Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Acta Neuropathol Commun ; 12(1): 5, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38172953

RESUMO

BACKGROUND: Parthanatos represents a critical molecular aspect of Parkinson's disease, wherein AIMP2 aberrantly activates PARP-1 through direct physical interaction. Although AIMP2 ought to be a therapeutic target for the disease, regrettably, it is deemed undruggable due to its non-enzymatic nature and predominant localization within the tRNA synthetase multi-complex. Instead, AIMP2 possesses an antagonistic splice variant, designated DX2, which counteracts AIMP2-induced apoptosis in the p53 or inflammatory pathway. Consequently, we examined whether DX2 competes with AIMP2 for PARP-1 activation and is therapeutically effective in Parkinson's disease. METHODS: The binding affinity of AIMP2 and DX2 to PARP-1 was contrasted through immunoprecipitation. The efficacy of DX2 in neuronal cell death was assessed under 6-OHDA and H2O2 in vitro conditions. Additionally, endosomal and exosomal activity of synaptic vesicles was gauged in AIMP2 or DX2 overexpressed hippocampal primary neurons utilizing optical live imaging with VAMP-vGlut1 probes. To ascertain the role of DX2 in vivo, rotenone-induced behavioral alterations were compared between wild-type and DX2 transgenic animals. A DX2-encoding self-complementary adeno-associated virus (scAAV) was intracranially injected into 6-OHDA induced in vivo animal models, and their mobility was examined. Subsequently, the isolated brain tissues were analyzed. RESULTS: DX2 translocates into the nucleus upon ROS stress more rapidly than AIMP2. The binding affinity of DX2 to PARP-1 appeared to be more robust compared to that of AIMP2, resulting in the inhibition of PARP-1 induced neuronal cell death. DX2 transgenic animals exhibited neuroprotective behavior in rotenone-induced neuronal damage conditions. Following a single intracranial injection of AAV-DX2, both behavior and mobility were consistently ameliorated in neurodegenerative animal models induced by 6-OHDA. CONCLUSION: AIMP2 and DX2 are proposed to engage in bidirectional regulation of parthanatos. They physically interact with PARP-1. Notably, DX2's cell survival properties manifest exclusively in the context of abnormal AIMP2 accumulation, devoid of any tumorigenic effects. This suggests that DX2 could represent a distinctive therapeutic target for addressing Parkinson's disease in patients.


Assuntos
Doença de Parkinson , Parthanatos , Animais , Humanos , Inibidores de Poli(ADP-Ribose) Polimerases , Proteínas Nucleares/metabolismo , Peróxido de Hidrogênio , Oxidopamina , Doença de Parkinson/genética , Doença de Parkinson/terapia , Rotenona , Linhagem Celular Tumoral
2.
Cells ; 11(5)2022 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-35269482

RESUMO

Evidence suggests that crosstalk occurs between microglial leucine-rich repeat kinase 2 (LRRK2)-a regulator of neuroinflammation-and neuron-released α-synuclein (αSyn)-a promoter of microglial activation and neuroinflammatory responses-in neuroinflammation-mediated Parkinson's disease (PD) progression. Therefore, we examined whether LRRK2 inhibition reduces the responses of microglia to neuroinflammation caused by neuron-released αSyn. We examined the neuroinflammatory responses provoked by Toll-like receptor 2 (TLR2)-positive αSyn of neuronal cells using an LRRK2 inhibitor in the mouse glioma cells, rat primary microglia, and human microglia cell line; and the effects of LRRK2 inhibitor in the co-culture of ectopic αSyn-expressing human neuroblastoma cells and human microglia cells and in mouse models by injecting αSyn. We analyzed the association between LRRK2 activity and αSyn oligomer and TLR2 levels in the substantia nigra tissues of human patients with idiopathic PD (iPD). The TLR2-specific αSyn elevated LRRK2 activity and neuroinflammation, and the LRRK2 inhibitor ameliorated neuroinflammatory responses in various microglia cells, alleviated neuronal degeneration along with neuroinflammation in the co-culture, and blocked the further progression of locomotor failure and dopaminergic neuronal degeneration caused by TLR2-specific αSyn in mice. Furthermore, LRRK2 phosphorylation was increased in patients with iPD showing αSyn-specific high TLR2 level. These results suggest the application of LRRK2 inhibitors as a novel therapeutic approach against αSyn-mediated PD progression.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Animais , Dopamina , Humanos , Inflamação/tratamento farmacológico , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Camundongos , Doenças Neuroinflamatórias , Doença de Parkinson/tratamento farmacológico , Ratos , Receptor 2 Toll-Like
3.
Anim Cells Syst (Seoul) ; 23(2): 106-111, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30949397

RESUMO

Leucine rich-repeat kinase 2 (LRRK2) is involved in the pathogenesis of Parkinson's disease (PD). LRRK2 has kinase and GTPase activities, and mediates several cell functions, including vesicle trafficking, apoptosis, autophagy, mitochondrial dynamics, and neuroinflammation. G2019S (GS) is the most prevalent mutation of LRRK2. The mutation increases kinase activity, suggesting that this activity is crucial for PD pathogenesis. The activation and inhibition of LRRK2 kinase increases and reduces the levels of proinflammatory cytokines, respectively suggesting that the role of LRRK2 in neuroinflammation is critical for the pathology of PD. Previously, we demonstrated that microglial activation by lipopolysaccharide (LPS) increases mitochondrial fission via the activation of LRRK2 kinase, while LRRK2 kinase inhibition diminishes the fission morphology and release of tumor necrosis factor-alpha (TNFα) in BV2 or rat primary microglia and the brains of GS transgenic mice. In this study, the ectopic expression of GS LRRK2 in BV2 cells significantly elevated the expression of Drp1 along the fragmented mitochondria and decreased mitochondria size compared with controls. GS LRRK2-transfected BV2 cells displayed significantly increased TNFα release and neuronal death. Inhibition of LRRK2 kinase alleviated these features. TNFα levels in brains of GS mice were significantly increased compared to those in their littermates. These data further support our previous findings concerning LPS-induced neuroinflammation and mitochondrial fission in microglia via LRRK2 kinase activation.

4.
Cell Cycle ; 18(4): 467-475, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30712480

RESUMO

Parkinson's disease (PD) is characterized by the degeneration of dopaminergic neurons in the substantia nigra and the presence of Lewy bodies (LB) in neurons. α-Synuclein (αSyn) is a major component of LB and promote the PD pathogenesis via its accumulation by the impaired proteasomal or autophagic clearance. Numerous studies have revealed that the reduction of proteasome activity and autophagy is accelerated by cellular senescence. Leucine-rich repeat kinase 2 (LRRK2) contributes to PD progression and its most prevalent mutation, G2019S LRRK2, increases its activity. Our previous report has shown that the G2019S LRRK2 mutant promoted p53-induced p21 expression and neuronal cytotoxicity. The p53-p21 pathway plays a role in cellular senescence. We hypothesized that the loss of dopaminergic neurons by the stimulated p53-p21 pathway via the G2019S LRRK2 mutation might be associated with cellular senescence, thereby promoting the accumulation of αSyn. We confirmed that the ectopic expression of the phosphomimetic p53 mutant, p21, or G2019 in differentiated SH-SY5Y cells increased the following: 1) the expression of ß-galactosidase, a marker of cellular senescence, and the activity of senescence-associated ß-galactosidase, 2) endogenous αSyn protein level, but not its mRNA level, and 3) αSyn fibril accumulation in dSH-SY5Y via low proteasome and cathepsin D activities. Elevated oligomeric αSyn and the increase in ß-galactosidase with induced p21 were observed in brain lysates of G2019S transgenic mice. Our results suggest that cellular senescence is promoted via the p53-p21 pathway due to the G2019S LRRK2 mutation. Eventually, decreased protein degradation by G2019S-mediated senescence could accelerate αSyn aggregate formation.


Assuntos
Senescência Celular , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima , alfa-Sinucleína/metabolismo , Animais , Encéfalo/metabolismo , Catepsina D/metabolismo , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/genética , Neurônios Dopaminérgicos/metabolismo , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Camundongos , Camundongos Transgênicos , Mutação , Neuroblastoma/patologia , Doença de Parkinson/metabolismo , Fosforilação , Plasmídeos/genética , Transfecção , Proteína Supressora de Tumor p53/genética , beta-Galactosidase/metabolismo
5.
Hum Mol Genet ; 26(1): 1-18, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27798112

RESUMO

Parkinson's disease (PD) is characterized by slow, progressive degeneration of dopaminergic neurons in the substantia nigra. The cause of neuronal death in PD is largely unknown, but several genetic loci, including leucine-rich repeat kinase 2 (LRRK2), have been identified. LRRK2 has guanosine triphosphatase (GTPase) and kinase activities, and mutations in LRRK2 are the major cause of autosomal-dominant familial PD. Histone deacetylases (HDACs) remove acetyl groups from lysine residues on histone tails, promoting transcriptional repression via condensation of chromatin. Here, we demonstrate that LRRK2 binds to and directly phosphorylates HDAC3 at Ser-424, thereby stimulating HDAC activity. Specifically, LRRK2 promoted the deacetylation of Lys-5 and Lys-12 on histone H4, causing repression of gene transcription. Moreover, LRRK2 stimulated nuclear translocation of HDAC3 via the phoshorylation of karyopherin subunit α2 and α6. HDAC3 phosphorylation and its nuclear translocation were increased in response to 6-hydroxydopamine (6-OHDA) treatment. LRRK2 also inhibited myocyte-specific enhancer factor 2D activity, which is required for neuronal survival. LRRK2 ultimately promoted 6-OHDA-induced cell death via positive modulation of HDAC3. These findings suggest that LRRK2 affects epigenetic histone modification and neuronal survival by facilitating HDAC3 activity and regulating its localization.


Assuntos
Encéfalo/patologia , Neurônios Dopaminérgicos/patologia , Histona Desacetilases/metabolismo , Histonas/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Neuritos/patologia , Neuroblastoma/patologia , Acetilação , Animais , Encéfalo/metabolismo , Sobrevivência Celular , Células Cultivadas , Neurônios Dopaminérgicos/metabolismo , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Células HEK293 , Histona Desacetilases/genética , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Neuritos/metabolismo , Neuroblastoma/metabolismo , Fosforilação , Ratos
6.
Biochem Biophys Res Commun ; 482(4): 1088-1094, 2017 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-27914807

RESUMO

Leucine-rich repeat kinase (LRRK2), a major causal gene of Parkinson's disease (PD), functions as a kinase. The most prevalent mutation of LRRK2 is G2019S. It exhibits increased kinase activity compared to the wildtype LRRK2. Previous studies have shown that LRRK2 can phosphorylate p53 at T304 and T377 of threonine-X-arginine (TXR) motif in neurons. Reduction of LRRK2 expression or inhibition of LRRK2 kinase activity has been shown to be able to alleviate LPS-induced neuroinflammation in microglia cells. In this study, we found that LRRK2 could also phosphorylate p53 in microglia model BV2 cells. Transfection of BV2 with phosphomimetic p53 T304/377D significantly increased the secretion of pro-inflammatory cytokine TNFα compared to BV2 transfected with p53 wild type after LPS treatment. In addition, conditioned media from these transfected cells increased the death of dopaminergic neuronal SN4741 cells. Moreover, such neurotoxic effect was rescued by co-treatment with the conditioned media and etanercept, a TNFα blocking antibody. Furthermore, TNFα secretion was significantly increased in primary microglia derived from G2019S transgenic mice treated with LPS compared to that in cells derived from their littermates. These results suggest that LRRK2 kinase activity in microglia can contribute to neuroinflammation in PD via phosphorylating p53 at T304 and T377 site.


Assuntos
Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Microglia/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Sobrevivência Celular , Meios de Cultivo Condicionados/química , Modelos Animais de Doenças , Etanercepte/química , Inflamação , Lipopolissacarídeos/química , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Doença de Parkinson/metabolismo , Fosforilação
7.
Mol Cell Neurosci ; 75: 71-80, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27364102

RESUMO

Autosomal-dominant mutations in the gene encoding leucine-rich repeat kinase 2 (LRRK2) account for the most common monogenic form of Parkinson's disease (PD). A link between autophagy dysregulation and LRRK2 has consistently been reported, but it remains poorly defined which step is targeted by LRRK2. Here, we sought to examine the effect of LRRK2 on the sequestration and degradation of aggregated protein complexes for autophagic clearance. Because two major intracellular protein degradation systems, the ubiquitin proteasome system and the autophagy, are functionally coupled, proteasome inhibition is suggested to activate autophagy. So, we induced protein quality control-associated autophagy using the proteasome inhibitor MG132 in differentiated SH-SY5Y cells and mice expressing G2019S mutant LRRK2 to uncover how the autophagy pathway is affected by LRRK2. We found that LRRK2 disrupted aggresome formation for autophagic clearance of accumulated protein aggregates. Specifically, we observed the following in differentiated SH-SY5Y cells with overexpressed wild-type and G2019S LRRK2: 1) large, clear, perinuclear aggresomes were not detected under MG132, instead, much smaller aggregates were broadly distributed in the cytosol; 2) enhanced accumulation of LC3-II and p62/ubiquitin-positive protein inclusions were noted; and 3) protein aggregates were not cleared even after a recovery period, which exacerbated the MG132-induced cytotoxicity. Notably, higher protein accumulation was detected in the brains of G2019S transgenic mice than in the brains of littermate control mice under proteasome inhibition. Our present findings provide insight into the precise mechanisms that underlie autophagy dysregulation in the brains of patients with PD with LRRK2 mutations.


Assuntos
Autofagia , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Animais , Encéfalo/metabolismo , Linhagem Celular Tumoral , Células HEK293 , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Leupeptinas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Agregados Proteicos , Proteína Sequestossoma-1/metabolismo , Ubiquitina/metabolismo
8.
Mol Biol Cell ; 27(2): 334-48, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26582392

RESUMO

The small GTPase Rab5 regulates the early endocytic pathway of transferrin (Tfn), and Rab5 deactivation is required for Tfn recycling. Rab5 deactivation is achieved by RabGAP5, a GTPase-activating protein, on the endosomes. Here we report that recruitment of RabGAP5 is insufficient to deactivate Rab5 and that developmentally regulated GTP-binding protein 2 (DRG2) is required for Rab5 deactivation and Tfn recycling. DRG2 was associated with phosphatidylinositol 3-phosphate-containing endosomes. It colocalized and interacted with EEA1 and Rab5 on endosomes in a phosphatidylinositol 3-kinase-dependent manner. DRG2 depletion did not affect Tfn uptake and recruitment of RabGAP5 and Rac1 to Rab5 endosomes. However, it resulted in impairment of interaction between Rab5 and RabGAP5, Rab5 deactivation on endosomes, and Tfn recycling. Ectopic expression of shRNA-resistant DRG2 rescued Tfn recycling in DRG2-depleted cells. Our results demonstrate that DRG2 is an endosomal protein and a key regulator of Rab5 deactivation and Tfn recycling.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Transferrina/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Sequência de Aminoácidos , Animais , Endocitose/fisiologia , Endossomos/metabolismo , Feminino , Proteínas de Ligação ao GTP/genética , Proteínas Ativadoras de GTPase/metabolismo , Células HeLa , Humanos , Células MCF-7 , Masculino , Fusão de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Estrutura Terciária de Proteína , Proteínas de Transporte Vesicular/metabolismo
9.
Mol Brain ; 8: 54, 2015 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-26384650

RESUMO

BACKGROUND: Leucine-rich repeat kinase 2 (LRRK2) is a gene in which a mutation causes Parkinson's disease (PD), and p53 is a prototype tumor suppressor. In addition, activation of p53 in patient with PD has been reported by several studies. Because phosphorylation of p53 is critical for regulating its activity and LRRK2 is a kinase, we tested whether p53 is phosphorylated by LRRK2. RESULTS: LRRK2 phosphorylates threonine (Thr) at TXR sites in an in vitro kinase assay, and the T304 and T377 were identified as putative phosphorylated residues. An increase of phospho-Thr in the p53 TXR motif was confirmed in the cells overexpressing G2019S, and human induced pluripotent stem (iPS) cells of a G2019S carrier. Interactions between LRRK2 and p53 were confirmed by co-immunoprecipitation of lysates of differentiated SH-SY5Y cells. LRRK2 mediated p53 phosphorylation translocalizes p53 predominantly to nucleus and increases p21(WAF1/CIP1) expression in SH-SY5Y cells based on reverse transcription-polymerase chain reaction and Western blot assay results. The luciferase assay using the p21(WAF1/CIP1) promoter-reporter also confirmed that LRRK2 kinase activity increases p21 expression. Exogenous expression of G2019S and the phosphomimetic p53 T304/377D mutants increased expression of p21(WAF1/CIP1) and cleaved PARP, and cytotoxicity in the same cells. We also observed increase of p21 expression in rat primary neuron cells after transient expression of p53 T304/377D mutants and the mid-brain lysates of the G2019S transgenic mice. CONCLUSION: p53 is a LRRK2 kinase substrate. Phosphorylation of p53 by LRRK2 induces p21(WAF1/CIP1) expression and apoptosis in differentiated SH-SY5Y cells and rat primary neurons.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Camundongos , Neurônios/metabolismo , Fosforilação , Fosfotreonina/metabolismo , Ligação Proteica , Transporte Proteico , Ratos , Transcrição Gênica
10.
Neuroreport ; 26(6): 380-6, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25807177

RESUMO

Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor relevant to the development of many mammalian organs including the brain. However, the molecular mechanisms by which signaling events mediate neuronal differentiation have not been fully elucidated. In the present study, we show for the first time that the orphan nuclear receptor estrogen-related receptor γ (ERRγ) is upregulated by HIF-1α and plays essential roles in HIF-1α-induced upregulation of dopaminergic marker molecules such as tyrosine hydroxylase and dopamine transporter. We found that deferoxamine upregulated HIF-1α and enhanced the dopaminergic phenotype and neurite outgrowth of SH-SY5Y cells. Deferoxamine activated transcription and protein expression of ERRγ, and deferoxamine-induced upregulation of tyrosine hydroxylase and dopamine transporter was attenuated by using the ERRγ inverse agonist or silencing ERRγ. Altogether, these results suggest that HIF-1α can positively regulate the dopaminergic phenotype through ERRγ. This study could provide new perspectives for understanding the mechanisms underlying the promotion of dopaminergic neuronal differentiation by hypoxia.


Assuntos
Diferenciação Celular , Neurônios Dopaminérgicos/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Receptores de Estrogênio/metabolismo , Linhagem Celular Tumoral , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Humanos , Transdução de Sinais , Tirosina 3-Mono-Oxigenase/metabolismo , Regulação para Cima
11.
J Biochem ; 157(6): 485-95, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25605758

RESUMO

Leucine-rich repeat kinase 2 (LRRK2) has been identified as a causative gene for Parkinson's disease (PD). LRRK2 contains a kinase and a GTPase domain, both of which provide critical intracellular signal-transduction functions. We showed previously that Rab5b, a small GTPase protein that regulates the motility and fusion of early endosomes, interacts with LRRK2 and co-regulates synaptic vesicle endocytosis. Using recombinant proteins, we show here that LRRK2 phosphorylates Rab5b at its Thr6 residue in in vitro kinase assays with mass spectrophotometry analysis. Phosphorylation of Rab5b by LRRK2 on the threonine residue was confirmed by western analysis using cells stably expressing LRRK2 G2019S. The phosphomimetic T6D mutant exhibited stronger GTPase activity than that of the wild-type Rab5b. In addition, phosphorylation of Rab5b by LRRK2 also exhibited GTPase activity stronger than that of the unphosphorylated Rab5b protein. Two assays testing Rab5's activity, neurite outgrowth analysis and epidermal growth factor receptor degradation assays, showed that Rab5b T6D exhibited phenotypes that were expected to be observed in the inactive Rab5b, including longer neurite length and less degradation of EGFR. These results suggest that LRRK2 kinase activity functions as a Rab5b GTPase activating protein and thus, negatively regulates Rab5b signalling.


Assuntos
Endossomos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Receptores ErbB/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Células HEK293 , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Mimetismo Molecular , Fosforilação , Especificidade por Substrato
12.
Biomed Res Int ; 2014: 704678, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25478574

RESUMO

Parkinson's disease (PD) is a difficult disease to diagnose although it is the second most common neurodegenerative disease. Recent studies show that exosome isolated from urine contains LRRK2 or DJ-1, proteins whose mutations cause PD. To investigate a potential use for urine exosomes as a tool for PD diagnosis, we compared levels of LRRK2, α-synuclein, and DJ-1 in urine exosomes isolated from Korean PD patients and non-PD controls. LRRK2 and DJ-1, but not α-synuclein, were detected in the urine exosome samples, as reported previously. We initially could not detect any significant difference in these protein levels between the patient and the control groups. However, when age, disease duration, L-dopa daily dose, and gender were considered as analytical parameters, LRRK2 and DJ-1 protein levels showed clear gender-dependent differences. In addition, DJ-1 level was significantly higher (1.7-fold) in male patients with PD than that in male non-PD controls and increased in an age-dependent manner in male patients with PD. Our observation might provide a clue to lead to a novel biomarker for PD diagnosis, at least in males.


Assuntos
Exossomos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/urina , Proteínas Oncogênicas/urina , Doença de Parkinson/urina , Idoso , Exossomos/patologia , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Masculino , Mutação , Proteínas Oncogênicas/biossíntese , Doença de Parkinson/diagnóstico , Doença de Parkinson/patologia , Proteína Desglicase DJ-1 , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/urina , República da Coreia , Caracteres Sexuais , alfa-Sinucleína/biossíntese , alfa-Sinucleína/urina
13.
Int J Neuropsychopharmacol ; 17(11): 1831-46, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24901414

RESUMO

Recent studies suggest that ketamine produces antidepressant actions via stimulation of mammalian target of rapamycin (mTOR), leading to increased levels of synaptic proteins in the prefrontal cortex. Thus, mTOR activation may be related to antidepressant action. However, the mTOR signalling underlying antidepressant drug action has not been well investigated. The aim of the present study was to determine whether alterations in mTOR signalling were observed following treatment with antidepressant drugs, using ketamine as a positive control. Using Western blotting, we measured changes in the mTOR-mediated proteins and synaptic proteins in rat hippocampal cultures. Dendritic outgrowth was determined by neurite assay. Our findings demonstrated that escitalopram, paroxetine and tranylcypromine significantly increased levels of phospho-mTOR and its down-stream regulators (phospho-4E-BP-1 and phospho-p70S6K); fluoxetine, sertraline and imipramine had no effect. All drugs tested increased up-stream regulators (phospho-Akt and phospho-ERK) levels. Increased phospho-mTOR induced by escitalopram, paroxetine or tranylcypromine was significantly blocked in the presence of specific PI3K, MEK or mTOR inhibitors, respectively. All drugs tested also increased hippocampal dendritic outgrowth and synaptic proteins levels. The mTOR inhibitor, rapamycin, significantly blocked these effects on escitalopram, paroxetine and tranylcypromine whereas fluoxetine, sertraline and imipramine effects were not affected. The effects of escitalopram, paroxetine and tranylcypromine paralleled those of ketamine. This study presents novel in vitro evidence indicating that some antidepressant drugs promote dendritic outgrowth and increase synaptic protein levels through mTOR signalling; however, other antidepressant drugs seem to act via a different pathway. mTOR signalling may be a promising target for the development of new antidepressant drugs.


Assuntos
Antidepressivos/farmacologia , Hipocampo/citologia , Neurônios/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Animais , Proteínas de Transporte/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Embrião de Mamíferos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Neuritos/efeitos dos fármacos , Neurônios/citologia , Proteína Oncogênica v-akt/metabolismo , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo
14.
PLoS One ; 7(4): e34693, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22496842

RESUMO

LRRK2, a Parkinson's disease associated gene, is highly expressed in microglia in addition to neurons; however, its function in microglia has not been evaluated. Using Lrrk2 knockdown (Lrrk2-KD) murine microglia prepared by lentiviral-mediated transfer of Lrrk2-specific small inhibitory hairpin RNA (shRNA), we found that Lrrk2 deficiency attenuated lipopolysaccharide (LPS)-induced mRNA and/or protein expression of inducible nitric oxide synthase, TNF-α, IL-1ß and IL-6. LPS-induced phosphorylation of p38 mitogen-activated protein kinase and stimulation of NF-κB-responsive luciferase reporter activity was also decreased in Lrrk2-KD cells. Interestingly, the decrease in NF-κB transcriptional activity measured by luciferase assays appeared to reflect increased binding of the inhibitory NF-κB homodimer, p50/p50, to DNA. In LPS-responsive HEK293T cells, overexpression of the human LRRK2 pathologic, kinase-active mutant G2019S increased basal and LPS-induced levels of phosphorylated p38 and JNK, whereas wild-type and other pathologic (R1441C and G2385R) or artificial kinase-dead (D1994A) LRRK2 mutants either enhanced or did not change basal and LPS-induced p38 and JNK phosphorylation levels. However, wild-type LRRK2 and all LRRK2 mutant variants equally enhanced NF-κB transcriptional activity. Taken together, these results suggest that LRRK2 is a positive regulator of inflammation in murine microglia, and LRRK2 mutations may alter the microenvironment of the brain to favor neuroinflammation.


Assuntos
Encéfalo/metabolismo , Encefalite/metabolismo , Microglia/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Linhagem Celular , Técnicas de Silenciamento de Genes , Interleucina-1beta/biossíntese , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Lipopolissacarídeos/farmacologia , MAP Quinase Quinase 4/metabolismo , Camundongos , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/biossíntese , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Fator de Necrose Tumoral alfa/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Neuropharmacology ; 61(4): 761-9, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21663752

RESUMO

Dopamine D(2) receptors (D(2)R) are the primary target of antipsychotic drugs and have been shown to regulate Akt/glycogen synthase kinase-3ß (GSK-3ß) signaling through scaffolding protein ß-arrestin 2. Amisulpride, an atypical antipsychotic drug, and haloperidol, a typical antipsychotic drug, are both potent D(2)R antagonists, but their therapeutic effects differ. In the present study, we compared the effects of amisulpride and haloperidol on the ß-arrestin 2-mediated Akt/GSK-3ß pathway in SH-SY5Y cells. To determine whether these drugs affected neuronal morphology in SH-SY5Y cells, we investigated the effects of amisulpride and haloperidol on neurite outgrowth using immunostaining. We examined the effects of these drugs on Akt and GSK-3ß and its well-known downstream regulators, cAMP response element-binding protein (CREB), brain-derived neurotrophic factor (BDNF), and Bcl-2 levels using Western blot analysis. Amisulpride, but not haloperidol, was found to enhance neurite outgrowth. Small interfering RNA (siRNA) for ß-arrestin 2 knockdown blocked the increase in amisulpride-induced neurite outgrowth. Furthermore, amisulpride increased the levels of Akt and GSK-3ß phosphorylation, while haloperidol had no effect. The elevation of Akt phosphorylation induced by amisulpride was reduced by ß-arrestin 2 siRNA. Moreover, amisulpride effectively increased the levels of phospho-CREB, BDNF, and Bcl-2. However, haloperidol had no effect on the levels of these proteins. Additionally, wortmannin, a phosphatidylinositol 3-kinase (PI3 K) inhibitor, blocked the stimulatory effect of amisulpride on phosphorylated Akt. Together, these results suggest that regulation of the ß-arrestin 2-dependent pathway via blockade of the D(2)R in SH-SY5Y cells is one mechanism underlying the neuroprotective effect of amisulpride, but not haloperidol.


Assuntos
Antagonistas dos Receptores de Dopamina D2 , Haloperidol/farmacologia , Receptores de Dopamina D2/fisiologia , Transdução de Sinais/efeitos dos fármacos , Sulpirida/análogos & derivados , Amissulprida , Arrestinas/fisiologia , Linhagem Celular Tumoral , Humanos , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Transdução de Sinais/fisiologia , Sulpirida/farmacologia , beta-Arrestina 2 , beta-Arrestinas
16.
Neurosignals ; 19(2): 86-96, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21474915

RESUMO

Genetic mutation of α-synuclein (α-SYN) is clearly verified as the causal factor of human and mouse Parkinson's disease. However, biological function of α-SYN has not been clearly demonstrated until now. In this investigation, we reveal that α-SYN is a co-regulator of growth factor-induced AKT activation. Elimination of SYN reduces the IGF-1-mediated AKT activation. Similarly, mutant SYN suppresses the IGF-1-induced AKT activation. Wild-type SYN can interact with AKT and enhance the solubility and plasma localization of AKT in response to IGF-1, whereas mutant α-SYNs do not interact with AKT. In addition, elevated expression of SYN blocks the AKT activation. We also find that si-RNA against α-SYN abolished the protective effect of IGF-1 against DNA damage-induced apoptosis. Our result strongly indicates that Parkinson's disease, induced by α-SYN mutation, is evoked by deregulation of the AKT-signaling cascade.


Assuntos
Fator de Crescimento Insulin-Like I/metabolismo , Doença de Parkinson/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , alfa-Sinucleína/metabolismo , Linhagem Celular , Humanos , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Doença de Parkinson/enzimologia , Doença de Parkinson/fisiopatologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , alfa-Sinucleína/genética
17.
Exp Cell Res ; 316(4): 649-56, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19769964

RESUMO

LRRK2 is an autosomal dominant gene whose mutations cause familial Parkinson's disease (PD). The LRRK2 protein contains a functional kinase and a GTPase domain. PD phenotypes caused by LRRK2 mutations are similar to those of idiopathic PD, implying that LRRK2 is an important participant in PD pathogenesis. Of LRRK2's PD-specific mutations, the G2019S is the most frequently observed one. Its over-expression is known to increase kinase activity and neurotoxicity compared to wild type (WT) LRRK2. Here, using a simple colorimetric cell viability assay, we analyzed LRRK2's neurotoxicity in dopaminergic SN4741 cells following treatment with hydrogen peroxide. When WT, G2019S, or empty vector was expressed in SN4741 cells, cell death was modestly and significantly increased in the order of G2019S>WT>vector. When these transfected cells were treated with hydrogen peroxide to mimic oxidative stress, cellular neurotoxicity was enhanced in the same order (i.e. G2019S>WT>vector). Moreover, incubation of SN4741 cells with conditioned medium from cells expressing G2019S and subjected to hydrogen peroxide treatment exhibited 10-15% more cell death than conditioned medium from cells transfected with vector or WT, suggesting that G2019S-expressing cells secrete a factor(s) affecting viability of neighboring cells. The kinase domain was mapped to be responsible for oxidative stress-induced neurotoxicity. In addition, over-expression of WT and G2019S LRRK2 lead to a weak, but significant, increase in intracellular reactive oxygen species (ROS) in the order of G2019S>WT as measured by DCFH-DA assay in both the presence and absence of H(2)O(2) treatment. Furthermore, in G2019S-expressing cells, co-expression of the anti-oxidant protein DJ-1 or ERK inhibitor treatment restored survival rate to a level similar to that of cells transfected with control vector under H(2)O(2) treatment. Taken together, our data suggest that the LRRK2 kinase domain increases the generation of ROS and causes enhanced neurotoxicity under H(2)O(2) treatment, which can be at least partially rescued by DJ-1 or the ERK inhibitor.


Assuntos
Neurônios/patologia , Estresse Oxidativo/genética , Fosfotransferases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Bioensaio/métodos , Western Blotting , Sobrevivência Celular , Células Cultivadas , Clonagem Molecular , Inibidores Enzimáticos/farmacologia , Vetores Genéticos , Humanos , Peróxido de Hidrogênio/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Mutação/genética , Proteínas Oncogênicas/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Plasmídeos , Proteína Desglicase DJ-1 , Proteínas Serina-Treonina Quinases/genética , Espécies Reativas de Oxigênio/análise , Transdução de Sinais
18.
Mol Cells ; 26(5): 454-8, 2008 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-18719354

RESUMO

Sex steroid hormone receptors play a central role in modulating telomerase activity, especially in cancer cells. However, information on the regulation of steroid hormone receptors and their distinct functions on telomerase activity within the mesenchymal stem cell are largely unavailable due to low telomerase activity in the cell. In this study, the effects of estrogen (E2) treatment and function of estrogen receptor alpha (ERalpha) and estrogen receptor beta (ERbeta) on telomerase activity were investigated in human mesenchymal stem cells (hMSCs). Telomerase activity and mRNA expression of the catalytic subunit of telomerase (hTERT) were upregulated by treatment of the cells with E2. The protein concentration of ERalpha was also increased by E2 treatment, and enhancement of ERalpha accumulation in the nucleus was clearly detected with immunocytochemistry. When ERalpha expression was reduced by siRNA transfection into hMSCs, the effect of E2 on the induction of hTERT expression and telomerase activity was diminished. In contrast, the transient overexpression of ERalpha increased the effect of E2 on the expression of hTERT mRNA. These findings indicate that the activation of hTERT expression and telomerase activity by E2 in hMSCs depends on ERalpha, but not on ERbeta.


Assuntos
Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/enzimologia , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Receptor beta de Estrogênio/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/metabolismo , Telomerase
19.
J Neurochem ; 98(2): 386-94, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16805833

RESUMO

Retinoic acid (RA), a derivative of vitamin A, critically controls brain patterning and neurogenesis during embryogenesis, and is known to regulate morphological differentiation of catecholaminergic neuronal cells. In this study, we investigated whether the retinoic acid receptor (RAR), a transcription factor specifically activated by all-trans-RA, could directly regulate transcription of tyrosine hydroxylase (TH), the first and rate-limiting step in the catecholamine biosynthesis pathway. First, treating TH-expressing human neuroblastoma SK-N-BE(2)C cells with all-trans RA resulted in an approximately 1.7-fold increase in endogenous TH mRNA expression, as determined by real-time PCR analysis. Second, when SK-N-BE(2)C cells were transiently co-transfected with the TH promoter-luciferase reporter construct, reporter gene expression was prominently activated by RAR in a ligand-dependent manner. Third, we identified a putative RAR responsive cis-regulatory element at - 1500 to - 1487 bp in the TH upstream promoter region by deletional and site-directed mutational analysis. Finally, we demonstrated that this putative motif directly interacts with RAR protein in a sequence-specific manner by means of an electrophoretic mobility shift assay. Taken together, our results indicate that the TH gene may be a direct downstream target of the RA signaling pathway and that RAR is able to activate TH transcription through interaction with an upstream sequence motif residing at - 1500 to - 1487 bp.


Assuntos
Regulação Enzimológica da Expressão Gênica/fisiologia , Receptores do Ácido Retinoico/fisiologia , Tirosina 3-Mono-Oxigenase/biossíntese , Tirosina 3-Mono-Oxigenase/genética , Animais , Catecolaminas/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Ensaio de Desvio de Mobilidade Eletroforética , Regulação Enzimológica da Expressão Gênica/genética , Luciferases/metabolismo , Plasmídeos/genética , Regiões Promotoras Genéticas/genética , RNA Mensageiro/biossíntese , Ratos , Receptores do Ácido Retinoico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Ativação Transcricional/genética , Ativação Transcricional/fisiologia , Transfecção
20.
Neurosci Lett ; 396(1): 57-61, 2006 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-16356647

RESUMO

Dopamine and the sex hormone testosterone are important factors regulating male sexual behavior. To investigate the possibility that these two factors are functionally interrelated, we investigated the potential role of the androgen receptor (AR) on transcriptional activity of the tyrosine hydroxylase (TH) gene that encodes the rate-limiting enzyme of the dopamine biosynthesis pathway. In this study, using transient co-transfection assays in TH-positive SK-N-BE(2)C and MN9D cells, we show that AR prominently transactivates TH promoter function in a ligand-dependent manner. Deletional and site-directed mutational analyses have mapped a putative androgen response element (ARE) in a region from -1562 to -1328 base pairs in the upstream TH promoter. We also found that DJ-1, one of recently identified genes whose mutations cause Parkinson's disease, down-regulated AR-dependent TH activation by approximately 50% in SK-N-BE(2)C cells. Based on these data, we propose that AR activates TH gene expression and that DJ-1 may modulate AR activity as a transcriptional co-repressor.


Assuntos
Encéfalo/metabolismo , Dopamina/biossíntese , Regulação Enzimológica da Expressão Gênica/fisiologia , Receptores Androgênicos/metabolismo , Comportamento Sexual Animal/fisiologia , Ativação Transcricional/fisiologia , Tirosina 3-Mono-Oxigenase/genética , Animais , Química Encefálica/genética , Linhagem Celular , Linhagem Celular Tumoral , Regulação para Baixo/genética , Humanos , Masculino , Camundongos , Mutação/genética , Proteínas Oncogênicas/genética , Peroxirredoxinas , Regiões Promotoras Genéticas/genética , Proteína Desglicase DJ-1 , Ratos , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Elementos de Resposta/genética , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA