Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Epilepsia ; 64(5): 1390-1402, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36808593

RESUMO

OBJECTIVE: Initiation and development of early seizures by chemical stimuli is associated with brain cell swelling resulting in edema of seizure-vulnerable brain regions. We previously reported that pretreatment with a nonconvulsive dose of glutamine (Gln) synthetase inhibitor methionine sulfoximine (MSO) mitigates the intensity of initial pilocarpine (Pilo)-induced seizures in juvenile rats. We hypothesized that MSO exerts its protective effect by preventing the seizure-initiating and seizure-propagating increase of cell volume. Taurine (Tau) is an osmosensitive amino acid, whose release reflects increased cell volume. Therefore, we tested whether the poststimulus rise of amplitude of Pilo-induced electrographic seizures and their attenuation by MSO are correlated with the release of Tau from seizure-affected hippocampus. METHODS: Lithium-pretreated animals were administered MSO (75 mg/kg ip) 2.5 h before the induction of convulsions by Pilo (40 mg/kg ip). Electroencephalographic (EEG) power was analyzed during 60 min post-Pilo, at 5-min intervals. Extracellular accumulation of Tau (eTau) served as a marker of cell swelling. eTau, extracellular Gln (eGln), and extracellular glutamate (eGlu) were assayed in the microdialysates of the ventral hippocampal CA1 region collected at 15-min intervals during the whole 3.5-h observation period. RESULTS: The first EEG signal became apparent at ~10 min post-Pilo. The EEG amplitude across most frequency bands peaked at ~40 min post-Pilo, and showed strong (r ~ .72-.96) temporal correlation with eTau, but no correlation with eGln or eGlu. MSO pretreatment delayed the first EEG signal in Pilo-treated rats by ~10 min, and depressed the EEG amplitude across most frequency bands, to values that remained strongly correlated with eTau (r > .92) and moderately correlated (r ~ -.59) with eGln, but not with eGlu. SIGNIFICANCE: Strong correlation between attenuation of Pilo-induced seizures and Tau release indicates that the beneficial effect of MSO is due to the prevention of cell volume increase concurrent with the onset of seizures.


Assuntos
Metionina Sulfoximina , Pilocarpina , Ratos , Animais , Pilocarpina/toxicidade , Metionina Sulfoximina/farmacologia , Metionina Sulfoximina/metabolismo , Taurina/farmacologia , Convulsões/induzido quimicamente , Convulsões/prevenção & controle , Convulsões/tratamento farmacológico , Hipocampo/metabolismo
3.
EBioMedicine ; 80: 104021, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35500536

RESUMO

BACKGROUND: Synucleinopathies such as Parkinson ́s disease (PD), Dementia with Lewy bodies (DLB) and Multiple System Atrophy (MSA) are characterized by deposition of misfolded and aggregated α-synuclein. Small aggregates (oligomers) of α-synuclein have been shown to be the most relevant neurotoxic species and are targeted by anle138b, an orally bioavailable small molecule compound which shows strong disease-modifying effects in animal models of synucleinopathies. METHODS: Anle138b was studied in a single-centre, double-blind, randomised, placebo-controlled single ascending dose (SAD) and multiple ascending dose (MAD) study in healthy subjects. Eligible participants were randomly assigned (1:1 for sentinel subjects and 1:5 for main group) to placebo or anle138b (dose range 50 mg to 300 mg per day), respectively. In addition, the effect of food on the pharmakokinetics of anle138b in healthy subjects was examined in doses of 150 mg per day. Participants were randomized to treatment sequence (fed→fasted) or (fasted→fed). Treatment was administered orally in hard gelatine capsules containing either 10 mg or 30 mg of anle138b or excipient only. The primary endpoints were safety and tolerability, the secondary endpoint was pharmakokinetics. Data from all randomized individuals were evaluated. CLINICALTRIALS: gov-identifier: NCT04208152. EudraCT-number: 2019-004218-33. FINDINGS: Between December 17th, 2019 and June 27th, 2020 196 healthy volunteers were screened and 68 participants were enrolled. Of these, all completed the study per protocol. There were no major protocol deviations. Adverse events in this healthy volunteer trial were mostly mild and all fully recovered or resolved prior to discharge. From baseline to completion of the trial no medically significant individual changes were observed in any system organ class. Already at multiple doses of 200 mg, exposure levels above the fully effective exposure in the MI2 mouse Parkinson model were observed. INTERPRETATION: The favourable safety and PK profile of anle138b in doses resulting in exposures above the fully effective plasma level in a mouse Parkinson model warrant further clinical trials in patients with synucleinopathies. FUNDING: This study was funded by MODAG GmbH and by the Michael J. Fox foundation for Parkinson's Research.


Assuntos
Doença de Parkinson , Sinucleinopatias , Animais , Benzodioxóis , Modelos Animais de Doenças , Método Duplo-Cego , Humanos , Camundongos , Doença de Parkinson/tratamento farmacológico , Pirazóis , alfa-Sinucleína
4.
Commun Biol ; 4(1): 1384, 2021 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-34893694

RESUMO

CD71+ erythroid cells (CECs) have been recently recognized in both neonates and cancer patients as potent immunoregulatory cells. Here, we show that in mice early-stage CECs expand in anemia, have high levels of arginase 2 (ARG2) and reactive oxygen species (ROS). In the spleens of anemic mice, CECs expansion-induced L-arginine depletion suppresses T-cell responses. In humans with anemia, CECs expand and express ARG1 and ARG2 that suppress T-cells IFN-γ production. Moreover, bone marrow CECs from healthy human donors suppress T-cells proliferation. CECs differentiated from peripheral blood mononuclear cells potently suppress T-cell activation, proliferation, and IFN-γ production in an ARG- and ROS-dependent manner. These effects are the most prominent for early-stage CECs (CD71highCD235adim cells). The suppressive properties disappear during erythroid differentiation as more differentiated CECs and mature erythrocytes lack significant immunoregulatory properties. Our studies provide a novel insight into the role of CECs in the immune response regulation.


Assuntos
Células Eritroides/imunologia , Tolerância Imunológica , Linfócitos T/imunologia , Adulto , Animais , Antígenos CD/metabolismo , Linhagem Celular , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Receptores da Transferrina/metabolismo , Adulto Jovem
5.
Brain ; 144(6): 1661-1669, 2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-33760024

RESUMO

α-Synuclein aggregation at the synapse is an early event in Parkinson's disease and is associated with impaired striatal synaptic function and dopaminergic neuronal death. The cysteine string protein (CSPα) and α-synuclein have partially overlapping roles in maintaining synaptic function and mutations in each cause neurodegenerative diseases. CSPα is a member of the DNAJ/HSP40 family of co-chaperones and like α-synuclein, chaperones the SNARE complex assembly and controls neurotransmitter release. α-Synuclein can rescue neurodegeneration in CSPαKO mice. However, whether α-synuclein aggregation alters CSPα expression and function is unknown. Here we show that α-synuclein aggregation at the synapse is associated with a decrease in synaptic CSPα and a reduction in the complexes that CSPα forms with HSC70 and STGa. We further show that viral delivery of CSPα rescues in vitro the impaired vesicle recycling in PC12 cells with α-synuclein aggregates and in vivo reduces synaptic α-synuclein aggregates increasing monomeric α-synuclein and restoring normal dopamine release in 1-120hαSyn mice. These novel findings reveal a mechanism by which α-synuclein aggregation alters CSPα at the synapse, and show that CSPα rescues α-synuclein aggregation-related phenotype in 1-120hαSyn mice similar to the effect of α-synuclein in CSPαKO mice. These results implicate CSPα as a potential therapeutic target for the treatment of early-stage Parkinson's disease.


Assuntos
Corpo Estriado/metabolismo , Dopamina/metabolismo , Proteínas de Choque Térmico HSP40/metabolismo , Proteínas de Membrana/metabolismo , Agregação Patológica de Proteínas/metabolismo , alfa-Sinucleína/metabolismo , Animais , Corpo Estriado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Agregação Patológica de Proteínas/patologia , Sinapses/metabolismo , Sinapses/patologia
6.
PLoS One ; 15(8): e0238075, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32833982

RESUMO

Parkinson disease is the most common neurodegenerative movement disorder, estimated to affect one in twenty-five individuals over the age of 80. Mutations in glucocerebrosidase 1 (GBA1) represent the most common genetic risk factor for Parkinson disease. The link between GBA1 mutations and α-synuclein accumulation, a hallmark of Parkinson disease, is not fully understood. Following our recent finding that Gba1 mutations lead to increased α-synuclein accumulation in mice, we have studied the effects of a single injection of mouse α-synuclein pre-formed fibrils into the striatum of Gba1 mice that carry a L444P knock-in mutation. We found significantly greater formation and spread of α-synuclein inclusions in Gba1-transgenic mice compared to wild-type controls. This indicates that the Gba1 L444P mutation accelerates α-synuclein pathology and spread.


Assuntos
Técnicas de Introdução de Genes , Glucosilceramidase/genética , Mutação , Agregados Proteicos/genética , alfa-Sinucleína/química , Animais , Humanos , Injeções , Camundongos , Neostriado/metabolismo , alfa-Sinucleína/metabolismo
7.
Acta Neuropathol ; 138(4): 575-595, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31165254

RESUMO

Parkinson's disease (PD) is characterized by the presence of α-synuclein aggregates known as Lewy bodies and Lewy neurites, whose formation is linked to disease development. The causal relation between α-synuclein aggregates and PD is not well understood. We generated a new transgenic mouse line (MI2) expressing human, aggregation-prone truncated 1-120 α-synuclein under the control of the tyrosine hydroxylase promoter. MI2 mice exhibit progressive aggregation of α-synuclein in dopaminergic neurons of the substantia nigra pars compacta and their striatal terminals. This is associated with a progressive reduction of striatal dopamine release, reduced striatal innervation and significant nigral dopaminergic nerve cell death starting from 6 and 12 months of age, respectively. In the MI2 mice, alterations in gait impairment can be detected by the DigiGait test from 9 months of age, while gross motor deficit was detected by rotarod test at 20 months of age when 50% of dopaminergic neurons in the substantia nigra pars compacta are lost. These changes were associated with an increase in the number and density of 20-500 nm α-synuclein species as shown by dSTORM. Treatment with the oligomer modulator anle138b, from 9 to 12 months of age, restored striatal dopamine release, prevented dopaminergic cell death and gait impairment. These effects were associated with a reduction of the inner density of large α-synuclein aggregates and an increase in dispersed small α-synuclein species as revealed by dSTORM. The MI2 mouse model recapitulates the progressive dopaminergic deficit observed in PD, showing that early synaptic dysfunction is associated to fine behavioral motor alterations, precedes dopaminergic axonal loss and neuronal death that become associated with a more consistent motor deficit upon reaching a certain threshold. Our data also provide new mechanistic insight for the effect of anle138b's function in vivo supporting that targeting α-synuclein aggregation is a promising therapeutic approach for PD.


Assuntos
Morte Celular/fisiologia , Neurônios Dopaminérgicos/patologia , Doença de Parkinson/patologia , Agregação Patológica de Proteínas/patologia , Substância Negra/patologia , alfa-Sinucleína/metabolismo , Animais , Modelos Animais de Doenças , Marcha/genética , Camundongos , Camundongos Transgênicos , Atividade Motora/genética , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Agregação Patológica de Proteínas/metabolismo , Substância Negra/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , alfa-Sinucleína/genética
8.
Brain ; 140(10): 2706-2721, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-28969384

RESUMO

Mutations in glucocerebrosidase 1 (GBA1) represent the most prevalent risk factor for Parkinson's disease. The molecular mechanisms underlying the link between GBA1 mutations and Parkinson's disease are incompletely understood. We analysed two aged (24-month-old) Gba1 mouse models, one carrying a knock-out mutation and the other a L444P knock-in mutation. A significant reduction of glucocerebrosidase activity was associated with increased total alpha-synuclein accumulation in both these models. Gba1 mutations alone did not alter the number of nigral dopaminergic neurons nor striatal dopamine levels. We then investigated the effect of overexpression of human alpha-synuclein in the substantia nigra of aged (18 to 21-month-old) L444P Gba1 mice. Following intraparenchymal injections of human alpha-synuclein carrying viral vectors, pathological accumulation of phosphorylated alpha-synuclein occurred within the transduced neurons. Stereological counts of nigral dopaminergic neurons revealed a significantly greater cell loss in Gba1-mutant than wild-type mice. These results indicate that Gba1 deficiency enhances neuronal vulnerability to neurodegenerative processes triggered by increased alpha-synuclein expression.


Assuntos
Dopamina/metabolismo , Glucosilceramidase/genética , Mutação/genética , Neurônios/patologia , Substância Negra/patologia , alfa-Sinucleína/metabolismo , Fatores Etários , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Glucosilceramidase/deficiência , Humanos , Leucina/genética , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Prolina/genética , Desempenho Psicomotor/fisiologia , Olfato/genética , Substância Negra/metabolismo , Transdução Genética , Tirosina 3-Mono-Oxigenase/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo
9.
Biochim Biophys Acta Mol Basis Dis ; 1863(6): 1596-1604, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28213125

RESUMO

Huntington's disease (HD) is caused by a mutation in the huntingtin gene (HTT), resulting in profound striatal neurodegeneration through an unknown mechanism. Perturbations in the urea cycle have been reported in HD models and in HD patient blood and brain. In neurons, arginase is a central urea cycle enzyme, and the metal manganese (Mn) is an essential cofactor. Deficient biological responses to Mn, and reduced Mn accumulation have been observed in HD striatal mouse and cell models. Here we report in vivo and ex vivo evidence of a urea cycle metabolic phenotype in a prodromal HD mouse model. Further, either in vivo or in vitro Mn supplementation reverses the urea-cycle pathology by restoring arginase activity. We show that Arginase 2 (ARG2) is the arginase enzyme present in these mouse brain models, with ARG2 protein levels directly increased by Mn exposure. ARG2 protein is not reduced in the prodromal stage, though enzyme activity is reduced, indicating that altered Mn bioavailability as a cofactor leads to the deficient enzymatic activity. These data support a hypothesis that mutant HTT leads to a selective deficiency of neuronal Mn at an early disease stage, contributing to HD striatal urea-cycle pathophysiology through an effect on arginase activity.


Assuntos
Corpo Estriado/metabolismo , Doença de Huntington/metabolismo , Manganês/metabolismo , Neurônios/metabolismo , Ureia/metabolismo , Animais , Arginase/metabolismo , Corpo Estriado/patologia , Modelos Animais de Doenças , Doença de Huntington/patologia , Masculino , Camundongos , Neurônios/patologia
10.
Mov Disord ; 31(2): 169-77, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26790375

RESUMO

Although the physiological function of α-synuclein is not fully understood, it has been suggested to primarily localize to the presynaptic terminals of mature neurons, where it fulfills roles in synaptic function and plasticity. Based on current knowledge, α-synuclein (αSYN) is thought to be involved in maintaining neurotransmitter homeostasis by regulating synaptic vesicle fusion, clustering, and trafficking between the reserve and ready-releasable pools, as well as interacting with neurotransmitter membrane transporters. In this review, we focus on evidence proposing synapses as the main site of αSYN pathology and its propagation in Parkinson's disease and dementia with Lewy bodies, which belong to a group of neurodegenerative diseases known as α-synucleinopathies. We provide an overview of the evidence supporting presynaptic dysfunction as the primary event in the pathogenesis of these conditions.


Assuntos
Doença por Corpos de Lewy/metabolismo , Doença de Parkinson/metabolismo , Sinapses/metabolismo , alfa-Sinucleína/metabolismo , Humanos , Doença por Corpos de Lewy/patologia , Doença de Parkinson/patologia , Sinapses/patologia
11.
Biol Psychiatry ; 79(5): 402-414, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26392130

RESUMO

BACKGROUND: Advanced Parkinson's disease (PD) is characterized by massive degeneration of nigral dopaminergic neurons, dramatic motor and cognitive alterations, and presence of nigral Lewy bodies, whose main constituent is α-synuclein (α-syn). However, the synaptic mechanisms underlying behavioral and motor effects induced by early selective overexpression of nigral α-syn are still a matter of debate. METHODS: We performed behavioral, molecular, and immunohistochemical analyses in two transgenic models of PD, mice transgenic for truncated human α-synuclein 1-120 and rats injected with the adeno-associated viral vector carrying wild-type human α-synuclein. We also investigated striatal synaptic plasticity by electrophysiological recordings from spiny projection neurons and cholinergic interneurons. RESULTS: We found that overexpression of truncated or wild-type human α-syn causes partial reduction of striatal dopamine levels and selectively blocks the induction of long-term potentiation in striatal cholinergic interneurons, producing early memory and motor alterations. These effects were dependent on α-syn modulation of the GluN2D-expressing N-methyl-D-aspartate receptors in cholinergic interneurons. Acute in vitro application of human α-syn oligomers mimicked the synaptic effects observed ex vivo in PD models. CONCLUSIONS: We suggest that striatal cholinergic dysfunction, induced by a direct interaction between α-syn and GluN2D-expressing N-methyl-D-aspartate receptors, represents a precocious biological marker of the disease.


Assuntos
Neurônios Colinérgicos/efeitos dos fármacos , Dopamina/fisiologia , Doença de Parkinson/tratamento farmacológico , Receptores de N-Metil-D-Aspartato/genética , alfa-Sinucleína/genética , Animais , Animais Geneticamente Modificados , Dependovirus , Modelos Animais de Doenças , Feminino , Humanos , Potenciação de Longa Duração , Masculino , Camundongos , Camundongos Transgênicos , Neostriado/fisiologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/genética , Transmissão Sináptica
12.
J Huntingtons Dis ; 4(1): 17-36, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26333255

RESUMO

BACKGROUND: Unusually large CAG repeat expansions (>60) in exon one of Huntingtin (HTT) are invariably associated with a juvenile-onset form of Huntington's disease (HD), characterized by a more extensive and rapidly progressing neuropathology than the more prevalent adult-onset form. However, existing mouse models of HD that express the full-length Htt gene with CAG repeat lengths associated with juvenile HD (ranging between ~75 to ~150 repeats in published models) exhibit selective neurodegenerative phenotypes more consistent with adult-onset HD. Objective: To determine if a very large CAG repeat (>200) in full-length Htt elicits neurodegenerative phenotypes consistent with juvenile HD. METHODS: Using a …bacterial artificial chromosome (BAC) system, we generated mice expressing full-length mouse Htt with ~225 CAG repeats under control of the mouse Htt promoter. Mice were characterized using behavioral, neuropathological, biochemical and brain imaging methods. RESULTS: BAC-225Q mice exhibit phenotypes consistent with a subset of features seen in juvenile-onset HD: very early motor behavior abnormalities, reduced body weight, widespread and progressive increase in Htt aggregates, gliosis, and neurodegeneration. Early striatal pathology was observed, including reactive gliosis and loss of dopamine receptors, prior to detectable volume loss. HD-related blood markers of impaired energy metabolism and systemic inflammation were also increased. Aside from an age-dependent progression of diffuse nuclear aggregates at 6 months of age to abundant neuropil aggregates at 12 months of age, other pathological and motor phenotypes showed little to no progression. CONCLUSIONS: The HD phenotypes present in animals 3 to 12 months of age make the BAC-225Q mice a unique and stable model of full-length mutant Htt associated phenotypes, including body weight loss, behavioral impairment and HD-like neurodegenerative phenotypes characteristic of juvenile-onset HD and/or late-stage adult-onset HD.


Assuntos
Comportamento Animal , Encéfalo/patologia , Doença de Huntington/genética , Camundongos , Neurônios/patologia , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Expansão das Repetições de Trinucleotídeos/genética , Animais , Atrofia , Encéfalo/metabolismo , Cromossomos Artificiais Bacterianos , Modelos Animais de Doenças , Progressão da Doença , Doença de Huntington/patologia , Doença de Huntington/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/metabolismo , Fenótipo , Regiões Promotoras Genéticas
13.
PLoS One ; 7(2): e31024, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22363539

RESUMO

YAC128 Huntington's disease (HD) transgenic mice accumulate less manganese (Mn) in the striatum relative to wild-type (WT) littermates. We hypothesized that Mn and mutant Huntingtin (HTT) would exhibit gene-environment interactions at the level of neurochemistry and neuronal morphology. Twelve-week-old WT and YAC128 mice were exposed to MnCl(2)-4H(2)O (50 mg/kg) on days 0, 3 and 6. Striatal medium spiny neuron (MSN) morphology, as well as levels of dopamine (DA) and its metabolites (which are known to be sensitive to Mn-exposure), were analyzed at 13 weeks (7 days from initial exposure) and 16 weeks (28 days from initial exposure). No genotype-dependent differences in MSN morphology were apparent at 13 weeks. But at 16 weeks, a genotype effect was observed in YAC128 mice, manifested by an absence of the wild-type age-dependent increase in dendritic length and branching complexity. In addition, genotype-exposure interaction effects were observed for dendritic complexity measures as a function of distance from the soma, where only YAC128 mice were sensitive to Mn exposure. Furthermore, striatal DA levels were unaltered at 13 weeks by genotype or Mn exposure, but at 16 weeks, both Mn exposure and the HD genotype were associated with quantitatively similar reductions in DA and its metabolites. Interestingly, Mn exposure of YAC128 mice did not further decrease DA or its metabolites versus YAC128 vehicle exposed or Mn exposed WT mice. Taken together, these results demonstrate Mn-HD disease-toxicant interactions at the onset of striatal dendritic neuropathology in YAC128 mice. Our results identify the earliest pathological change in striatum of YAC128 mice as being between 13 to 16 weeks. Finally, we show that mutant HTT suppresses some Mn-dependent changes, such as decreased DA levels, while it exacerbates others, such as dendritic pathology.


Assuntos
Dopamina/metabolismo , Exposição Ambiental , Doença de Huntington/patologia , Manganês/toxicidade , Neostriado/patologia , Neurônios/patologia , Animais , Dendritos/efeitos dos fármacos , Dendritos/metabolismo , Dendritos/patologia , Genótipo , Doença de Huntington/genética , Masculino , Manganês/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise Multivariada , Neostriado/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo
14.
J Proteome Res ; 11(2): 1118-32, 2012 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-22191580

RESUMO

Huntington's disease (HD) is a neurodegenerative disorder caused by expansion of a CAG repeat within the Huntingtin (HTT) gene, though the clinical presentation of disease and age-of-onset are strongly influenced by ill-defined environmental factors. We recently reported a gene-environment interaction wherein expression of mutant HTT is associated with neuroprotection against manganese (Mn) toxicity. Here, we are testing the hypothesis that this interaction may be manifested by altered protein expression patterns in striatum, a primary target of both neurodegeneration in HD and neurotoxicity of Mn. To this end, we compared striatal proteomes of wild-type and HD (YAC128Q) mice exposed to vehicle or Mn. Principal component analysis of proteomic data revealed that Mn exposure disrupted a segregation of WT versus mutant proteomes by the major principal component observed in vehicle-exposed mice. Identification of altered proteins revealed novel markers of Mn toxicity, particularly proteins involved in glycolysis, excitotoxicity, and cytoskeletal dynamics. In addition, YAC128Q-dependent changes suggest that axonal pathology may be an early feature in HD pathogenesis. Finally, for several proteins, genotype-specific responses to Mn were observed. These differences include increased sensitivity to exposure in YAC128Q mice (UBQLN1) and amelioration of some mutant HTT-induced alterations (SAE1, ENO1). We conclude that the interaction of Mn and mutant HTT may suppress proteomic phenotypes of YAC128Q mice, which could reveal potential targets in novel treatment strategies for HD.


Assuntos
Manganês/toxicidade , Neostriado/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteoma/metabolismo , Análise de Variância , Animais , Western Blotting , Eletroforese em Gel Bidimensional , Interação Gene-Ambiente , Proteína Huntingtina , Masculino , Camundongos , Camundongos Transgênicos , Neostriado/química , Neostriado/efeitos dos fármacos , Análise de Componente Principal , Proteoma/efeitos dos fármacos , Proteoma/genética , Proteômica
15.
Neurotoxicology ; 32(6): 896-906, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21641932

RESUMO

Gender differences in sensitivity and toxicokinetics of multiple metals have been identified in humans. A recent study suggested that young girls performed worse on intellectual exams than young boys exposed to manganese (Mn) in the environment. Animal studies have shown that Mn exposure causes differential effects on behavior in male compared to female mice. We hypothesized that in response to Mn exposure striatal Mn accumulation and/or striatal medium spiny neuron (MSN) morphology show gender-dependent effects. We evaluated the contribution of gender to neuropathology by examining striatal MSN morphology in male and female mice exposed to Mn. We found that gender played a significant role in alterations of striatal MSN morphology in mice exposed to Mn. Gender-dependent changes were strongest when striatal Mn levels were elevated 24h following the final Mn exposure. Nevertheless, gender-dependent alterations in neuron morphology were still present 3 weeks after the final Mn exposure. Gender differences in neuron morphology were not due to differential striatal Mn accumulation between genders. We conclude that although gender does not affect striatal Mn accumulation, MSN morphology is differentially sensitive to elevated Mn levels.


Assuntos
Gânglios da Base/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Cloretos/toxicidade , Poluentes Ambientais/toxicidade , Neurônios/efeitos dos fármacos , Análise de Variância , Animais , Gânglios da Base/metabolismo , Gânglios da Base/patologia , Cloretos/metabolismo , Dendritos/efeitos dos fármacos , Dendritos/patologia , Poluentes Ambientais/metabolismo , Feminino , Masculino , Compostos de Manganês/metabolismo , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Medição de Risco , Fatores de Risco , Fatores Sexuais , Fatores de Tempo
16.
Toxicol Pathol ; 39(1): 115-23, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21075920

RESUMO

Astrocytes assume multiple roles in maintaining an optimally suited milieu for neuronal function. Select astrocytic functions include the maintenance of redox potential, the production of trophic factors, the regulation of neurotransmitter and ion concentrations, and the removal of toxins and debris from the cerebrospinal fluid (CSF). Impairments in these and other functions, as well as physiological reactions of astrocytes to injury, can trigger or exacerbate neuronal dysfunction. This review addresses select metabolic interactions between neurons and astrocytes and emphasizes the role of astrocytes in mediating and amplifying the progression of several neurodegenerative disorders, such as Parkinson's disease (PD), hepatic encephalopathy (HE), hyperammonemia (HA), Alzheimer's disease (AD), and ischemia.


Assuntos
Astrócitos/metabolismo , Encéfalo/patologia , Fenômenos Fisiológicos do Sistema Nervoso , Neurotransmissores , Doença de Alzheimer/patologia , Animais , Encefalopatia Hepática/patologia , Humanos , Neurônios/metabolismo , Neurotransmissores/metabolismo , Doença de Parkinson/patologia
17.
Toxicol Sci ; 117(1): 163-8, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20530233

RESUMO

We endeavored here to shed light on the supply of glutathione (GSH) precursors from glial cells to neurons and on the interference of ammonia with this process. Administration of ammonium chloride (ammonia) via a microdialysis probe to the rat prefrontal cortex rapidly increased GSH content in the microdialysates. The increase was abrogated by the inhibitor of astrocytic energy metabolism fluoroacetate and the inhibitor of glutathione synthesis buthionine sulfoximine. GSH in the microdialysates was significantly elevated in rats with simple hyperammonemia (HA) or hepatic encephalopathy (HE) (three ip administrations of ammonium acetate or thioacetamide, respectively, at 24-h intervals), only when microdialysis was carried out in the presence of a gamma-glutamyltranspeptidase (gammaGT) inhibitor acivicin. Extracellular GSH increased in cultured rat cortical astrocytes treated with 5mM ammonia for 1 h, but not for 3-72 h, which was the period of increased gammaGT activity. GSH remained increased during the whole 72-h incubation with 5 or 10mM ammonia in C6 glioma cells, where gammaGT activity is intrinsically low and was not increased by ammonia. Collectively, the results suggest that in rats with HA or HE ammonia specifically promote GSH synthesis and export from astrocytes and increase its extracellular degradation, which may improve the availability of precursors for GSH synthesis in neurons and their resistance to ammonia toxicity.


Assuntos
Amônia/toxicidade , Astrócitos/efeitos dos fármacos , Glutationa/metabolismo , Hiperamonemia/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Amônia/sangue , Animais , Astrócitos/metabolismo , Linhagem Celular Tumoral , Espaço Extracelular/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley
18.
Toxicol Sci ; 117(1): 169-79, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20547568

RESUMO

Expansion of a polyglutamine tract in Huntingtin (Htt) leads to the degeneration of medium spiny neurons in Huntington's disease (HD). Furthermore, the HTT gene has been functionally linked to iron (Fe) metabolism, and HD patients show alterations in brain and peripheral Fe homeostasis. Recently, we discovered that expression of mutant HTT is associated with impaired manganese (Mn) uptake following overexposure in a striatal neuronal cell line and mouse model of HD. Here we test the hypothesis that the transferrin receptor (TfR)-mediated Fe uptake pathway is responsible for the HD-associated defects in Mn uptake. Western blot analysis showed that TfR levels are reduced in the mutant STHdh(Q111/Q111) striatal cell line, whereas levels of the Fe and Mn transporter, divalent metal transporter 1 (DMT1), are unchanged. To stress the Fe transport system, we exposed mutant and wild-type cells to elevated Fe(III), which revealed a subtle impairment in net Fe uptake only at the highest Fe exposures. In contrast, the HD mutant line exhibited substantial deficits in net Mn uptake, even under basal conditions. Finally, to functionally evaluate a role for Fe transporters in the Mn uptake deficit, we examined Mn toxicity in the presence of saturating Fe(III) levels. Although Fe(III) exposure decreased Mn neurotoxicity, it did so equally for wild-type and mutant cells. Therefore, although Fe transporters contribute to Mn uptake and toxicity in the striatal cell lines, functional alterations in this pathway are insufficient to explain the strong Mn resistance phenotype of this HD cell model.


Assuntos
Corpo Estriado/efeitos dos fármacos , Modelos Animais de Doenças , Homeostase , Doença de Huntington/metabolismo , Ferro/metabolismo , Manganês/metabolismo , Manganês/toxicidade , Animais , Transporte Biológico , Proteínas de Transporte de Cátions/metabolismo , Corpo Estriado/metabolismo , Humanos , Proteína Huntingtina , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética
19.
J Neurochem ; 112(1): 227-37, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19845833

RESUMO

Recognizing the similarities between Huntington's disease (HD) pathophysiology and the neurotoxicology of various metals, we hypothesized that they may exhibit disease-toxicant interactions revealing cellular pathways underlying neurodegeneration. Here, we utilize metals and the STHdh mouse striatal cell line model of HD to perform a gene-environment interaction screen. We report that striatal cells expressing mutant Huntingtin exhibit elevated sensitivity to cadmium toxicity and resistance to manganese toxicity. This neuroprotective gene-environment interaction with manganese is highly specific, as it does not occur with iron, copper, zinc, cobalt, cadmium, lead, or nickel ions. Analysis of the Akt cell stress signaling pathway showed diminished activation with manganese exposure and elevated activation after cadmium exposure in the mutant cells. Direct examination of intracellular manganese levels found that mutant cells have a significant impairment in manganese accumulation. Furthermore, YAC128Q mice, a HD model, showed decreased total striatal manganese levels following manganese exposure relative to wild-type mice. Thus, this disease-toxicant interaction screen has revealed that expression of mutant Huntingtin results in heightened sensitivity to cadmium neurotoxicity and a selective impairment of manganese accumulation.


Assuntos
Doença de Huntington/genética , Doença de Huntington/prevenção & controle , Manganês/toxicidade , Animais , Bovinos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Proteína Huntingtina , Doença de Huntington/induzido quimicamente , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética
20.
Neurochem Res ; 33(2): 267-72, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17726645

RESUMO

Ammonia neurotoxicity is associated with overactivation of N-methyl-D-aspartate (NMDA) receptors leading to enhanced nitric oxide and cyclic GMP synthesis and to accumulation of reactive oxygen and nitrogen species. Ammonia is detoxified in the brain via synthesis of glutamine, which if accumulated in excess contributes to astrocytic swelling, mitochondrial dysfunction and cerebral edema. This study was aimed at testing the hypothesis that the activity of the NMDA/NO/cGMP pathway is controlled by the ammonia-induced production of Gln in the brain. Ammonium chloride (final concentration 5 mM), infused for 40 min to the rat striatum via a microdialysis probe, caused a significant increase in Gln (by 40%), NO oxidation products (nitrite+nitrate=NOx) (by 35%) and cGMP (by 50%) concentration in the microdialysate. A Gln synthetase inhibitor, methionine sulfoximine (MSO, 5 mM), added directly to the microdialysate, completely prevented ammonia-mediated production of Gln, and paradoxically, it increased ammonia-mediated production of NOx and cGMP by 230% and 250%, respectively. Of note, MSO given alone significantly reduced basal Gln concentration in the rat striatum, had no effect on the basal NOx concentration, and attenuated basal concentration of cGMP in the microdialysate by 50%. The results of the present study suggest that Gln, at physiological concentrations, may ameliorate excessive activation of the NO-cGMP pathway by neurotoxic concentrations of ammonia. However, in view of potential direct interference of MSO with the pathway, exogenously added Gln and less toxic modulators of Gln content and/or transport will have to be employed in further studies on the underlying mechanisms.


Assuntos
Amônia/farmacologia , Corpo Estriado/efeitos dos fármacos , Glutamina/biossíntese , Guanosina Monofosfato/metabolismo , Metionina Sulfoximina/farmacologia , Óxido Nítrico/metabolismo , Animais , Corpo Estriado/metabolismo , Glutamina/antagonistas & inibidores , Masculino , Microdiálise , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA