Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 545
Filtrar
1.
Nature ; 629(8010): 235-243, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38499039

RESUMO

Biogenic monoamines-vital transmitters orchestrating neurological, endocrinal and immunological functions1-5-are stored in secretory vesicles by vesicular monoamine transporters (VMATs) for controlled quantal release6,7. Harnessing proton antiport, VMATs enrich monoamines around 10,000-fold and sequester neurotoxicants to protect neurons8-10. VMATs are targeted by an arsenal of therapeutic drugs and imaging agents to treat and monitor neurodegenerative disorders, hypertension and drug addiction1,8,11-16. However, the structural mechanisms underlying these actions remain unclear. Here we report eight cryo-electron microscopy structures of human VMAT1 in unbound form and in complex with four monoamines (dopamine, noradrenaline, serotonin and histamine), the Parkinsonism-inducing MPP+, the psychostimulant amphetamine and the antihypertensive drug reserpine. Reserpine binding captures a cytoplasmic-open conformation, whereas the other structures show a lumenal-open conformation stabilized by extensive gating interactions. The favoured transition to this lumenal-open state contributes to monoamine accumulation, while protonation facilitates the cytoplasmic-open transition and concurrently prevents monoamine binding to avoid unintended depletion. Monoamines and neurotoxicants share a binding pocket that possesses polar sites for specificity and a wrist-and-fist shape for versatility. Variations in this pocket explain substrate preferences across the SLC18 family. Overall, these structural insights and supporting functional studies elucidate the mechanism of vesicular monoamine transport and provide the basis to develop therapeutics for neurodegenerative diseases and substance abuse.


Assuntos
Monoaminas Biogênicas , Interações Medicamentosas , Proteínas Vesiculares de Transporte de Monoamina , Humanos , 1-Metil-4-fenilpiridínio/química , 1-Metil-4-fenilpiridínio/metabolismo , 1-Metil-4-fenilpiridínio/farmacologia , Anfetamina/química , Anfetamina/farmacologia , Anfetamina/metabolismo , Sítios de Ligação , Monoaminas Biogênicas/química , Monoaminas Biogênicas/metabolismo , Microscopia Crioeletrônica , Dopamina/química , Dopamina/metabolismo , Modelos Moleculares , Norepinefrina/química , Norepinefrina/metabolismo , Ligação Proteica , Prótons , Reserpina/farmacologia , Reserpina/química , Reserpina/metabolismo , Serotonina/química , Serotonina/metabolismo , Especificidade por Substrato , Proteínas Vesiculares de Transporte de Monoamina/química , Proteínas Vesiculares de Transporte de Monoamina/metabolismo , Proteínas Vesiculares de Transporte de Monoamina/ultraestrutura
2.
Immun Inflamm Dis ; 12(3): e1194, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38501544

RESUMO

BACKGROUND: Parkinson's disease (PD) is a common neurodegenerative disorder. Microglia-mediated neuroinflammation has emerged as an involving mechanism at the initiation and development of PD. Activation of adenosine triphosphate (ATP)-sensitive potassium (KATP ) channels can protect dopaminergic neurons from damage. Sodium butyrate (NaB) shows anti-inflammatory and neuroprotective effects in some animal models of brain injury and regulates the KATP channels in islet ß cells. In this study, we aimed to verify the anti-inflammatory effect of NaB on PD and further explored potential molecular mechanisms. METHODS: We established an in vitro PD model in BV2 cells using 1-methyl-4-phenylpyridinium (MPP+ ). The effects of MPP+ and NaB on BV2 cell viability were detected by cell counting kit-8 assays. The morphology of BV2 cells with or without MPP+ treatment was imaged via an optical microscope. The expression of Iba-1 was examined by the immunofluorescence staining. The intracellular ATP content was estimated through the colorimetric method, and Griess assay was conducted to measure the nitric oxide production. The expression levels of pro-inflammatory cytokines and KATP channel subunits were evaluated by reverse transcription-quantitative polymerase chain reaction and western blot analysis. RESULTS: NaB (5 mM) activated the KATP channels through elevating Kir6.1 and Kir6.1 expression in MPP+ -challenged BV2 cells. Both NaB and pinacidil (a KATP opener) suppressed the MPP+ -induced activation of BV2 cells and reduced the production of nitrite and pro-inflammatory cytokines in MPP+ -challenged BV2 cells. CONCLUSION: NaB treatment alleviates the MPP+ -induced inflammatory responses in microglia via activation of KATP channels.


Assuntos
Doença de Parkinson , Animais , Doença de Parkinson/metabolismo , Ácido Butírico/farmacologia , Ácido Butírico/metabolismo , Microglia/metabolismo , 1-Metil-4-fenilpiridínio/metabolismo , 1-Metil-4-fenilpiridínio/farmacologia , Inflamação/metabolismo , Citocinas/metabolismo , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/metabolismo
3.
Exp Gerontol ; 188: 112387, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38431178

RESUMO

OBJECTIVE: Dysregulation of covalently closed circular RNAs (circRNAs) has been associated with neurological disorders, the role of circHIVP2 in Parkinson's disease (PD) and its molecular mechanism is not well understood. METHODS: 127 patients with PD and 85 healthy people were enrolled. RT-qPCR was employed to examine the levels of circHIVEP2. ROC curve to explore the diagnostic. Mpp+ induced the SH-SY5Y to construct an in vitro PD cell model. Cell viability, apoptosis, and secretion levels of inflammatory factors were analyzed by CCK-8, flow cytometry, and ELISA assay. CircHIVEP2 targets miRNA predicted by bioinformatics database and validated by the dual luciferase reporter and RIP assays. RESULTS: CircHIVEP2 was typically lower in PD patients than in controls. CircHIVEP2 has certain specificity and sensitivity to recognize PD patients from healthy individuals. miR-485-3p, a target miRNA of circHIVEP2, was significantly elevated in PD patients. Additionally, MPP+ induction reduced cell viability and promoted apoptosis and inflammatory factor overproduction. However, overexpression of circHIVEP2 significantly inhibited the effects of MPP+, but this inhibition was significantly attenuated by elevated miR-485-3p. CONCLUSION: circHIVEP2 is a potential diagnostic biomarker for PD, and its upregulation mitigated MPP+-induced nerve damage and inflammation and this may be through targeted by the miR-485-3p.


Assuntos
MicroRNAs , Neuroblastoma , Doença de Parkinson , Humanos , Doença de Parkinson/genética , 1-Metil-4-fenilpiridínio/farmacologia , Linhagem Celular Tumoral , MicroRNAs/genética , Apoptose
4.
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
5.
Int J Mol Sci ; 24(13)2023 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-37445592

RESUMO

Parkinson's disease is a neurodegenerative disorder characterized by oxidative stress and immune activation in the nigro-striatal pathway. Simvastatin regulates cholesterol metabolism and protects from atherosclerosis disease. Simvastatin-tween 80 was administered 7 days before sterotaxic intrastriatal administration of MPP+ (1-methyl-4-phenylpyridine) in rats. Fluorescent lipidic product formation, dopamine levels, and circling behavior were considered damage markers. Twenty-four hours and six days after, the animal group lesioned with MPP+ showed significant damage in relation to the control group. Animals pretreated with simvastatin significantly reduced the MPP+-induced damage compared to the MPP+ treated group. As apoptosis promotes neuroinflammation and neuronal degeneration in Parkinson's disease, and since there is not currently a proteomic map of the nigro-striatum of rats and assuming a high homology among the identified proteins in other rat tissues, we based the search for rat protein homologs related to the establishment of inflammation response. We demonstrate that most proteins related to inflammation decreased in the simvastatin-treated rats. Furthermore, differential expression of antioxidant enzymes in striated tissue of rat brains was found in response to simvastatin. These results suggest that simvastatin could prevent striatal MPP+-induced damage and, for the first time, suggest that the molecular mechanisms involved in this have a protective effect.


Assuntos
Doença de Parkinson , Ratos , Animais , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/etiologia , Doença de Parkinson/metabolismo , Sinvastatina/farmacologia , Sinvastatina/uso terapêutico , Sinvastatina/metabolismo , Proteômica , Substância Negra/metabolismo , Dopamina/metabolismo , 1-Metil-4-fenilpiridínio/farmacologia , Corpo Estriado/metabolismo , Modelos Animais de Doenças
6.
Cell Biol Int ; 47(9): 1502-1518, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37208975

RESUMO

The transient receptor potential channel (TRP) channels are expressed in neuronal tissues and involved in neurological diseases such as pain, epilepsy, neuronal apoptosis, and neurodegenerative diseases. Formerly, we have investigated how neuronal differentiation changes TRP channels expression profile and how Parkinson's disease model is related with this expression levels. We have found that transient receptor potential channel melastatin subtype 7 (TRPM7), transient receptor potential channel melastatin subtype 8 and transient receptor potential channel vanilloid subtype 1 (TRPV1) channels have pivotal effects on differentiation and 1-Methyl-4-phenylpyridinium (MPP+ )-induced Parkinson's disease model in SH-SY5Y cells. In this study, we have investigated that downregulation of the TRP channels to evaluate how differentiation status changes to Parkinson's disease pathological hallmarks. We have also performed to other analyses to elucidate these TRP channels' function in MPP+ -induced neurotoxicity related apoptosis, cell viability, caspase 3 and 9 enzyme activities, intracellular reactive oxygen species production, mitochondrial depolarization levels, Ca2+ signaling, Alpha-synuclein and Dopamine levels, mono amino oxidase A and B enzymatic activities, both in differentiated and undifferentiated neuronal cells. Herein we have concluded that especially TRPM7 and TRPV1 channels have distinct role in Parkinson's disease pathology via their activity changings in pathological state, and downregulation of these channels or specific antagonists can be useful for the possible treatment strategy for Parkinson's disease and related markers.


Assuntos
Neuroblastoma , Doença de Parkinson , Canais de Cátion TRPM , Canais de Potencial de Receptor Transitório , Humanos , Canais de Potencial de Receptor Transitório/metabolismo , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo , Regulação para Baixo , Apoptose , 1-Metil-4-fenilpiridínio/farmacologia , Canais de Cátion TRPV/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Membrana/metabolismo
7.
Comb Chem High Throughput Screen ; 26(14): 2476-2486, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37073660

RESUMO

AIM AND OBJECTIVE: Long intergenic non-coding RNA-p21 (lincRNA-p21) plays a critical role in various senescence-associated physiological and pathological conditions. We aimed to explore the senescence-associated effects of lincRNA-p21 in 1-methyl-4-phenylpyridinium (MPP+) treated neuroblastoma SH-SY5Y cell line as a therapeutic target. MATERIALS AND METHODS: The RNA expression levels of lincRNA-p21, p53, p16, and telomere length were examined with reverse transcription-quantitative polymerase chain reaction (RTqPCR). The Telo TAGGG™ Telomerase PCR ELISA PLUS Kit was used to determine telomerase activity. Cellular viability was evaluated with the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and lactate dehydrogenase (LDH) assay. Western blot was performed to analyze ß-catenin protein expression. Besides, oxidative stress was evaluated by Jaggregate- forming delocalized lipophilic cation, 5,5',6,6'-tetrachloro-1,1',3,3'- tetraethylbenzimidazolocarbocyanine++ + iodide (JC­1) stain, fluorescence spectrophotometry, colorimetric assay, and malondialdehyde (MDA) formation. RESULTS: This research demonstrated that MPP+ caused a distinct increase in the expression of LincRNA- p21 in SH-SY5Y cells. MPP+ induced cellular senescence with decreasing cellular proliferation and viability, increasing expression levels of senescence-associated makers such as genes p53 and p16, accompanied by significantly decreasing telomere length and telomerase activity. At the same time, these effects were abolished by silencing lincRNA-p21 with small interfering RNA (siRNA). On the contrary, ß-catenin silencing contributes to reversing anti-senescent effects caused by lincRNA-p21 silencing. Moreover, modifying lincRNA-p21 exerted an anti-senescent influence depending on decreasing oxidant stress. CONCLUSION: Our study showed that in the treatment of MPP+, lincRNA-p21 might serve a role in the SH-SY5Y cell senescence by modulating the Wnt/ß-catenin pathway, as well as increasing oxidant stress. Thus, trying to target lincRNA-p21 may have important therapeutic and practical implications for PD.


Assuntos
Neuroblastoma , RNA Longo não Codificante , Telomerase , Humanos , 1-Metil-4-fenilpiridínio/farmacologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Telomerase/metabolismo , Telomerase/farmacologia , beta Catenina/metabolismo , Proteína Supressora de Tumor p53/genética , Apoptose , Senescência Celular , Oxidantes/farmacologia , Linhagem Celular Tumoral
8.
Mol Biol Rep ; 50(5): 4423-4433, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36977807

RESUMO

BACKGROUND: Growing evidence indicates that cannabinoid type 2 (CB2) receptor activation inhibits neuroinflammation in the pathogenesis of Parkinson's disease (PD). Nonetheless, the precise mechanisms of CB2 receptor-mediated neuroprotection have not been fully elucidated. The differentiation of microglia from the M1 to M2 phenotype plays a vital role in neuroinflammation. METHODS: In the present study, we investigated the effect of CB2 receptor activation on the M1/M2 phenotypic transformation of microglia treated with 1-methyl-4-phenylpyridinium (MPP+). The M1 phenotype microglia markers, including inducible nitric oxide (iNOS), interleukin 6 (IL-6), and CD86, and the M2 phenotype microglia markers, including arginase-1 (Arg-1), IL-10, and CD206, were detected by western blots and flow cytometry. The levels of phosphoinositide-3-kinase (PI3K)/Akt and nuclear factor erythroid 2-related factor 2 (Nrf2) were determined by Western blots. Subsequent addition of Nrf2 inhibitors initially revealed the specific mechanism by which CB2 receptors affect phenotypic changes in microglia. RESULTS: Our results showed that pretreatment with JWH133 significantly inhibited the MPP+-induced up-regulation of M1 phenotype microglia markers. Meanwhile, JWH133 increased the levels of M2 phenotype microglia markers. JWH133-mediated effects were blocked by co-treatment with AM630. Mechanism studies found that MPP+ treatment downregulated PI3K, Akt phosphorylated proteins, and nuclear Nrf2 protein. JWH133 pretreatment promoted PI3K/Akt activation and facilitated nuclear translocation of Nrf2, which was reversed by the PI3K inhibitor. Further studies showed that Nrf2 inhibitors inverted the effect of JWH133 on microglia polarization. CONCLUSION: The results indicate that CB2 receptor activation promotes MPP+-induced microglia transformation from M1 to M2 phenotype through PI3K/Akt/Nrf2 signaling pathway.


Assuntos
Canabinoides , Microglia , Humanos , Microglia/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , 1-Metil-4-fenilpiridínio/farmacologia , 1-Metil-4-fenilpiridínio/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Doenças Neuroinflamatórias , Receptor CB2 de Canabinoide/genética , Transdução de Sinais , Canabinoides/farmacologia , Canabinoides/metabolismo
9.
Mitochondrion ; 69: 95-103, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36758857

RESUMO

Mitochondrial dysfunction is closely linked with the pathophysiology of several neurodegenerative disorders including Parkinson's disease (PD). Despite several therapeutic advancements related to symptomatic modification of PD pathology, strategies targeting mitochondrial dysfunctions remain largely elusive. Recently, transient receptor potential (TRP) channels have been shown to play a pivotal role in the control of mitochondrial and neuronal functioning in PD. In this study, the effect of 2-aminoethoxydiphenyl borate (2-APB), TRP channel blocker was investigated in the context of mitochondrial dysfunctions in 1-methyl-4-phenylpyridinium (MPP+)-treated SH-SY5Y cells and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-administered Sprague Dawley rats. MPP+-treated SH-SY5Y cells exhibited reductions in cell viability, generation of reactive oxygen species (ROS) and loss of mitochondrial membrane potential. Co-treatment with 2-APB led to an increase in cell viability, reduction in intracellular and mitochondrial ROS and improvement in mitochondrial membrane potential compared to MPP+-treated SH-SY5Y cells. In addition, intranigral administration of MPTP led to a significant reduction in motor function in the rats. Fourteen days of 2-APB (3 and 10 mg/kg, i.p.) treatment improved behavioural parameters. MPTP-induced decrease in complex I activity and mitochondrial potential were also blocked by 2-APB in the mitochondria isolated from the brain regions i.e. midbrain and striatum. MPTP-induced decrease in tyrosine hydroxylase levels were also restored by 2-APB. Moreover, MPTP-induced reduction in proteins involved in mitochondrial biogenesis, viz. peroxisome proliferator-activated-receptor-gamma coactivator and mitochondrial transcription factor-A were increased after 2-APB treatment in vivo. In summary, 2-APB has a promising neuroprotective role in the MPP+/MPTP models of PD via targeting mitochondrial dysfunctions and biogenesis.


Assuntos
Neuroblastoma , Doença de Parkinson , Humanos , Ratos , Animais , Camundongos , 1-Metil-4-fenilpiridínio/metabolismo , 1-Metil-4-fenilpiridínio/farmacologia , Doença de Parkinson/tratamento farmacológico , Espécies Reativas de Oxigênio/metabolismo , Ratos Sprague-Dawley , Neuroblastoma/metabolismo , Mitocôndrias/metabolismo , Camundongos Endogâmicos C57BL , Linhagem Celular Tumoral , Neurônios Dopaminérgicos
10.
Eur Rev Med Pharmacol Sci ; 27(2): 570-579, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36734700

RESUMO

OBJECTIVE: In recent years, botanical medicines alone or in conjunction with existing therapies have attracted considerable popularity as an alternative treatment for Parkinson's Disease (PD). For instance, Geranium robertianum L. (Geraniaceae family) has been used in folk medicine for its antioxidant and anti-inflammatory properties. However, its neuroprotective potential has not been well demonstrated. MATERIALS AND METHODS: Herein and for the first time, we have investigated the in vitro neuroprotective effects of leaf extract of G. Robertianum over a wide dose range (0-200 µg/mL) on the PD model using retinoic acid (RA)-differentiated SHSY-5Y cells and 1-methyl-4-phenylpyridinium (MPP+)-induced neurotoxicity. The neuroprotective effects were determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and lactate dehydrogenase (LDH) release assays. The antioxidant activity of the extract was measured by total antioxidant status (TAS) and total oxidant status (TOS). The effect of leaf aqueous extracts on acetylcholinesterase (AChE) activity was also determined. Finally, cell death mechanisms were analyzed by flow cytometry. RESULTS: Our results showed that G. Robertianum leaf extract ameliorated cytotoxicity and oxidative damage by MPP+. Moreover, G. Robertianum extract exhibited a protective activity against MPP+ induced apoptosis. CONCLUSIONS: The current findings could lead to a promising new candidate for a possible cure of Parkinson's disease through neuroprotective mechanisms with respect to antioxidant and apoptosis inhibitory properties of G. Robertianum water extracts.


Assuntos
Geranium , Fármacos Neuroprotetores , Doença de Parkinson , 1-Metil-4-fenilpiridínio/farmacologia , Acetilcolinesterase , Antioxidantes/farmacologia , Apoptose , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Extratos Vegetais/farmacologia
11.
Int J Mol Sci ; 24(1)2023 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-36614176

RESUMO

The low effectiveness of symptomatic pharmacotherapy for Parkinson's disease (PD), which compensates for dopamine (DA) deficiency under degeneration of nigrostriatal dopaminergic (DAergic) neurons, could apparently be improved with neuroprotective therapy, which slows down neurodegeneration and PD progression. For this, it is necessary to have a DAergic cell line for the development of a PD model to screen neuroprotectors. We used immortalized human embryonic mesencephalon LUHMES cells (LCs) differentiated into DAergic neurons. The aim of this study was to characterize the phenotype of differentiated LCs and develop an 1-methyl-4-phenylpyridinium iodide (MPP+)-based test system for screening neuroprotectors. Using polymerase chain reaction (PCR) and immunocytochemistry, it has been shown that all differentiated LCs express genes and synthesize proteins characteristic of all neurons (microtubule-associated protein 2, bIII-tubulin, synaptotagmin 1) and specifically of DAergic neurons (tyrosine hydroxylase, aromatic L-amino acid decarboxylase, DA transporter, vesicular monoamine transporter 2). Furthermore, LCs are able to produce a small amount of DA, but under special conditions. To assess the mechanisms of neurodegeneration and neuroplasticity under the influence of toxins and antiparkinsonian drugs, including neuroprotectors, we have developed an LCs-based MPP+ PD model and proposed an original panel of markers for testing functional and structural cell disorders.


Assuntos
1-Metil-4-fenilpiridínio , Doença de Parkinson , Humanos , 1-Metil-4-fenilpiridínio/farmacologia , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Antiparkinsonianos/metabolismo , Fenótipo
12.
Environ Toxicol ; 38(4): 857-866, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36629037

RESUMO

Parkinson's disease (PD), a chronic and progressive neurodegenerative disease, can reduce the population of dopaminergic neurons in the substantia nigra. The cause of this neuronal death remains unclear. 1-Methyl-4-phenylpyridinium ion (MPP+) is a potent neurotoxin that can destroy dopaminergic (DA) neurons and promote PD. Garcinol, a polyisoprenylated benzophenone derivative, was extracted from Garcinia indica and is an important active compound it has been used as an anticancer, antioxidant, and anti-inflammatory, agent and it can suppress reactive oxygen species (ROS) mediated cell death in a PD model. Human neuroblastoma (SH-SY5Y) cells (1 × 105 cells) were treated with MPP+ (1 mM) for 24 h to induce cellular ROS production. The formation of ROS was suppressed by pretreatment with different concentrations of garcinol (0.5 and 1.0 µM) for 3 h in SH-SY5Y cells. The present study found that MPP+ treatment increased the formation of reactive oxygen species (ROS), and the increased ROS began to promote cell death in SH-SY5Y cells. However, our natural compound garcinol effectively blocked MPP+-mediated ROS formation by activating the DJ-1/SIRT1 and PGC-1α mediated antioxidant pathway. Further findings indicate that the activated SIRT1 can also regulate p-AMPK-mediated autophagy to protect the neurons from the damage it concludes that garcinol sub-sequential regulates intracellular autophagy in this model, and the productive efficacy of garcinol was confirmed by western blot analysis and MitoSOX DCFDA and MTT assays. The results showed garcinol increased protection due to the prevention of MPP+-induced ROS and the promotion of cell survival.


Assuntos
Neuroblastoma , Doenças Neurodegenerativas , Doença de Parkinson , Humanos , Antioxidantes/metabolismo , 1-Metil-4-fenilpiridínio/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Estresse Oxidativo , Sirtuína 1/metabolismo , Linhagem Celular Tumoral , Morte Celular , Autofagia , Sobrevivência Celular , Apoptose
13.
Immun Inflamm Dis ; 11(1): e756, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36705403

RESUMO

INTRODUCTION: The inflammation mediated by microglial cells plays an important role in the process of neurodegenerative diseases. Recent evidence indicates that semaphorin 7A (SEMA7A) is implicated in various neurodegenerative diseases, but whether it plays a role in Parkinson's disease (PD) remains unclear. METHODS: In this study, 1.0 mmol/L 1-methyl-4-phenylpyridinium (MPP+ )-stimulated mouse microglia (BV2) cells were used as an in vitro model of PD. The expression of SEMA7A was detected by quantitative polymerase chain reaction. Cell Counting Kit-8 and apoptosis kits were used to analyze the viability and apoptosis of BV-2 cells. The content of IL-6, IL-ß, and tumor necrosis factor-α was determined by ELISA (enzyme-linked immunosorbent assay) kit. Western blot was used to detect the protein expression level of the inducible NO synthase and cyclooxygenase-2. RESULTS: Our findings indicated that SEMA7A expression in BV2 cells was upregulated after MPP+ stimulation. Knockdown of SEMA7A promoted cell viability while it inhibited apoptosis and the expression of proinflammatory enzymes and proinflammatory cytokines. Silencing SEMA7A-induced peroxisome proliferator-activated receptor-gamma (PPAR-γ) activation and mitogen-activated protein kinase (MAPK) signaling pathway inactivation. Furthermore, a PPAR-γ inhibitor and an MAPK activator promoted the effect of MPP+ on cell viability, apoptosis, and inflammation of BV2 cells; what is more, the PPAR-γ inhibitor and MAPK activator blocked the inhibitory effect of SEMA7A downregulation on MPP+ -induced injury. CONCLUSION: In general, knockdown of SEMA7A inhibits MPP+ -induced BV2 cell apoptosis and inflammation via PPAR-γ activation and MAPK inactivation, which may provide a new therapy target for PD.


Assuntos
Proteínas Quinases Ativadas por Mitógeno , Semaforinas , Camundongos , Animais , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/farmacologia , Microglia/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , 1-Metil-4-fenilpiridínio/metabolismo , 1-Metil-4-fenilpiridínio/farmacologia , Inflamação/genética , Inflamação/metabolismo , Apoptose/genética , Antígenos CD/metabolismo , Semaforinas/genética , Semaforinas/metabolismo , Semaforinas/farmacologia
14.
Int J Mol Sci ; 23(12)2022 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-35742931

RESUMO

Particulate matter (PM), a component of air pollution, has been epidemiologically associated with a variety of diseases. Recent reports reveal that PM has detrimental effects on the brain. In this study, we aimed to investigate the biological effects of ambient particles on the neurodegenerative disease Parkinson's disease (PD). We exposed mice to coarse particles (PM10: 2.5-10 µm) for short (5 days) and long (8 weeks) durations via intratracheal instillation. Long-term PM10 exposure exacerbated motor impairment and dopaminergic neuron death in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse models. Short-term PM10 exposure resulted in both pulmonary and systemic inflammatory responses in mice. We further investigated the mechanism underlying PM10-induced neurotoxicity in cocultures of lung LA-4 epithelial cells and RAW264.7 macrophages. PM10 treatment elicited a dramatic increase in proinflammatory mediators in LA-4/RAW264.7 coculture. Treating BV2 microglial cells with PM10-treated conditioned medium induced microglial activation. Furthermore, 1-methyl-4-phenylpyridinium (MPP+) treatment caused notable cell death in N2A neurons cocultured with activated BV2 cells in PM10-conditioned medium. Altogether, our results demonstrated that PM10 plays a role in the neurodegeneration associated with PD. Thus, the impact of PM10 on neurodegeneration could be related to detrimental air pollution-induced systemic effects on the brain.


Assuntos
Doenças Neurodegenerativas , Doença de Parkinson , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , 1-Metil-4-fenilpiridínio/farmacologia , Animais , Meios de Cultivo Condicionados/farmacologia , Modelos Animais de Doenças , Neurônios Dopaminérgicos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Doenças Neurodegenerativas/metabolismo , Doença de Parkinson/etiologia , Doença de Parkinson/metabolismo , Material Particulado/toxicidade
15.
Neurosci Lett ; 781: 136667, 2022 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-35490904

RESUMO

Parkinson disease (PD) is a prevalent neurodegenerative disorder that is characterized by motor and behavioral disturbances, including resting tremors, rigidity, bradykinesia, and postural instability. The primary cause of PD is the degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) region that subsequently reduces the dopamine content in the striatum (ST); this is a promising therapeutic target for PD. Resilin is an elastomeric protein with high strain, low stiffness, and high resilience that is found in insect cuticles. However, scant evidence supports the application of resilin in neurodegenerative diseases, including PD. Herein, we investigated the protective effects of resilin on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD in mouse models and explored the mechanisms underlying its action. Resilin significantly and concentration-dependently reduced 1-methyl-4-phenylpyridinium+ (MPP+)-induced apoptotic neurotoxicity in differentiated PC12 and SH-SY5Y cells. Moreover, resilin prevented dopamine depletion in ST, and immunohistochemical findings indicated that resilin protects against dopaminergic neuronal loss induced by MPTP in the SNpc and ST. Behavioral studies using pole and rotarod tests showed significantly improved PD-related motor impairment in mice treated with resilin. We then explored the molecular mechanisms underlying the apoptosis of dopaminergic neurons using protein arrays and discovered that resilin inhibits dopaminergic neuronal death through the apoptosis signaling factors cytochrome c and caspases-9 and -3 in the SNpc. Thus, resilin has potential in treating PD by controlling apoptosis signals.


Assuntos
Neuroblastoma , Fármacos Neuroprotetores , Doença de Parkinson , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , 1-Metil-4-fenilpiridínio/farmacologia , Animais , Modelos Animais de Doenças , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Humanos , Proteínas de Insetos , Camundongos , Camundongos Endogâmicos C57BL , Neuroblastoma/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson/metabolismo , Substância Negra/metabolismo
16.
Bioengineered ; 13(4): 10889-10901, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35481549

RESUMO

This study aimed to analyze the function and latent mechanism of long noncoding RNA BACE1-antisense transcript (lncRNA BACE1-AS) in MPP+-induced SH-SY5Y cells. SH-SY5Y cells were cultivated in 1 mM MPP+ for 24 h to establish Parkinson's disease (PD) model in vitro. TargetScan and luciferase reporter assay were conducted to predict and verify the interaction between microRNA (miR)-214-3p and CDIP1 (Cell death-inducing p53-target protein 1). Cell viability, lactate dehydrogenase (LDH) release, and cell apoptosis were evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2 H-tetrazolium bromide (MTT), LDH, and flow cytometer. The secretion of inflammatory factors and representative biomarkers of oxidative stress, including reactive oxygen species (ROS) and superoxide dismutase (SOD) were assessed using enzyme-linked immunosorbent assay (ELISA) and specific assay kits. Results suggested that lncRNA BACE1-AS was over-expressed and miR-214-3p was under-expressed in MPP+-stimulated SH-SY5Y cells. Further analyses revealed that MPP+ inhibited cell viability; enhanced cell apoptosis, Cleaved Caspase-3 expression and Cleaved Caspase-3/GAPDH ratio; induced oxidative stress and inflammation in SH-SY5Y cells were inhibited by lncRNA BACE1-AS-siRNA transfection; and all these inhibitions were reversed by miR-214-3p inhibitor. In addition, we found that CDIP1 was directly targeted by miR-214-3p and up-regulated in MPP+-stimulated SH-SY5Y cells. Further functional assays suggested that CDIP1-plasmid reversed the effects of miR-214-3p mimic on MPP+-stimulated SH-SY5Y cells. In conclusion, lncRNA BACE1-AS regulates SH-SY5Y cell proliferation, apoptosis, inflammatory response, and oxidative stress through direct regulation of miR-214-3p/CDIP1 signaling axis, and could be a potential candidate associated with the diagnosis and treatment of PD.


Assuntos
MicroRNAs , Doença de Parkinson , RNA Longo não Codificante , 1-Metil-4-fenilpiridínio/farmacologia , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Secretases da Proteína Precursora do Amiloide/farmacologia , Apoptose/genética , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Ácido Aspártico Endopeptidases/farmacologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Humanos , MicroRNAs/metabolismo , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , RNA Longo não Codificante/metabolismo , Proteína Supressora de Tumor p53
17.
Eur Neurol ; 85(3): 235-244, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35108712

RESUMO

BACKGROUND: At present, symptomatic treatment may improve the life quality of Parkinson's disease (PD) patients to a certain extent but cannot completely cure PD. Therefore, it is urgent medical problem to be solved for improving the efficacy and safety of PD treatment. METHODS: SH-SY5Y and SK-N-SH cells were treated with 1-methyl-4-phenylpyridinium (MPP+) to establish PD model cells. miR-126-5p and specific protein-1 (SP1) expression levels were detected by quantitative Real-Time PCR (qRT-PCR). Western blot was applied to measure protein levels of SP1, Bax, and Bcl-2. The viabilities and apoptosis rates of treated cells were measured using cell counting kit-8 assay and flow cytometry analysis. Enzyme-linked immunosorbent assay was performed to measure TNF-α and IL-1ß releases. Interaction between miR-126-5p and SP1 was examined by dual-luciferase reporter assay. RESULTS: MPP+ treatment greatly downregulated miR-126-5p expression while upregulated SP1 expression in SH-SY5Y and SK-N-SH cells in a time- and does-dependent manner. Overexpression of miR-126-5p facilitated cell viability, while reduced cell apoptosis and inflammatory responses induced by MPP+ treatment. Moreover, SP1 was a target of miR-126-5p and could be negatively regulated by miR-126-5p. Overexpression of SP1 could reverse the effects of miR-126-5p on MPP+-administrated cells. CONCLUSION: Our results suggested that miR-126-5p attenuated the neurotoxicity induced by MPP+ in vitro through targeting SP1 (Graphical abstract), which further enhanced our understanding of the pathological mechanism of PD.


Assuntos
MicroRNAs , Doença de Parkinson , Fator de Transcrição Sp1 , 1-Metil-4-fenilpiridínio/farmacologia , Apoptose/genética , Linhagem Celular Tumoral , Humanos , MicroRNAs/genética , Doença de Parkinson/patologia , Fator de Transcrição Sp1/genética
18.
Brain Res ; 1782: 147814, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35123924

RESUMO

BACKGROUND: Abnormal expression of long non-coding RNA (lncRNA) is associated with the progression of Parkinson's disease (PD). LINC00943 has been proved to play an important role in the development of PD, so its role and mechanism in PD progression are worth further exploration. METHODS: MPTP was used to construct PD mice model, and its active ingredient MPP+ was used to construct PD cell model. Cell proliferation and apoptosis were determined by MTT assay, EdU staining and flow cytometry. The protein levels of Cyclin D1, Bcl-2 and specificity protein 1 (SP1) were tested by western blot analysis. The concentrations of inflammation factors were examined by ELISA assay. The expression levels of LINC00943, microRNA (miR)-338-3p and SP1 were measured using quantitative real-time PCR. The interaction between miR-338-3p and LINC00943 or SP1 was confirmed using dual-luciferase reporter assay and RIP assay. RESULTS: Our data showed that LINC00943 was highly expressed in the brain tissues of MPTP-treated mice and MPP+-induced SK-N-SH cells. Knockdown of LINC00943 could promote the proliferation, while inhibit the apoptosis and inflammation of MPP+-induced SK-N-SH cells to alleviate cell injury. In terms of mechanism, we pointed out that LINC00943 could sponge miR-338-3p, and miR-338-3p could target SP1. The negative regulation of si-LINC00943 on MPP+-induced SK-N-SH cell injury could be reversed by miR-338-3p inhibitor. Moreover, miR-338-3p had a protective effect on SK-N-SH cells from MPP+-induced injury, which could be reversed by SP1 overexpression. Additionally, we confirmed that LINC00943 positively regulated SP1 via sponging miR-338-3p. CONCLUSION: To sum up, our data revealed that knockdown LINC00943 might alleviate PD progression through regulating the miR-338-3p/SP1 axis.


Assuntos
MicroRNAs , Doença de Parkinson , RNA Longo não Codificante , Fator de Transcrição Sp1 , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , 1-Metil-4-fenilpiridínio/farmacologia , Animais , Apoptose , Linhagem Celular Tumoral , Inflamação/metabolismo , Camundongos , MicroRNAs/genética , Doença de Parkinson/genética , RNA Longo não Codificante/genética , Fator de Transcrição Sp1/metabolismo
19.
Nitric Oxide ; 120: 44-52, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35033681

RESUMO

We previously demonstrated different expression patterns of the neuronal nitric oxide synthase (nNOS) splicing variants, nNOS-µ and nNOS-α, in the rat brain; however, their exact functions have not been fully elucidated. In this study, we compared the enzymatic activities of nNOS-µ and nNOS-α and investigated intracellular redox signaling in nNOS-expressing PC12 cells, stimulated with a neurotoxicant, 1-methyl-4-phenylpyridinium ion (MPP+), to enhance the nNOS uncoupling reaction. Using in vitro studies, we show that nNOS-µ produced nitric oxide (NO), as did nNOS-α, in the presence of tetrahydrobiopterin (BH4), an important cofactor for the enzymatic activity. However, nNOS-µ generated more NO and less superoxide than nNOS-α in the absence of BH4. MPP + treatment induced more reactive oxygen species (ROS) production in nNOS-α-expressing PC12 cells than in those expressing nNOS-µ, which correlated with the intracellular production of 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP), a downstream messenger of nNOS redox signaling, and apoptosis in these cells. Furthermore, post-treatment with 8-nitro-cGMP aggravated MPP+-induced cytotoxicity via activation of the H-Ras/extracellular signal-regulated kinase signaling pathway. In conclusion, our results provide strong evidence that nNOS-µ exhibits distinctive enzymatic properties of NO/ROS production, contributing to the regulation of intracellular redox signaling, including the downstream production of 8-nitro-cGMP.


Assuntos
Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico/metabolismo , Superóxidos/metabolismo , 1-Metil-4-fenilpiridínio/farmacologia , Animais , Apoptose/efeitos dos fármacos , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Oxirredução , Células PC12 , Fosforilação/efeitos dos fármacos , Isoformas de Proteínas/metabolismo , Ratos
20.
Neurotox Res ; 39(6): 1771-1781, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34773593

RESUMO

Parkinson's disease (PD) is a multi-factorial neurodegenerative disease. Long noncoding RNAs (lncRNAs) have been revealed to be involved in the process of PD. Herein, this study aimed to investigate the potential function and mechanism of JHDM1D-AS1 (JHDM1D antisense 1) in PD process. 1-Methyl-4-phenylpyridinium (MPP +)-induced SK-N-SH cells were used to conduct expression and function analyses. Levels of genes and proteins were examined using real-time reverse transcription PCR (RT-qPCR) and Western blot. Cell viability and apoptosis were determined using CCK-8 assay, flow cytometry, and Western blot, respectively. ELISA analysis was performed for the detection of interleukin (IL)-1ß, IL-6, and tumor necrosis factor (TNF)-α. The contents of lactate dehydrogenase (LDH), superoxide dismutase (SOD), and malondialdehyde (MDA) were measured using commercial kits. The direct interactions between miR-134-5p and PIK3R3 (Phosphoinositide-3-Kinase Regulatory Subunit 3) or JHDM1D-AS1 were verified by dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. JHDM1D-AS1 expression was decreased by MPP + in SK-N-SH cells in a dose- or time-dependent manner. Functionally, JHDM1D-AS1 overexpression attenuated MPP + -evoked neuronal apoptosis, inflammation, and oxidative stress. Mechanistically, JHDM1D-AS1 competitively bound to miR-134-5p to upregulate the expression of its target PIK3R3. Rescue experiments suggested that miR-134-5p upregulation reversed the inhibitory effects of JHDM1D-AS1 on MPP + -induced neuronal injury. Moreover, inhibition of miR-134-5p protected neurons against MPP + -induced neuronal apoptosis, inflammation, and oxidative stress, which were abolished by PIK3R3 silencing. JHDM1D-AS1 protected against MPP + -induced neuron injury via miR-134-5p/PIK3R3 axis, suggesting the potential involvement of this axis in PD process.


Assuntos
Histona Desmetilases com o Domínio Jumonji/metabolismo , MicroRNAs/metabolismo , Neurônios/efeitos dos fármacos , Doença de Parkinson/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , RNA Longo não Codificante/metabolismo , 1-Metil-4-fenilpiridínio/antagonistas & inibidores , 1-Metil-4-fenilpiridínio/farmacologia , Western Blotting , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Doença de Parkinson/tratamento farmacológico , Reação em Cadeia da Polimerase em Tempo Real
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA