Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Biomed Pharmacother ; 174: 116442, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38513596

RESUMEN

Parkinson's disease (PD) is a complex neurodegenerative disorder with an unclear etiology. Despite significant research efforts, developing disease-modifying treatments for PD remains a major unmet medical need. Notably, drug repositioning is becoming an increasingly attractive direction in drug discovery, and computational approaches offer a relatively quick and resource-saving method for identifying testable hypotheses that promote drug repositioning. We used an artificial intelligence (AI)-based drug repositioning strategy to screen an extensive compound library and identify potential therapeutic agents for PD. Our AI-driven analysis revealed that efavirenz and nevirapine, approved for treating human immunodeficiency virus infection, had distinct profiles, suggesting their potential effects on PD pathophysiology. Among these, efavirenz attenuated α-synuclein (α-syn) propagation and associated neuroinflammation in the brain of preformed α-syn fibrils-injected A53T α-syn Tg mice and α-syn propagation and associated behavioral changes in the C. elegans BiFC model. Through in-depth molecular investigations, we found that efavirenz can modulate cholesterol metabolism and mitigate α-syn propagation, a key pathological feature implicated in PD progression by regulating CYP46A1. This study opens new avenues for further investigation into the mechanisms underlying PD pathology and the exploration of additional drug candidates using advanced computational methodologies.


Asunto(s)
Alquinos , Inteligencia Artificial , Benzoxazinas , Ciclopropanos , Reposicionamiento de Medicamentos , Enfermedad de Parkinson , alfa-Sinucleína , Ciclopropanos/farmacología , Ciclopropanos/uso terapéutico , Alquinos/farmacología , Benzoxazinas/farmacología , Reposicionamiento de Medicamentos/métodos , Animales , alfa-Sinucleína/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Ratones , Caenorhabditis elegans/efectos de los fármacos , Ratones Transgénicos , Humanos , Nevirapina/farmacología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
2.
medRxiv ; 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-37398476

RESUMEN

Neuroinflammation through enhanced innate immunity is thought play a role in the pathogenesis of Parkinson's disease (PD). Methods for monitoring neuroinflammation in living patients with PD are currently limited to positron emission tomography (PET) ligands that lack specificity in labeling immune cells in the nervous system. The colony stimulating factor 1 receptor (CSF1R) plays a crucial role in microglial function, an important cellular contributor to the nervous system's innate immune response. Using immunologic methods, we show that CSF1R in human brain is colocalized with the microglial marker, ionized calcium binding adaptor molecule 1 (Iba1). In PD, CSF1R immunoreactivity is significantly increased in PD across multiple brain regions, with the largest differences in the midbrain versus controls. Autoradiography revealed significantly increased [3H]JHU11761 binding in the inferior parietal cortex of PD patients. PET imaging demonstrated that higher [11C]CPPC binding in the striatum was associated with greater motor disability in PD. Furthermore, increased [11C]CPPC binding in various regions correlated with more severe motor disability and poorer verbal fluency. This study finds that CSF1R expression is elevated in PD and that [11C]CPPC-PET imaging of CSF1R is indicative of motor and cognitive impairments in the early stages of the disease. Moreover, the study underscores the significance of CSF1R as a promising biomarker for neuroinflammation in Parkinson's disease, suggesting its potential use for non-invasive assessment of disease progression and severity, leading to earlier diagnosis and targeted interventions.

3.
Proc Natl Acad Sci U S A ; 119(36): e2204835119, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-36044549

RESUMEN

Physical activity provides clinical benefit in Parkinson's disease (PD). Irisin is an exercise-induced polypeptide secreted by skeletal muscle that crosses the blood-brain barrier and mediates certain effects of exercise. Here, we show that irisin prevents pathologic α-synuclein (α-syn)-induced neurodegeneration in the α-syn preformed fibril (PFF) mouse model of sporadic PD. Intravenous delivery of irisin via viral vectors following the stereotaxic intrastriatal injection of α-syn PFF cause a reduction in the formation of pathologic α-syn and prevented the loss of dopamine neurons and lowering of striatal dopamine. Irisin also substantially reduced the α-syn PFF-induced motor deficits as assessed behaviorally by the pole and grip strength test. Recombinant sustained irisin treatment of primary cortical neurons attenuated α-syn PFF toxicity by reducing the formation of phosphorylated serine 129 of α-syn and neuronal cell death. Tandem mass spectrometry and biochemical analysis revealed that irisin reduced pathologic α-syn by enhancing endolysosomal degradation of pathologic α-syn. Our findings highlight the potential for therapeutic disease modification of irisin in PD.


Asunto(s)
Cuerpo Estriado , Fibronectinas , Enfermedad de Parkinson , alfa-Sinucleína , Animales , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Fibronectinas/administración & dosificación , Fibronectinas/genética , Fibronectinas/metabolismo , Ratones , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/terapia , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(29): e2110746119, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35858361

RESUMEN

Intraneuronal inclusions of misfolded α-synuclein (α-syn) and prion-like spread of the pathologic α-syn contribute to progressive neuronal death in Parkinson's disease (PD). Despite the pathologic significance, no efficient therapeutic intervention targeting α-synucleinopathy has been developed. In this study, we provide evidence that astrocytes, especially those cultured from the ventral midbrain (VM), show therapeutic potential to alleviate α-syn pathology in multiple in vitro and in vivo α-synucleinopathic models. Regulation of neuronal α-syn proteostasis underlies the therapeutic function of astrocytes. Specifically, VM-derived astrocytes inhibited neuronal α-syn aggregation and transmission in a paracrine manner by correcting not only intraneuronal oxidative and mitochondrial stresses but also extracellular inflammatory environments, in which α-syn proteins are prone to pathologic misfolding. The astrocyte-derived paracrine factors also promoted disassembly of extracellular α-syn aggregates. In addition to the aggregated form of α-syn, VM astrocytes reduced total α-syn protein loads both by actively scavenging extracellular α-syn fibrils and by a paracrine stimulation of neuronal autophagic clearance of α-syn. Transplantation of VM astrocytes into the midbrain of PD model mice alleviated α-syn pathology and protected the midbrain dopamine neurons from neurodegeneration. We further showed that cografting of VM astrocytes could be exploited in stem cell-based therapy for PD, in which host-to-graft transmission of α-syn pathology remains a critical concern for long-term cell therapeutic effects.


Asunto(s)
Astrocitos , Trasplante de Tejido Encefálico , Enfermedad de Parkinson , Proteostasis , alfa-Sinucleína , Animales , Astrocitos/trasplante , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Mesencéfalo/patología , Mesencéfalo/cirugía , Ratones , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/terapia , alfa-Sinucleína/metabolismo
5.
Mol Brain ; 14(1): 122, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34321069

RESUMEN

Parkinson's disease (PD) is the second most prevalent neurodegenerative disease, with aging being considered the greatest risk factor for developing PD. Caveolin-1 (Cav-1) is known to participate in the aging process. Recent evidence indicates that prion-like propagation of misfolded α-synuclein (α-syn) released from neurons to neighboring neurons plays an important role in PD progression. In the present study, we demonstrated that cav-1 expression in the brain increased with age, and considerably increased in the brain of A53T α-syn transgenic mice. Cav-1 overexpression facilitated the uptake of α-syn into neurons and formation of additional Lewy body-like inclusion bodies, phosphorylation of cav-1 at tyrosine 14 was found to be crucial for this process. This study demonstrates the relationship between age and α-syn spread and will facilitate our understanding of the molecular mechanism of the cell-to-cell transmission of α-syn.


Asunto(s)
Envejecimiento/metabolismo , Caveolina 1/metabolismo , Neuronas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Encéfalo/patología , Línea Celular Tumoral , Células Cultivadas , Endocitosis , Humanos , Cuerpos de Inclusión/metabolismo , Cuerpos de Lewy/metabolismo , Masculino , Microdominios de Membrana , Ratones Endogámicos C57BL , Modelos Biológicos , Fosforilación , Fosfotirosina/metabolismo , Proteolisis , Ratas Sprague-Dawley
6.
FEBS J ; 288(23): 6593-6602, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33332736

RESUMEN

The pathogenesis of Parkinson's disease (PD), which is a progressive neurodegenerative disease, is associated with the formation of protein inclusion bodies called Lewy bodies (LB) or Lewy neurites (LN). α-Synuclein (α-Syn) is a major component of LB and LN. The formation of LB or LN is mediated by formation of α-Syn fibrils, which are formed from α-Syn monomers and oligomers. Additionally, intercellular α-Syn propagation has been proposed to be important for the progression of PD. Thus, various studies have focused on elucidating the role of α-Syn propagation in the pathogenesis of PD. Previous studies have reported that α-Syn species are released from the cells through various pathways, including the unconventional secretion pathways. The released α-Syn species are internalized by the cells through multiple mechanisms, including receptor-mediated endocytosis. Some molecular processes involved in intercellular α-Syn propagation have been recently elucidated. This review discusses the current studies on the molecular mechanisms underlying the release and uptake of α-Syn and their physiological relevance.


Asunto(s)
Astrocitos/metabolismo , Cuerpos de Lewy/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Animales , Astrocitos/citología , Exosomas/metabolismo , Humanos , Microglía/citología , Neuronas/citología , Enfermedad de Parkinson/patología , Transporte de Proteínas
8.
EMBO Rep ; 21(7): e48950, 2020 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-32372484

RESUMEN

Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons located in the substantia nigra pars compacta and the presence of proteinaceous inclusions called Lewy bodies and Lewy neurites in numerous brain regions. Increasing evidence indicates that Lewy pathology progressively involves additional regions of the nervous system as the disease advances, and the prion-like propagation of α-synuclein (α-syn) pathology promotes PD progression. Accordingly, the modulation of α-syn transmission may be important for the development of disease-modifying therapies in patients with PD. Here, we demonstrate that α-syn fibrils induce c-src activation in neurons, which depends on the FcγRIIb-SHP-1/-2-c-src pathway and enhances signals for the uptake of α-syn into neurons. Blockade of c-src activation inhibits the uptake of α-syn and the formation of Lewy body-like inclusions. Furthermore, the blockade of c-src activation also inhibits the release of α-syn via activation of autophagy. The brain-permeable c-src inhibitor, saracatinib, efficiently reduces α-syn propagation into neighboring regions in an in vivo model system. These results suggest a new therapeutic target against progressive PD.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Encéfalo/metabolismo , Neuronas Dopaminérgicas/metabolismo , Humanos , Cuerpos de Lewy/metabolismo , Enfermedad de Parkinson/genética , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
9.
Cell Rep ; 22(1): 136-148, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29298416

RESUMEN

Recent evidence of prion-like propagation of α-synuclein (α-syn) into neighboring neurons set up a paradigm to elucidate the mechanism of progression of Parkinson's disease (PD) and to develop therapeutic strategies. Here, we show that FcγRIIB expressed in neurons functions as a receptor for α-syn fibrils and mediates cell-to-cell transmission of α-syn. SHP-1 and 2 are activated downstream by α-syn fibrils through FcγRIIB and play an important role in cell-to-cell transmission of α-syn. Also, taking advantage of a co-culture system, we show that cell-to-cell transmission of α-syn induces intracellular Lewy body-like inclusion body formation and that the FcγRIIB/SHP-1/2 signaling pathway is involved in it. Therefore, the FcγRIIB-SHP-1/-2 signaling pathway may be a therapeutic target for the progression of PD. The in vitro system is an efficient tool for further high-throughput screening that can be used for developing a therapeutic intervention in PD.


Asunto(s)
Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Priones , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Receptores de IgG/metabolismo , Transducción de Señal , alfa-Sinucleína/metabolismo , Línea Celular , Humanos , Neuronas/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Transporte de Proteínas/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Receptores de IgG/genética , alfa-Sinucleína/genética
10.
Sci Rep ; 6: 28823, 2016 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-27346864

RESUMEN

Parkinson's disease (PD) is a common chronic and progressive neurodegenerative disorder. Although the cause of PD is still poorly understood, mutations in many genes including SNCA, parkin, PINK1, LRRK2, and DJ-1 have been identified in the familial forms of PD. It was recently proposed that alterations in lipid rafts may cause the neurodegeneration shown in PD. Here, we observe that DJ-1 deficiency decreased the expression of flotillin-1 (flot-1) and caveolin-1 (cav-1), the main protein components of lipid rafts, in primary astrocytes and MEF cells. As a mechanism, DJ-1 regulated flot-1 stability by direct interaction, however, decreased cav-1 expression may not be a direct effect of DJ-1, but rather as a result of decreased flot-1 expression. Dysregulation of flot-1 and cav-1 by DJ-1 deficiency caused an alteration in the cellular cholesterol level, membrane fluidity, and alteration in lipid rafts-dependent endocytosis. Moreover, DJ-1 deficiency impaired glutamate uptake into astrocytes, a major function of astrocytes in the maintenance of CNS homeostasis, by altering EAAT2 expression. This study will be helpful to understand the role of DJ-1 in the pathogenesis of PD, and the modulation of lipid rafts through the regulation of flot-1 or cav-1 may be a novel therapeutic target for PD.


Asunto(s)
Astrocitos/metabolismo , Caveolina 1/metabolismo , Ácido Glutámico/metabolismo , Proteínas de la Membrana/metabolismo , Proteína Desglicasa DJ-1/deficiencia , Proteína Desglicasa DJ-1/genética , Animales , Encéfalo/patología , Sistema Nervioso Central , Colesterol/metabolismo , Endocitosis , Transportador 2 de Aminoácidos Excitadores/metabolismo , Regulación de la Expresión Génica , Ácido Glutámico/farmacocinética , Homeostasis , Microdominios de Membrana , Ratones , Ratones Noqueados , Mutación , Unión Proteica
11.
Mol Neurodegener ; 10: 63, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26627850

RESUMEN

BACKGROUND: Parkinson's disease (PD) is characterized by progressive loss of midbrain dopaminergic neurons, resulting in motor dysfunctions. While most PD is sporadic in nature, a significant subset can be linked to either autosomal dominant or recessive mutations. PARK2, encoding the E3 ubiquitin ligase, parkin, is the most frequently mutated gene in autosomal recessive early onset PD. It has recently been reported that PD-associated gene products such as PINK1, α-synuclein, LRRK2, and DJ-1, as well as parkin associate with lipid rafts, suggesting that the dysfunction of these proteins in lipid rafts may be a causal factor of PD. Therefore here, we examined the relationship between lipid rafts-related proteins and parkin. RESULTS: We identified caveolin-1 (cav-1), which is one of the major constituents of lipid rafts at the plasma membrane, as a substrate of parkin. Loss of parkin function was found to disrupt the ubiquitination and degradation of cav-1, resulting in elevated cav-1 protein level in cells. Moreover, the total cholesterol level and membrane fluidity was altered by parkin deficiency, causing dysregulation of lipid rafts-dependent endocytosis. Further, cell-to-cell transmission of α-synuclein was facilitated by parkin deficiency. CONCLUSIONS: Our results demonstrate that alterations in lipid rafts by the loss of parkin via cav-1 may be a causal factor of PD, and cav-1 may be a novel therapeutic target for PD.


Asunto(s)
Caveolina 1/genética , Endocitosis/fisiología , Microdominios de Membrana/metabolismo , Mutación/genética , Enfermedad de Parkinson/metabolismo , Ubiquitina-Proteína Ligasas/genética , Animales , Caveolina 1/metabolismo , Neuronas Dopaminérgicas/metabolismo , Ratones , Enfermedad de Parkinson/genética , alfa-Sinucleína/metabolismo
12.
Neurobiol Dis ; 83: 90-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26342897

RESUMEN

Parkinson's disease (PD) is the second most prevalent neurodegenerative disease. Although the etiology of PD has not yet been fully understood, accumulating evidence indicates that neuroinflammation plays a critical role in the progression of PD. α-Synuclein (α-Syn) has been considered to be a key player of the pathogenesis of PD, and recent reports that prion-like propagation of misfolded α-syn released from neurons may play an important role in the progression of PD have led to increased attention to the studies elucidating the roles of extracellular α-syn in the CNS. Extracellular α-syn has also been reported to regulate microglial inflammatory response. In this study, we demonstrated that aggregated α-syn inhibited microglial phagocytosis by activating SHP-1. SHP-1 activation was also observed in A53T α-syn transgenic mice. In addition, aggregated α-syn bound to FcγRIIB on microglia, inducing SHP-1 activation, further inhibiting microglial phagocytosis. Aggregated α-syn upregulated FcγRIIB expression in microglia and upregulated FcγRIIB was also observed in A53T α-syn transgenic mice. These data suggest that aggregated α-syn released from neurons dysregulates microglial immune response through inhibiting microglial phagocytosis, further causing neurodegeneration observed in PD. The interaction of aggregated α-syn and FcγRIIB and further SHP-1 activation can be a new therapeutic target against PD.


Asunto(s)
Encéfalo/metabolismo , Microglía/metabolismo , Microglía/fisiología , Fagocitosis , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Receptores de IgG/metabolismo , alfa-Sinucleína/metabolismo , Animales , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Agregado de Proteínas/fisiología , Ratas , Ratas Sprague-Dawley
13.
J Biol Chem ; 287(30): 24862-72, 2012 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-22619171

RESUMEN

Parkinson disease (PD) is the second most common neurodegenerative disease characterized by a progressive dopaminergic neuronal loss in association with Lewy body inclusions. Gathering evidence indicates that α-synuclein (α-syn), a major component of the Lewy body, plays an important role in the pathogenesis of PD. Although α-syn is considered to be a cytoplasmic protein, it has been detected in extracellular biological fluids, including human cerebrospinal fluid and blood plasma of healthy and diseased individuals. In addition, a prion-like spread of α-syn aggregates has been recently proposed to contribute to the propagation of Lewy bodies throughout the nervous system during progression of PD, suggesting that the metabolism of extracellular α-syn might play a key role in the pathogenesis of PD. In the present study, we found that plasmin cleaved and degraded extracellular α-syn specifically in a dose- and time- dependent manner. Aggregated forms of α-syn as well as monomeric α-syn were also cleaved by plasmin. Plasmin cleaved mainly the N-terminal region of α-syn and also inhibited the translocation of extracellular α-syn into the neighboring cells in addition to the activation of microglia and astrocytes by extracellular α-syn. Further, extracellular α-syn regulated the plasmin system through up-regulation of plasminogen activator inhibitor-1 (PAI-1) expression. These findings help to understand the molecular mechanism of PD and develop new therapeutic targets for PD.


Asunto(s)
Astrocitos/metabolismo , Fibrinolisina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/metabolismo , Enfermedad de Parkinson/metabolismo , Proteolisis , alfa-Sinucleína/metabolismo , Animales , Astrocitos/patología , Línea Celular , Fibrinolisina/genética , Humanos , Cuerpos de Lewy/genética , Cuerpos de Lewy/metabolismo , Cuerpos de Lewy/patología , Proteínas del Tejido Nervioso/genética , Neuroglía/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Inhibidor 1 de Activador Plasminogénico/biosíntesis , Inhibidor 1 de Activador Plasminogénico/genética , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/genética , alfa-Sinucleína/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA