Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 187
Filtrar
1.
Mov Disord ; 39(5): 766-767, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38627965

RESUMO

Sinus infection of Saccharomyces cerevisiae accelerates the aggregation of α-synuclein (α-syn) in A53T mice, which was caused by prion protein Sup35. Sup35 promotes α-syn aggregation in vitro and in vivo and leads to Parkinson's disease (PD)-like motor impairment in wildtype mice, suggesting that the yeast Sup35 triggers α-syn pathology in PD.


Assuntos
Doença de Parkinson , Fatores de Terminação de Peptídeos , Proteínas de Saccharomyces cerevisiae , alfa-Sinucleína , Animais , Humanos , Camundongos , alfa-Sinucleína/metabolismo , alfa-Sinucleína/genética , Modelos Animais de Doenças , Camundongos Transgênicos , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Fatores de Terminação de Peptídeos/metabolismo , Fatores de Terminação de Peptídeos/genética , Príons/metabolismo , Príons/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
2.
Neurochem Res ; 49(2): 466-476, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37917337

RESUMO

Parkinson's disease (PD) is a prevalent neurodegenerative disorder characterized by the loss of dopaminergic neurons and the accumulation of iron in the substantia nigra. While iron accumulation and inflammation are implicated in PD pathogenesis, their impact on oligodendrocytes, the brain's myelin-forming cells, remains elusive. This study investigated the influence of interleukin-1ß (IL-1ß), an elevated proinflammatory cytokine in PD, on iron-related proteins in MO3.13 oligodendrocytes. We found that IL-1ß treatment in undifferentiated MO3.13 oligodendrocytes increased iron regulatory protein 1 and transferrin receptor 1 (TfR1) expression while decreasing ferroportin 1 (FPN1) expression. Consequently, iron uptake was enhanced, and iron release was reduced, leading to intracellular iron accumulation. Conversely, IL-1ß treatment in differentiated MO3.13 oligodendrocytes exhibited the opposite effect, with decreased TfR1 expression, increased FPN1 expression, and reduced iron uptake. These findings suggest that IL-1ß-induced dysregulation of iron metabolism in oligodendrocytes may contribute to the pathological processes observed in PD. IL-1ß can increase the iron content in undifferentiated oligodendrocytes, thus facilitating the differentiation of undifferentiated MO3.13 oligodendrocytes. In differentiated oligodendrocytes, IL-1ß may facilitate iron release, providing a potential source of iron for neighboring dopaminergic neurons, thereby initiating a cascade of deleterious events. This study provides valuable insights into the intricate interplay between inflammation, abnormal iron accumulation, and oligodendrocyte dysfunction in PD. Targeting the IL-1ß-mediated alterations in iron metabolism may hold therapeutic potential for mitigating neurodegeneration and preserving dopaminergic function in PD.


Assuntos
Proteína 1 Reguladora do Ferro , Doença de Parkinson , Humanos , Interleucina-1beta/metabolismo , Proteína 1 Reguladora do Ferro/metabolismo , Doença de Parkinson/metabolismo , Ferro/metabolismo , Inflamação/metabolismo , Oligodendroglia/metabolismo
3.
Acta Pharmacol Sin ; 45(1): 52-65, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37674043

RESUMO

Gut microbiota disturbance and systemic inflammation have been implicated in the degeneration of dopaminergic neurons in Parkinson's disease (PD). How the alteration of gut microbiota results in neuropathological events in PD remains elusive. In this study, we explored whether and how environmental insults caused early neuropathological events in the substantia nigra (SN) of a PD mouse model. Aged (12-month-old) mice were orally administered rotenone (6.25 mg·kg-1·d-1) 5 days per week for 2 months. We demonstrated that oral administration of rotenone to ageing mice was sufficient to establish a PD mouse model and that microglial activation and iron deposition selectively appeared in the SN of the mice prior to loss of motor coordination and dopaminergic neurons, and these events could be fully blocked by microglial elimination with a PLX5622-formulated diet. 16 S rDNA sequencing analysis showed that the gut microbiota in rotenone-treated mice was altered, and mice receiving faecal microbial transplantation (FMT) from ageing mice treated with rotenone for 2 months exhibited the same pathology in the SN. We demonstrated that C-X-C motif chemokine ligand-1 (CXCL1) was an essential molecule, as intravenous injection of CXCL1 mimicked almost all the pathology in serum and SN induced by oral rotenone and FMT. Using metabolomics and transcriptomics analyses, we identified the PPAR pathway as a key pathway involved in rotenone-induced neuronal damage. Inhibition of the PPARγ pathway was consistent in the above models, whereas its activation by linoleic acid (60 mg·kg-1·d-1, i.g. for 1 week) could block these pathological events in mice intravenously injected with CXCL1. Altogether, these results reveal that the altered gut microbiota resulted in neuroinflammation and iron deposition occurring early in the SN of ageing mice with oral administration of rotenone, much earlier than motor symptoms and dopaminergic neuron loss. We found that CXCL1 plays a crucial role in this process, possibly via PPARγ signalling inhibition. This study may pave the way for understanding the "brain-gut-microbiota" molecular regulatory networks in PD pathogenesis. The aged C57BL/6 male mice with rotenone intragastric administration showed altered gut microbiota, which caused systemic inflammation, PPARγ signalling inhibition and neuroinflammation, brain iron deposition and ferroptosis, and eventually dopaminergic neurodegeneration in PD.


Assuntos
Microbioma Gastrointestinal , Doença de Parkinson , Camundongos , Animais , Masculino , Rotenona/toxicidade , Doenças Neuroinflamatórias , PPAR gama , Camundongos Endogâmicos C57BL , Doença de Parkinson/patologia , Substância Negra/patologia , Neurônios Dopaminérgicos/patologia , Inflamação/patologia , Ferro , Modelos Animais de Doenças
4.
Acta Pharmacol Sin ; 45(2): 268-281, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37674042

RESUMO

Cell senescence has been implicated in the pathology of Parkinson's disease (PD). Both abnormal α-synuclein aggregation and iron deposition are suggested to be the triggers, facilitators, and aggravators during the development of PD. In this study, we investigated the involvement of α-synuclein and iron in the process of cell senescence in a mouse model of PD. In order to overexpress α-syn-A53T in the substantia nigra pars compacta (SNpc), human α-syn-A53T was microinjected into both sides of the SNpc in mice. We found that overexpression of α-syn-A53T for one week induced significant pro-inflammatory senescence-associated secretory phenotype (SASP), increased cell senescence-related proteins (ß-gal, p16, p21, H2A.X and γ-H2A.X), mitochondrial dysfunction accompanied by dysregulation of iron-related proteins (L-ferritin, H-ferritin, DMT1, IRP1 and IRP2) in the SNpc. In contrast, significant loss of nigral dopaminergic neurons and motor dysfunction were only observed after overexpression of α-syn-A53T for 4 weeks. In PC12 cells stably overexpressing α-syn-A53T, iron overload (ferric ammonium citrate, FAC, 100 µM) not only increased the level of reactive oxygen species (ROS), p16 and p21, but also exacerbated the processes of oxidative stress and cell senescence signalling induced by α-syn-A53T overexpression. Interestingly, reducing the iron level with deferoxamine (DFO) or knockdown of transferrin receptor 1 (TfR1) significantly improved both the phenotypes and dysregulated proteins of cell senescence induced by α-syn-A53T overexpression. All these evidence highlights the toxic interaction between iron and α-synuclein inducing cell senescence, which precedes nigral dopaminergic neuronal loss in PD. Further investigation on cell senescence may yield new therapeutic agents for the prevention or treatment of PD.


Assuntos
Doença de Parkinson , Ratos , Camundongos , Animais , Humanos , Doença de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Neurônios Dopaminérgicos/metabolismo , Ferro/metabolismo , Substância Negra/metabolismo , Substância Negra/patologia , Dopamina/metabolismo , Senescência Celular , Modelos Animais de Doenças
5.
J Neurophysiol ; 2023 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-37584076

RESUMO

Alpha-synuclein (α-syn) is a major component of lewy bodies, which is biomarker of Parkinson's disease (PD). It accumulates in substantia nigra pars compacts (SNpc) to form insoluble aggregates and cause neurotoxicity, which is often accompanied by iron deposition. In this study, we compared the iron reductase activity between monomeric α-syn (M-α-syn) and oligomeric α-syn (O-α-syn), investigated the effect of α-syn on iron metabolism of BV2 microglia cells as well. We found that α-syn had ferric reductase activity, and O-α-syn had stronger enzyme activity than M-α-syn. M-α-syn upregulated iron uptake protein, divalent metal transporter1 (DMT1) expression and iron influx, but did not regulate iron release protein, ferroportin1 (FPN1) expression and iron efflux. O-α-syn elevated the expression of both DMT1 and FPN1, thus increased the iron influx and efflux in BV2 microglial cells, but the expressions of iron regulatory protein1 and hypoxia inducible factor2α have no significant change. Moreover, both M-α-syn and O-α-syn could increase the mRNA expressions of TNF-α and IL-1ß in BV2 microglia cells. Taken together, our data suggest that both types of α-syn can activate microglia, which leads to increased expressions of pro-inflammatory factors. α-syn can affect DMT1 and FPN1 expressions in BV2 microglia cells, which might be through its ferric reductase activity.

6.
J Neurochem ; 167(3): 347-361, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37746863

RESUMO

Heme oxygenase-1 (HO-1) is the only way for cells to decompose heme. It can cleave heme to produce carbon monoxide (CO), ferrous iron (Fe2+ ), and biliverdin (BV). BV is reduced to bilirubin (BR) by biliverdin reductase(BVR). In previous studies, HO-1 was considered to have protective effects because of its anti-inflammatory, anti-apoptosis, and antiproliferation functions. However, emerging experimental studies have found that the metabolites derived from HO-1 can cause increase iin intracellular oxidative stress, mitochondrial damage, iron death, and autophagy. Because of its particularity, it is very meaningful to understand its exact mechanism. In this review, we summarized the protective and toxic effects of HO-1, its potential mechanism, its role in neurodegenerative diseases and related drug research. This knowledge may be beneficial to the development of new therapies for neurodegenerative diseases and is crucial to the development of new therapeutic strategies and biomarkers.

7.
Neurochem Res ; 48(3): 830-838, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36352276

RESUMO

Oligodendrocytes are the most iron-rich cells in the brain. Studies have shown that oligodendrocytes are very sensitive to oxidative stress, and iron overload is more likely to cause damage to oligodendrocytes. The purpose of this experiment was to investigate the damaging effect and mechanism of ferric ammonium citrate (FAC) on MO3.13 oligodendrocytes. In FAC treatment group, the intracellular iron concentration and intracellular reactive oxygen species were increased. There were no obvious changes in nucleus and chromatin, but increased mitochondrial membrane density, decreased mitochondrial cristae and mitochondrial length were observed. Glutathione peroxidase 4 (GPX4) expression was decreased, but the ratio of Bcl-2/Bax protein levels and cleaved caspase-3 expression did not change. Moreover, the iron chelator deferoxamine (DFO) and the ferroptosis inhibitor ferrostatin-1(Fer-1) could inhibit the upregulation of GPX4, which indicating that DFO and Fer-1 could inhibit ferroptosis in MO3.13 oligodendrocytes induced by iron overload. Furthermore, the phosphorylation level of p53 was not changed, while the ratio of protein expressions of p-Erk1/2/Erk1/2 were markedly increased. Taken together, our data suggest that iron overload induces ferroptosis but not apoptosis in oligodendrocytes. The mechanism may be related to mitogen-activated protein kinase pathway activation rather than p53 pathway activation.


Assuntos
Ferroptose , Sobrecarga de Ferro , Humanos , Apoptose , Sobrecarga de Ferro/metabolismo , Ferro/metabolismo , Espécies Reativas de Oxigênio/metabolismo
8.
Neurochem Res ; 48(6): 1707-1715, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36602724

RESUMO

Various pharmacological blockers targeting K+ channel have been identified to be related to the treatment of Parkinson's disease (PD). Previous studies showed that 4-Aminopyridine (4-AP), a wide-spectrum K+ channel blocker, was able to attenuate apomorphine-induced rotation in parkinsonism rats, indicating the possible beneficial effects in attenuation of PD motor symptoms. However, it is unclear whether 4-AP exhibits neuroprotective effects against the neurodegeneration of substantia nigra (SN)-striatum system in PD. In this study, the 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mouse model was employed to evaluate the neuroprotective effects of 4-AP. Results showed that 4-AP inhibited MPTP-induced dopaminergic neuronal loss in the SN as well as dopamine depletion in the striatum. Behavior indexes of open field test and rotarod test confirmed that 4-AP attenuated MPTP-induced motor deficits. We also showed that 4-AP treatment could significantly attenuate the MPTP-induced increase in malonaldehyde (MDA) levels and decrease in superoxide dismutase (SOD) levels. Additionally, MPTP significantly reduced the Bcl-2 expression and promoted the Caspase-3 activation; 4-AP protected dopaminergic neurons against MPTP-induced neurotoxicity by reversing these changes. These results indicate that 4-AP exerts a neuroprotective effect on dopaminergic neurons against MPTP by decreasing oxidative stress and apoptosis. This provides a promising therapeutic target for the treatment of PD.


Assuntos
Intoxicação por MPTP , Fármacos Neuroprotetores , Doença de Parkinson , Animais , Camundongos , Ratos , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Neurônios Dopaminérgicos , Camundongos Endogâmicos C57BL , Intoxicação por MPTP/tratamento farmacológico , Intoxicação por MPTP/prevenção & controle , Intoxicação por MPTP/induzido quimicamente , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Fármacos Neuroprotetores/metabolismo , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Substância Negra , 4-Aminopiridina/farmacologia
9.
Sheng Li Xue Bao ; 75(2): 197-204, 2023 Apr 25.
Artigo em Chinês | MEDLINE | ID: mdl-37089094

RESUMO

The purpose of this study was to establish a suitable method for extracting cerebrospinal fluid (CSF) from C57BL/6 mice. A patch clamp electrode puller was used to draw a glass micropipette, and a brain stereotaxic device was used to fix the mouse's head at an angle of 135° from the body. Under a stereoscopic microscope, the skin and muscle tissue on the back of the mouse's head were separated, and the dura mater at the cerebellomedullary cistern was exposed. The glass micropipette (with an angle of 20° to 30° from the dura mater) was used to puncture at a point 1 mm inboard of Y-shaped dorsal vertebral artery for CSF sampling. After the first extraction, the glass micropipette was connected with a 1 mL sterile syringe to form a negative pressure device for the second extraction. The results showed that the successful rate of CSF extraction was 83.33% (30/36). Average CSF extraction amount was (7.16 ± 0.43) µL per mouse. In addition, C57BL/6 mice were given intranasally ferric ammonium citrate (FAC) to establish a model of brain iron accumulation, and the CSF extraction technique established in the present study was used for sampling. The results showed that iron content in the CSF from the normal saline control group was not detected, while the iron content in the CSF from FAC-treated group was (76.24 ± 38.53) µmol/L, and the difference was significant. These results suggest that glass micropipette vacuum technique of CSF sampling established in the present study has the advantages of simplicity, high success rate, large extraction volume, and low bleeding rate, and is suitable for the research on C57BL/6 mouse neurological disease models.


Assuntos
Encéfalo , Cisterna Magna , Camundongos , Animais , Vácuo , Camundongos Endogâmicos C57BL , Líquido Cefalorraquidiano
10.
Neurochem Res ; 47(12): 3659-3669, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35829942

RESUMO

Estrogen is a steroid hormone produced mainly by the ovaries. It has been found that estrogen could regulate iron metabolism in neurons and astrocytes in different ways. The role of estrogen on iron metabolism in microglia is currently unknown. In this study, we investigated the effect and mechanism of 17ß-estrogen (E2) on iron transport proteins. We found that following E2 treatment for 24h in BV2 microglial cell lines, the iron importer divalent metal transporter 1 (DMT1) and iron exporter ferroportin 1 (FPN1) were up-regulated , iron storage protein ferritin (FT) was increased. The protein levels of iron regulatory proteins (IRPs) and hepcidin remained unchanged, but hypoxia inducible factor 1 alpha (HIF-1α) was up-regulated. Two kinds of estrogen receptor ß (ERß) antagonist G15 and G protein estrogen receptor (GPER) antagonist PHTPPcould block the effects of E2 in BV2 microglial cell lines. These results suggest that estrogen could increase the protein expressions of DMT1, FPN1, FT-L and FT-H in BV2 microglia cells, which were not related to the regulation of IRP1 and hepcidin, but to the upregulation of HIF-1α. In addition, estrogen might regulate the expressions of iron-related proteins through both ER ß and GPER in BV2 microglia cells.


Assuntos
Hepcidinas , Microglia , Hepcidinas/metabolismo , Microglia/metabolismo , Ferro/metabolismo , Receptor beta de Estrogênio/metabolismo , Receptores de Estrogênio , Estrogênios/farmacologia , Estrogênios/metabolismo , Proteínas de Ligação ao GTP/metabolismo
11.
Cell Commun Signal ; 19(1): 120, 2021 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-34922574

RESUMO

Regulated cell death (RCD) is a ubiquitous process in living organisms that is essential for tissue homeostasis or to restore biological balance under stress. Over the decades, various forms of RCD have been reported and are increasingly being found to involve in human pathologies and clinical outcomes. We focus on five high-profile forms of RCD, including apoptosis, pyroptosis, autophagy-dependent cell death, necroptosis and ferroptosis. Cumulative evidence supports that not only they have different features and various pathways, but also there are extensive cross-talks between modes of cell death. As the understanding of RCD pathway in evolution, development, physiology and disease continues to improve. Here we review an updated classification of RCD on the discovery and features of processes. The prominent focus will be placed on key mechanisms of RCD and its critical role in neurodegenerative disease. Video abstract.


Assuntos
Doenças Neurodegenerativas
12.
Neurochem Res ; 46(6): 1502-1513, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33704649

RESUMO

Alpha-synuclein plays a vital role in the pathology of Parkinson's disease (PD). Spreading of α-synuclein in neighboring cells was believed to contribute to progression in PD. How α-synuclein transmission affects adjacent cells is not full elucidated. Here, we used recombinant α-synuclein to mimic intercellular transmitted α-synuclein in MES23.5 dopaminergic cells, to investigate whether and how it could modulate iron metabolism. The results showed that α-synuclein treatment up-regulated divalent metal transporter 1 (DMT1) and down-regulated iron transporter (FPN), also up-regulated iron regulatory protein 1 (IRP1) protein levels and hepcidin mRNA levels. Endocytosis inhibitor dynasore pretreatment completely abolished and even reversed the upregulation of DMT1 and IRP1 induced by α-synuclein, however, FPN down-regulation was partially blocked by dynasore. Autophagy-inducing agent rapamycin reversed DMT1 up-regulation and FPN down-regulation, and fully blocked the upregulation of IRP1. Elevated hepcidin levels induced by α-synuclein was fully blocked by dynasore pretreatment, however, even higher with rapamycin pretreatment. Alpha-synuclein treatment triggered endoplasmic reticulum (ER) stress. ER stress inducer thapsigargin induced similar responses elicited by α-synuclein. ER stress inhibitor salubrinal blocked the up-regulation of IRP1 and hepcidin, as well as DMT1 up-regulation and FPN down-regulation, also dramatically abolished cAMP-response elements binding protein phosphorylation induced by α-synuclein. Taken together, these finding indicated that extracellular α-synuclein could regulate cellular iron metabolism, probably mediated by ER stress. It provides novel evidence to elucidate the relationships between transmitted α-synuclein and iron metabolism disturbance in PD.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Proteína 1 Reguladora do Ferro/metabolismo , alfa-Sinucleína/farmacologia , Animais , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/química , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Regulação para Baixo/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Hepcidinas/metabolismo , Hidrazonas/farmacologia , Camundongos , Fosforilação/efeitos dos fármacos , Ratos , Sirolimo/farmacologia , Regulação para Cima/efeitos dos fármacos
13.
Sheng Li Xue Bao ; 73(2): 315-328, 2021 Apr 25.
Artigo em Chinês | MEDLINE | ID: mdl-33903893

RESUMO

As a member of the Ras superfamily, Rab proteins are small GTP-binding proteins. In the process of endocytosis of macromolecules and substances delivery between organelles, Rab proteins act on vesicle formation, transport, tethering and fusion by recruiting their effectors, therefore being key regulatory factors in vesicle trafficking. Disturbance of localizations and functions of Rab proteins and their effectors are involved in the pathogenesis of several diseases. This review focuses on the main functions of Rab proteins and their possible roles in the onset and progression of neurodegenerative diseases including Parkinson's disease, Alzheimer's disease, and Huntington's disease.


Assuntos
Doenças Neurodegenerativas , Movimento Celular , Endocitose , Humanos , Transporte Proteico , Proteínas rab de Ligação ao GTP/metabolismo
14.
Sheng Li Xue Bao ; 73(1): 89-102, 2021 Feb 25.
Artigo em Chinês | MEDLINE | ID: mdl-33665664

RESUMO

Parkinson's disease (PD), one of the most frequent neurodegenerative disorders, is characterized by the selective loss of dopaminergic neurons in the substantia nigra (SN). Genetic vulnerability, aging, environmental insults are believed to contribute to the pathogenesis of PD. However, the cellular and molecular mechanism of dopaminergic neurons degeneration remains incompletely understood. Dopamine (DA) metabolism is a cardinal physiological process in dopaminergic neurons, which is closely related to the loss of dopaminergic neurons in the SN. DA metabolism takes part in several pathological processes of PD neurodegeneration, such as iron metabolism disturbance, α-synuclein mis-folding, endoplasmic reticulum stress, protein degradation dysfunction, neuroinflammatory response, etc. In this review, we will describe altered DA metabolism and its contributions to PD pathogenesis.


Assuntos
Doença de Parkinson , Dopamina , Neurônios Dopaminérgicos , Humanos , Doença de Parkinson/etiologia , Substância Negra , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
15.
Int J Mol Sci ; 21(21)2020 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-33171641

RESUMO

Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta. Several brain-gut peptides are able to exert neuroprotective effects on the nigrostriatal dopaminergic system. Apelin-13 is a neuropeptide, conveying potential neuroprotective activities. However, whether, and how, apelin-13 could antagonize rotenone-induced neurotoxicity has not yet been elucidated. In the present study, rotenone-treated SH-SY5Y cells and rats were used to clarify whether apelin-13 has protective effects on dopaminergic neurons, both in vivo and in vitro. The results showed that apelin-13 could protect SH-SY5Y cells from rotenone-induced injury and apoptosis. Apelin-13 was able to activate autophagy, and restore rotenone induced autophagy impairment in SH-SY5Y cells, which could be blocked by the autophagy inhibitor 3-Methyladenine. Apelin-13 activated AMPK/mTOR/ULK-1 signaling, AMPKα inhibitor compound C, as well as apelin receptor blockage via siRNA, which could block apelin-13-induced signaling activation, autophagy activation, and protective effects, in rotenone-treated SH-SY5Y cells. These results indicated that apelin-13 exerted neuroprotective properties against rotenone by stimulating AMPK/mTOR/ULK-1 signaling-mediated autophagy via the apelin receptor. We also observed that intracerebroventricular injection of apelin-13 could alleviate nigrostriatal dopaminergic neuron degeneration in rotenone-treated rats. Our findings provide new insights into the mechanism by which apelin-13 might attenuate neurotoxicity in PD.


Assuntos
Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Rotenona/toxicidade , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Receptores de Apelina/antagonistas & inibidores , Receptores de Apelina/genética , Receptores de Apelina/metabolismo , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Linhagem Celular , Neurônios Dopaminérgicos/patologia , Humanos , Masculino , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Neurotoxinas/toxicidade , Transtornos Parkinsonianos/tratamento farmacológico , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/patologia , RNA Interferente Pequeno/genética , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo
16.
J Cell Physiol ; 234(4): 4232-4242, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30132882

RESUMO

Previous studies have demonstrated an effect of estrogen on iron metabolism in peripheral tissues. The role of estrogen on brain iron metabolism is currently unknown. In this study, we investigated the effect and mechanism of estrogen on iron transport proteins. We demonstrated that the iron exporter ferroportin 1 (FPN1) and iron importer divalent metal transporter 1 (DMT1) were upregulated and iron content was decreased after estrogen treatment for 12 hr in primary cultured astrocytes. Hypoxia-inducible factor-1 alpha (HIF-1α) was upregulated, but HIF-2α remained unchanged after estrogen treatment for 12 hr in primary cultured astrocytes. In primary cultured neurons, DMT1 was downregulated, FPN1 was upregulated, iron content decreased, iron regulatory protein (IRP1) was downregulated, but HIF-1α and HIF-2α remained unchanged after estrogen treatment for 12 hr. These results suggest that the regulation of iron metabolism by estrogen in astrocytes and neurons is different. Estrogen increases FPN1 and DMT1 expression by inducing HIF-1α in astrocytes, whereas decreased expression of IRP1 may account for the decreased DMT1 and increased FPN1 expression in neurons.


Assuntos
Astrócitos/efeitos dos fármacos , Estradiol/farmacologia , Ferro/metabolismo , Neurônios/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Astrócitos/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Células Cultivadas , Idade Gestacional , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteína 1 Reguladora do Ferro/metabolismo , Neurônios/metabolismo , Ratos Wistar , Fatores de Tempo
17.
FASEB J ; : fj201800060RR, 2018 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-29897813

RESUMO

Increasing evidence has confirmed that nigral iron accumulation and activation of NMDA receptors (NRs) contribute to the neurodegeneration of dopamine (DA) neurons in Parkinson's disease (PD). Earlier work indicated that activation of NRs participated in iron metabolism in the hippocampus. However, the relationship between activation of NRs and iron accumulation in DA neurons of the substantia nigra in PD was unknown. In this study, our results showed that NRs inhibitors MK-801 and AP5 protected nigrostriatal projection system and reduced nigral iron levels of 6-hydroxydopamine (6-OHDA)-induced PD rats. In vitro studies demonstrated that NMDA treatment increased the expression of iron importer divalent metal transporter 1 (DMT1) and decreased the expression of iron exporter ferropotin 1 (Fpn1), which were dependent on iron regulatory protein 1 (IRP1). This led to increased intracellular iron levels and intensified the decrease in mitochondrial transmembrane potential in MES23.5 dopaminergic neurons. In addition, we reported that MK801 and neuronal nitric oxide synthase inhibitor could antagonize 6-OHDA-induced up-regulation of IRP1 and DMT1 and down-regulation of Fpn1, thus attenuating 6-OHDA-induced iron accumulation in MES23.5 cells. This suggested that 6-OHDA-induced activation of NRs might modulate the expression of DMT1 and Fpn1 via the neuronal nitric oxide synthase-IRP1 pathway.-Xu, H., Liu, X., Xia, J., Yu, T., Qu, Y., Jiang, H., Xie, J., Activation of NMDA receptors mediated iron accumulation via modulating iron transporters in Parkinson's disease.

18.
Sheng Li Xue Bao ; 71(3): 439-453, 2019 Jun 25.
Artigo em Chinês | MEDLINE | ID: mdl-31218335

RESUMO

Exosomes are extracellular membranous vesicles with a diameter of 30-100 nm derived from a variety of eukaryocytes. The cargo of exosomes includes proteins, lipids, nucleic acids, and substances of the cells from which they originate. They can transfer functional cargo to neighboring and distal cells, therefore contributing to intercellular communication in both physiological and pathological processes. In recent years, it was shown that exosomes in several neurodegenerative diseases are closely related to the transmission of disease-related misfolded proteins (such as α-synuclein, tau, amyloid ß-protein, etc). These proteins are transported by exosomes, thus promoting the propagation to unaffected cells or areas and accelerating the progression of neurodegenerative diseases. This review focuses on the origin and composition, biological synthesis, secretion, function of exosomes, as well as their roles in the pathogenesis and progression of neurodegenerative diseases. In addition, we also discuss that exosomes can serve as biomarkers and drug delivery vehicles, and play a role in the diagnosis and treatment of neurodegenerative diseases.


Assuntos
Exossomos/patologia , Doenças Neurodegenerativas/patologia , Peptídeos beta-Amiloides , Biomarcadores , Comunicação Celular , Humanos , alfa-Sinucleína , Proteínas tau
19.
Biochim Biophys Acta Mol Basis Dis ; 1864(3): 967-973, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29317336

RESUMO

Understandings of the disturbed iron metabolism in Parkinson's disease (PD) are largely from the perspectives of neurons. Neurodegenerative processes in PD trigger universal and conserved astroglial dysfunction and microglial activation. In this review, we start with astroglia and microglia in PD with an emphasis on their roles in spreading α-synuclein pathology, and then focus on their contributions in iron metabolism under normal conditions and the diseased state of PD. Elevated iron in the brain regions affects glial features, meanwhile, glial effects on neuronal iron metabolism are largely dependent on their releasing factors. These advances might be valuable for better understanding and modulating iron metabolism disturbance in PD.


Assuntos
Astrócitos/fisiologia , Distúrbios do Metabolismo do Ferro/etiologia , Ferro/metabolismo , Microglia/fisiologia , Doença de Parkinson/complicações , Doença de Parkinson/metabolismo , Animais , Astrócitos/patologia , Humanos , Distúrbios do Metabolismo do Ferro/metabolismo , Microglia/patologia
20.
Sheng Li Xue Bao ; 70(3): 294-300, 2018 Jun 25.
Artigo em Chinês | MEDLINE | ID: mdl-29926071

RESUMO

Parkinson's disease (PD) is a common neurodegenerative disease characterized by the degeneration of dopaminergic neurons in the substantia nigra and the intraneuronal Lewy bodies in this area. Genetic mutations in PD pathogenesis have been explored and better understood in recent years. GBA variants are now considered to be the single largest risk factor for PD. Gaucher disease (GD) is a lysosomal storage disorder disease and an inherited deficiency of lysosomal glucocerebrosidase (GCase) arising from mutations in the gene GBA. A group of patients with GD exhibit parkinsonian symptoms, meanwhile, GBA mutations are more frequently observed in patients with PD. These lines of evidence suggest a close relationship between GBA mutations and PD. GBA mutations are associated with an earlier onset age and a distinct cognitive decline in PD. GCase loss-of-function caused by GBA mutations interferes with the degradation of α-synuclein, and α-synuclein pathology in turn inhibits normal GCase function in PD, which forms a vicious cycle. However, the exact mechanisms for this bidirectional pathogenic loop have not to be fully elucidated. In this review, we summarize the current understandings on the potential link between GBA mutations and PD pathogenesis, which may show novel insights into PD etiology and therapeutics.


Assuntos
Glucosilceramidase/genética , Mutação , Doença de Parkinson/genética , Disfunção Cognitiva , Neurônios Dopaminérgicos/patologia , Doença de Gaucher/genética , Humanos , Transtornos Parkinsonianos/genética , Fatores de Risco , Substância Negra , alfa-Sinucleína
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa