Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
1.
J Integr Neurosci ; 23(9): 175, 2024 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-39344237

RESUMEN

BACKGROUND: The abnormal aggregation of α-synuclein (α-syn) in the substantia nigra pars compacta (SNpc) region of the brain is characteristic of Parkinson's disease (PD), leading to the selective demise of neurons. Modifications in the post-translational processing of α-syn, phosphorylation at Ser129 in particular, are implicated in α-syn aggregation and are considered key hallmarks of PD. Furthermore, dysregulated Wnt/ß-catenin signaling, influenced by glycogen synthase kinase-3 beta (GSK-3ß), is implicated in PD pathogenesis. Inhibition of GSK-3ß holds promise in promoting neuroprotection by enhancing the Wnt/ß-catenin pathway. METHODS: In our previous study utilizing 1-methyl-4-phenylpyridinium (MPP+)-administered differentiated SH-SY5Y cells and a PD mouse model, we explored Vanillin's neuroprotective properties and related mechanisms against neuronal loss induced by MPP+/1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration. In the current study, we elucidated the mitigating effects of Vanillin on motor impairments, P-Ser129-α-syn expression, Wnt/ß-catenin signaling, and autophagic neuron death induced by MPTP in a mouse model of PD by performing motor function tests, western blot analysis and immunostaining. RESULTS: Our results show that Vanillin effectively modulated the motor dysfunctions, GSK-3ß expression, and activity, activated the Wnt/ß-catenin signaling, and reduced autophagic neuronal demise in the MPTP-lesioned mice, highlighting its neuroprotective effects. CONCLUSIONS: These findings underscore the complex interplay between α-syn pathology, GSK-3ß, Wnt/ß-catenin signaling, and autophagic-cell death in PD pathogenesis. Targeting these pathways, particularly with Vanillin, can be a promising therapeutic strategy for restoring dopaminergic (DA-ergic) neuronal homeostasis and slowing the progression of PD. Further research is crucial to resolving existing disputes and translating these discoveries into effective therapeutic interventions for PD patients.


Asunto(s)
Benzaldehídos , Modelos Animales de Enfermedad , Fármacos Neuroprotectores , Vía de Señalización Wnt , alfa-Sinucleína , Animales , Masculino , Ratones , alfa-Sinucleína/metabolismo , alfa-Sinucleína/efectos de los fármacos , Benzaldehídos/farmacología , Benzaldehídos/administración & dosificación , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glucógeno Sintasa Quinasa 3 beta/efectos de los fármacos , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/administración & dosificación , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/tratamiento farmacológico , Sinucleinopatías/metabolismo , Sinucleinopatías/tratamiento farmacológico , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/fisiología
2.
Toxicol Appl Pharmacol ; 435: 115853, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34973289

RESUMEN

Parkinson's disease (PD) is the second most common progressive neurodegenerative disorder. Although mounting studies have been conducted, no effective therapy is available to halt its progression. Indole-3-carbinol (I3C) is a naturally occurring compound obtained by ß-thioglucosidase-mediated autolysis of glucobrassicin in cruciferous vegetables. Besides its powerful antioxidant activity, I3C has shown neuroprotection against depression and chemically induced neurotoxicity via its anti-inflammatory and antiapoptotic effects. This study aimed to investigate the neuroprotective effects of I3C against rotenone (ROT)-induced PD in male albino rats. The possible protective mechanisms were also explored. PD was induced by subcutaneous administration of ROT (2 mg/kg) for 28 days. The effects of I3C (25, 50, and 100 mg/kg/day) were assessed by catalepsy test (bar test), spontaneous locomotor activity, rotarod test, weight change, tyrosine hydroxylase (TH) expression, α-synuclein (α-Syn) expression, striatal dopamine (DA) content, and histological examination. The highest dose of I3C (100 mg/kg) was the most effective to prevent ROT-mediated motor dysfunctions and amend striatal DA decrease, weight loss, neurodegeneration, TH expression reduction, and α-Syn expression increase in both the midbrain and striatum. Further mechanistic investigations revealed that the neuroprotective effects of I3C are partially attributed to its anti-inflammatory and antiapoptotic effects and the activation of the sirtuin 1/AMP-activated protein kinase pathway. Altogether, these results suggested that I3C could attenuate biochemical, molecular, and functional changes in a rat PD model with following repeated rotenone exposures.


Asunto(s)
Indoles/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/prevención & control , Rotenona , Sirtuina 1/metabolismo , Desacopladores , Animales , Peso Corporal/efectos de los fármacos , Catalepsia/inducido químicamente , Catalepsia/prevención & control , Dopamina/metabolismo , Masculino , Actividad Motora/efectos de los fármacos , Neostriado/efectos de los fármacos , Neostriado/metabolismo , Enfermedad de Parkinson Secundaria/psicología , Equilibrio Postural/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Sirtuina 1/efectos de los fármacos , Tirosina 3-Monooxigenasa/metabolismo , alfa-Sinucleína/biosíntesis , alfa-Sinucleína/efectos de los fármacos
3.
Neurobiol Dis ; 151: 105256, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33429042

RESUMEN

Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are neurodegenerative disorders characterized by the misfolding and aggregation of alpha-synuclein (aSyn). Doxycycline, a tetracyclic antibiotic shows neuroprotective effects, initially proposed to be due to its anti-inflammatory properties. More recently, an additional mechanism by which doxycycline may exert its neuroprotective effects has been proposed as it has been shown that it inhibits amyloid aggregation. Here, we studied the effects of doxycycline on aSyn aggregation in vivo, in vitro and in a cell free system using real-time quaking induced conversion (RT-QuiC). Using H4, SH-SY5Y and HEK293 cells, we found that doxycycline decreases the number and size of aSyn aggregates in cells. In addition, doxycycline inhibits the aggregation and seeding of recombinant aSyn, and attenuates the production of mitochondrial-derived reactive oxygen species. Finally, we found that doxycycline induces a cellular redistribution of aggregates in a C.elegans animal model of PD, an effect that is associated with a recovery of dopaminergic function. In summary, we provide strong evidence that doxycycline treatment may be an effective strategy against synucleinopathies.


Asunto(s)
Doxiciclina/farmacología , Fármacos Neuroprotectores/farmacología , Agregación Patológica de Proteínas/patología , Sinucleinopatías/patología , alfa-Sinucleína/efectos de los fármacos , Animales , Caenorhabditis elegans , Línea Celular , Humanos , Cuerpos de Inclusión/efectos de los fármacos , Cuerpos de Inclusión/metabolismo
4.
Neurobiol Dis ; 148: 105218, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33296726

RESUMEN

Parkinson's disease (PD) is the second most common neurodegenerative disease. Pathologically, PD is characterized by the formation of Lewy bodies (LBs) in the brain, which mainly comprises phosphorylated and aggregated α-synuclein (α-syn). The aberrant aggregation of α-syn is believed to play a key role in the pathogenesis of PD. While α-syn expression can be reduced by antisense oligonucleotides (ASOs), the challenge to deliver ASOs safely and effectively into the neurons remains unresolved. Here, we developed a safe and highly effective ASO delivery method by using exosomes. We first identified the ASO sequence that selectively reduced α-syn expression: ASO4. Exosome-mediated delivery of ASO4 (exo-ASO4) showed high cellular uptake and low toxicity in primary neuronal cultures. Exo-ASO4 also significantly attenuated α-syn aggregation induced by pre-formed α-syn fibrils in vitro. Exo-ASO4 intracerebroventricular injection into the brains of α-syn A53T mice, a transgenic model of PD, significantly decreased the expression of α-syn and attenuated its aggregation. Furthermore, exo-ASO4 ameliorated the degeneration of dopaminergic neurons in these mice. Finally, the α-syn A53T mice showed significantly improved locomotor functions after exo-ASO4 injection. Overall, this study demonstrates that exosome-mediated ASO4 delivery may be an effective treatment option for PD.


Asunto(s)
Neuronas Dopaminérgicas/efectos de los fármacos , Exosomas , Locomoción/efectos de los fármacos , Oligonucleótidos Antisentido/farmacología , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Portadores de Fármacos , Humanos , Inyecciones Intraventriculares , Ratones , Ratones Transgénicos , Enfermedad de Parkinson/genética , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
5.
Neurobiol Dis ; 159: 105507, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34509608

RESUMEN

Mutations in the lysosomal enzyme glucocerebrosidase (GCase, GBA1 gene) are the most common genetic risk factor for developing Parkinson's disease (PD). GCase metabolizes the glycosphingolipids glucosylceramide (GlcCer) and glucosylsphingosine (GlcSph). Mutations in GBA1 reduce enzyme activity and the resulting accumulation of glycosphingolipids may contribute to the underlying pathology of PD, possibly via altering lysosomal function. While reduction of GCase activity exacerbates α-synuclein (α-syn) aggregation, it has not been determined that this effect is the result of altered glycosphingolipid levels and lysosome function or some other effect of altering GCase. The glycosphingolipid GlcCer is synthesized by a single enzyme, glucosylceramide synthase (GCS), and small molecule inhibitors (GCSi) reduce cellular glycosphingolipid levels. In the present studies, we utilize a preformed fibril (PFF) rodent primary neuron in vitro model of α-syn pathology to investigate the relationship between glycosphingolipid levels, α-syn pathology, and lysosomal function. In primary cultures, pharmacological inhibition of GCase and D409V GBA1 mutation enhanced accumulation of glycosphingolipids and insoluble phosphorylated α-syn. Administration of a novel small molecule GCSi, benzoxazole 1 (BZ1), significantly decreased glycosphingolipid concentrations in rodent primary neurons and reduced α-syn pathology. BZ1 rescued lysosomal deficits associated with the D409V GBA1 mutation and α-syn PFF administration, and attenuated α-syn induced neurodegeneration of dopamine neurons. In vivo studies revealed BZ1 had pharmacological activity and reduced glycosphingolipids in the mouse brain to a similar extent observed in neuronal cultures. These data support the hypothesis that reduction of glycosphingolipids through GCS inhibition may impact progression of synucleinopathy and BZ1 is useful tool to further examine this important biology.


Asunto(s)
Benzoxazoles/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Glucosilceramidasa/genética , Glucosiltransferasas/antagonistas & inhibidores , Glicoesfingolípidos/metabolismo , Lisosomas/efectos de los fármacos , Sinucleinopatías/metabolismo , alfa-Sinucleína/efectos de los fármacos , Animales , Neuronas Dopaminérgicas/metabolismo , Técnicas In Vitro , Lisosomas/metabolismo , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Cultivo Primario de Células , Agregado de Proteínas , Ratas , Sinucleinopatías/genética , alfa-Sinucleína/metabolismo
6.
FASEB J ; 34(5): 6718-6728, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32259355

RESUMEN

α-synuclein (aSyn) is a major player in Parkinson's disease and a group of other disorders collectively known as synucleinopathies, but the precise molecular mechanisms involved are still unclear. aSyn, as virtually all proteins, undergoes a series of posttranslational modifications during its lifetime, which can affect its biology and pathobiology. We recently showed that glycation of aSyn by methylglyoxal (MGO) potentiates its oligomerization and toxicity, induces dopaminergic neuronal cell loss in mice, and affects motor performance in flies. Small heat-shock proteins (sHsps) are molecular chaperones that facilitate the folding of proteins or target misfolded proteins for clearance. Importantly, sHsps were shown to prevent aSyn aggregation and cytotoxicity. Upon treating cells with increasing amounts of methylglyoxal, we found that the levels of Hsp27 decreased in a dose-dependent manner. Therefore, we hypothesized that restoring the levels of Hsp27 in glycating environments could alleviate the pathogenicity of aSyn. Consistently, we found that Hsp27 reduced MGO-induced aSyn aggregation in cells, leading to the formation of nontoxic aSyn species. Remarkably, increasing the levels of Hsp27 suppressed the deleterious effects induced by MGO. Our findings suggest that in glycating environments, the levels of Hsp27 are important for modulating the glycation-associated cellular pathologies in synucleinopathies.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Proteínas de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Agregado de Proteínas/efectos de los fármacos , Piruvaldehído/farmacología , alfa-Sinucleína/química , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Glioma/tratamiento farmacológico , Glioma/genética , Glioma/metabolismo , Glicosilación , Proteínas de Choque Térmico/genética , Humanos , Chaperonas Moleculares/genética , Células Tumorales Cultivadas , alfa-Sinucleína/efectos de los fármacos
7.
Biochem J ; 477(3): 645-670, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31939603

RESUMEN

The aggregation of the protein alpha synuclein (α-Syn), a known contributor in Parkinson's disease (PD) pathogenesis is triggered by transition metal ions through occupational exposure and disrupted metal ion homeostasis. Naturally occurring small molecules such as polyphenols have emerged as promising inhibitors of α-Syn fibrillation and toxicity and could be potential therapeutic agents against PD. Here, using an array of biophysical tools combined with cellular assays, we demonstrate that the novel polyphenolic compound scutellarin efficiently inhibits the uninduced and metal-induced fibrillation of α-Syn by acting at the nucleation stage and stabilizes a partially folded intermediate of α-Syn to form SDS-resistant, higher-order oligomers (∼680 kDa) and also disaggregates preformed fibrils of α-Syn into similar type of higher-order oligomers. ANS binding assay, fluorescence lifetime measurements and cell-toxicity experiments reveal scutellarin-generated oligomers as compact, low hydrophobicity structures with modulated surface properties and significantly reduced cytotoxicity than the fibrillation intermediates of α-Syn control. Fluorescence spectroscopy and isothermal titration calorimetry establish the binding between scutellarin and α-Syn to be non-covalent in nature and of moderate affinity (Ka ∼ 105 M-1). Molecular docking approaches suggest binding of scutellarin to the residues present in the NAC region and C-terminus of monomeric α-Syn and the C-terminal residues of fibrillar α-Syn, demonstrating inhibition of fibrillation upon binding to these residues and possible stabilization of the autoinhibitory conformation of α-Syn. These findings reveal interesting insights into the mechanism of scutellarin action and establish it as an efficient modulator of uninduced as well as metal-induced α-Syn fibrillation and toxicity.


Asunto(s)
Apigenina/farmacología , Glucuronatos/farmacología , Enfermedad de Parkinson , Agregado de Proteínas/efectos de los fármacos , Agregación Patológica de Proteínas/tratamiento farmacológico , alfa-Sinucleína/metabolismo , Amiloide/efectos de los fármacos , Amiloide/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Agregación Patológica de Proteínas/metabolismo , alfa-Sinucleína/efectos de los fármacos
8.
Int J Mol Sci ; 22(21)2021 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-34768886

RESUMEN

Exposure to heavy metals, including arsenic and cadmium, is associated with neurodegenerative disorders such as Parkinson's disease. However, the mechanistic details of how these metals contribute to pathogenesis are not well understood. To search for underlying mechanisms involving α-synuclein, the protein that forms amyloids in Parkinson's disease, we here assessed the effects of arsenic and cadmium on α-synuclein amyloid formation in vitro and in Saccharomyces cerevisiae (budding yeast) cells. Atomic force microscopy experiments with acetylated human α-synuclein demonstrated that amyloid fibers formed in the presence of the metals have a different fiber pitch compared to those formed without metals. Both metal ions become incorporated into the amyloid fibers, and cadmium also accelerated the nucleation step in the amyloid formation process, likely via binding to intermediate species. Fluorescence microscopy analyses of yeast cells expressing fluorescently tagged α-synuclein demonstrated that arsenic and cadmium affected the distribution of α-synuclein aggregates within the cells, reduced aggregate clearance, and aggravated α-synuclein toxicity. Taken together, our in vitro data demonstrate that interactions between these two metals and α-synuclein modulate the resulting amyloid fiber structures, which, in turn, might relate to the observed effects in the yeast cells. Whilst our study advances our understanding of how these metals affect α-synuclein biophysics, further in vitro characterization as well as human cell studies are desired to fully appreciate their role in the progression of Parkinson's disease.


Asunto(s)
Metales Pesados/toxicidad , Enfermedades Neurodegenerativas , alfa-Sinucleína/metabolismo , Amiloide/metabolismo , Arsenitos/toxicidad , Cadmio/toxicidad , Línea Celular/metabolismo , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/metabolismo , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Saccharomyces cerevisiae/metabolismo , alfa-Sinucleína/efectos de los fármacos
9.
Learn Mem ; 27(9): 395-413, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32817306

RESUMEN

A set of common-acting iron-responsive 5'untranslated region (5'UTR) motifs can fold into RNA stem loops that appear significant to the biology of cognitive declines of Parkinson's disease dementia (PDD), Lewy body dementia (LDD), and Alzheimer's disease (AD). Neurodegenerative diseases exhibit perturbations of iron homeostasis in defined brain subregions over characteristic time intervals of progression. While misfolding of Aß from the amyloid-precursor-protein (APP), alpha-synuclein, prion protein (PrP) each cause neuropathic protein inclusions in the brain subregions, iron-responsive-like element (IRE-like) RNA stem-loops reside in their transcripts. APP and αsyn have a role in iron transport while gene duplications elevate the expression of their products to cause rare familial cases of AD and PDD. Of note, IRE-like sequences are responsive to excesses of brain iron in a potential feedback loop to accelerate neuronal ferroptosis and cognitive declines as well as amyloidosis. This pathogenic feedback is consistent with the translational control of the iron storage protein ferritin. We discuss how the IRE-like RNA motifs in the 5'UTRs of APP, alpha-synuclein and PrP mRNAs represent uniquely folded drug targets for therapies to prevent perturbed iron homeostasis that accelerates AD, PD, PD dementia (PDD) and Lewy body dementia, thus preventing cognitive deficits. Inhibition of alpha-synuclein translation is an option to block manganese toxicity associated with early childhood cognitive problems and manganism while Pb toxicity is epigenetically associated with attention deficit and later-stage AD. Pathologies of heavy metal toxicity centered on an embargo of iron export may be treated with activators of APP and ferritin and inhibitors of alpha-synuclein translation.


Asunto(s)
Regiones no Traducidas 5'/efectos de los fármacos , Péptidos beta-Amiloides/metabolismo , Ferritinas/metabolismo , Ferroptosis/fisiología , Intoxicación por Metales Pesados/metabolismo , Proteínas Reguladoras del Hierro/metabolismo , Trastornos Neurocognitivos/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Fármacos Neuroprotectores/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , alfa-Sinucleína/metabolismo , Péptidos beta-Amiloides/efectos de los fármacos , Animales , Ferritinas/efectos de los fármacos , Ferroptosis/efectos de los fármacos , Intoxicación por Metales Pesados/tratamiento farmacológico , Intoxicación por Metales Pesados/fisiopatología , Humanos , Proteínas Reguladoras del Hierro/efectos de los fármacos , Trastornos Neurocognitivos/tratamiento farmacológico , Enfermedades Neurodegenerativas/tratamiento farmacológico , alfa-Sinucleína/efectos de los fármacos
10.
Angew Chem Int Ed Engl ; 60(10): 5173-5178, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33180342

RESUMEN

Proteins reconfigure their 3D-structure, and consequently their function, under the control of specific molecular interactions that sense, process and transmit information from the surrounding environment. When this fundamental process is hampered, many pathologies occur as in the case of protein misfolding diseases. In this work, we follow the early steps of α-synuclein (aS) aggregation, a process associated with Parkinson's disease etiopathogenesis, that is promptly promoted by a light-mediated binding between the protein and a photoactive foldamer. The latter can switch between two conformations, one of which generates supramolecular fibrillar seeds that act as molecular templates able to induce a fast ß-sheet transition for aS monomers that successively undergo fibrillar polymerization. The proposed method represents a powerful tool to study protein aggregation relevant to misfolding diseases in a controlled and inducible system.


Asunto(s)
Peptidomiméticos/química , Multimerización de Proteína/efectos de los fármacos , alfa-Sinucleína/metabolismo , Humanos , Peptidomiméticos/efectos de la radiación , Conformación Proteica/efectos de la radiación , alfa-Sinucleína/efectos de los fármacos
11.
Neurobiol Dis ; 144: 105049, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32800998

RESUMEN

Kinase activating missense mutations in leucine-rich repeat kinase 2 (LRRK2) predispose to Parkinson's disease. Consequently, there is much interest in delineating LRRK2 biology, both in terms of gaining further insight into disease causes, and also determining whether or not LRRK2 is a potential Parkinson's disease therapeutic target. Indeed, many potent and selective small molecule inhibitors of LRRK2 have been developed and are currently being used for pre-clinical testing in cell and animal models. In the current study, we have obtained fibroblasts from four subjects with the common LRRK2 mutation, G2019S. Fibroblasts were reprogrammed to induced pluripotent stem cells and then to neural stem cells and ultimately neurons. Two clones for each of the human neural cell lines were then chronically treated with and without either of two distinct inhibitors of LRRK2 and effects on toxicity and Parkinson's disease related phenotypes were assessed. Cells with the G2019S mutation had a propensity to accumulate the pathological Parkinson's disease protein α-synuclein. Moreover, α-synuclein accumulation in the G2019S cells was significantly reduced with both LRRK2 inhibitors in seven of the eight cell lines studied. LRRK2 inhibitors also improved the nuclear morphology of G2019S cells and impacted on measures of autophagy and endoplasmic reticulum stress. Lastly, we did not find evidence of inhibitor toxicity under the chronic treatment conditions. These results add to evidence that LRRK2 inhibitors may have utility in the treatment of Parkinson's disease via reducing α-synuclein.


Asunto(s)
Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/antagonistas & inhibidores , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Neuronas/efectos de los fármacos , Enfermedad de Parkinson/genética , Agregado de Proteínas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , alfa-Sinucleína/efectos de los fármacos , Línea Celular , Fibroblastos , Humanos , Técnicas In Vitro , Células Madre Pluripotentes Inducidas , Células-Madre Neurales , Neuronas/metabolismo , Neuronas/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , alfa-Sinucleína/metabolismo
12.
J Neural Transm (Vienna) ; 127(2): 131-147, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31993732

RESUMEN

Parkinson's disease has been considered as a motor neuron disease with dopamine (DA) deficit caused by neuronal loss in the substantia nigra, but now proposed as a multi-system disorder associated with α-synuclein accumulation in neuronal and non-neuronal systems. Neuroprotection in Parkinson's disease has intended to halt or reverse cell death of nigro-striatal DA neurons and prevent the disease progression, but clinical studies have not presented enough beneficial results, except the trial of rasagiline by delayed start design at low dose of 1 mg/day only. Now strategy of disease-modifying therapy should be reconsidered taking consideration of accumulation and toxicity of α-synuclein preceding the manifest of motor symptoms. Hitherto neuroprotective therapy has been aimed to mitigate non-specific risk factors; oxidative stress, mitochondrial dysfunction, apoptosis, deficits of neurotrophic factors (NTFs), inflammation and accumulation of pathogenic protein. Future disease-modify therapy should target more specified pathogenic factors, including deregulated mitochondrial homeostasis, deficit of NTFs and α-synuclein toxicity. Selegiline and rasagiline, inhibitors of type B monoamine oxidase, have been proved to exhibit potent neuroprotective function: regulation of mitochondrial apoptosis system, maintenance of mitochondrial function, increased expression of genes coding antioxidant enzymes, anti-apoptotic Bcl-2 and pro-survival NTFs, and suppression of oligomerization and aggregation of α-synuclein and the toxicity in cellular and animal experiments. However, the present available pharmacological therapy starts too late to reverse disease progression, and future disease-modifying therapy should include also non-pharmacological complementary therapy during the prodromal stage.


Asunto(s)
Apoptosis/efectos de los fármacos , Homeostasis/efectos de los fármacos , Indanos/farmacología , Mitocondrias/efectos de los fármacos , Inhibidores de la Monoaminooxidasa/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Selegilina/farmacología , alfa-Sinucleína/efectos de los fármacos , Animales , Humanos
13.
Int J Mol Sci ; 21(21)2020 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-33114222

RESUMEN

The intrinsically disordered protein α-synuclein plays a major role in Parkinson's disease. The protein can oligomerize resulting in the formation of various aggregated species in neuronal cells, leading to neurodegeneration. The interaction of α-synuclein with biological cell membranes plays an important role for specific functions of α-synuclein monomers, e.g., in neurotransmitter release. Using different types of detergents to mimic lipid molecules present in biological membranes, including the presence of Ca2+ ions as an important structural factor, we aimed to gain an understanding of how α-synuclein interacts with membrane models and how this affects the protein conformation and potential oligomerization. We investigated detergent binding stoichiometry, affinity and conformational changes of α-synuclein taking detergent concentration, different detergent structures and charges into account. With native nano-electrospray ionization ion mobility-mass spectrometry, we were able to detect unique conformational patterns resulting from binding of specific detergents to α-synuclein. Our data demonstrate that α-synuclein monomers can interact with detergent molecules irrespective of their charge, that protein-micelle interactions occur and that micelle properties are an important factor.


Asunto(s)
Detergentes/farmacología , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Humanos , Modelos Biológicos , Modelos Moleculares , Nanotecnología , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Espectrometría de Masa por Ionización de Electrospray , alfa-Sinucleína/efectos de los fármacos
14.
Int J Mol Sci ; 21(23)2020 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-33255444

RESUMEN

The formation of amyloid fibrils is linked to multiple neurodegenerative disorders, including Alzheimer's and Parkinson's disease. Despite years of research and countless studies on the topic of such aggregate formation, as well as their resulting structure, the current knowledge is still fairly limited. One of the main aspects prohibiting effective aggregation tracking is the environment's effect on amyloid-specific dyes, namely thioflavin-T (ThT). Currently, there are only a few studies hinting at ionic strength being one of the factors that modulate the dye's binding affinity and fluorescence intensity. In this work we explore this effect under a range of ionic strength conditions, using insulin, lysozyme, mouse prion protein, and α-synuclein fibrils. We show that ionic strength is an extremely important factor affecting both the binding affinity, as well as the fluorescence intensity of ThT.


Asunto(s)
Amiloide/efectos de los fármacos , Benzotiazoles/farmacología , Agregado de Proteínas/efectos de los fármacos , Agregación Patológica de Proteínas/prevención & control , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Animales , Benzotiazoles/química , Sitios de Unión/efectos de los fármacos , Fluorescencia , Humanos , Insulina/química , Cinética , Ratones , Concentración Osmolar , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/prevención & control , Proteínas Priónicas/química , Proteínas Priónicas/efectos de los fármacos , Agregación Patológica de Proteínas/metabolismo , Unión Proteica/efectos de los fármacos , alfa-Sinucleína/química , alfa-Sinucleína/efectos de los fármacos
15.
Neurobiol Dis ; 129: 1-12, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31051233

RESUMEN

We recently found that in mouse dopaminergic neurons, the heteromer formed by the dopamine D3 receptor (D3R) and the ß2 subunit of acetylcholine nicotinic receptor (nAChR) exerts neurotrophic effects when activated by nicotine, leading to neurons with enlarged cell bodies and increased dendrite arborization. Beside this action, we now show that nicotine, by activating the D3R-nAChR heteromer, protects dopaminergic neurons against neuronal injury. In primary cultures of mouse dopaminergic neurons, in fact, the ability of nicotine to inhibit both the pathological accumulation of alpha-synuclein induced by glucose deprivation and the consequent morphological defects were strongly prevented by disrupting the D3R-nAChR heteromer with specific interfering TAT-peptides; the relevance of the phosphoinositide 3-kinase (PI3K) intracellular signaling in mediating nicotine prevention of alpha-synuclein aggregation has been also demonstrated. Moreover, the ability of nicotine in restoring the ubiquitin-proteasome system has been found as a mechanism contributing to the neuroprotective properties of nicotine. By using the proximity ligation assay, we have shown that the D3R-nAChR heteromer is also expressed in human dopaminergic neurons derived from induced pluripotent stem cells. In this human cell model, nicotine exerts neuroprotective effects specifically acting through the D3R-nAChR complex thus indicating that this heteromer is a relevant molecular effector involved in the protection of human dopaminergic neurons.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Nicotina/farmacología , Receptores de Dopamina D3/metabolismo , Receptores Nicotínicos/metabolismo , alfa-Sinucleína/efectos de los fármacos , Animales , Células Cultivadas , Neuronas Dopaminérgicas/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fármacos Neuroprotectores/farmacología , Receptores de Dopamina D3/efectos de los fármacos , Receptores Nicotínicos/efectos de los fármacos
16.
Neuropathol Appl Neurobiol ; 45(2): 157-173, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29679389

RESUMEN

AIMS: The accumulation of α-synuclein is a hallmark in the pathogenesis of Parkinson's disease (PD). Natural resistance-associated macrophage protein-1 (Nramp1) was previously shown to contribute to the degradation of extracellular α-synuclein in microglia under conditions of iron overload. This study was aimed at investigating the role of Nramp1 in α-synuclein pathology in the neurone under 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)/1-methyl-4-phenylpyridinium (MPP+ ) treatment. METHODS: The expression of Nramp1 and pathological features (including iron and α-synuclein accumulation) were examined in the dopaminergic neurones of humans (with and without PD) and of mice [with and without receiving chronic MPTP intoxication]. The effects of Nramp1 expression on low-dose MPP+ -induced α-synuclein expression and neurotoxicity were determined in human dopaminergic neuroblastoma SH-SY5Y cells. RESULTS: Similar to the findings in the substantia nigra of human PD, lower expression of Nramp1 but higher levels of iron and α-synuclein were identified in the dopaminergic neurones of mice receiving chronic MPTP intoxication, compared to controls. In parallel to the loss of dopaminergic neurones, the numbers of glial fibrillary acidic protein- and ionized calcium-binding adapter molecule-1-positive cells were significantly increased in the substantia nigra of MPTP-treated mice. Likewise, in human neuroblastoma SH-SY5Y cells exposed to low-dose MPP+ , Nramp1 expression and cathepsin D activity were decreased, along with an increase in α-synuclein protein expression and aggregation. Overexpression of functional Nramp1 restored cathepsin D activity and attenuated α-synuclein up-regulation and neuronal cell death caused by MPP+ treatment. CONCLUSIONS: These data suggest that the neuronal expression of Nramp1 is important for protecting against the development of MPTP/MPP+ -induced α-synuclein pathology and neurotoxicity.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Proteínas de Transporte de Catión/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , alfa-Sinucleína/efectos de los fármacos , Anciano , Anciano de 80 o más Años , Animales , Proteínas de Transporte de Catión/metabolismo , Línea Celular Tumoral , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Masculino , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Enfermedad de Parkinson/patología , Sustancia Negra/efectos de los fármacos , Sustancia Negra/patología , alfa-Sinucleína/metabolismo
17.
Mov Disord ; 34(2): 255-263, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30452793

RESUMEN

BACKGROUND: MSA is a fatal neurodegenerative disease characterized by autonomic failure and severe motor impairment. Its main pathological hallmark is the accumulation of α-synuclein in oligodendrocytes, leading to glial and neuronal dysfunction and neurodegeneration. These features are recapitulated in the PLP-hαSyn mouse model expressing human α-synuclein in oligodendrocytes. At present, there is no effective disease-modifying therapy. Previous experiments have shown that the aggregation inhibitor, anle138b, reduces neurodegeneration and behavioral deficits in mouse models of other proteinopathies. OBJECTIVES: To test the therapeutic potential of anle138b in a mouse model of MSA. METHODS: Two-month-old PLP-hαSyn mice were fed over a period of 4 months with pellets containing anle138b at two different doses (0.6 and 2 g/kg) and compared to healthy controls and PLP-hαSyn mice fed with placebo pellets. At the end of the treatment, behavioral and histological analyses were performed. RESULTS: We observed a reversal of motor function to healthy control levels when PLP-hαSyn mice were treated with both doses of anle138b. Histological and molecular analyses showed a significant reduction in α-synuclein oligomers and glial cytoplasmic inclusions in animals fed with anle138b compared to nontreated mice. These animals also present preservation of dopaminergic neurons and reduction in microglial activation in SN correlating with the α-synuclein reduction observed. CONCLUSIONS: Anle138b reduces α-synuclein accumulation in PLP-hαSyn mice, leading to neuroprotection, reduction of microglial activation, and preservation of motor function supporting the use of anle138b in a future clinical trial for MSA. © 2018 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Benzodioxoles/farmacología , Atrofia de Múltiples Sistemas/tratamiento farmacológico , Degeneración Nerviosa/prevención & control , Pirazoles/farmacología , alfa-Sinucleína/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Ratones Transgénicos , Trastornos del Movimiento/patología , Atrofia de Múltiples Sistemas/patología , Degeneración Nerviosa/tratamiento farmacológico , Neuroglía/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , alfa-Sinucleína/metabolismo
18.
Dig Dis Sci ; 64(12): 3502-3517, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31187328

RESUMEN

BACKGROUND: Gastrointestinal (GI) motility dysfunction is the most common non-motor symptom of Parkinson's disease (PD). Studies have indicated that GI motility functions are impaired before the onset of PD. AIMS: To investigate the underlying mechanism of PD-induced GI dysmotility in MPTP (1-methyl 4-phenyl 1,2,3,6-tetrahydropyridine)-induced animal model. METHODS: C57BL/6 mice were administered with or without a selective dopamine neurotoxin, MPTP, to induce parkinsonian symptoms. In addition to in vivo studies, in vitro experiments were also conducted in colon specimens using l-methyl-4-phenylpyridinium (MPP+), a metabolic product of MPTP. Gastric emptying, colon motility, nitrergic relaxation, and western blot experiments were performed as reported. RESULTS: MPTP-induced PD mice showed decreased expression of nuclear factor erythroid 2-related factor (Nrf2) and its target phase II genes in gastric and colon neuromuscular tissues. Decreased levels of tetrahydrobiopterin (BH4, a critical cofactor for nNOS dimerization) associated with uncoupling of nNOS in gastric and colon tissues exposed to MPTP. Impaired enteric nitrergic system led to delayed gastric emptying and slower colonic motility compared to the control mice. In vitro results in colon specimens confirm that activation of Nrf2 restored MPP+-induced suppression of alpha-synuclein, tyrosine hydroxylase (TH), Nrf2, and heme oxygenase-1. In vitro exposure to L-NAME [N(w)-nitro-L-arginine methyl ester], a NOS synthase inhibitor, reduced protein expression of TH in colon tissue homogenates. CONCLUSIONS: Loss of Nrf2/BH4/nNOS expression in PD impairs antioxidant gene expression, which deregulates NO synthesis, thereby contributing to the development of GI dysmotility and constipation. Nitric oxide appears to be important to maintain dopamine synthesis in the colon.


Asunto(s)
Motilidad Gastrointestinal/fisiología , Intoxicación por MPTP/genética , Factor 2 Relacionado con NF-E2/genética , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , 1-Metil-4-fenilpiridinio/farmacología , Animales , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Western Blotting , Colon/efectos de los fármacos , Colon/metabolismo , Colon/fisiopatología , Estreñimiento , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Vaciamiento Gástrico/fisiología , Regulación de la Expresión Génica , Hemo-Oxigenasa 1/efectos de los fármacos , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Intoxicación por MPTP/metabolismo , Intoxicación por MPTP/fisiopatología , Masculino , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Enfermedad de Parkinson/fisiopatología , Trastornos Parkinsonianos , Tirosina 3-Monooxigenasa/efectos de los fármacos , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo , alfa-Sinucleína/efectos de los fármacos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
19.
Int J Mol Sci ; 21(1)2019 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-31861692

RESUMEN

Heat shock proteins play roles in assisting other proteins to fold correctly and in preventing the aggregation and accumulation of proteins in misfolded conformations. However, the process of aging significantly degrades this ability to maintain protein homeostasis. Consequently, proteins with incorrect conformations are prone to aggregate and accumulate in cells, and this aberrant aggregation of misfolded proteins may trigger various neurodegenerative diseases, such as Parkinson's disease. Here, we investigated the possibilities of suppressing α-synuclein aggregation by using a mutant form of human chaperonin Hsp60, and a derivative of the isolated apical domain of Hsp60 (Hsp60 AD(Cys)). In vitro measurements were used to detect the effects of chaperonin on amyloid fibril formation, and interactions between Hsp60 proteins and α-synuclein were probed by quartz crystal microbalance analysis. The ability of Hsp60 AD(Cys) to suppress α-synuclein intracellular aggregation and cytotoxicity was also demonstrated. We show that Hsp60 mutant and Hsp60 AD(Cys) both effectively suppress α-synuclein amyloid fibril formation, and also demonstrate for the first time the ability of Hsp60 AD(Cys) to function as a mini-chaperone inside cells. These results highlight the possibility of using Hsp60 AD as a method of prevention and treatment of neurodegenerative diseases.


Asunto(s)
Chaperonina 60/química , Chaperonina 60/farmacología , Proteínas Mitocondriales/química , Proteínas Mitocondriales/farmacología , Agregado de Proteínas/efectos de los fármacos , alfa-Sinucleína/metabolismo , Sitios de Unión , Línea Celular , Chaperonina 60/genética , Humanos , Proteínas Mitocondriales/genética , Modelos Moleculares , Mutación , Unión Proteica , Dominios Proteicos , Tecnicas de Microbalanza del Cristal de Cuarzo , alfa-Sinucleína/química , alfa-Sinucleína/efectos de los fármacos
20.
BMC Neurosci ; 19(1): 17, 2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29614969

RESUMEN

BACKGROUND: Accumulation of the α-synuclein (α-syn) protein is a hallmark of a group of brain disorders collectively known as synucleinopathies. The mechanisms responsible for α-syn accumulation are not well understood. Several studies suggest a link between synucleinopathies and the cholesterol metabolite 27-hydroxycholesterol (27-OHC). 27-OHC is the major cholesterol metabolite in the blood that crosses the blood brain barrier, and its levels can increase following hypercholesterolemia, aging, and oxidative stress, which are all factors for increased synucleinopathy risk. In this study, we determined the extent to which 27-OHC regulates α-syn levels in human dopaminergic neurons, the cell type in which α-syn accumulates in PD, a major synucleinopathy disorder. RESULTS: Our results show that 27-OHC significantly increases the protein levels, not the mRNA expression of α-syn. The effects of 27-OHC appear to be independent of an action through liver X receptors (LXR), its cognate receptors, as the LXR agonist, GW3965, or the LXR antagonist ECHS did not affect α-syn protein or mRNA levels. Furthermore, our data strongly suggest that the 27-OHC-induced increase in α-syn protein levels emanates from inhibition of the proteasomal degradation of this protein and a decrease in the heat shock protein 70 (HSP70). CONCLUSIONS: Identifying 27-OHC as a factor that can increase α-syn levels and the inhibition of the proteasomal function and reduction in HSP70 levels as potential cellular mechanisms involved in regulation of α-syn. This may help in targeting the correct degradation of α-syn as a potential avenue to preclude α-syn accumulation.


Asunto(s)
Colesterol/sangre , Neuronas Dopaminérgicas/efectos de los fármacos , Hidroxicolesteroles/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , alfa-Sinucleína/metabolismo , Humanos , ARN/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , alfa-Sinucleína/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA