Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Headache Pain ; 23(1): 39, 2022 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-35350973

RESUMO

BACKGROUND: Restless legs syndrome is a highly prevalent comorbidity of migraine; however, its genetic contributions remain unclear. OBJECTIVES: To identify the genetic variants of restless legs syndrome in migraineurs and to investigate their potential pathogenic roles. METHODS: We conducted a two-stage genome-wide association study (GWAS) to identify susceptible genes for restless legs syndrome in 1,647 patients with migraine, including 264 with and 1,383 without restless legs syndrome, and also validated the association of lead variants in normal controls unaffected with restless legs syndrome (n = 1,053). We used morpholino translational knockdown (morphants), CRISPR/dCas9 transcriptional knockdown, transient CRISPR/Cas9 knockout (crispants) and gene rescue in one-cell stage embryos of zebrafish to study the function of the identified genes. RESULTS: We identified two novel susceptibility loci rs6021854 (in VSTM2L) and rs79823654 (in CCDC141) to be associated with restless legs syndrome in migraineurs, which remained significant when compared to normal controls. Two different morpholinos targeting vstm2l and ccdc141 in zebrafish demonstrated behavioural and cytochemical phenotypes relevant to restless legs syndrome, including hyperkinetic movements of pectoral fins and decreased number in dopaminergic amacrine cells. These phenotypes could be partially reversed with gene rescue, suggesting the specificity of translational knockdown. Transcriptional CRISPR/dCas9 knockdown and transient CRISPR/Cas9 knockout of vstm2l and ccdc141 replicated the findings observed in translationally knocked-down morphants. CONCLUSIONS: Our GWAS and functional analysis suggest VSTM2L and CCDC141 are highly relevant to the pathogenesis of restless legs syndrome in migraineurs.


Assuntos
Síndrome das Pernas Inquietas , Animais , Estudo de Associação Genômica Ampla , Humanos , Síndrome das Pernas Inquietas/complicações , Síndrome das Pernas Inquietas/genética , Peixe-Zebra/genética
2.
PLoS One ; 15(9): e0232729, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32915786

RESUMO

Zinc ions (Zn2+) are important messenger molecules involved in various physiological functions. To maintain the homeostasis of cytosolic Zn2+ concentration ([Zn2+]c), Zrt/Irt-related proteins (ZIPs) and Zn2+ transporters (ZnTs) are the two families of proteins responsible for decreasing and increasing the [Zn2+]c, respectively, by fluxing Zn2+ across the membranes of the cell and intracellular compartments in opposite directions. Most studies focus on the cytotoxicity incurred by a high concentration of [Zn2+]c and less investigate the [Zn2+]c at physiological levels. Zinc oxide-nanoparticle (ZnO-NP) is blood brain barrier-permeable and elevates the [Zn2+]c to different levels according to the concentrations of ZnO-NP applied. In this study, we mildly elevated the [Zn2+]c by ZnO-NP at concentrations below 1 µg/ml, which had little cytotoxicity, in cultured human neuroblastoma SH-SY5Y cells and characterized the importance of Zn2+ transporters in 6-hydroxy dopamine (6-OHDA)-induced cell death. The results show that ZnO-NP at low concentrations elevated the [Zn2+]c transiently in 6 hr, then declined gradually to a basal level in 24 hr. Knocking down the expression levels of ZnT1 (located mostly at the plasma membrane) and ZIP8 (present in endosomes and lysosomes) increased and decreased the ZnO-NP-induced elevation of [Zn2+]c, respectively. ZnO-NP treatment reduced the basal levels of reactive oxygen species and Bax/Bcl-2 mRNA ratios; in addition, ZnO-NP decreased the 6-OHDA-induced ROS production, p53 expression, and cell death. These results show that ZnO-NP-induced mild elevation in [Zn2+]c activates beneficial effects in reducing the 6-OHDA-induced cytotoxic effects. Therefore, brain-delivery of ZnO-NP can be regarded as a potential therapy for neurodegenerative diseases.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Nanopartículas Metálicas , Óxido de Zinco/farmacologia , Zinco/metabolismo , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular , Expressão Gênica/efeitos dos fármacos , Humanos , Oxidopamina/metabolismo , Espécies Reativas de Oxigênio/metabolismo
3.
Sci Rep ; 9(1): 19301, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31848379

RESUMO

Alzheimer's disease (AD) is the most common type of dementia and also one of the leading causes of death worldwide. However, the underlying mechanisms remain unclear, and currently there is no drug treatment that can prevent or cure AD. Here, we have applied the advantages of using induced pluripotent stem cell (iPSC)-derived neurons (iNs) from AD patients, which are able to offer human-specific drug responsiveness, in order to evaluate therapeutic candidates for AD. Using approach involving an inducible neurogenin-2 transgene, we have established a robust and reproducible protocol for differentiating human iPSCs into glutamatergic neurons. The AD-iN cultures that result have mature phenotypic and physiological properties, together with AD-like biochemical features that include extracellular ß-amyloid (Aß) accumulation and Tau protein phosphorylation. By screening using a gene set enrichment analysis (GSEA) approach, Graptopetalum paraguayense (GP) has been identified as a potential therapeutic agent for AD from among a range of Chinese herbal medicines. We found that administration of a GP extract caused a significantly reduction in the AD-associated phenotypes of the iNs, including decreased levels of extracellular Aß40 and Aß42, as well as reduced Tau protein phosphorylation at positions Ser214 and Ser396. Additionally, the effect of GP was more prominent in AD-iNs compared to non-diseased controls. These findings provide valuable information that suggests moving extracts of GP toward drug development, either for treating AD or as a health supplement to prevent AD. Furthermore, our human iN-based platform promises to be a useful strategy when it is used for AD drug discovery.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/genética , Crassulaceae/química , Fragmentos de Peptídeos/genética , Proteínas tau/genética , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/efeitos dos fármacos , Descoberta de Drogas , Medicamentos de Ervas Chinesas/química , Medicamentos de Ervas Chinesas/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas do Tecido Nervoso/genética , Neurônios/efeitos dos fármacos , Neurônios/patologia
4.
PLoS Biol ; 17(10): e3000508, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31593566

RESUMO

CDGSH iron-sulfur domain-containing protein 2 (Cisd2) is pivotal to mitochondrial integrity and intracellular Ca2+ homeostasis. In the heart of Cisd2 knockout mice, Cisd2 deficiency causes intercalated disc defects and leads to degeneration of the mitochondria and sarcomeres, thereby impairing its electromechanical functioning. Furthermore, Cisd2 deficiency disrupts Ca2+ homeostasis via dysregulation of sarco/endoplasmic reticulum Ca2+-ATPase (Serca2a) activity, resulting in an increased level of basal cytosolic Ca2+ and mitochondrial Ca2+ overload in cardiomyocytes. Most strikingly, in Cisd2 transgenic mice, a persistently high level of Cisd2 is sufficient to delay cardiac aging and attenuate age-related structural defects and functional decline. In addition, it results in a younger cardiac transcriptome pattern during old age. Our findings indicate that Cisd2 plays an essential role in cardiac aging and in the heart's electromechanical functioning. They highlight Cisd2 as a novel drug target when developing therapies to delay cardiac aging and ameliorate age-related cardiac dysfunction.


Assuntos
Senilidade Prematura/genética , Envelhecimento/fisiologia , Bloqueio Atrioventricular/genética , Proteínas Relacionadas à Autofagia/genética , Coração/fisiopatologia , Proteínas do Tecido Nervoso/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Senilidade Prematura/metabolismo , Senilidade Prematura/fisiopatologia , Animais , Bloqueio Atrioventricular/diagnóstico por imagem , Bloqueio Atrioventricular/metabolismo , Bloqueio Atrioventricular/fisiopatologia , Proteínas Relacionadas à Autofagia/deficiência , Cálcio/metabolismo , Eletrocardiografia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Coração/fisiologia , Homeostase/fisiologia , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias Cardíacas/genética , Mitocôndrias Cardíacas/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Proteínas do Tecido Nervoso/deficiência , Sarcômeros/fisiologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Transcriptoma
5.
Mol Neurobiol ; 56(12): 8475-8476, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31317492

RESUMO

The original version of this article unfortunately contained a mistake. The authors observed inadvertent error in Fig. 7d, in which the image of the GFAP/DAPI in the WT saline treated mice was rotated left 90-degree by mistake. The corrected representative image is given below.

6.
Mol Neurobiol ; 56(12): 8451-8474, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31257558

RESUMO

Astrocytes play pivotal roles in regulating glutamate homeostasis at tripartite synapses. Inhibition of soluble epoxide hydrolase (sEHi) provides neuroprotection by blocking the degradation of 14,15-epoxyeicosatrienoic acid (14,15-EET), a lipid mediator whose synthesis can be activated downstream from group 1 metabotropic glutamate receptor (mGluR) signaling in astrocytes. However, it is unclear how sEHi regulates glutamate excitotoxicity. Here, we used three primary rat cortical culture systems, neuron-enriched (NE), astrocyte-enriched glia-neuron mix (GN), and purified astrocytes, to delineate the underlying mechanism by which sEHi and 14,15-EET attenuate excitotoxicity. We found that sEH inhibitor 12-(3-adamantan-1-yl-ureido)-dodecanoic acid (AUDA) and 14,15-EET both attenuated N-methyl-D-aspartate (NMDA)-induced neurite damage and cell death in GN, not NE, cortical cultures. The anti-excitotoxic effects of 14,15-EET and AUDA were both blocked by the group 1 mGluR5 antagonist 2-methyl-6-(phenylethynyl)pyridine (MPEP), as were their protective effects against NMDA-disrupted perineuronal astrocyte processes expressing glutamate transporter-1 (GLT-1) and subsequent glutamate uptake. Knockdown of sEH expression also attenuated NMDA neurotoxicity in mGluR5- and GLT-1-dependent manners. The 14,15-EET/AUDA-preserved astroglial integrity was confirmed in glutamate-stimulated primary astrocytes along with the reduction of the c-Jun N-terminal kinase 1 phosphorylation, in which the 14,15-EET effect is mGluR5-dependent. In vivo studies validated that sEHi and genetic deletion of sEH (Ephx2-KO) ameliorated excitotoxic kainic acid-induced seizure, memory impairment, and neuronal loss while preserving GLT-1-expressing perineuronal astrocytes in hippocampal CA3 subregions. These results suggest that 14,15-EET mediates mGluR5-dependent anti-excitotoxicity by protecting astrocytes to maintain glutamate homeostasis, which may account for the beneficial effect of sEH inhibition in excitotoxic brain injury and diseases.


Assuntos
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Astrócitos/patologia , Inibidores Enzimáticos/farmacologia , Epóxido Hidrolases/antagonistas & inibidores , Ácido Glutâmico/metabolismo , Homeostase , Plasticidade Neuronal/efeitos dos fármacos , Neurotoxinas/toxicidade , Ácido 8,11,14-Eicosatrienoico/farmacologia , Adamantano/análogos & derivados , Adamantano/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Epóxido Hidrolases/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Hipocampo/metabolismo , Ácido Caínico , Ácidos Láuricos/farmacologia , Camundongos Endogâmicos C57BL , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Modelos Biológicos , N-Metilaspartato , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos Sprague-Dawley , Receptor de Glutamato Metabotrópico 5/antagonistas & inibidores , Receptor de Glutamato Metabotrópico 5/metabolismo , Solubilidade
7.
Mol Neurobiol ; 56(9): 6095-6105, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30721447

RESUMO

Mutations in RAB18, a member of small G protein, cause Warburg micro syndrome (WARBM), whose clinical features include vision impairment, postnatal microcephaly, and lower limb spasticity. Previously, our Rab18-/- mice exhibited hind limb weakness and spasticity as well as signs of axonal degeneration in the spinal cord and lumbar spinal nerves. However, the cellular and molecular function of RAB18 and its roles in the pathogenesis of WARBM are still not fully understood. Using immunofluorescence staining and expression of Rab18 and organelle markers, we find that Rab18 associates with lysosomes and actively traffics along neurites in cultured neurons. Interestingly, Rab18-/- neurons exhibit impaired lysosomal transport. Using autophagosome marker LC3-II, we show that Rab18 dysfunction leads to aberrant autophagy activities in neurons. Electron microscopy further reveals accumulation of lipofuscin-like granules in the dorsal root ganglion of Rab18-/- mice. Surprisingly, Rab18 colocalizes, cofractionates, and coprecipitates with the lysosomal regulator Rab7, mutations of which cause Charcot-Marie-Tooth (CMT) neuropathy type 2B. Moreover, Rab7 is upregulated in Rab18-deficient neurons, suggesting a compensatory effect. Together, our results suggest that the functions of RAB18 and RAB7 in lysosomal and autophagic activities may constitute an overlapping mechanism underlying WARBM and CMT pathogenesis in the nervous system.


Assuntos
Anormalidades Múltiplas/metabolismo , Autofagia , Catarata/congênito , Doença de Charcot-Marie-Tooth/metabolismo , Córnea/anormalidades , Hipogonadismo/metabolismo , Deficiência Intelectual/metabolismo , Lisossomos/metabolismo , Microcefalia/metabolismo , Sistema Nervoso/metabolismo , Atrofia Óptica/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Catarata/metabolismo , Córnea/metabolismo , Epistasia Genética , Células HEK293 , Humanos , Laminopatias , Camundongos , Neurônios/metabolismo , Células PC12 , Ligação Proteica , Ratos , Ratos Sprague-Dawley
8.
J Cell Sci ; 131(23)2018 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-30404828

RESUMO

α-Synuclein is associated with Parkinson's disease, and is mainly localized in presynaptic terminals and regulates exocytosis, but its physiological roles remain controversial. Here, we studied the effects of soluble and aggregated α-synuclein on exocytosis, and explored the molecular mechanism by which α-synuclein interacts with regulatory proteins, including Rab3A, Munc13-1 (also known as Unc13a) and Munc18-1 (also known as STXBP1), in order to regulate exocytosis. Through fluorescence recovery after photobleaching experiments, overexpressed α-synuclein in PC12 cells was found to be in a monomeric form, which promotes exocytosis. In contrast, aggregated α-synuclein induced by lactacystin treatment inhibits exocytosis. Our results show that α-synuclein is involved in vesicle priming and fusion. α-Synuclein and phorbol 12-myristate 13-acetate (PMA), which is known to enhance vesicle priming mediated by Rab3A, Munc13-1 and Munc18-1, act on the same population of vesicles, but regulate priming independently. Furthermore, the results show a novel effects of α-synuclein on mobilizing Ca2+ release from thapsigargin-sensitive Ca2+ pools to enhance the ATP-induced [Ca2+]i increase, which enhances vesicle fusion. Our results provide a detailed understanding of the action of α-synuclein during the final steps of exocytosis.


Assuntos
Cálcio/metabolismo , Exocitose/fisiologia , Tapsigargina/farmacologia , alfa-Sinucleína/metabolismo , Animais , Fusão de Membrana/fisiologia , Células PC12 , Ratos , Tapsigargina/metabolismo , Transfecção , Proteína rab3A de Ligação ao GTP/genética , Proteína rab3A de Ligação ao GTP/metabolismo
9.
Cephalalgia ; 38(3): 466-475, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28952330

RESUMO

Background Susceptibility genes for migraine, despite it being a highly prevalent and disabling neurological disorder, have not been analyzed in Asians by genome-wide association study (GWAS). Methods We conducted a two-stage case-control GWAS to identify susceptibility genes for migraine without aura in Han Chinese residing in Taiwan. In the discovery stage, we genotyped 1005 clinic-based Taiwanese migraine patients and 1053 population-based sex-matched controls using Axiom Genome-Wide CHB Array. In the replication stage, we genotyped 27 single-nucleotide polymorphisms with p < 10-4 in 1120 clinic-based migraine patients and 604 sex-matched normal controls by using Sequenom. Variants at LRP1, TRPM8, and PRDM, which have been replicated in Caucasians, were also genotyped. Results We identified a novel susceptibility locus (rs655484 in DLG2) that reached GWAS significance level for migraine risk in Han Chinese ( p = 1.45 × 10-12, odds ratio [OR] = 2.42), and also another locus (rs3781545in GFRA1) with suggestive significance ( p = 1.27 × 10-7, OR = 1.38). In addition, we observed positive association signals with a similar trend to the associations identified in Caucasian GWASs for rs10166942 in TRPM8 (OR = 1.33, 95% confidence interval [CI] = 1.14-1.54, Ppermutation = 9.99 × 10-5; risk allele: T) and rs1172113 in LRP1 (OR = 1.23, 95% CI = 1.04-1.45, Ppermutation = 2.9 × 10-2; risk allele: T). Conclusion The present study is the first migraine GWAS conducted in Han-Chinese and Asians. The newly identified susceptibility genes have potential implications in migraine pathogenesis. DLG2 is involved in glutamatergic neurotransmission, and GFRA1 encodes GDNF receptors that are abundant in CGRP-containing trigeminal neurons. Furthermore, positive association signals for TRPM8 and LRP1 suggest the possibility for common genetic contributions across ethnicities.


Assuntos
Predisposição Genética para Doença/genética , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Guanilato Quinases/genética , Transtornos de Enxaqueca/genética , Proteínas Supressoras de Tumor/genética , Adulto , Povo Asiático/genética , Feminino , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Taiwan
10.
Pflugers Arch ; 470(1): 29-38, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28762161

RESUMO

Adrenal medullary chromaffin cells in mammals are innervated by sympathetic preganglionic nerve fibers, as are sympathetic ganglion neurons. Acetylcholine in the ganglion neurons is well established as mediating fast and slow excitatory postsynaptic potentials through nicotinic and muscarinic acetylcholine receptors (AChRs), respectively. The role of muscarinic AChRs during neuronal transmission in chromaffin cells varies among different mammals. Furthermore, the ion channel mechanisms associated with the muscarinic AChR-mediated increase in excitability of chromaffin cells are complicated and different from the excitation of ganglion neurons, which has been ascribed to the inhibition of M-type K+ channels. In this review, we focus on muscarinic receptor-mediated excitation in rodent and guinea pig chromaffin cells, in particular, on the role of muscarinic receptors in neuronal transmission, the muscarinic receptor subtypes involved in excitation and secretion, and the muscarinic regulation of ion channels including TWIK-related acid-sensitive K+ channels. Finally, we discuss prospectively the future of muscarinic receptor research in adrenal chromaffin cells.


Assuntos
Medula Suprarrenal/citologia , Células Cromafins/metabolismo , Canais de Potássio/metabolismo , Receptores Muscarínicos/metabolismo , Canais de Cátion TRPC/metabolismo , Potenciais de Ação , Medula Suprarrenal/metabolismo , Animais , Células Cromafins/fisiologia , Humanos , Receptores Muscarínicos/genética
11.
Cell Rep ; 21(8): 2198-2211, 2017 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-29166610

RESUMO

CISD2 is located within the chromosome 4q region frequently deleted in hepatocellular carcinoma (HCC). Mice with Cisd2 heterozygous deficiency develop a phenotype similar to the clinical manifestation of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). Cisd2 haploinsufficiency causes a low incidence (20%) of spontaneous HCC and promotes HBV-associated and DEN-induced HCC; conversely, 2-fold overexpression of Cisd2 suppresses HCC in these models. Mechanistically, Cisd2 interacts with Serca2b and mediates its Ca2+ pump activity via modulation of Serca2b oxidative modification, which regulates ER Ca2+ uptake and maintains intracellular Ca2+ homeostasis in the hepatocyte. CISD2 haploinsufficiency disrupts calcium homeostasis, causing ER stress and subsequent NAFLD and NASH. Hemizygous deletion and decreased expression of CISD2 are detectable in a substantial fraction of human HCC specimens. These findings substantiate CISD2 as a haploinsufficient tumor suppressor and highlights Cisd2 as a drug target when developing therapies to treat NAFLD/NASH and prevent HCC.


Assuntos
Cálcio/metabolismo , Carcinoma Hepatocelular/patologia , Proteínas de Transporte/metabolismo , Haploinsuficiência/genética , Neoplasias Hepáticas/patologia , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Proteínas Relacionadas à Autofagia , Carcinoma Hepatocelular/metabolismo , Proteínas de Transporte/genética , Homeostase/fisiologia , Humanos , Neoplasias Hepáticas/genética , Proteínas de Membrana/genética , Camundongos , Proteínas do Tecido Nervoso/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia
12.
Mol Cell Neurosci ; 82: 35-45, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28427888

RESUMO

Zinc ion (Zn2+), the second most abundant transition metal after iron in the body, is essential for neuronal activity and also induces toxicity if the concentration is abnormally high. Our previous results show that exposure of cultured cortical neurons to dopamine elevates intracellular Zn2+ concentrations ([Zn2+]i) and induces autophagosome formation but the mechanism is not clear. In this study, we characterized the signaling pathway responsible for the dopamine-induced elevation of [Zn2+]i and the effect of [Zn2+]i in modulating the autophagy in cultured rat embryonic cortical neurons. N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine (TPEN), a membrane-permeable Zn2+ chelator, could rescue the cell death and suppress the autophagosome puncta number induced by dopamine. Dopamine treatment increased the lipidation level of the endogenous microtubule-associated protein 1A/1B-light chain 3 (LC3 II), an autophagosome marker. TPEN added 1h before, but not after, dopamine treatment suppressed the dopamine-induced elevation of LC3 II level. Inhibitors of the dopamine D1-like receptor, protein kinase A (PKA), and NOS suppressed the dopamine-induced elevation of [Zn2+]i. PKA activators and NO generators directly increased [Zn2+]i in cultured neurons. Through cell fractionation, proteins with m.w. values between 5 and 10kD were found to release Zn2+ following NO stimulation. In addition, TPEN pretreatment and an inhibitor against PKA could suppress the LC3 II level increased by NO and dopamine, respectively. Therefore, our results demonstrate that dopamine-induced elevation of [Zn2+]i is mediated by the D1-like receptor-PKA-NO pathway and is important in modulating the cell death and autophagy.


Assuntos
Dopamina/metabolismo , Neurônios/metabolismo , Transdução de Sinais , Zinco/metabolismo , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Células Cultivadas , Quelantes/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Etilenodiaminas/farmacologia , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/efeitos dos fármacos , Óxido Nítrico/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos
13.
J Neurochem ; 139(1): 120-33, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27385273

RESUMO

The pathogenesis of Parkinson's disease (PD) is not completely understood, Zinc (Zn(2+) ) and dopamine (DA) have been shown to involve in the degeneration of dopaminergic cells. By microarray analysis, we identified Gadd45b as a candidate molecule that mediates Zn(2+) and DA-induced cell death; the mRNA and protein levels of Gadd45b are increased by Zn(2+) treatment and raised to an even higher level by Zn(2+) plus DA treatment. Zn(2+) plus DA treatment-induced PC12 cell death was enhanced when there was over-expression of Gadd45b and was decreased by knock down of Gadd45b. MAPK p38 and JNK signaling was able to cross-talk with Gadd45b during Zn(2+) and DA treatment. The synergistic effects of Zn(2+) and DA on PC12 cell death can be accounted for by an activation of the Gadd45b-induced cell death pathway and an inhibition of p38/JNK survival pathway. Furthermore, the in vivo results show that the levels of Gadd45b protein expression and phosphorylation of p38 were increased in the substantia nigra by the infusion of Zn(2+) /DA in the mouse brain and the level of Gadd45b mRNA is significantly higher in the substantia nigra of male PD patients than normal controls. The novel role of Gadd45b and its interactions with JNK and p38 will help our understanding of the pathogenesis of PD and help the development of future treatments for PD. Zinc and dopamine are implicated in the degeneration of dopaminergic neurons. We previously demonstrated that zinc and dopamine induced synergistic effects on PC12 cell death. Results from this study show that these synergistic effects can be accounted for by activation of the Gadd45b-induced cell death pathway and inhibition of the p38/JNK survival pathway. We provide in vitro and in vivo evidence to support a novel role for Gadd45b in the pathogenesis of Parkinson's disease.


Assuntos
Antígenos de Diferenciação/efeitos dos fármacos , Antígenos de Diferenciação/genética , Dopamina/toxicidade , Doença de Parkinson/genética , Doença de Parkinson/patologia , Zinco/toxicidade , Acetilcisteína/farmacologia , Animais , Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Morte Celular/efeitos dos fármacos , Sinergismo Farmacológico , Sequestradores de Radicais Livres/farmacologia , Técnicas de Silenciamento de Genes , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Necrose/patologia , Proteínas Nucleares/genética , Células PC12 , Ratos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Cell Calcium ; 59(6): 299-311, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27020658

RESUMO

Midbrain dopaminergic (DA) neurons are the key to finely tune the voluntary movement, habit and motivation. The progressive and selective degeneration of these neurons is a pathological hallmark of Parkinson's disease (PD). The susceptibility of DA neurons in the SNpc may result from differences in how Ca(2+) is handled. However, very little information is available about the mechanisms involved in the regulation of intracellular Ca(2+) concentration ([Ca(2+)]i) in DA neurons. In this study, the relative contributions of various Na(+)/Ca(2+) exchangers and their interplay with internal Ca(2+) stores, endoplasmic reticulum (ER) and the mitochondria, in the regulation of the [Ca(2+)]i of mouse mesencephalic neurons were characterized. Both the K(+)-dependent Na(+)/Ca(2+) exchanger (NCKX) and the K(+)-independent Na(+)/Ca(2+) exchanger (NCX) can be detected and are functional in DA and non-DA neurons. NCX accounts for the larger component of Na(+)/Ca(2+) exchange activity. Single-cell RT-PCR analysis showed each individual neuron expressed a distinct set of the Na(+)/Ca(2+) exchangers. Furthermore, the Na(+)/Ca(2+) exchangers play prominent roles in removing [Ca(2+)]i induced by glutamate but not [Ca(2+)]i induced by depolarization. The mitochondria serve as a major Ca(2+) sink and are functionally located close to NCX. In contrast, the ER is functionally located close to NCKX and acts primarily as a Ca(2+) source with marginal effects. This study reveals that the Na(+)/Ca(2+) exchangers, the ER and the mitochondria, which cooperate interactively, act similarly when regulating [Ca(2+)]i in mesencephalic DA and non-DA neurons. The heterogeneous expression of multiple types of Na(+)/Ca(2+) exchangers and the quantitative differences found in [Ca(2+)]i regulation, together with other risk factors specific to DA neurons such as dopamine oxidation resulting in oxidative stress, may drive these cells to undergo selective degeneration.


Assuntos
Cálcio/metabolismo , Retículo Endoplasmático/metabolismo , Mesencéfalo/citologia , Mitocôndrias/metabolismo , Neurônios/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Canais de Cálcio/metabolismo , Células Cultivadas , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Retículo Endoplasmático/efeitos dos fármacos , Glutamatos/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Modelos Biológicos , Neurônios/efeitos dos fármacos , Potássio/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Trocador de Sódio e Cálcio/genética
15.
J Neurosci ; 36(6): 2027-43, 2016 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-26865625

RESUMO

Growth-associated protein 43 (GAP43), a protein kinase C (PKC)-activated phosphoprotein, is often implicated in axonal plasticity and regeneration. In this study, we found that GAP43 can be induced by the endotoxin lipopolysaccharide (LPS) in rat brain astrocytes both in vivo and in vitro. The LPS-induced astrocytic GAP43 expression was mediated by Toll-like receptor 4 and nuclear factor-κB (NF-κB)- and interleukin-6/signal transducer and activator of transcription 3 (STAT3)-dependent transcriptional activation. The overexpression of the PKC phosphorylation-mimicking GAP43(S41D) (constitutive active GAP43) in astrocytes mimicked LPS-induced process arborization and elongation, while application of a NF-κB inhibitory peptide TAT-NBD or GAP43(S41A) (dominant-negative GAP43) or knockdown of GAP43 all inhibited astrogliosis responses. Moreover, GAP43 knockdown aggravated astrogliosis-induced microglial activation and expression of proinflammatory cytokines. We also show that astrogliosis-conditioned medium from GAP43 knock-down astrocytes inhibited GAP43 phosphorylation and axonal growth, and increased neuronal damage in cultured rat cortical neurons. These proneurotoxic effects of astrocytic GAP43 knockdown were accompanied by attenuated glutamate uptake and expression of the glutamate transporter excitatory amino acid transporter 2 (EAAT2) in LPS-treated astrocytes. The regulation of EAAT2 expression involves actin polymerization-dependent activation of the transcriptional coactivator megakaryoblastic leukemia 1 (MKL1), which targets the serum response elements in the promoter of rat Slc1a2 gene encoding EAAT2. In sum, the present study suggests that astrocytic GAP43 mediates glial plasticity during astrogliosis, and provides beneficial effects for neuronal plasticity and survival and attenuation of microglial activation. SIGNIFICANCE STATEMENT: Astrogliosis is a complex state in which injury-stimulated astrocytes exert both protective and harmful effects on neuronal survival and plasticity. In this study, we demonstrated for the first time that growth-associated protein 43 (GAP43), a well known growth cone protein that promotes axonal regeneration, can be induced in rat brain astrocytes by the proinflammatory endotoxin lipopolysaccharide via both nuclear factor-κB and signal transducer and activator of transcription 3-mediated transcriptional activation. Importantly, LPS-induced GAP43 mediates plastic changes of astrocytes while attenuating astrogliosis-induced microglial activation and neurotoxicity. Hence, astrocytic GAP43 upregulation may serve to indicate beneficial astrogliosis after CNS injury.


Assuntos
Astrócitos/efeitos dos fármacos , Proteína GAP-43/biossíntese , Proteína GAP-43/genética , Gliose/genética , Microglia/efeitos dos fármacos , NF-kappa B/genética , Síndromes Neurotóxicas/genética , Síndromes Neurotóxicas/patologia , Fator de Transcrição STAT3/genética , Receptor 4 Toll-Like/genética , Animais , Citocinas/biossíntese , Transportador 2 de Aminoácido Excitatório/biossíntese , Transportador 2 de Aminoácido Excitatório/genética , Ativação de Macrófagos/efeitos dos fármacos , Neurônios , Fosforilação , Ratos , Ratos Sprague-Dawley , Transativadores/biossíntese , Transativadores/genética , Fatores de Transcrição
16.
Cephalalgia ; 36(11): 1028-1037, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26643377

RESUMO

Objective Several genetic variants have been found to increase the risk of restless legs syndrome (RLS). The aim of the present study was to determine if these genetic variants were also associated with the comorbidity of RLS and migraine in patients. Methods Thirteen single-nucleotide polymorphisms (SNPs) at six RLS risk loci ( MEIS1, BTBD9, MAP2K5, PTPRD, TOX3, and an intergenic region on chromosome 2p14) were genotyped in 211 migraine patients with RLS and 781 migraine patients without RLS. Association analyses were performed for the overall cohort, as well as for the subgroups of patients who experienced migraines with and without aura and episodic migraines (EMs) vs. chronic migraines (CMs). In order to verify which genetic markers were potentially related to the incidence of RLS in migraine patients, multivariate regression analyses were also performed. Results Among the six tested loci, only MEIS1 was significantly associated with RLS. The most significant SNP of MEIS1, rs2300478, increased the risk of RLS by 1.42-fold in the overall cohort ( p = 0.0047). In the subgroup analyses, MEIS1 augmented the risk of RLS only in the patients who experienced EMs (odds ratio (OR) = 1.99, p = 0.0004) and not those experiencing CMs. Multivariate regression analyses further showed that rs2300478 in MEIS1 (OR = 1.39, p = 0.018), a CM diagnosis (OR = 1.52, p = 0.022), and depression (OR = 1.86, p = 0.005) were independent predictors of RLS in migraine. Conclusions MEIS1 variants were associated with an increased risk of RLS in migraine patients. It is possible that an imbalance in iron homeostasis and the dopaminergic system may represent a link between RLS incidence and migraines.


Assuntos
Predisposição Genética para Doença/epidemiologia , Predisposição Genética para Doença/genética , Transtornos de Enxaqueca/epidemiologia , Transtornos de Enxaqueca/genética , Proteína Meis1/genética , Síndrome das Pernas Inquietas/epidemiologia , Síndrome das Pernas Inquietas/genética , Adulto , Distribuição por Idade , Causalidade , Comorbidade , Feminino , Estudos de Associação Genética , Marcadores Genéticos/genética , Humanos , Masculino , Transtornos de Enxaqueca/diagnóstico , Polimorfismo de Nucleotídeo Único/genética , Prevalência , Síndrome das Pernas Inquietas/diagnóstico , Distribuição por Sexo , Taiwan/epidemiologia
17.
Exp Neurol ; 267: 143-51, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25779931

RESUMO

Mutations in the gene of RAB18, a member of Ras superfamily of small G-proteins, cause Warburg Micro Syndrome (WARBM) which is characterized by defective neurodevelopmental and ophthalmological phenotypes. Despite loss of Rab18 had been reported to induce disruption of the endoplasmic reticulum structure and neuronal cytoskeleton organization, parts of the pathogenic mechanism caused by RAB18 mutation remain unclear. From the N-ethyl-N-nitrosourea (ENU)-induced mutagenesis library, we identified a mouse line whose Rab18 was knocked out. This Rab18(-/-) mouse exhibited stomping gait, smaller testis and eyes, mimicking several features of WARBM. Rab18(-/-) mice were obviously less sensitive to pain and touch than WT mice. Histological examinations on Rab18(-/-) mice revealed progressive axonal degeneration in the optic nerves, dorsal column of the spinal cord and sensory roots of the spinal nerves while the motor roots were spared. All the behavioral and pathological changes that resulted from abnormalities in the sensory axons were prevented by introducing an extra copy of Rab18 transgene in Rab18(-/-) mice. Our results reveal that sensory axonal degeneration is the primary cause of stomping gait and progressive weakness of the hind limbs in Rab18(-/-) mice, and optic nerve degeneration should be the major pathology of progressive optic atrophy in children with WARBM. Our results indicate that the sensory nervous system is more vulnerable to Rab18 deficiency and WARBM is not only a neurodevelopmental but also neurodegenerative disease.


Assuntos
Anormalidades Múltiplas , Catarata/congênito , Córnea/anormalidades , Etilnitrosoureia/farmacologia , Hipogonadismo , Deficiência Intelectual , Microcefalia , Mutagênese/efeitos dos fármacos , Degeneração Neural/etiologia , Atrofia Óptica , Deleção de Sequência/genética , Proteínas rab de Ligação ao GTP/deficiência , Anormalidades Múltiplas/induzido quimicamente , Anormalidades Múltiplas/genética , Fatores Etários , Animais , Axônios/patologia , Axônios/ultraestrutura , Catarata/induzido quimicamente , Catarata/complicações , Catarata/genética , Modelos Animais de Doenças , Retículo Endoplasmático/patologia , Retículo Endoplasmático/ultraestrutura , Olho/patologia , Hipogonadismo/induzido quimicamente , Hipogonadismo/complicações , Hipogonadismo/genética , Deficiência Intelectual/induzido quimicamente , Deficiência Intelectual/complicações , Deficiência Intelectual/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microcefalia/induzido quimicamente , Microcefalia/complicações , Microcefalia/genética , Microftalmia/etiologia , Microftalmia/genética , Degeneração Neural/patologia , Atrofia Óptica/induzido quimicamente , Atrofia Óptica/complicações , Atrofia Óptica/genética , Doenças do Nervo Óptico/etiologia , Doenças do Nervo Óptico/genética , Desempenho Psicomotor/efeitos dos fármacos , Testículo/patologia , Percepção do Tato/efeitos dos fármacos , Percepção do Tato/genética , Proteínas rab de Ligação ao GTP/genética
18.
Neurology ; 83(10): 903-12, 2014 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-25098539

RESUMO

OBJECTIVE: To describe a novel mutation in TRK-fused gene (TFG) as a new cause of dominant axonal Charcot-Marie-Tooth disease (CMT) identified by exome sequencing and further characterized by in vitro functional studies. METHODS: Exome sequencing and linkage analysis were utilized to investigate a large Taiwanese family with a dominantly inherited adult-onset motor and sensory axonal neuropathy in which mutations in common CMT2-implicated genes had been previously excluded. Functional effects of the mutant gene products were investigated in vitro. RESULTS: Exome sequencing of 2 affected individuals in this family revealed a novel heterozygous mutation, c.806G>T (p.Gly269Val), in TFG that perfectly cosegregates with the CMT2 phenotype in all 27 family members. This mutation occurs at an evolutionarily conserved residue and is absent in the 1,140 ethnically matched control chromosomes. Genome-wide linkage study also supported its disease-causative role. Cell transfection studies showed that the TFG p.Gly269Val mutation increased the propensity of TFG proteins to form aggregates, resulting in sequestration of both mutant and wild-type TFG proteins and might thus deplete functional TFG molecules. The secreted Gaussia luciferase reporter assay demonstrated that inhibition of endogenous TFG compromised the protein secretion pathways, which could only be rescued by expressing wild-type TFG but not the p.Gly269Val altered proteins. TFG mutation was not found in 55 additional unrelated patients with CMT2, suggesting its rarity. CONCLUSION: This study identifies a new cause of dominant CMT2 and highlights the importance of TFG in the protein secretory pathways that are essential for proper functioning of the human peripheral nervous system.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Proteínas/genética , Adulto , Doença de Charcot-Marie-Tooth/patologia , Doença de Charcot-Marie-Tooth/fisiopatologia , Exoma , Feminino , Ligação Genética , Humanos , Imageamento por Ressonância Magnética , Masculino , Redes e Vias Metabólicas/genética , Pessoa de Meia-Idade , Mutação/genética , Linhagem , Fenótipo , Proteínas/metabolismo , Taiwan
19.
Hum Mol Genet ; 23(18): 4770-85, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24833725

RESUMO

CISD2 is a causative gene associated with Wolfram syndrome (WFS). However, it remains a mystery as to how the loss of CISD2 causes metabolic defects in patients with WFS. Investigation on the role played by Cisd2 in specific cell types may help us to resolve these underlying mechanisms. White adipose tissue (WAT) is central to the maintenance of energy metabolism and glucose homeostasis in humans. In this study, adipocyte-specific Cisd2 knockout (KO) mice showed impairment in the development of epididymal WAT (eWAT) in the cell autonomous manner. A lack of Cisd2 caused defects in the biogenesis and function of mitochondria during differentiation of adipocytes in vitro. Insulin-stimulated glucose uptake and secretion of adiponectin by the Cisd2 KO adipocytes were decreased. Moreover, Cisd2 deficiency increased the cytosolic level of Ca(2+) and induced Ca(2+)-calcineurin-dependent signaling that inhibited adipogenesis. Importantly, Cisd2 was found to interact with Gimap5 on the mitochondrial and ER membranes and thereby modulate mitochondrial Ca(2+) uptake associated with the maintenance of intracellular Ca(2+) homeostasis in adipocytes. Thus, it would seem that Cisd2 plays an important role in intracellular Ca(2+) homeostasis, which is required for the differentiation and functioning of adipocytes as well as the regulation of glucose homeostasis in mice.


Assuntos
Adipócitos/citologia , Adipócitos/metabolismo , Cálcio/metabolismo , Proteínas de Transporte/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Adiponectina/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Proteínas Relacionadas à Autofagia , Proteínas de Transporte/genética , Diferenciação Celular , Citosol/metabolismo , Proteínas de Ligação ao GTP , Glucose/metabolismo , Células HEK293 , Homeostase , Humanos , Camundongos , Camundongos Knockout , Mitocôndrias/fisiologia , Proteínas do Tecido Nervoso/genética
20.
PLoS One ; 9(2): e85909, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24498266

RESUMO

Levosimendan, a known calcium sensitizer with positive inotropic and vasodilating properties, might also be cardioprotective during ischemia-reperfusion (I/R) insult. Its effects on calcium homeostasis and apoptosis in I/R injury remain unclear. Na(+)/Ca(2+) exchanger (NCX) is a critical mediator of calcium homeostasis in cardiomyocytes, with reverse-mode NCX activity responsible for intracellular calcium overload and apoptosis of cardiomyocytes during I/R. We probed effects and underlying mechanisms of levosimendan on apoptosis and NCX activity in cultured human cardiomyocyte progenitor cells (CPC)-derived cardiomyocytes undergoing anoxia-reoxygenation (A/R), simulating I/R in vivo. Administration of levosimendan decreased apoptosis of CPC-derived cardiomyocytes induced by A/R. The increase in reverse-mode NCX activity after A/R was curtailed by levosimendan, and NCX1 was translocated away from the cell membrane. Concomitantly, endoplasmic reticulum (ER) stress response induced by A/R was attenuated in CPC-derived cardiomycytes treated with NCX-targeted siRNA or levosimendan, with no synergistic effect between treatments. Results indicated levosimendan inhibited reverse-mode NCX activity to protect CPC-derived cardiomyocytes from A/R-induced ER stress and cell death.


Assuntos
Apoptose/efeitos dos fármacos , Hidrazonas/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Piridazinas/farmacologia , Trocador de Sódio e Cálcio/antagonistas & inibidores , Células-Tronco/efeitos dos fármacos , Adolescente , Western Blotting , Cardiotônicos/farmacologia , Hipóxia Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Criança , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Feminino , Citometria de Fluxo , Células HEK293 , Humanos , Masculino , Microscopia Confocal , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Oxigênio/farmacologia , Interferência de RNA , Simendana , Trocador de Sódio e Cálcio/genética , Trocador de Sódio e Cálcio/metabolismo , Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...