Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Ecotoxicol Environ Saf ; 271: 115995, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38245935

RESUMO

Fenpropathrin (Fen), a volatile pyrethroid insecticide, is used widely for agricultural applications and has been reported to increase the risk of Parkinson's disease (PD). However, the molecular basis, underlying mechanisms, and pathophysiology of Fen-exposed Parkinsonism remain unknown. Recent studies have revealed epigenetic mechanisms underlying PD-related pathway regulation, including DNA methylation. Epigenetic mechanisms are potential targets for therapeutic intervention in neurodegenerative diseases. After whole-genome bisulfite sequencing (WGBS) of midbrain tissues from a Fen-exposed PD-like mouse model, we performed an association analysis of DNA methylation and gene expression. Then we successfully screened for the DNA methylation differential gene Ambra1, which is closely related to PD. The hypermethylation-low expression Ambra1 gene aggravated DA neuron damage in vitro and in vivo through the Ambra1/Parkin/LC3B-mediated mitophagy pathway. We administered 5-aza-2'-deoxycytidine (5-Aza-dC) to upregulate Ambra1 expression, thereby reducing Ambra1-mediated mitophagy and protecting DA neurons against Fen-induced damage. In conclusion, these findings elucidate the potential function of Ambra1 under the regulation of DNA methylation, suggesting that the inhibition of DNA methylation may alleviate Fen-exposed neuron damage.


Assuntos
Doença de Parkinson , Piretrinas , Camundongos , Animais , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Neurônios Dopaminérgicos/metabolismo , Metilação de DNA , Regulação para Baixo , Piretrinas/toxicidade , Piretrinas/metabolismo , Modelos Animais de Doenças , Decitabina , Proteínas Adaptadoras de Transdução de Sinal/genética
2.
J Transl Med ; 21(1): 747, 2023 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-37875930

RESUMO

BACKGROUND: The pathogenesis of Parkinson's disease (PD) has not been fully elucidated, and there are no effective disease-modifying drugs for the treatment of PD. Mesenchymal stem cells have been used to treat several diseases, but are not readily available. METHODS: Here, we used phenotypically uniform trophoblast stage-derived mesenchymal stem cells (T-MSCs) from embryonic stem cells, which are capable of stable production, and their exosomes (T-MSCs-Exo) to explore the molecular mechanisms involved in dopaminergic (DA) neuron protection in PD models using experimental assays (e.g., western blotting, immunofluorescence and immunohistochemistry staining). RESULTS: We assessed the levels of DA neuron injury and oxidative stress in MPTP-induced PD mice and MPP+-induced MN9D cells after treating them with T-MSCs or T-MSCs-Exo. Furthermore, T-MSCs-Exo miRNA sequencing analysis revealed that miR-100-5p-enriched T-MSCs-Exo directly targeted the 3' UTR of NOX4, which could protect against the loss of DA neurons, maintain nigro-striatal system function, ameliorate motor deficits, and reduce oxidative stress via the Nox4-ROS-Nrf2 axis in PD models. CONCLUSIONS: The study suggests that miR-100-5p-enriched T-MSCs-Exo may be a promising biological agent for the treatment of PD. Schematic summary of the mechanism underlying the neuroprotective actions of T-MSCs-Exo in PD. T-MSCs Exo may inhibit the expression level of the target gene NOX4 by delivering miR-100-5p, thereby reducing ROS production and alleviating oxidative stress via the Nox4-ROS-Nrf2 axis, thus improving DA neuron damage in PD.


Assuntos
Exossomos , Células-Tronco Mesenquimais , MicroRNAs , Doença de Parkinson , Camundongos , Animais , MicroRNAs/genética , MicroRNAs/metabolismo , Antioxidantes/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Exossomos/metabolismo , Doença de Parkinson/genética , Doença de Parkinson/terapia , Células-Tronco Mesenquimais/metabolismo , NADPH Oxidase 4/genética , NADPH Oxidase 4/metabolismo
3.
J Cell Mol Med ; 25(5): 2530-2548, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33523598

RESUMO

Excitatory amino acid transporter 2 (EAAT2), the gene of which is known as solute carrier family 1 member 2 (SLC1A2), is an important membrane-bound transporter that mediates approximately 90% of the transport and clearance of l-glutamate at synapses in the central nervous system (CNS). Transmembrane domain 2 (TM2) of EAAT2 is close to hairpin loop 2 (HP2) and far away from HP1 in the inward-facing conformation. In the present study, 14 crucial amino acid residues of TM2 were identified via alanine-scanning mutations. Further analysis in EAAT2-transfected HeLa cells in vitro showed that alanine substitutions of these residues resulted in a decrease in the efficiency of trafficking/targeting to the plasma membrane and/or reduced functionality of membrane-bound, which resulted in impaired transporter activity. After additional mutations, the transporter activities of some alanine-substitution mutants recovered. Specifically, the P95A mutant decreased EAAT2-associated anion currents. The Michaelis constant (Km ) values of the mutant proteins L85A, L92A and L101A were increased significantly, whereas R87 and P95A were decreased significantly, indicating that the mutations L85A, L92A and L101A reduced the affinity of the transporter and the substrate, whereas R87A and P95A enhanced this affinity. The maximum velocity (Vmax) values of all 14 alanine mutant proteins were decreased significantly, indicating that all these mutations reduced the substrate transport rate. These results suggest that critical residues in TM2 affect not only the protein expression and membrane-bound localization of EAAT2, but also its interactions with substrates. Additionally, our findings elucidate that the P95A mutant decreased EAAT2-related anion currents. Our results indicate that the TM2 of EAAT2 plays a vital role in the transport process. The key residues in TM2 affect protein expression in the membrane, substrate transport and the anion currents of EAAT2.


Assuntos
Aminoácidos , Ânions/metabolismo , Transportador 2 de Aminoácido Excitatório/química , Transportador 2 de Aminoácido Excitatório/metabolismo , Domínios e Motivos de Interação entre Proteínas , Aminoácidos/metabolismo , Transporte Biológico , Membrana Celular/metabolismo , Transportador 2 de Aminoácido Excitatório/genética , Células HeLa , Humanos , Cinese , Modelos Moleculares , Mutação , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade
4.
Neurobiol Dis ; 134: 104617, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31669733

RESUMO

As the most common cause of progressive cognitive decline in humans, Alzheimer's disease (AD) has been intensively studied, but the mechanisms underlying its profound synaptic dysfunction remain unclear. Here we confirm that exposing wild-type mice to an enriched environment (EE) facilitates signaling in the hippocampus that promotes long-term potentiation (LTP). Exposing the hippocampus of mice kept in standard housing to soluble Aß oligomers impairs LTP, but EE can fully prevent this. Mechanistically, the key molecular features of the EE benefit are an upregulation of miRNA-132 and an inhibition of histone deacetylase (HDAC) signaling. Specifically, soluble Aß oligomers decreased miR-132 expression and increased HDAC3 levels in cultured primary neurons. Further, we provide evidence that HDAC3 is a direct target of miR-132. Overexpressing miR-132 or injecting an HDAC3 inhibitor into mice in standard housing mimics the benefits of EE in enhancing hippocampal LTP and preventing hippocampal impairment by Aß oligomers in vivo. We conclude that EE enhances hippocampal synaptic plasticity by upregulating miRNA-132 and reducing HDAC3 signaling in a way that counteracts the synaptotoxicity of human Aß oligomers. Our findings provide a rationale for prolonged exposure to cognitive novelty and/or epigenetic modulation to lessen the progressive effects of Aß accumulation during human brain aging.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides/toxicidade , Histona Desacetilases/metabolismo , Abrigo para Animais , Potenciação de Longa Duração/fisiologia , MicroRNAs/metabolismo , Animais , Feminino , Regulação da Expressão Gênica/fisiologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Transdução de Sinais/fisiologia
5.
Neural Plast ; 2020: 8872296, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33281897

RESUMO

Astrocytes are a major constituent of the central nervous system (CNS). Astrocytic oxidative stress contributes to the development of Parkinson's disease (PD). Maintaining production of antioxidant and detoxification of reactive oxygen and nitrogen species (ROS/RNS) in astrocytes is critical to prevent PD. Study has illuminated that ascorbic acid (AA) stimulates dopamine synthesis and expression of tyrosine hydroxylase in human neuroblastoma cells. However, the role and regulatory mechanisms of AA on detoxification of astrocytes are still unclear. The purpose of our study is in-depth study of the regulatory mechanism of AA on detoxification of astrocytes. We found that AA pretreatment decreased the expression of ROS and inducible nitric oxide synthase (iNOS) in MPP+-treated astrocytes. In contrast, the expression levels of antioxidative substances-including superoxide dismutase (SOD), glutathione (GSH), and glutamate-cysteine ligase modifier (GCLM) subunit-were upregulated after AA pretreatment in MPP+-treated astrocytes. However, inhibition of NF-κB prevented such AA induced increases in antioxidative substances following MPP+ treatment in astrocytes, suggesting that AA improved antioxidative function of astrocytes through inhibiting NF-κB-mediated oxidative stress. Furthermore, in vivo studies revealed that AA preadministration also suppressed NF-κB and upregulated the expression levels of antioxidative substances in the midbrain of MPTP-treated mice. Additionally, pretreatment of AA alleviated MPTP-induced PD-like pathology in mice. Taken together, our results demonstrate that preadministration of AA improves the antioxidative function of astrocytes through suppressing NF-κB signaling, following alleviated the pathogenesis of PD which induced by MPTP. Hence, our findings elucidate a novel protective mechanism of AA in astrocytes.


Assuntos
Ácido Ascórbico/farmacologia , Astrócitos/efeitos dos fármacos , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Transtornos Parkinsonianos/mortalidade , Transdução de Sinais/efeitos dos fármacos , Animais , Astrócitos/metabolismo , Glutationa/metabolismo , Camundongos , Fármacos Neuroprotetores/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo
6.
Mol Pharmacol ; 95(1): 33-42, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30348896

RESUMO

To maintain efficient synaptic communication, glutamate transporters reuptake glutamate from the synaptic cleft and prevent glutamate concentrations from reaching neurotoxic levels. The number of amino acid residues of the transmembrane (TM) domain 4b-4c loop of mammalian excitatory amino acid transporters (EAATs) is 50 amino acids more than that of the prokaryotic homolog. To investigate the spatial proximity and functional significance of residues in glutamate transporters, cysteine pairs were introduced at positions A243 of the TM4b-4c loop and T396 or A414 of TM7, respectively. The transport activity of double mutants A243C/T396C and A243C/A414C was inhibited by Cu(II) (1,10-phenanthroline)3 [copper phenanthroline (CuPh)] and cadmium ions, but the uptake activity of corresponding single mutants remained unchanged. Treatment with dithiothreitol after CuPh restored much of the transport activity. The inhibitory effects of CuPh and cadmium could only be detected when cysteine pairs are in the same polypeptide. Therefore, we suggest that the formation of these disulfide bonds occurs intramolecularly. Glutamate, potassium, and DL-threo-ß-benzyloxyaspartate facilitated crosslinking in the A243C/T396C transporter and this suggests that the TM4b-4c loop and ß-bridge region in TM7 were drawn into close proximity to each other in the inward- and outward-facing conformation of EAAT1. Thus, these data provide evidence that substrate-induced structural rearrangements occur between the TM4b-4c loop and TM7 during the transport cycle.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Cisteína/metabolismo , Transportador 1 de Aminoácido Excitatório/metabolismo , Ácido Glutâmico/metabolismo , Mutagênese/fisiologia , Sequência de Aminoácidos , Ácido Aspártico/farmacologia , Transporte Biológico , Cádmio/farmacologia , Linhagem Celular Tumoral , Ditiotreitol/farmacologia , Células HeLa , Humanos , Proteínas de Membrana/metabolismo , Fenantrolinas/farmacologia , Domínios Proteicos
7.
Mol Pharmacol ; 94(1): 713-721, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29654220

RESUMO

Glutamatergic synaptic transmitters are cleared from the synaptic cleft through excitatory amino acid transporters (EAATs) that are responsible for recycling glutamate and transporting it into neurons and glial cells. To probe the structural role of the TM4b-4c loop of EAAT1 (Rattus norvegicus), each of the 57 amino acid residues was mutated to cysteine. Thirteen of the single mutants have very low transport activity. Aqueous accessibility of the introduced cysteines from the remaining mutants was then explored by membrane-permeant and membrane-impermeant sulfhydryl reagents in different conditions. F190C, V238C, and A243C were affected by MTSET, whereas Q189C, F190C, V238C, A243C, and L244C were sensitive to MTSEA. Q189C and L244C transport activity was diminished in the presence of potassium, which is expected to favor the inward-facing conformation of the transporter. Inversely, L244C was protected by glutamate. The modification of A243C by MTSEA was enhanced by either potassium and glutamate or dl-threo-ß-benzyloxyaspartate. From these results, we suggest that residues F190C, V238C, and A243C may be located near the extracellular surface, and the TM4b-4c loop forms multiple reentrant membrane loops on the cell surface. Alternatively, F190C, V238C, and A243C may function in the transport pathway, which is exposed to MTSET. In addition, Q189C, A243C, and L244C are conformationally sensitive and may play a role in the transport cycle.


Assuntos
Transporte Biológico/fisiologia , Transportador 1 de Aminoácido Excitatório/metabolismo , Tetraspaninas/metabolismo , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Ácido Aspártico/metabolismo , Cisteína/metabolismo , Ácido Glutâmico/metabolismo , Células HeLa , Humanos , Mutagênese/fisiologia , Potássio/metabolismo , Estrutura Secundária de Proteína , Ratos , Relação Estrutura-Atividade
8.
J Neurochem ; 145(1): 34-50, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29364516

RESUMO

Recent studies have strongly shown that cell-to-cell transmission of neuropathogenic proteins is a common mechanism for the development of neurodegenerative diseases. However, the underlying cause is complex and little is known. Although distinct processes are involved in the pathogenesis of various diseases, they all share the common feature of iron accumulation, an attribute that is particularly prominent in synucleinopathies. However, whether iron is a cofactor in facilitating the spread of α-synuclein remains unclear. Here, we constructed a cell-to-cell transmission model of α-synuclein using SN4741 cell line based on adenovirus vectors. Cells were treated with FeCl2, and α-synuclein aggregation and transmission were then evaluated. In addition, the possible mechanisms were investigated through gene knockdown or over-expression. Our results demonstrated that iron promoted α-synuclein aggregation and transmission by inhibiting autophagosome-lysosome fusion. Furthermore, iron decreased the expression of nuclear transcription factor EB (TFEB), a master transcriptional regulator of autophagosome-lysosome fusion, and inhibited its nuclear translocation through activating AKT/mTORC1 signaling. After silencing TFEB, ratios of α-synuclein aggregation and transmission were not significantly altered by the presence of iron; on the other hand, when TFEB was over-expressed, the transmission of α-synuclein induced by iron was obviously reversed; suggesting the mechanism by which iron promotes α-synuclein transmission may be mediated by TFEB. Taken together, our data reveal a previously unknown relationship between iron and α-synuclein, and identify TFEB as not only a potential target for preventing α-synuclein transmission, but also a critical factor for iron-induced α-synuclein aggregation and transmission. Indeed, this newly discovered role of iron and TFEB in synucleinopathies may provide novel targets for developing therapeutic strategies to prevent α-synuclein transmission in Parkinson's disease.


Assuntos
Autofagossomos/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Ferro/metabolismo , Lisossomos/metabolismo , Agregação Patológica de Proteínas/metabolismo , alfa-Sinucleína/metabolismo , Animais , Autofagossomos/efeitos dos fármacos , Comunicação Celular/efeitos dos fármacos , Linhagem Celular , Humanos , Ferro/farmacologia , Lisossomos/efeitos dos fármacos , Camundongos
9.
J Gene Med ; 19(4)2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28221705

RESUMO

BACKGROUND: Accumulating evidence suggests that insulin-like growth factor 1 (IGF1) plays an important role in Parkinson's disease (PD) pathogenesis. However, it is not clear whether IGF1 polymorphism contributes to PD risk. METHODS: We performed a case-control study in a Han Chinese population that included 512 sporadic PD cases and 535 matched controls. All participants were genotyped for rs972936 using the Sequenom MassARRAY iPLEX platform. Serum IGF1 levels of 61 de novo, drug-naïve PD patients and 55 age- and sex-matched controls were also measured using an enzyme-linked immunosorbent assay. RESULTS: Genotype frequency of rs972936-CC was significantly associated with an increased PD risk (p = 0.009), especially in males (p = 0.024) and late-onset patients (p = 0.013). Serum IGF1 levels were significantly increased in de novo, drug-naïve PD patients compared to controls (p = 0.036), although they were not correlated with motor dysfunction in PD patients (p = 0.220). CONCLUSIONS: The present study shows that rs972936 polymorphism may increase susceptibility to PD, especially in males and late-onset patients. Furthermore, high serum IGF1 levels may be a potential diagnostic biomarker for PD in the Han Chinese population, although they do not correlate with a more severe motor dysfunction.


Assuntos
Povo Asiático/genética , Predisposição Genética para Doença , Fator de Crescimento Insulin-Like I/genética , Doença de Parkinson/genética , Polimorfismo Genético , Idade de Início , Idoso , Alelos , Biomarcadores , Estudos de Casos e Controles , China , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Doença de Parkinson/sangue , Doença de Parkinson/epidemiologia , Polimorfismo de Nucleotídeo Único , Vigilância da População
10.
J Gene Med ; 19(11): 360-365, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28990350

RESUMO

BACKGROUND: Parkinson's disease (PD) is the second most common neurodegenerative disorder worldwide. Epigenetic modifications, specifically DNA methylation, have been implicated in the development of this disease. Genetic variants of DNA methyltransferase 3b (DNMT3b), one of the most important DNA methyltransferases, were shown to be associated with PD in a Brazilian population. However, it is unclear whether genetic variants of DNMT3b increase the risk of PD in the Chinese Han people. The present study aimed to investigate the association of the DNMT3b variants rs2424913, rs998382 and rs2424932 with PD in a Chinese Han population. METHODS: We studied 487 Chinese Han patients with sporadic PD and 485 healthy age-, sex- and ethnicity-matched controls. DNA was extracted from peripheral blood leukocytes and the individual genotypes were determined using the SNaPshot method. RESULTS: We found that the rs2424932 and rs998382 variants were significantly associated with an increased risk of PD compared to the controls [rs2424932: odds ratio (OR) = 1.632, 95% confidence interval (CI) = 1.108-2.406, p = 0.013; rs998382: OR = 1.612, 95% CI = 1.103-2.382, p = 0.014]. Subgroup analysis suggested that female patients carrying the rs2424932 or rs998382 variants were more likely to develop PD than female controls (rs2424932: OR = 3.863, 95% CI = 2.004-7.445, p < 0.001; rs998382: OR = 3.679, 95% CI = 1.943-6.964, p < 0.001). Haplotype analysis indicated that the three variants comprised one block and that the Trs2424913 -Crs998382 -A rs2424932 haplotype was correlated with an increased risk of PD (p = 0.0046), especially for Chinese Han females (p < 0.0001). CONCLUSIONS: The results of the present study strongly suggest that DNMT3b variants are associated with PD in the Chinese Han people, especially females.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , Predisposição Genética para Doença/genética , Doença de Parkinson/genética , Polimorfismo de Nucleotídeo Único , Idoso , Alelos , Povo Asiático/genética , China , Feminino , Frequência do Gene , Predisposição Genética para Doença/etnologia , Genótipo , Haplótipos , Humanos , Masculino , Pessoa de Meia-Idade , Doença de Parkinson/enzimologia , Doença de Parkinson/etnologia , Fatores de Risco , Fatores Sexuais , DNA Metiltransferase 3B
11.
Neural Plast ; 2016: 8941327, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26981287

RESUMO

Parkinson's disease (PD) is the most common movement disorder disease in the elderly and is characterized by degeneration of dopamine neurons and formation of Lewy bodies. Glutamate is the major excitatory neurotransmitter in the central nervous system (CNS). If glutamate is not removed promptly in the synaptic cleft, it will excessively stimulate the glutamate receptors and induce excitotoxic effects on the CNS. With lack of extracellular enzyme to decompose glutamate, glutamate uptake in the synaptic cleft is mainly achieved by the excitatory amino acid transporters (EAATs, also known as high-affinity glutamate transporters). Current studies have confirmed that decreased expression and function of EAATs appear in PD animal models. Moreover, single unilateral administration of EAATs inhibitor in the substantia nigra mimics several PD features and this is a solid evidence supporting that decreased EAATs contribute to the process of PD. Drugs or treatments promoting the expression and function of EAATs are shown to attenuate dopamine neurons death in the substantia nigra and striatum, ameliorate the behavior disorder, and improve cognitive abilities in PD animal models. EAATs are potential effective drug targets in treatment of PD and thus study of relationship between EAATs and PD has predominant medical significance currently.


Assuntos
Encéfalo/metabolismo , Proteínas de Transporte de Glutamato da Membrana Plasmática/metabolismo , Ácido Glutâmico/metabolismo , Doença de Parkinson/metabolismo , Transmissão Sináptica , Animais , Humanos , Neurônios/metabolismo , Sinapses/metabolismo
12.
Mol Pharmacol ; 86(6): 657-64, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25267718

RESUMO

Excitatory amino acid transporter 1 plays an important role in keeping the synaptic glutamate concentration below neurotoxic levels by translocating this neurotransmitter into the cell. Both reentrant hairpin loops, HP1 and -2, have been shown to take part in binding the substrate and the more deeply buried sodium ion, and might therefore be a part of the intra- or extracellular gate of the transporter. However, the shape of the motion of either loop relative to transmembrane domain (TM) 4 during the transport cycle has not yet been fully resolved. Using copper(II) (1,10-phenanthroline)3 (CuPh) for cross-linking cysteine pairs, we found strong inhibition of transport when A243C (TM4) was combined with S366C (HP1), I453C (HP2), or T456C (HP2). These findings were reinforced by the impact of cadmium on transport activity, and both approaches consistently showed that proximity was exclusively intramonomeric. Under conditions that promote the inward-facing state, inhibition by CuPh in A243C/S366C was reduced, while the opposite was seen when the outward-facing one was stabilized, suggesting that the two positions are farther apart in the former conformation than in the latter. Surprisingly, maximal cross-linking of A243C with I453C or T456C was not observed under conditions that promote the inward-facing state. Altogether, our data suggest that the transporter may undergo complex relative movement between these positions on TM4 and HP1/HP2 during the transport cycle.


Assuntos
Transportador 1 de Aminoácido Excitatório/química , Ácido Aspártico/farmacologia , Cádmio/farmacologia , Reagentes de Ligações Cruzadas/farmacologia , Transportador 1 de Aminoácido Excitatório/fisiologia , Ácido Glutâmico/farmacologia , Células HeLa , Humanos , Estrutura Terciária de Proteína
13.
Amino Acids ; 46(7): 1697-705, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24692063

RESUMO

Excitatory amino acid transporter 1 (EAAT1) plays an important role in restricting the neurotoxicity of glutamate. Previous structure-function studies have provided evidence that reentrant helical hairpin loop (HP) 1 has predominant function during the transport cycle. The proposed internal gate HP1 is packed against transmembrane domain (TM) 2 and TM5 in its closed state, and two residues located in TM2 and HP2 of EAAT1 are in close proximity. However, the spatial relationship between TM2 and HP1 during the transport cycle remains unknown. In this study, we used chemical cross-linking of introduced cysteine pair (V96C and S366C) in a cysteine-less version of EAAT1 to assess the proximity of TM2 and HP1. Here, we show that inhibition of transport by copper(II)(1,10-phenanthroline)3 (CuPh) and cadmium ion (Cd(2+)) were observed in the V96C/S366C mutant. Glutamate or potassium significantly protected against the inhibition of transport activity of V96C/S366C by CuPh, while TBOA potentiated the inhibition of transport activity of V96C/S366C by CuPh. We also checked the kinetic parameters of V96C/S366C treated with or without CuPh in the presence of NaCl, NaCl + L-glutamate, NaCl + TBOA, and KCl, respectively. The sensitivity of V96C and S366C to membrane-impermeable sulfhydryl reagent MTSET [(2-trimethylammonium) methanethiosulfonate] was attenuated by glutamate or potassium. TBOA had no effect on the sensitivity of V96C and S366C to MTSET. These data suggest that the spatial relationship between Val-96 of TM2 and Ser-366 of HP1 is altered in the transport cycle.


Assuntos
Cisteína/genética , Transportador 1 de Aminoácido Excitatório/química , Transportador 1 de Aminoácido Excitatório/genética , Sequência de Aminoácidos , Ácido Aspártico/farmacologia , Cloreto de Cádmio/farmacologia , Reagentes de Ligações Cruzadas , Cisteína/química , Cisteína/metabolismo , Ditiotreitol/farmacologia , Transportador 1 de Aminoácido Excitatório/metabolismo , Ácido Glutâmico/metabolismo , Ácido Glutâmico/farmacocinética , Células HeLa/efeitos dos fármacos , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fenantrolinas/farmacologia , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
14.
Zool Res ; 45(1): 108-124, 2024 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-38114437

RESUMO

Parkinson's disease (PD) is a neurodegenerative condition that results in dyskinesia, with oxidative stress playing a pivotal role in its progression. Antioxidant peptides may thus present therapeutic potential for PD. In this study, a novel cathelicidin peptide (Cath-KP; GCSGRFCNLFNNRRPGRLTLIHRPGGDKRTSTGLIYV) was identified from the skin of the Asiatic painted frog ( Kaloula pulchra). Structural analysis using circular dichroism and homology modeling revealed a unique αßß conformation for Cath-KP. In vitro experiments, including free radical scavenging and ferric-reducing antioxidant analyses, confirmed its antioxidant properties. Using the 1-methyl-4-phenylpyridinium ion (MPP +)-induced dopamine cell line and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice, Cath-KP was found to penetrate cells and reach deep brain tissues, resulting in improved MPP +-induced cell viability and reduced oxidative stress-induced damage by promoting antioxidant enzyme expression and alleviating mitochondrial and intracellular reactive oxygen species accumulation through Sirtuin-1 (Sirt1)/Nuclear factor erythroid 2-related factor 2 (Nrf2) pathway activation. Both focal adhesion kinase (FAK) and p38 were also identified as regulatory elements. In the MPTP-induced PD mice, Cath-KP administration increased the number of tyrosine hydroxylase (TH)-positive neurons, restored TH content, and ameliorated dyskinesia. To the best of our knowledge, this study is the first to report on a cathelicidin peptide demonstrating potent antioxidant and neuroprotective properties in a PD model by targeting oxidative stress. These findings expand the known functions of cathelicidins, and hold promise for the development of therapeutic agents for PD.


Assuntos
Discinesias , Fármacos Neuroprotetores , Doença de Parkinson , Animais , Camundongos , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/uso terapêutico , 1-Metil-4-fenilpiridínio/farmacologia , 1-Metil-4-fenilpiridínio/uso terapêutico , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Catelicidinas/metabolismo , Discinesias/tratamento farmacológico , Discinesias/veterinária , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Estresse Oxidativo , Doença de Parkinson/veterinária
15.
J Biol Chem ; 287(21): 17198-17205, 2012 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-22493292

RESUMO

Transporters of the major excitatory neurotransmitter glutamate play a crucial role in glutamatergic neurotransmission by removing their substrate from the synaptic cleft. The transport mechanism involves co-transport of glutamic acid with three Na(+) ions followed by countertransport of one K(+) ion. Structural work on the archeal homologue Glt(Ph) indicates a role of a conserved asparagine in substrate binding. According to a recent proposal, this residue may also participate in a novel Na(+) binding site. In this study, we characterize mutants of this residue from the neuronal transporter EAAC1, Asn-451. None of the mutants, except for N451S, were able to exhibit transport. However, the K(m) of this mutant for l-aspartate was increased ∼30-fold. Remarkably, the increase for d-aspartate and l-glutamate was 250- and 400-fold, respectively. Moreover, the cation specificity of N451S was altered because sodium but not lithium could support transport. A similar change in cation specificity was observed with a mutant of a conserved threonine residue, T370S, also implicated to participate in the novel Na(+) site together with the bound substrate. In further contrast to the wild type transporter, only l-aspartate was able to activate the uncoupled anion conductance by N451S, but with an almost 1000-fold reduction in apparent affinity. Our results not only provide experimental support for the Na(+) site but also suggest a distinct orientation of the substrate in the binding pocket during the activation of the anion conductance.


Assuntos
Asparagina/química , Asparagina/metabolismo , Transportador 3 de Aminoácido Excitatório/química , Transportador 3 de Aminoácido Excitatório/metabolismo , Substituição de Aminoácidos , Animais , Asparagina/genética , Sítios de Ligação , Transportador 3 de Aminoácido Excitatório/genética , Transporte de Íons/fisiologia , Mutação de Sentido Incorreto , Coelhos , Especificidade por Substrato , Xenopus laevis
16.
ACS Chem Neurosci ; 14(11): 2112-2122, 2023 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-37203180

RESUMO

Human exposure to fenpropathrin, a widely used pesticide, is linked to Parkinson's-like symptoms in the body. However, a specific pathogenic mechanism is still unclear. This study found that fenpropathrin increased the expression of murine double minute 2 (Mdm2) and reduced the expression of p53. Fenpropathrin stimulated the expression of neural precursor cell expressed, developmentally down-regulated 4-like (Nedd4L) and promoted the secretion of the inflammatory cytokine interleukin-6 (IL-6) through the Mdm2-p53 pathway. Nedd4L, a ubiquitin ligase, mediated the ubiquitination degradation of glutamate transporter 1 (GLT-1), resulting in glutamate accumulation and excitotoxicity aggravation. Our findings elucidate part of the pathogenic mechanism of fenpropathrin toxicity and provide scientific evidence to help develop guidance for pesticide control and environmental protection.


Assuntos
Praguicidas , Proteínas Proto-Oncogênicas c-mdm2 , Humanos , Animais , Camundongos , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Interleucina-6 , Ubiquitinação
17.
ACS Nano ; 17(20): 19961-19980, 2023 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-37807265

RESUMO

Parkinson's disease (PD) is a neurodegenerative disorder characterized by the degeneration of dopamine (DA) neurons in the midbrain substantia nigra pars compacta (SNpc). While existing therapeutic strategies can alleviate PD symptoms, they cannot inhibit DA neuron loss. Herein, a tailor-made human serum albumin (HSA)-based selenium nanosystem (HSA/Se nanoparticles, HSA/Se NPs) to treat PD that can overcome the intestinal epithelial barrier (IEB) and blood-brain barrier (BBB) is described. HSA, a transporter for drug delivery, has superior biological characteristics that make it an ideal potential drug delivery substance. Findings reveal that HSA/Se NPs have lower toxicity and higher efficacy than other selenium species and the ability to overcome the IEB and BBB to enrich DA neurons, which then protect MN9D cells from MPP+-induced neurotoxicity and ameliorate both behavioral deficits and DA neuronal death in MPTP-model mice. Thus, a therapeutic drug delivery system composed of orally gavaged HSA/Se NPs for the treatment of PD is described.


Assuntos
Nanopartículas , Doença de Parkinson , Selênio , Humanos , Camundongos , Animais , Doença de Parkinson/tratamento farmacológico , Neurônios Dopaminérgicos , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças
18.
Proc Natl Acad Sci U S A ; 106(34): 14297-302, 2009 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-19706515

RESUMO

Glutamate transporters maintain low synaptic concentrations of neurotransmitter by coupling uptake to flux of other ions. Their transport cycle consists of two separate translocation steps, namely cotransport of glutamic acid with three Na(+) followed by countertransport of K(+). Two Tl(+) binding sites, presumed to serve as sodium sites, were observed in the crystal structure of a related archeal homolog and the side chain of a conserved aspartate residue contributed to one of these sites. We have mutated the corresponding residue of the eukaryotic glutamate transporters GLT-1 and EAAC1 to asparagine, serine, and cysteine. Remarkably, these mutants exhibited significant sodium-dependent radioactive acidic amino acid uptake when expressed in HeLa cells. Reconstitution experiments revealed that net uptake by the mutants in K(+)-loaded liposomes was impaired. However, with Na(+) and unlabeled L-aspartate inside the liposomes, exchange levels were around 50-90% of those by wild-type. In further contrast to wild-type, where either substrate or K(+) stimulated the anion conductance by the transporter, substrate but not K(+) modulated the anion conductance of the mutants expressed in oocytes. Both with wild-type EAAC1 and EAAC1-D455N, not only sodium but also lithium could support radioactive acidic amino acid uptake. In contrast, with D455S and D455C, radioactive uptake was only observed in the presence of sodium. Thus the conserved aspartate is required for transporter-cation interactions in each of the two separate translocation steps and likely participates in an overlapping sodium and potassium binding site.


Assuntos
Encéfalo/metabolismo , Cátions/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Tálio/metabolismo , Animais , Proteínas Arqueais/genética , Proteínas Arqueais/metabolismo , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Ácido Aspártico/farmacologia , Sítios de Ligação , Transportador 2 de Aminoácido Excitatório/genética , Transportador 2 de Aminoácido Excitatório/fisiologia , Transportador 3 de Aminoácido Excitatório/genética , Transportador 3 de Aminoácido Excitatório/metabolismo , Transportador 3 de Aminoácido Excitatório/fisiologia , Feminino , Células HeLa , Humanos , Transporte de Íons/efeitos dos fármacos , Lipossomos , Lítio/metabolismo , Lítio/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Mutação , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Oócitos/fisiologia , Potássio/metabolismo , Potássio/farmacologia , Ligação Proteica , Coelhos , Ratos , Sódio/metabolismo , Sódio/farmacologia , Xenopus laevis
19.
Proc Natl Acad Sci U S A ; 106(49): 20752-7, 2009 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-19926849

RESUMO

Glutamate transporters regulate synaptic concentrations of this neurotransmitter by coupling its flux to that of sodium and other cations. Available crystal structures of an archeal homologue of these transporters, GltPh, resemble an extracellular-facing state, in which the bound substrate is occluded only by a small helical hairpin segment called HP2. However, a pathway to the cytoplasmic side of the membrane is not clearly apparent. We previously modeled an alternate state of a transporter from the neurotransmitter:sodium symporter family, which has an entirely different fold, solely on the presence of inverted-topology structural repeats. In GltPh, we identified two distinct sets of inverted-topology repeats and used these repeats to model an inward-facing conformation of the protein. To test this model, we introduced pairs of cysteines into the neuronal glutamate transporter EAAC1, at positions that are >27 A apart in the crystal structures of GltPh, but approximately = 10 A apart in the inward-facing model. Transport by these mutants was activated by pretreatment with the reducing agent dithithreitol. Subsequent treatment with the oxidizing agent copper(II)(1,10-phenantroline)(3) abolished this activation. The inhibition of transport was potentiated under conditions thought to promote the inward-facing conformation of the transporter. By contrast, the inhibition was reduced in the presence of the nontransportable substrate analogue D,L-threo-beta-benzyloxyaspartate, which favors the outward-facing conformation. Other conformation-sensitive accessibility measurements are also accommodated by our inward-facing model. These results suggest that the inclusion of inverted-topology repeats in transporters may provide a general solution to the requirement for two symmetry-related states in a single protein.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/química , Proteínas Arqueais/química , Proteínas Arqueais/metabolismo , Pyrococcus horikoshii/metabolismo , Sequências Repetitivas de Aminoácidos , Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Proteínas Arqueais/antagonistas & inibidores , Transporte Biológico/efeitos dos fármacos , Reagentes de Ligações Cruzadas/farmacologia , Cisteína/metabolismo , Citoplasma/química , Citoplasma/efeitos dos fármacos , Ditiotreitol/farmacologia , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fenantrolinas/farmacologia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Pyrococcus horikoshii/efeitos dos fármacos , Coelhos , Especificidade por Substrato/efeitos dos fármacos , Xenopus
20.
Front Immunol ; 13: 907047, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35812414

RESUMO

Respiratory syncytial virus (RSV) infection is the most frequent cause of hospitalization in pediatric patients. Current systemic treatment and vaccines are not curative and re-infection is often associated with a more drastic incidence of the disease. Baicalin is a flavonoid isolated from Scutellaria baicalensis with potent anti-viral characteristics, namely against RSV. However, its precise mechanism of action remains unclear. Here, using in vitro methods and an in vivo murine model of RSV infection, we showed that baicalin inhibits RSV replication induces translational upregulation of type I interferons (IFNs), IFN-α and IFN-ß, and reverses epithelial thickening in lung tissues. Moreover, baicalin inhibits transcription of the RSV non-structural proteins NS1 and NS2. Molecular docking and surface plasmon resonance-based affinity analysis showed that baicalin also binds to the α3 helix of the NS1 protein with an affinity constant of 1.119 × 10-5 M. Polysome profiling showed that baicalin inhibits translation of the RSV matrix protein (M) RNA. Baicalin mediates increased release of the ribosomal protein L13a from the large ribosomal subunit, where the extra ribosomal subunit L13a inhibits M RNA translation. These results comprehensively establish the multiple mechanisms by which baicalin induces a potent innate immune response against RSV infection.


Assuntos
Infecções por Vírus Respiratório Sincicial , Vírus Sincicial Respiratório Humano , Animais , Criança , Flavonoides/farmacologia , Humanos , Imunidade Inata , Camundongos , Simulação de Acoplamento Molecular , RNA/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA