Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cell Mol Neurobiol ; 42(5): 1321-1339, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33528716

RESUMEN

Parkinson's disease (PD) is a severe neurodegenerative disorder caused by the progressive loss of dopaminergic neurons in the substantia nigra and affects millions of people. Currently, mitochondrial dysfunction is considered as a central role in the pathogenesis of both sporadic and familial forms of PD. Mitophagy, a process that selectively targets damaged or redundant mitochondria to the lysosome for elimination via the autophagy devices, is crucial in preserving mitochondrial health. So far, aberrant mitophagy has been observed in the postmortem of PD patients and genetic or toxin-induced models of PD. Except for mitochondrial dysfunction, mitophagy is involved in regulating several other PD-related pathological mechanisms as well, e.g., oxidative stress and calcium imbalance. So far, the mitophagy mechanisms induced by PD-related proteins, PINK1 and Parkin, have been studied widely, and several other PD-associated genes, e.g., DJ-1, LRRK2, and alpha-synuclein, have been discovered to participate in the regulation of mitophagy as well, which further strengthens the link between mitophagy and PD. Thus, in this view, we reviewed mitophagy pathways in belief and discussed the interactions between mitophagy and several PD's pathological mechanisms and how PD-related genes modulate the mitophagy process.


Asunto(s)
Mitofagia , Enfermedad de Parkinson , Autofagia , Humanos , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Enfermedad de Parkinson/metabolismo
2.
Neurochem Res ; 47(6): 1721-1735, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35229270

RESUMEN

Parkinson's disease (PD) is the second most common neurodegenerative disorder. Progressive loss of dopaminergic neurons in the substantia nigra (SN) is one of the major pathological changes. However, the reasons for the dopaminergic neuron loss are still ambiguous and further studies are needed to evaluate the in-depth mechanisms of neuron death. Oxidative stress is a significant factor causing neuronal damage. Dopaminergic neurons in the SN are susceptible to oxidative stress, which is closely associated with iron dyshomeostasis in the brain. Ginsenoside Rg1 from ginseng plays a crucial role in neuroprotective effects through anti-inflammation and attenuating the aggregation of abnormal α-synuclein. In our study, we established a chronic PD mouse model by 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine combined with probenecid and explored the effect of Rg1 on the oxidative stress and brain iron homeostasis. Rg1 was verified to improve the level of tyrosine hydroxylase and anti-oxidant stress. In addition, Rg1 maintained the iron-regulated protein homeostasis by increasing the expression of ferritin heavy chain and decreasing ferritin light chain in oligodendrocytes, especially the mature oligodendrocytes (OLs). Furthermore, Rg1 had a positive effect on the myelin sheath protection and increased the number of mature oligodendrocytes, proved by the increased staining of myelin basic protein and CC-1. In conclusion, Rg1 could play a neuroprotective role through remitting the iron-regulated protein dyshomeostasis by ferritin and against lipid peroxidation stress in oligodendrocytes.


Asunto(s)
Ginsenósidos , Fármacos Neuroprotectores , Enfermedad de Parkinson , Animales , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas , Ginsenósidos/farmacología , Ginsenósidos/uso terapéutico , Hierro/metabolismo , Peroxidación de Lípido , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Oligodendroglía/metabolismo
3.
Neurochem Res ; 47(12): 3627-3634, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35348944

RESUMEN

Parkinson's disease (PD) is a chronic neurodegenerative disease mainly characterized by movement disorders and other non-motor symptoms, including the loss of dopaminergic neurons in the substantia nigra parts. Abnormal α-synuclein aggregation in the brain is closely associated with the loss of dopaminergic neurons. α-synuclein can propagate in the central nervous system (CNS) and periphery under pathological conditions. Many researches have focused on its aggregation and distribution in the CNS and explored its relationship with PD. But in recent years, the distribution of α-synuclein in peripheral tissues have been paid much attention. This review summarized the distribution of α-synuclein in the choroid plexus, blood, saliva, gastrointestine and other tissues, and discussed the potential mechanism of α-synuclein aggregation, providing a basis for the early diagnosis and intervention of PD.


Asunto(s)
Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Humanos , alfa-Sinucleína/metabolismo , Enfermedades Neurodegenerativas/patología , Enfermedad de Parkinson/patología , Sustancia Negra/metabolismo , Neuronas Dopaminérgicas/metabolismo
4.
Acta Pharmacol Sin ; 43(9): 2253-2266, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35132190

RESUMEN

Neuroinflammation is closely related to the pathogenesis of neurodegenerative diseases. Activation of microglia, the resident immune cells in CNS, induces inflammatory responses, resulting in the release of neurotoxic molecules, which favors neuronal death and neurodegeneration. Nuclear receptor-related 1 (Nurr1) protein, one of the orphan nuclear receptor superfamilies, is an emerging target for neuroprotective therapy. In addition, the anti-inflammatory function of cannabinoid (CB) receptors has attracted increasing interest. As both CB receptors (especially CB2 receptor) and Nurr1 exist in microglia, and regulate a number of same molecular points such as NF-κB, we herein explored the interplay between the CB2 receptor and Nurr1 as well as the regulatory mechanisms in microglial cells. We showed that the application of CB2 receptor agonists JWH015 (1, 10 µM) significantly increased the nuclear Nurr1 protein in BV-2 cells and primary midbrain microglia. Overexpression of Nurr1 or application of Nurr1 agonist C-DIM12 (10 µM) significantly increased the mRNA level of CB2 receptor in BV-2 cells, suggesting that positive expression feedback existing between the CB2 receptor and Nurr1. After 2-AG and JWH015 activated the CB2 receptors, the levels of p-ERK, p-AKT, p-GSK-3ß in BV-2 cells were significantly increased. Using ERK1/2 inhibitor U0126 and PI3K/AKT inhibitor LY294002, we revealed that the amount of Nurr1 in the nucleus was upregulated through ß-arrestin2/ERK1/2 and PI3K/AKT/GSK-3ß signaling pathways. With these inhibitors, we found a cross-talk interaction between the two pathways, and the ERK1/2 signaling pathway played a more dominant regulatory role. Furthermore, we demonstrated that when the CB2 receptor was activated, the phagocytic function of BV-2 cells was significantly weakened; the activation of Nurr1 also inhibited the phagocytic function of BV-2 cells. Pretreatment with the signaling pathway inhibitors, especially U0126, reversed the inhibitory effect of 2-AG on phagocytosis, suggesting that CB2 receptor may regulate the phagocytic function of microglia by activating Nurr1. In conclusion, CB2 receptor or/and Nurr1-mediated signal pathways play instrumental roles in the progress of phagocytosis, which are expected to open up new treatment strategies for neurodegenerative diseases.


Asunto(s)
Microglía , Proteínas Proto-Oncogénicas c-akt , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Lipopolisacáridos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor Cannabinoide CB2/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal
5.
Molecules ; 27(21)2022 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-36364248

RESUMEN

Accumulating evidence has shown that Parkinson's disease (PD) is a systemic disease other than a mere central nervous system (CNS) disorder. One of the most important peripheral symptoms is gastrointestinal dysfunction. The enteric nervous system (ENS) is regarded as an essential gateway to the environment. The discovery of the prion-like behavior of α-synuclein makes it possible for the neurodegenerative process to start in the ENS and spread via the gut-brain axis to the CNS. We first confirmed that synucleinopathies existed in the stomachs of chronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)/probenecid (MPTP/p)-induced PD mice, as indicated by the significant increase in abnormal aggregated and nitrated α-synuclein in the TH-positive neurons and enteric glial cells (EGCs) of the gastric myenteric plexus. Next, we attempted to clarify the mechanisms in single MPTP-injected mice. The stomach naturally possesses high monoamine oxidase-B (MAO-B) activity and low superoxide dismutase (SOD) activity, making the stomach susceptible to MPTP-induced oxidative stress, as indicated by the significant increase in reactive oxygen species (ROS) in the stomach and elevated 4-hydroxynonenal (4-HNE) in the EGCs after MPTP exposure for 3 h. Additionally, stomach synucleinopathies appear before those of the nigrostriatal system, as determined by Western blotting 12 h after MPTP injection. Notably, nitrated α-synuclein was considerably increased in the EGCs after 3 h and 12 h of MPTP exposure. Taken together, our work demonstrated that the EGCs could be new contributors to synucleinopathies in the stomach. The early-initiated synucleinopathies might further influence neighboring neurons in the myenteric plexus and the CNS. Our results offer a new experimental clue for interpreting the etiology of PD.


Asunto(s)
Intoxicación por MPTP , Enfermedad de Parkinson , Trastornos Parkinsonianos , Sinucleinopatías , Ratones , Animales , alfa-Sinucleína , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/efectos adversos , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Neuroglía , Estómago
6.
Eur J Neurosci ; 53(9): 2946-2959, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32031280

RESUMEN

The critical role of mitochondrial dysfunction in the pathological mechanisms of neurodegenerative disorders, particularly Parkinson's disease (PD), is well established. Compelling evidence indicates that Parkinson's proteins (e.g., α-synuclein, Parkin, PINK1, DJ-1, and LRRK2) are associated with mitochondrial dysfunction and oxidative stress in PD. Significantly, there is a possible central role of alpha-synuclein (α-Syn) in the occurrence of mitochondrial dysfunction and oxidative stress by the mediation of different signaling pathways. Also, tau, traditionally considered as the main component of neurofibrillary tangles, aggregates and amplifies the neurotoxic effects on mitochondria by interacting with α-Syn. Moreover, oxidative stress caused by mitochondrial dysfunction favors assembly of both α-Syn and tau and also plays a key role in the formation of protein aggregates. In this review, we provide an overview of the relationship between these two pathological proteins and mitochondrial dysfunction in PD, and also summarize the underlying mechanisms in the interplay of α-Syn aggregation and phosphorylated tau targeting the mitochondria, to find new strategies to prevent PD processing.


Asunto(s)
Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Humanos , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Estrés Oxidativo , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo
7.
Cell Mol Neurobiol ; 41(7): 1395-1411, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32623547

RESUMEN

Parkinson's disease (PD), as one of the complex neurodegenerative disorders, affects millions of aged people. Although the precise pathogenesis remains mostly unknown, a significant number of studies have demonstrated that mitochondrial dysfunction acts as a major role in the pathogeny of PD. Both nuclear and mitochondrial DNA mutations can damage mitochondrial integrity. Especially, mutations in several genes that PD-linked have a closed association with mitochondrial dysfunction (e.g., Parkin, PINK1, DJ-1, alpha-synuclein, and LRRK2). Parkin, whose mutation causes autosomal-recessive juvenile parkinsonism, plays an essential role in mitochondrial quality control of mitochondrial biogenesis, mitochondrial dynamics, and mitophagy. Therefore, we summarized the advanced studies of Parkin's role in mitochondrial quality control and hoped it could be studied further as a therapeutic target for PD.


Asunto(s)
Mitocondrias/metabolismo , Enfermedad de Parkinson/metabolismo , Trastornos Parkinsonianos/genética , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Humanos , Mitocondrias/patología , Mitofagia/genética , Mitofagia/fisiología , Mutación/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Trastornos Parkinsonianos/metabolismo , Ubiquitina-Proteína Ligasas/genética
8.
Pharmacol Res ; 170: 105541, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33711434

RESUMEN

Diversiform ways of intercellular communication are vital links in maintaining homeostasis and disseminating physiological states. Among intercellular bridges, tunneling nanotubes (TNTs) discovered in 2004 were recognized as potential pharmacology targets related to the pathogenesis of common or infrequent neurodegenerative disorders. The neurotoxic aggregates in neurodegenerative diseases including scrapie prion protein (PrPSc), mutant tau protein, amyloid-beta (Aß) protein, alpha-synuclein (α-syn) as well as mutant Huntington (mHTT) protein could promote TNT formation via certain physiological mechanisms, in turn, mediating the intercellular transmission of neurotoxicity. In this review, we described in detail the skeleton, the formation, the physicochemical properties, and the functions of TNTs, while paying particular attention to the key role of TNTs in the transport of pathological proteins during neurodegeneration.


Asunto(s)
Comunicación Celular , Estructuras de la Membrana Celular/metabolismo , Degeneración Nerviosa , Enfermedades Neurodegenerativas/metabolismo , Animales , Estructuras de la Membrana Celular/efectos de los fármacos , Estructuras de la Membrana Celular/patología , Humanos , Nanotubos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/patología , Fármacos Neuroprotectores/uso terapéutico , Agregado de Proteínas , Agregación Patológica de Proteínas , Transporte de Proteínas
9.
Acta Pharmacol Sin ; 42(9): 1409-1421, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33214696

RESUMEN

Huntington's disease (HD) is one of main neurodegenerative diseases, characterized by striatal atrophy, involuntary movements, and motor incoordination. Ginsenoside Rg1 (Rg1), an active ingredient in ginseng, possesses a variety of neuroprotective effects with low toxicity and side effects. In this study, we investigated the potential therapeutic effects of Rg1 in a mouse model of HD and explored the underlying mechanisms. HD was induced in mice by injection of 3-nitropropionic acid (3-NP, i.p.) for 4 days. From the first day of 3-NP injection, the mice were administered Rg1 (10, 20, 40 mg·kg-1, p.o.) for 5 days. We showed that oral pretreatment with Rg1 alleviated 3-NP-induced body weight loss and behavioral defects. Furthermore, pretreatment with Rg1 ameliorated 3-NP-induced neuronal loss and ultrastructural morphological damage in the striatum. Moreover, pretreatment with Rg1 reduced 3-NP-induced apoptosis and inhibited the activation of microglia, inflammatory mediators in the striatum. We revealed that Rg1 exerted neuroprotective effects by suppressing 3-NP-induced activation of the MAPKs and NF-κΒ signaling pathways in the striatum. Thus, our results suggest that Rg1 exerts therapeutic effects on 3-NP-induced HD mouse model via suppressing MAPKs and NF-κΒ signaling pathways. Rg1 may be served as a novel therapeutic option for HD.


Asunto(s)
Cuerpo Estriado/metabolismo , Ginsenósidos/farmacología , Enfermedad de Huntington/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Fármacos Neuroprotectores/farmacología , Animales , Apoptosis , Cuerpo Estriado/patología , Cuerpo Estriado/ultraestructura , Enfermedad de Huntington/inducido químicamente , Enfermedad de Huntington/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Neuronas/efectos de los fármacos , Nitrocompuestos , Propionatos , Transducción de Señal/efectos de los fármacos
10.
Pharmacol Res ; 151: 104553, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31760107

RESUMEN

Parkinson's disease (PD) that afflicts millions of individuals worldwide is associated with deposits of aggregate-prone proteins (e.g., α-synuclein) and with mitochondrial dysfunction in neuronal cells. Mitochondria are the main source of reactive oxygen species, provide energy for neuronal cells, and are regarded as dynamic organelles that are determined by mitochondrial fission, fusion, and mitophagy to maintain mitochondrial homeostasis. Growing evidence reveals that several dynamics-related proteins, such as dynamin-related protein 1 (Drp1), mediate mitochondrial fission, fusion, and mitophagy, to protect against neurodegeneration in PD. More importantly, not only is Drp1-mediated fission required for mitophagy that exerts a protective effect on neurons, but abnormal mitochondrial fission and mitophagy can drive neuronal survival or cell death (i.e., autophagy, apoptosis, and necroptosis), suggesting that Drp1 may play a pivotal role in the pathogenesis of PD. Also, PD-related proteins such as α-synuclein, leucine-rich repeat kinase-2, PTEN-induced putative kinase 1, and Parkin have been proven to interact with Drp1, thus contributing to mitochondrial dynamics and clearance, as well as neuronal fate. Here, we review the roles of Drp1 in mitochondrial fission, dynamics, mitophagy, bulk autophagy, apoptosis, and necroptosis for a better understanding of mitochondrial disturbances in PD-associated neurodegeneration and summarize the advances of novel chemical compounds targeting Drp1 to provide new insight into potential PD therapies.


Asunto(s)
Dinaminas/metabolismo , Dinámicas Mitocondriales , Mitofagia , Neuronas/patología , Enfermedad de Parkinson/patología , Animales , Antiparkinsonianos/farmacología , Antiparkinsonianos/uso terapéutico , Muerte Celular/efectos de los fármacos , Descubrimiento de Drogas , Dinaminas/análisis , Humanos , Dinámicas Mitocondriales/efectos de los fármacos , Mitofagia/efectos de los fármacos , Terapia Molecular Dirigida , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo
11.
Bioorg Chem ; 97: 103659, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32078940

RESUMEN

Seven flavonoid dimers, biflavocochins A-G, together with six known compounds were isolated from the red resins of Dracaena cochinchinensis (Chinese dragon's blood). Their structures were elucidated based on extensive spectroscopic analysis. The absolute configurations of 1-7 was assigned by experimental and quantum chemical calculated ECD spectra, and that of 4 was further established by X-ray diffraction analysis using Cu Kα radiation. Compounds 1-3 are novel dimers of homoisoflavonoid and dihydrochalcone with a unique dibenzopyran ring. Compounds 2, 6, 7 exhibited moderate PTP1B inhibitory activities in an enzyme assay. Compound 1 showed neuroprotective effect on serum deficiency-induced cellular damage in PC12 cells.


Asunto(s)
Dracaena/química , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Fármacos Neuroprotectores/farmacología , Extractos Vegetales/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Animales , Cristalografía por Rayos X , Dimerización , Inhibidores Enzimáticos/química , Flavonoides/química , Humanos , Modelos Moleculares , Fármacos Neuroprotectores/química , Células PC12 , Extractos Vegetales/química , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Ratas
13.
Pharmacol Res ; 146: 104336, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31271846

RESUMEN

Alzheimer's disease (AD) and Parkinson's disease (PD) are recognized as the universal neurodegenerative diseases, with the involvement of misfolded proteins pathology, leading to oxidative stress, glial cells activation, neuroinflammation, mitochondrial dysfunction, and cellular apoptosis. Several discoveries indicate that accumulation of pathogenic proteins, i.e. amyloid ß (Aß), the microtubule-binding protein tau, and α-synuclein, are parallel with oxidative stress, neuroinflammation, and mitochondrial dysfunction. Whether the causative factors are misfolded proteins or these pathophysiological changes, leading to neurodegeneration still remain ambiguous. Importantly, directing pharmacological researches towards the prevention of AD and PD seem a promising approach to detect these complicating mechanisms, and provide new insight into therapy for AD and PD patients. Mangiferin (MGF, 2-C-ß-D-glucopyranosyl-1, 3, 6, 7-tetrahydroxyxanthone), well-known as a natural product, is detached from multiple plants, including Mangifera indica L. With the structure of C-glycosyl and phenolic moiety, MGF possesses multipotent properties starting from anti-oxidant effects, to the alleviation of mitochondrial dysfunction, neuroinflammation, and cellular apoptosis. In particular, MGF can cross the blood-brain barrier to exert neuronal protection. Different researches implicate that MGF is able to protect the central nervous system from oxidative stress, mitochondrial dysfunction, neuroinflammation, and apoptosis under in vitro and in vivo models. Additional facts support that MGF plays a role in improving the declined memory and cognition of rat models. Taken together, the neuroprotective capacity of MGF may stand out as an agent candidate for AD and PD therapy.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Xantonas/farmacología , Xantonas/uso terapéutico , Animales , Antioxidantes/fisiología , Antioxidantes/uso terapéutico , Humanos , Estrés Oxidativo/efectos de los fármacos
14.
Acta Pharmacol Sin ; 40(12): 1503-1512, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31388087

RESUMEN

Parkinson's disease (PD) is a multifactorial disorder characterized by progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) and the presence of Lewy bodies (LBs) consisting of misfolded α-synuclein protein. The etiology of PD is still not clear but systemic inflammation is proved to trigger and exacerbate DA neurons degeneration. Toll-like receptor 4 (TLR4) is a pattern-recognition receptor (PRR) and plays a major role in promoting the host immune. TLR4-mediated signal pathways induce the release of many inflammatory cytokines. It is reasonable to hypothesize that TLR4 is the mediator in microglia contributing to the damage of DA neurons in the SNpc. In this study, we evaluated the role of TLR4 in the chronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)/probenecid mouse model. Both TLR4-deficient and wild-type (WT) mice were injected with probenecid (250 mg/kg, i.p.) followed by injection of MPTP (25 mg/kg, s.c.) every 4 days for 10 times. From D43 to D47, the behavioral performance in pole test and wire hang test was assessed. Then the mice were euthanized, and SN and striatum were dissected out for biochemical tests. We showed that compared with MPTP-treated WT mice, TLR4 deficiency significantly attenuated MPTP-induced motor deficits and TH-protein expression reduction in SNpc and striatum, suppressed MPTP-induced α-synuclein abnormality and neuroinflammation mediated through oxidative stress, glial activation, NF-κB and the NLRP3 inflammasome signaling pathways. These findings highlight the neuroprotective effect of TLR4-pathways in the chronic MPTP-induced PD mouse model.


Asunto(s)
Enfermedad de Parkinson Secundaria/fisiopatología , Receptor Toll-Like 4/fisiología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Animales , Astrocitos/metabolismo , Astrocitos/patología , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Inflamasomas/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Microglía/patología , Subunidad p50 de NF-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/genética , Porción Compacta de la Sustancia Negra/metabolismo , Porción Compacta de la Sustancia Negra/patología , Probenecid , Transducción de Señal/fisiología , Receptor Toll-Like 4/genética , Tirosina 3-Monooxigenasa/metabolismo , alfa-Sinucleína/metabolismo
15.
Acta Pharmacol Sin ; 40(8): 991-998, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30728466

RESUMEN

Olfactory bulb, as one of sensory organs opening to the outside, is susceptible to toxic environment and easy to deteriorate. Recent studies in Parkinson's disease (PD) patients and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated monkeys have shown that abnormal α-synuclein is accumulated in the olfactory glomeruli, suggesting that the lesions of PD are not only confined to the substantia nigra (SN) but also located in the olfactory bulb. Thus, olfactory bulb might be the region of onset in PD pathogenesis and a targeted region for diagnosis and treatment of PD. However, the relationship between olfactory bulb and pathogenesis of PD remains unclear. In the present study, we investigated the inflammatory pathological alterations in olfactory bulb and the underlying mechanisms in chronic MPTP mice. Mice were treated with MPTP/P, i.e., MPTP (25 mg/kg, s.c.) plus probenecid (250 mg/kg, i.p.) every 4 days, for ten times. The mice displayed typical parkinsonian syndrome. Then we examined their olfactory function and the pathologic changes in olfactory bulb. The mice showed obvious olfactory dysfunction in a buried pellet test. Immunohistochemical studies revealed that tyrosine hydroxylase (TH) protein levels were significantly decreased, whereas abnormal α-synuclein was significantly increased in the olfactory bulbs. Furthermore, the olfactory bulbs in MPTP/P-treated mice showed significantly increased levels of interleukin-1ß (IL-1ß), caspase-1, glial fibrillary acidic protein (GFAP), Toll receptor 4 (TLR4), phosphorylation of p65, as well as activated molecules of NOD-like receptor protein 3 (NLRP3) that were associated with neuroinflammation. Our results demonstrate that MPTP/P-caused olfactory bulb damage might be related to NLRP3-mediated inflammation.


Asunto(s)
Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Bulbo Olfatorio/efectos de los fármacos , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Animales , Masculino , Ratones Endogámicos C57BL , Bulbo Olfatorio/patología , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/etiología , Probenecid/farmacología , Multimerización de Proteína/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , alfa-Sinucleína/metabolismo
17.
Molecules ; 23(11)2018 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-30388770

RESUMEN

Genetic studies have revealed that rare mutations and multiplications of the gene locus in α-synuclein (α-syn) are implicated in the pathogenesis of Parkinson's disease (PD). However, the pathological effects of α-syn are still obscure. The neurotoxicity of α-syn is mainly determined by its protein levels, which depend on a balance between synthesis and degradation. Therefore, verifying the possible routes contributing to the clearance of α-syn is important for PD therapy. In this study, we established stable lines overexpressing human wild-type (WT) and E46K mutant α-syn in rat PC12 cells and investigated the degradation pathways of α-syn by using a panel of inhibitors and inducers of lysosome and proteasome function. We also monitored the degradation kinetics of α-syn by using cycloheximide to block protein synthesis. Our data showed that both proteasome and chaperon-mediated autophagy (CMA) are responsible for the degradation of the WT α-syn. Meanwhile, E46K mutant α-syn is mainly degraded by the proteasome and macroautophagy pathway. Compared with the WT protein, E46K mutant α-syn turned over more slowly in PC12 cells. In addition, overexpression of E46K mutant α-syn increased vulnerability of PC12 cells to apoptosis insults when compared with WT α-syn. Our findings may verify the possible routes contributing to the degradation of the E46K mutant α-syn.


Asunto(s)
Autofagia , Mutación , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Animales , Apoptosis , Expresión Génica , Humanos , Células PC12 , Proteolisis , Ratas
18.
J Pharmacol Exp Ther ; 363(2): 284-292, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28912345

RESUMEN

The novel bibenzyl compound 2-[4-hydroxy-3-(4- hydroxyphenyl) benzyl]-4-(4- hydroxyphenyl) phenol (20C) plays a neuroprotective role in vitro, but its effects in vivo have not yet been elucidated. In this study, we estimated the efficacy of 20C in vivo using a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine/probenecid (MPTP/p) mouse model from behavior, dopamine, and neuron and then the possible mechanisms for these effects were further investigated. The experimental results showed that 20C improved behavioral deficits, attenuated dopamine depletion, reduced dopaminergic neuron loss, protected the blood-brain barrier (BBB) structure, ameliorated α-synuclein dysfunction, suppressed glial activation, and regulated both nuclear factor-κB (NF-κB) signaling and the NOD-like receptor protein (NLRP) 3 inflammasome pathway. Our results indicated that 20C may prevent neurodegeneration in the MPTP/p mouse model by targeting α-synuclein and regulating α-synuclein-related inflammatory responses, including BBB damage, glial activation, NF-κB signaling, and the NLRP3 inflammasome pathway.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Bibencilos/farmacología , Intoxicación por MPTP/prevención & control , Fármacos Neuroprotectores/farmacología , Fenoles/farmacología , Probenecid/toxicidad , Animales , Astrocitos/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Dopamina/metabolismo , Inflamación/metabolismo , Intoxicación por MPTP/metabolismo , Intoxicación por MPTP/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Transmisión Sináptica/efectos de los fármacos
19.
Acta Pharmacol Sin ; 38(10): 1317-1328, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28649132

RESUMEN

1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model remains the most commonly used animal model of Parkinson's disease (PD). There are three MPTP-treatment schemes: acute, subacute and chronic. Considering the advantages of the period and similarity to PD, the subacute model was often chosen to assess the validity of new candidates, but the changes caused by the subacute MPTP treatment and the appropriate positive control for this model remain to be further confirmed. The aim of this study was: to estimate the value of the subacute MPTP mouse model in aspects of behavioral performance, biochemical changes and pathological abnormalities, and to find effective positive drugs. Male C57BL/6 mice were injected with MPTP (30 mg·kg-1·d-1, ip) for 5 consecutive days. Three days before MPTP injection, the mice were orally administered selegiline (3 mg·kg-1·d-1), pramipexole (3 mg·kg-1·d-1), or medopar (100 mg·kg-1·d-1) for 18 days. Behavioral performance was assessed in the open field test, pole test and rotarod test. Neurotransmitters in the striatum were detected using HPLC. Protein levels were measured by Western blot. Pathological characteristics were examined by immunohistochemistry. Ultrastructure changes were observed by electron microscopy. The subacute MPTP treatment did not induce evident motor defects despite severe injuries in the dopaminergic system. Additionally, MPTP significantly increased the α-synuclein levels and the number of astrocytes in the striatum, and destroyed the blood-brain barrier (BBB) in the substantia nigra pars compacta. Both selegiline and pramipexole were able to protect the mice against MPTP injuries. We conclude that the subacute MPTP mouse model does not show visible motor defects; it is not enough to evaluate the validity of a candidate just based on behavioral examination, much attention should also be paid to the alterations in neurotransmitters, astrocytes, α-synuclein and the BBB. In addition, selegiline or pramipexole is a better choice than medopar as an effective positive control for the subacute MPTP model.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/administración & dosificación , Antiparkinsonianos/farmacología , Modelos Animales de Enfermedad , Trastornos Parkinsonianos/fisiopatología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Conducta Animal/efectos de los fármacos , Benserazida/farmacología , Benzotiazoles/farmacología , Barrera Hematoencefálica/metabolismo , Cromatografía Líquida de Alta Presión/métodos , Cuerpo Estriado/metabolismo , Combinación de Medicamentos , Levodopa/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Pramipexol , Selegilina/farmacología , alfa-Sinucleína/metabolismo
20.
Acta Pharmacol Sin ; 37(6): 731-40, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27180985

RESUMEN

AIM: Our preliminary study shows that a bibenzyl compound isolated from Gastrodia elata, 2-[4-hydroxy-3-(4-hydroxybenzyl)benzyl]-4-(4-hydroxybenzyl)phenol (designated 20C), protects PC12 cells against H2O2-induced injury. In this study we investigated whether 20C exerted neuroprotective action in a cell model of Parkinson's disease. METHODS: A cell model of Parkinson's disease was established in PC12 cells by exposure to rotenone (4 µmol/L) for 48 h. Cell viability and apoptosis were assessed, and intracellular ROS level and the mitochondrial membrane potential (MMP) were detected. The expression of apoptosis-related proteins Bax, Bcl-2, cytochrome c, cleaved caspase-3, and oxidative stress-related proteins Nrf2, HO-1 and NQO1 were examined using Western blotting. The mRNA levels of HO-1 and NQO1 were determined with RT-PCR. The nuclear translocation of Nrf2 was observed with immunofluorescence staining. RESULTS: Treatment with rotenone significantly increased the number of apoptotic cells, accompanied by marked increases in the Bax/Bcl-2 ratio, cytochrome c release and caspase-3 activation. Rotenone also increased ROS accumulation, reduced MMP, and increased the nuclear translocation of Nrf2 as well as the mRNA and protein levels of the Nrf2 downstream target genes HO-1 and NQO1 in PC12 cells. Co-treatment with 20C (0.01-1 µmol/L) dose-dependently attenuated rotenone-induced apoptosis and oxidative stress in PC12 cells. Nrf2 knockdown by siRNA partially reversed the protective effects of 20C in rotenone-treated PC12 cells. CONCLUSION: The bibenzyl compound 20C protects PC12 cells from rotenone-induced apoptosis, at least in part, via activation of the Nrf2/ARE/HO-1 signaling pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Bibencilos/farmacología , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson Secundaria/tratamiento farmacológico , Rotenona , Transducción de Señal/efectos de los fármacos , Animales , Elementos de Respuesta Antioxidante/efectos de los fármacos , Bibencilos/química , Gastrodia/química , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Fármacos Neuroprotectores/química , Estrés Oxidativo/efectos de los fármacos , Células PC12 , Enfermedad de Parkinson Secundaria/genética , Enfermedad de Parkinson Secundaria/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Ratas , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA