologia , Proteínas I-kappa B/efeitos dos fármacos , Inflamação/tratamento farmacológico , Lipopolissacarídeos/farmacologia , Camundongos , Microglia/metabolismo , Transdução de Sinais/efeitos dos fármacos
5.
Mol Pharmacol ; 95(3): 269-285, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30567956

RESUMO

Quinone reductase 2 (QR2, E.C. 1.10.5.1) is an enzyme with a feature that has attracted attention for several decades: in standard conditions, instead of recognizing NAD(P)H as an electron donor, it recognizes putative metabolites of NADH, such as N-methyl- and N-ribosyl-dihydronicotinamide. QR2 has been particularly associated with reactive oxygen species and memory, strongly suggesting a link among QR2 (as a possible key element in pro-oxidation), autophagy, and neurodegeneration. In molecular and cellular pharmacology, understanding physiopathological associations can be difficult because of a lack of specific and powerful tools. Here, we present a thorough description of the potent, nanomolar inhibitor [2-(2-methoxy-5H-1,4b,9-triaza(indeno[2,1-a]inden-10-yl)ethyl]-2-furamide (S29434 or NMDPEF; IC50 = 5-16 nM) of QR2 at different organizational levels. We provide full detailed syntheses, describe its cocrystallization with and behavior at QR2 on a millisecond timeline, show that it penetrates cell membranes and inhibits QR2-mediated reactive oxygen species (ROS) production within the 100 nM range, and describe its actions in several in vivo models and lack of actions in various ROS-producing systems. The inhibitor is fairly stable in vivo, penetrates cells, specifically inhibits QR2, and shows activities that suggest a key role for this enzyme in different pathologic conditions, including neurodegenerative diseases.


Assuntos
Piridinas/farmacologia , Alcaloides de Pirrolizidina/farmacologia , Quinona Redutases/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Hep G2 , Humanos , Masculino , Camundongos , NAD(P)H Desidrogenase (Quinona)/metabolismo , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
6.
Glia ; 66(11): 2353-2365, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30394585

RESUMO

When activated, microglial cells have the potential not only to secrete typical proinflammatory mediators but also to release the neurotransmitter glutamate in amounts that may promote excitotoxicity. Here, we wished to determine the potential of the Parkinson's disease (PD) protein α-Synuclein (αS) to stimulate glutamate release using cultures of purified microglial cells. We established that glutamate release was robustly increased when microglial cultures were treated with fibrillary aggregates of αS but not with the native monomeric protein. Promotion of microglial glutamate release by αS aggregates (αSa) required concomitant engagement of TLR2 and P2X7 receptors. Downstream to cell surface receptors, the release process was mediated by activation of a signaling cascade sequentially involving phosphoinositide 3-kinase (PI3K) and NADPH oxidase, a superoxide-producing enzyme. Inhibition of the Xc- antiporter, a plasma membrane exchange system that imports extracellular l-cystine and exports intracellular glutamate, prevented the release of glutamate induced by αSa, indicating that system Xc- was the final effector element in the release process downstream to NADPH oxidase activation. Of interest, the stimulation of glutamate release by αSa was abrogated by dopamine through an antioxidant effect requiring D1 dopamine receptor activation and PI3K inhibition. Altogether, present data suggest that the activation of microglial cells by αSa may possibly result in a toxic build-up of extracellular glutamate contributing to excitotoxic stress in PD. The deficit in dopamine that characterizes this disorder may further aggravate this process in a vicious circle mechanism.


Assuntos
Dopamina/farmacologia , Ácido Glutâmico/metabolismo , Microglia/efeitos dos fármacos , Agregados Proteicos/efeitos dos fármacos , alfa-Sinucleína/metabolismo , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Animais , Animais Recém-Nascidos , Anticorpos Monoclonais/farmacologia , Isótopos de Carbono/farmacocinética , Células Cultivadas , Cistina/farmacocinética , Humanos , Lipopeptídeos/farmacologia , Lipopolissacarídeos/farmacologia , Camundongos , Microglia/ultraestrutura , Inibidores da Agregação Plaquetária/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor 2 Toll-Like/imunologia , alfa-Sinucleína/farmacologia
7.
Glia ; 66(8): 1736-1751, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29665074

RESUMO

Neuroinflammation and mitochondrial dysfunction, key mechanisms in the pathogenesis of Parkinson's disease (PD), are usually explored independently. Loss-of-function mutations of PARK2 and PARK6, encoding the E3 ubiquitin protein ligase Parkin and the mitochondrial serine/threonine kinase PINK1, account for a large proportion of cases of autosomal recessive early-onset PD. PINK1 and Parkin regulate mitochondrial quality control and have been linked to the modulation of innate immunity pathways. We report here an exacerbation of NLRP3 inflammasome activation by specific inducers in microglia and bone marrow-derived macrophages from Park2-/- and Pink1-/- mice. The caspase 1-dependent release of IL-1ß and IL-18 was, therefore, enhanced in Park2-/- and Pink1-/- cells. This defect was confirmed in blood-derived macrophages from patients with PARK2 mutations and was reversed by MCC950, which specifically inhibits NLRP3 inflammasome complex formation. Enhanced NLRP3 signaling in Parkin-deficient cells was accompanied by a lack of induction of A20, a well-known negative regulator of the NF-κB pathway recently shown to attenuate NLRP3 inflammasome activity. We also found an inverse correlation between A20 abundance and IL-1ß release, in human macrophages challenged with NLRP3 inflammasome inducers. Overall, our observations suggest that the A20/NLRP3-inflammasome axis participates in the pathogenesis of PARK2-linked PD, paving the way for the exploration of its potential as a biomarker and treatment target.


Assuntos
Retroalimentação Fisiológica/fisiologia , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Ubiquitina-Proteína Ligases/deficiência , Adulto , Humanos , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Microglia/metabolismo , Pessoa de Meia-Idade , Mitocôndrias/metabolismo , NF-kappa B/metabolismo
8.
Hum Mol Genet ; 25(14): 2972-2984, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27206984

RESUMO

Mutations in PARK2, encoding the E3 ubiquitin protein ligase Parkin, are a common cause of autosomal recessive Parkinson's disease (PD). Loss of PARK2 function compromises mitochondrial quality by affecting mitochondrial biogenesis, bioenergetics, dynamics, transport and turnover. We investigated the impact of PARK2 dysfunction on the endoplasmic reticulum (ER)-mitochondria interface, which mediates calcium (Ca2+) exchange between the two compartments and is essential for Parkin-dependent mitophagy. Confocal and electron microscopy analyses showed the ER and mitochondria to be in closer proximity in primary fibroblasts from PARK2 knockout (KO) mice and PD patients with PARK2 mutations than in controls. Ca2+ flux to the cytosol was also modified, due to enhanced ER-to-mitochondria Ca2+ transfers, a change that was also observed in neurons derived from induced pluripotent stem cells of a patient with PARK2 mutations. Subcellular fractionation showed the abundance of the Parkin substrate mitofusin 2 (Mfn2), which is known to modulate the ER-mitochondria interface, to be specifically higher in the mitochondrion-associated ER membrane compartment in PARK2 KO tissue. Mfn2 downregulation or the exogenous expression of normal Parkin restored cytosolic Ca2+ transients in fibroblasts from patients with PARK2 mutations. In contrast, a catalytically inactive PD-related Parkin variant had no effect. Overall, our data suggest that Parkin is directly involved in regulating ER-mitochondria contacts and provide new insight into the role of the loss of Parkin function in PD development.


Assuntos
Retículo Endoplasmático/metabolismo , GTP Fosfo-Hidrolases/genética , Mitocôndrias/metabolismo , Doença de Parkinson/genética , Ubiquitina-Proteína Ligases/genética , Animais , Sinalização do Cálcio/genética , Citosol/metabolismo , Retículo Endoplasmático/patologia , Fibroblastos , GTP Fosfo-Hidrolases/biossíntese , Regulação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias/patologia , Mitofagia/genética , Mutação , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia
9.
J Neurochem ; 142(1): 14-28, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28398653

RESUMO

Despite its low chemical reactivity, the noble gas xenon possesses a remarkable spectrum of biological effects. In particular, xenon is a strong neuroprotectant in preclinical models of hypoxic-ischemic brain injury. In this study, we wished to determine whether xenon retained its neuroprotective potential in experimental settings that model the progressive loss of midbrain dopamine (DA) neurons in Parkinson's disease. Using rat midbrain cultures, we established that xenon was partially protective for DA neurons through either direct or indirect effects on these neurons. So, when DA neurons were exposed to l-trans-pyrrolidine-2,4-dicarboxylic acid so as to increase ambient glutamate levels and generate slow and sustained excitotoxicity, the effect of xenon on DA neurons was direct. The vitamin E analog Trolox also partially rescued DA neurons in this setting and enhanced neuroprotection by xenon. However, in the situation where DA cell death was spontaneous, the protection of DA neurons by xenon appeared indirect as it occurred through the repression of a mechanism mediated by proliferating glial cells, presumably astrocytes and their precursor cells. Xenon also exerted trophic effects for DA neurons in this paradigm. The effects of xenon were mimicked and improved by the N-methyl-d-aspartate glutamate receptor antagonist memantine and xenon itself appeared to work by antagonizing N-methyl-d-aspartate receptors. Note that another noble gas argon could not reproduce xenon effects. Overall, present data indicate that xenon can provide protection and trophic support to DA neurons that are vulnerable in Parkinson's disease. This suggests that xenon might have some therapeutic value for this disorder.


Assuntos
Anestésicos Inalatórios/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Mesencéfalo/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Xenônio/farmacologia , Animais , Antioxidantes/farmacologia , Morte Celular/efeitos dos fármacos , Células Cultivadas , Cromanos/farmacologia , Ácidos Dicarboxílicos/antagonistas & inibidores , Ácidos Dicarboxílicos/toxicidade , Antagonistas de Aminoácidos Excitatórios/farmacologia , Memantina/farmacologia , Técnicas de Cultura de Órgãos , Pirrolidinas/antagonistas & inibidores , Pirrolidinas/toxicidade , Ratos , Ratos Wistar
10.
Glia ; 64(11): 1912-24, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27452488

RESUMO

Purified microglial cells in culture are frequently used to model brain inflammatory responses but obtaining large yields of these cells on a routine basis can be quite challenging. Here, we demonstrate that it is possible to achieve high-yield isolation of pure microglial (MAC-1(+) /Fcrls(+) /Ccr2(-) ) cells from postnatal brain tissue through a simple culture procedure that mainly relies on the adhesion preference of these cells to the polycation polyethyleneimine (PEI) in serum-supplemented DMEM medium. Accordingly, other synthetic or biological substrates failed to mimic PEI effects under the same culture conditions. Replacement of DMEM by DMEM/F12 nutrient mixture did not permit microglial cell isolation on PEI coating, indicating that PEI effects were context-dependent. Remarkably, the lack of culture feeding during progression of microglial cell isolation strongly improved cell yield, suggesting that nutritional deprivation was required to optimize this process. When generated in large culture flasks coated with PEI, cultures of microglial cells were easily recovered by trypsin proteolysis to produce subcultures for functional studies. These cultures responded to lipopolysaccharide (LPS, 1-10 ng/ml) treatment by secreting pro-inflammatory cytokines such as TNF-α, IL-6, IL-1ß and by generating nitric oxide and reactive oxygen species. Most interestingly, this response was curtailed by appropriate reference drugs. Microglial cells were also strongly responsive to the mitogenic cytokine GM-CSF, which confirms that the functional repertoire of these cells was well preserved. Because of its high yield and simplicity, we believe that the present method will prove to be especially convenient for mechanistic studies or screening assays. GLIA 2016;64:1912-1924.


Assuntos
Citocinas/metabolismo , Microglia/fisiologia , Animais , Animais Recém-Nascidos , Antineoplásicos/farmacologia , Encéfalo/citologia , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Dexametasona/farmacologia , Proteína Glial Fibrilar Ácida/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Laminina/farmacologia , Lipopolissacarídeos/farmacologia , Antígeno de Macrófago 1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/metabolismo , Microglia/efeitos dos fármacos , Óxido Nítrico/metabolismo , Oligopeptídeos/farmacologia , Polietilenoimina/farmacologia , Espécies Reativas de Oxigênio/metabolismo
11.
Mol Pharmacol ; 87(3): 525-32, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25552485

RESUMO

To determine whether orexinergic hypothalamic peptides can influence the survival of brainstem dopamine (DA) neurons, we used a model system of rat midbrain cultures in which DA neurons degenerate spontaneously and progressively as they mature. We established that orexin (OX)-B provides partial but significant protection to spontaneously dying DA neurons, whereas the homologous peptide OXA has only marginal effects. Importantly, DA neurons rescued by OXB accumulated DA efficiently by active transport, suggesting that they were functional. G-protein-coupled OX1 and OX2 receptors were both present on DA neurons, but the protective effect of OXB was attributable solely to OX2 receptors; a selective inhibitor of this receptor subtype, N-ethyl-2-[(6-methoxy-3-pyridinyl)[(2-methylphenyl)sulfonyl]amino]-N-(3-pyridinylmethyl)-acetamide (EMPA), suppressed this effect, whereas a selective agonist, [Ala(11), d-Leu(15)]OXB, reproduced it. Survival promotion by OXB required intracellular calcium mobilization via inositol-1,4,5-triphosphate and ryanodine receptors. Nicotine, a well known neuroprotective molecule for DA neurons, improved OXB-mediated rescue through the activation of α-bungarotoxin-sensitive (presumably α7) nicotinic receptors, although nicotine had no effect on its own. Altogether, our data suggest that the loss of hypothalamic orexinergic neurons that occurs in Parkinson's disease might confer an increased vulnerability to midbrain DA neurons in this disorder.


Assuntos
Neurônios Dopaminérgicos/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/administração & dosagem , Mesencéfalo/efeitos dos fármacos , Degeneração Neural/prevenção & controle , Neuropeptídeos/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Nicotina/administração & dosagem , Sono , Animais , Células Cultivadas , Neurônios Dopaminérgicos/patologia , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Mesencéfalo/patologia , Degeneração Neural/patologia , Orexinas , Ratos , Ratos Wistar , Sono/fisiologia
12.
Eur J Neurosci ; 41(1): 129-36, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25328140

RESUMO

Several studies conducted in patients with Parkinson's disease have reported that the degeneration of substantia nigra dopaminergic neurons, which are essential for motor control, is associated with the loss of hypothalamic orexin neurons, which are involved in sleep regulation. In order to better explore the mutual interactions between these two systems, we wished to determine in macaques: (i) if the two orexin peptides, orexin-A and orexin-B, are distributed in the same hypothalamic cells and if they are localized in nerve terminals that project onto nigral dopaminergic neurons, and (ii) if there is a loss of orexin neurons in the hypothalamus and of orexin fibers innervating nigral dopaminergic neurons in macaques rendered parkinsonian by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) intoxication. We showed that virtually all cells stained for orexin-A in the hypothalamus co-expressed orexin-B. Numerous terminals stained for both orexin-A and orexin-B immunoreactivity that innervated the whole extent of the ventral tegmental area and substantia nigra pars compacta were found in close proximity to tyrosine hydroxylase-immunoreactive dendrites. These data indicate that orexin-A and orexin-B peptides are in a position to play a role in controlling the activity of nigral dopaminergic neurons. However, no loss of orexin-A or orexin-B neurons in the hypothalamus and no loss of orexin fibers in the substantia nigra pars compacta was found in MPTP-treated macaques when compared with control macaques. We conclude that a relatively selective dopaminergic lesion, such as that performed in MPTP-treated macaques, is not sufficient to induce a loss of hypothalamic orexin neurons.


Assuntos
Hipotálamo/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Intoxicação por MPTP/patologia , Neurônios/patologia , Neuropeptídeos/metabolismo , Substância Negra/patologia , Animais , Contagem de Células , Morte Celular , Hipotálamo/metabolismo , Imuno-Histoquímica , Intoxicação por MPTP/metabolismo , Macaca fascicularis , Vias Neurais/metabolismo , Vias Neurais/patologia , Neurônios/metabolismo , Orexinas , Parte Compacta da Substância Negra/metabolismo , Parte Compacta da Substância Negra/patologia , Fotomicrografia , Substância Negra/metabolismo , Área Tegmentar Ventral/metabolismo , Área Tegmentar Ventral/patologia
13.
J Neuroinflammation ; 11: 86, 2014 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-24886419

RESUMO

BACKGROUND: Increasing evidence suggests that inflammation associated with microglial cell activation in the substantia nigra (SN) of patients with Parkinson disease (PD) is not only a consequence of neuronal degeneration, but may actively sustain dopaminergic (DA) cell loss over time. We aimed to study whether the intracellular chaperone heat shock protein 60 (Hsp60) could serve as a signal of CNS injury for activation of microglial cells. METHODS: Hsp60 mRNA expression in the mesencephalon and the striatum of C57/BL6 mice treated with MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) and the Hsp60/TH mRNA ratios in the SN of PD patients and aged-matched subjects were measured. To further investigate a possible link between the neuronal Hsp60 response and PD-related cellular stress, Hsp60 immunoblot analysis and quantification in cell lysates from SH-SY5Y after treatment with 100 µM MPP+ (1-methyl-4-phenylpyridinium) at different time points (6, 12, 24 and 48 hours) compared to control cells were performed. Additional MTT and LDH assay were used. We next addressed the question as to whether Hsp60 influences the survival of TH+ neurons in mesencephalic neuron-glia cultures treated either with MPP+ (1 µM), hHsp60 (10 µg/ml) or a combination of both. Finally, we measured IL-1ß, IL-6, TNF-α and NO-release by ELISA in primary microglial cell cultures following treatment with different hHsp60 preparations. Control cultures were exposed to LPS. RESULTS: In the mesencephalon and striatum of mice treated with MPTP and also in the SN of PD patients, we found that Hsp60 mRNA was up-regulated. MPP+, the active metabolite of MPTP, also caused an increased expression and release of Hsp60 in the human dopaminergic cell line SH-SY5Y. Interestingly, in addition to being toxic to DA neurons in primary mesencephalic cultures, exogenous Hsp60 aggravated the effects of MPP+. Yet, although we demonstrated that Hsp60 specifically binds to microglial cells, it failed to stimulate the production of pro-inflammatory cytokines or NO by these cells. CONCLUSIONS: Overall, our data suggest that Hsp60 is likely to participate in DA cell death in PD but via a mechanism unrelated to cytokine release.


Assuntos
Chaperonina 60/metabolismo , Corpo Estriado/patologia , Neurônios Dopaminérgicos/metabolismo , Intoxicação por MPTP/patologia , Mesencéfalo/patologia , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Células Cultivadas , Chaperonina 60/genética , Modelos Animais de Doenças , Dopaminérgicos/farmacologia , L-Lactato Desidrogenase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Óxido Nítrico/metabolismo , Ligação Proteica/efeitos dos fármacos , RNA Mensageiro/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
14.
FASEB J ; 27(9): 3414-23, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23699175

RESUMO

Parkinson disease (PD) is a degenerative brain disorder characterized by motor symptoms that are unequivocally associated with the loss of dopaminergic (DA) neurons in the substantia nigra (SN). Although our knowledge of the mechanisms that contribute to DA cell death in both hereditary and sporadic forms of the disease has advanced significantly, the nature of the pathogenic process remains poorly understood. In this review, we present evidence that neurodegeneration occurs when the electrical activity and excitability of these neurons is reduced. In particular, we will focus on the specific need these neurons may have for stimulation in order to survive and on the molecular and cellular mechanisms that may be compromised when this need is no longer met in PD.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Doença de Parkinson/metabolismo , Animais , Neurônios Dopaminérgicos/fisiologia , Humanos , Modelos Biológicos
15.
Mol Pharmacol ; 84(6): 888-98, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24077968

RESUMO

Previous studies on postmortem human brain tissue have shown that the iron-binding glycoprotein lactoferrin is upregulated in dopamine (DA) neurons resistant to degeneration in Parkinson disease (PD). To study how this could possibly relate to disease progression, we used midbrain cultures and experimental settings that model the progressive loss of DA neurons in this disorder. Human lactoferrin of either recombinant or natural origin provided robust protection to vulnerable DA neurons in a culture paradigm in which these neurons die spontaneously and selectively as they mature. The efficacy of lactoferrin was comparable to that of glial cell line-derived neurotrophic factor, a prototypical neurotrophic factor for DA neurons. Neuroprotection by lactoferrin was attributable to its binding to heparan sulfate proteoglycans on the cell surface of DA neurons and subsequently to partial inactivation of focal adhesion kinase (FAK), a major effector kinase of integrins. We established that FAK inactivation served to unmask a prosurvival phosphoinositide 3-kinase/AKT-dependent signaling pathway that stimulates calcium shuttling from endoplasmic reticulum to mitochondria. DA neurons exposed to the mitochondrial toxin 1-methyl-4-phenylpyridinium were also partially protected by lactoferrin, further supporting the view that mitochondria may represent a downstream target for lactoferrin protective actions. Finally, we found that the iron binding capability of lactoferrin intervened in DA cell rescue only when neurodegeneration was consecutive to iron-catalyzed oxidative stress. Overall, our data suggest that the accumulation of lactoferrin in PD brains might be evidence of an attempt by the brain to minimize the consequences of neurodegeneration.


Assuntos
Cálcio/metabolismo , Dopamina/metabolismo , Lactoferrina/farmacologia , Degeneração Neural/metabolismo , Neurônios/metabolismo , 1-Metil-4-fenilpiridínio/toxicidade , Animais , Sítios de Ligação , Morte Celular/efeitos dos fármacos , Células Cultivadas , Quinase 1 de Adesão Focal/antagonistas & inibidores , Quinase 1 de Adesão Focal/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Homeostase , Humanos , Lactoferrina/metabolismo , Mesencéfalo/citologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Degeneração Neural/patologia , Neuroglia/efeitos dos fármacos , Neuroglia/patologia , Neurônios/patologia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Ratos , Ratos Wistar , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia
16.
J Neurochem ; 127(6): 782-92, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23802648

RESUMO

The uricosuric agent probenecid is co-administered with the dopaminergic neurotoxin MPTP to produce a chronic mouse model of Parkinson's disease. It has been proposed that probenecid serves to elevate concentrations of MPTP in the brain by reducing renal elimination of the toxin. However, this mechanism has never been formally demonstrated to date and is questioned by our previous data showing that intracerebral concentrations of MPP(+), the active metabolite of MPTP, are not modified by co-injection of probenecid. In this study, we investigated the potentiating effects of probenecid in vivo and in vitro arguing against the possibility of altered metabolism or impaired renal elimination of MPTP. We find that probenecid (i) is toxic in itself to several neuronal populations apart from dopaminergic neurons, and (ii) that it also potentiates the effects of other mitochondrial complex I inhibitors such as rotenone. On a mechanistic level, we show that probenecid is able to lower intracellular ATP concentrations and that its toxic action on neuronal cells can be reversed by extracellular ATP. Probenecid can potentiate the effect of mitochondrial toxins due to its impact on ATP metabolism and could therefore be useful to model atypical parkinsonian syndromes.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , 1-Metil-4-fenilpiridínio/metabolismo , Dopaminérgicos/toxicidade , Neurotoxinas/toxicidade , Doença de Parkinson/patologia , Probenecid/toxicidade , Uricosúricos/toxicidade , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/metabolismo , Animais , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Modelos Animais de Doenças , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Sinergismo Farmacológico , Complexo I de Transporte de Elétrons/antagonistas & inibidores , Metabolismo Energético , Camundongos , Doença de Parkinson/etiologia , Doença de Parkinson/metabolismo , Rotenona/toxicidade
17.
Cells ; 12(9)2023 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-37174736

RESUMO

Chlordecone (CLD) is an organochlorine pesticide (OCP) that is currently banned but still contaminates ecosystems in the French Caribbean. Because OCPs are known to increase the risk of Parkinson's disease (PD), we tested whether chronic low-level intoxication with CLD could reproduce certain key characteristics of Parkinsonism-like neurodegeneration. For that, we used culture systems of mouse midbrain dopamine (DA) neurons and glial cells, together with the nematode C. elegans as an in vivo model organism. We established that CLD kills cultured DA neurons in a concentration- and time-dependent manner while exerting no direct proinflammatory effects on glial cells. DA cell loss was not impacted by the degree of maturation of the culture. The use of fluorogenic probes revealed that CLD neurotoxicity was the consequence of oxidative stress-mediated insults and mitochondrial disturbances. In C. elegans worms, CLD exposure caused a progressive loss of DA neurons associated with locomotor deficits secondary to alterations in food perception. L-DOPA, a molecule used for PD treatment, corrected these deficits. Cholinergic and serotoninergic neuronal cells were also affected by CLD in C. elegans, although to a lesser extent than DA neurons. Noticeably, CLD also promoted the phosphorylation of the aggregation-prone protein tau (but not of α-synuclein) both in midbrain cell cultures and in a transgenic C. elegans strain expressing a human form of tau in neurons. In summary, our data suggest that CLD is more likely to promote atypical forms of Parkinsonism characterized by tau pathology than classical synucleinopathy-associated PD.


Assuntos
Clordecona , Doença de Parkinson , Transtornos Parkinsonianos , Praguicidas , Animais , Humanos , Camundongos , Caenorhabditis elegans/metabolismo , Clordecona/metabolismo , Praguicidas/toxicidade , Ecossistema , Transtornos Parkinsonianos/patologia , Doença de Parkinson/metabolismo , Neurônios Dopaminérgicos/metabolismo , Mesencéfalo/patologia
18.
FASEB J ; 25(8): 2563-73, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21507900

RESUMO

Epidemiological and experimental evidence indicates that nicotine is protective for Parkinson disease vulnerable dopamine neurons, but the underlying mechanism of this effect remains only partly characterized. To address this question, we established rat midbrain cultures maintained in experimental conditions that favor the selective and spontaneous loss of dopamine neurons. We report here that nicotine afforded neuroprotection to dopamine neurons (EC(50)=0.32 µM) but only in a situation where cytosolic Ca(2+) (Ca(2+)(cyt)) was slightly and chronically elevated above control levels by concurrent depolarizing treatments. By a pharmacological approach, we demonstrated that the rise in Ca(2+)(cyt) was necessary to sensitize dopamine neurons to the action of nicotine through a mechanism involving α-bungarotoxin-sensitive (presumably α7) nicotinic acetylcholine receptors (nAChRs) and secondarily T-type voltage-gated calcium channels. Confirming the role played by α7 nAChRs in this effect, nicotine had no protective action in midbrain cultures prepared from genetically engineered mice lacking this receptor subtype. Signaling studies revealed that Ca(2+)(cyt) elevations evoked by nicotine and concomitant depolarizing treatments served to activate a survival pathway involving the calcium effector protein calmodulin and phosphatidylinositol 3-kinase. Collectively, our data support the idea that the protective action of nicotine for dopamine neurons is activity-dependent and gated by Ca(2+)(cyt).


Assuntos
Cálcio/metabolismo , Dopamina/metabolismo , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Fármacos Neuroprotetores/farmacologia , Nicotina/farmacologia , Animais , Canais de Cálcio Tipo T/metabolismo , Calmodulina/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Citoplasma/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Mesencéfalo/citologia , Camundongos , Modelos Neurológicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Receptores Nicotínicos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tetraetilamônio/farmacologia , Técnicas de Cultura de Tecidos , Receptor Nicotínico de Acetilcolina alfa7
19.
Cells ; 11(10)2022 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-35626675

RESUMO

To model α-Synuclein (αS) aggregation and neurodegeneration in Parkinson's disease (PD), we established cultures of mouse midbrain dopamine (DA) neurons and chronically exposed them to fibrils 91 (F91) generated from recombinant human αS. We found that F91 have an exquisite propensity to seed the aggregation of endogenous αS in DA neurons when compared to other neurons in midbrain cultures. Until two weeks post-exposure, somal aggregation in DA neurons increased with F91 concentrations (0.01-0.75 µM) and the time elapsed since the initiation of seeding, with, however, no evidence of DA cell loss within this time interval. Neither toxin-induced mitochondrial deficits nor genetically induced loss of mitochondrial quality control mechanisms promoted F91-mediated αS aggregation or neurodegeneration under these conditions. Yet, a significant loss of DA neurons (~30%) was detectable three weeks after exposure to F91 (0.5 µM), i.e., at a time point where somal aggregation reached a plateau. This loss was preceded by early deficits in DA uptake. Unlike αS aggregation, the loss of DA neurons was prevented by treatment with GDNF, suggesting that αS aggregation in DA neurons may induce a form of cell death mimicking a state of trophic factor deprivation. Overall, our model system may be useful for exploring PD-related pathomechanisms and for testing molecules of therapeutic interest for this disorder.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Animais , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Mesencéfalo/metabolismo , Camundongos , Doença de Parkinson/metabolismo , alfa-Sinucleína/metabolismo
20.
ACS Chem Neurosci ; 13(23): 3303-3313, 2022 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-36347018

RESUMO

A tetrahydroisoquinoline identified in Mucuna pruriens ((1R,3S)-6,7-dihydroxy-1-methyl-1,2,3,4-tetrahydroisoquinoline-1,3-dicarboxylic acid, compound 4) was synthesized and assessed for its in vitro pharmacological profile and in vivo effects in two animal models of Parkinson's disease. Compound 4 inhibits catechol-O-methyltransferase (COMT) with no affinity for the dopaminergic receptors or the dopamine transporter. It restores dopamine-mediated motor behavior when it is co-administered with L-DOPA to C. elegans worms with 1-methyl-4-phenylpyridinium-damaged dopaminergic neurons. In a 6-hydroxydopamine rat model of Parkinson's disease, its co-administration at 30 mg/kg with L-DOPA enhances the effect of L-DOPA with an intensity similar to that of tolcapone 1 at 30 mg/kg but for a shorter duration. The effect is not dose-dependent. Compound 4 seems not to cross the blood-brain barrier and thus acts as a peripheral COMT inhibitor. COMT inhibition by compound 4 further validates the traditional use of M. pruriens for the treatment of Parkinson's disease, and compound 4 can thus be considered as a promising drug candidate for the development of safe, peripheral COMT inhibitors.


Assuntos
Levodopa , Doença de Parkinson , Animais , Ratos , Levodopa/farmacologia , Levodopa/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Catecol O-Metiltransferase , Caenorhabditis elegans , Personalidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA