Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Mol Ther ; 26(2): 550-567, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29273501

RESUMEN

Progressive neuronal death in brainstem nuclei and widespread accumulation of α-synuclein are neuropathological hallmarks of Parkinson's disease (PD). Reduction of α-synuclein levels is therefore a potential therapy for PD. However, because α-synuclein is essential for neuronal development and function, α-synuclein elimination would dramatically impact brain function. We previously developed conjugated small interfering RNA (siRNA) sequences that selectively target serotonin (5-HT) or norepinephrine (NE) neurons after intranasal administration. Here, we used this strategy to conjugate inhibitory oligonucleotides, siRNA and antisense oligonucleotide (ASO), with the triple monoamine reuptake inhibitor indatraline (IND), to selectively reduce α-synuclein expression in the brainstem monoamine nuclei of mice after intranasal delivery. Following internalization of the conjugated oligonucleotides in monoamine neurons, reduced levels of endogenous α-synuclein mRNA and protein were found in substantia nigra pars compacta (SNc), ventral tegmental area (VTA), dorsal raphe nucleus (DR), and locus coeruleus (LC). α-Synuclein knockdown by ∼20%-40% did not cause monoaminergic neurodegeneration and enhanced forebrain dopamine (DA) and 5-HT release. Conversely, a modest human α-synuclein overexpression in DA neurons markedly reduced striatal DA release. These results indicate that α-synuclein negatively regulates monoamine neurotransmission and set the stage for the testing of non-viral inhibitory oligonucleotides as disease-modifying agents in α-synuclein models of PD.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Oligonucleótidos/genética , alfa-Sinucleína/genética , Administración Intranasal , Animales , Células Cultivadas , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Técnicas de Transferencia de Gen , Terapia Genética , Humanos , Ratones , Vías Nerviosas , Oligonucleótidos/administración & dosificación , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/terapia , Prosencéfalo/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Serotonina/metabolismo , Transducción de Señal , Sustancia Negra/metabolismo , Sustancia Negra/fisiopatología , Transmisión Sináptica/genética
2.
Ann Neurol ; 75(3): 351-62, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24243558

RESUMEN

OBJECTIVE: Mounting evidence suggests that α-synuclein, a major protein component of Lewy bodies (LB), may be responsible for initiating and spreading the pathological process in Parkinson disease (PD). Supporting this concept, intracerebral inoculation of synthetic recombinant α-synuclein fibrils can trigger α-synuclein pathology in mice. However, it remains uncertain whether the pathogenic effects of recombinant synthetic α-synuclein may apply to PD-linked pathological α-synuclein and occur in species closer to humans. METHODS: Nigral LB-enriched fractions containing pathological α-synuclein were purified from postmortem PD brains by sucrose gradient fractionation and subsequently inoculated into the substantia nigra or striatum of wild-type mice and macaque monkeys. Control animals received non-LB fractions containing soluble α-synuclein derived from the same nigral PD tissue. RESULTS: In both mice and monkeys, intranigral or intrastriatal inoculations of PD-derived LB extracts resulted in progressive nigrostriatal neurodegeneration starting at striatal dopaminergic terminals. No neurodegeneration was observed in animals receiving non-LB fractions from the same patients. In LB-injected animals, exogenous human α-synuclein was quickly internalized within host neurons and triggered the pathological conversion of endogenous α-synuclein. At the onset of LB-induced degeneration, host pathological α-synuclein diffusely accumulated within nigral neurons and anatomically interconnected regions, both anterogradely and retrogradely. LB-induced pathogenic effects required both human α-synuclein present in LB extracts and host expression of α-synuclein. INTERPRETATION: α-Synuclein species contained in PD-derived LB are pathogenic and have the capacity to initiate a PD-like pathological process, including intracellular and presynaptic accumulations of pathological α-synuclein in different brain areas and slowly progressive axon-initiated dopaminergic nigrostriatal neurodegeneration.


Asunto(s)
Neuronas Dopaminérgicas/patología , Cuerpos de Lewy/química , Degeneración Nerviosa/patología , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/patología , Extractos de Tejidos/toxicidad , alfa-Sinucleína/toxicidad , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/patología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Femenino , Humanos , Cuerpos de Lewy/metabolismo , Cuerpos de Lewy/patología , Macaca mulatta , Ratones , Ratones Noqueados , Microinyecciones , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/metabolismo , Enfermedad de Parkinson/metabolismo , Sustancia Negra/efectos de los fármacos , Sustancia Negra/patología , Extractos de Tejidos/química , alfa-Sinucleína/administración & dosificación , alfa-Sinucleína/genética , alfa-Sinucleína/aislamiento & purificación
3.
Sci Rep ; 10(1): 7118, 2020 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-32346016

RESUMEN

Chitinase 3-like 1 (CHI3L1) is known to play a role as prognostic biomarker in the early stages of multiple sclerosis (MS), and patients with high cerebrospinal fluid CHI3L1 levels have an increased risk for the development of neurological disability. Here, we investigated its potential neurotoxic effect by adding recombinant CHI3L1 in vitro to primary cultures of mouse cortical neurons and evaluating both neuronal functionality and survival by immunofluorescence. CHI3L1 induced a significant neurite length retraction after 24 and 48 hours of exposure and significantly reduced neuronal survival at 48 hours. The cytotoxic effect of CHI3L1 was neuron-specific and was not observed in mouse immune or other central nervous system cells. These results point to a selective neurotoxic effect of CHI3L1 in vitro and suggest a potential role of CHI3L1 as therapeutic target in MS patients.


Asunto(s)
Proteína 1 Similar a Quitinasa-3/administración & dosificación , Esclerosis Múltiple/tratamiento farmacológico , Neuronas/efectos de los fármacos , Animales , Biomarcadores/líquido cefalorraquídeo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Proteína 1 Similar a Quitinasa-3/metabolismo , Humanos , Ratones , Esclerosis Múltiple/enzimología , Neuronas/enzimología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/metabolismo
4.
ACS Chem Neurosci ; 11(17): 2679-2687, 2020 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-32786306

RESUMEN

Dopamine is a key neurotransmitter in the pathophysiology of various neurological disorders such as addiction or Parkinson's disease. Disturbances in its metabolism could lead to dopamine accumulation in the cytoplasm and an increased production of o-quinones and their derivatives, which have neurotoxic potential and act as precursors in neuromelanin synthesis. Thus, quantification of the dopaminergic metabolism is essential for monitoring changes that may contribute to disease development. Here, we developed and validated an UPLC-MS/MS method to detect and quantify a panel of eight dopaminergic metabolites, including the oxidation product aminochrome. Our method was validated in differentiated SH-SY5Y cells and mouse brain tissue and was then employed in brain samples from humans and rats to ensure method reliability in different matrices. Finally, to prove the biological relevance of our method, we determined metabolic changes in an in vitro cellular model of dopamine oxidation/neuromelanin production and in human postmortem samples from Parkinson's disease patients. The current study provides a validated method to simultaneously monitor possible alterations in dopamine degradation and o-quinone production pathways that can be applied to in vitro and in vivo experimental models of neurological disorders and human brain samples.


Asunto(s)
Dopamina , Espectrometría de Masas en Tándem , Animales , Encéfalo , Cromatografía Líquida de Alta Presión , Cromatografía Liquida , Humanos , Ratones , Ratas , Reproducibilidad de los Resultados
5.
Autophagy ; 15(11): 2028-2030, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31480882

RESUMEN

In Parkinson disease (PD), there is a preferential degeneration of neurons that contain the dark-brown cytoplasmic pigment neuromelanin, in particular dopaminergic neurons of the substantia nigra (SN), the loss of which leads to the typical motor symptoms of the disease and constitutes the cardinal pathological diagnostic criterion for PD. Neuromelanin is generally considered a byproduct of dopamine oxidative metabolism and, in humans, it is first detected in early childhood and accumulates progressively with age until occupying most of the neuronal cytoplasm, as neurons apparently lack the means to degrade or eliminate this pigment. Aging is the main risk factor for developing PD, but the molecular substrate underlying this link remains unknown. Despite the close and long-established association between neuromelanin and PD, the potential contribution of neuromelanin to PD pathogenesis has remained elusive because, in contrast to humans, common laboratory animal species, such as rodents, lack neuromelanin. To overcome this major limitation, we have recently generated the first experimental in vivo rodent model exhibiting age-dependent production and accumulation of human-like neuromelanin within PD-vulnerable dopaminergic nigral neurons, at levels up to those reached in elderly humans.


Asunto(s)
Autofagia , Enfermedad de Parkinson , Anciano , Animales , Niño , Preescolar , Citoplasma , Humanos , Melaninas , Proteostasis , Sustancia Negra
6.
Nat Commun ; 10(1): 973, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30846695

RESUMEN

In Parkinson's disease (PD) there is a selective degeneration of neuromelanin-containing neurons, especially substantia nigra dopaminergic neurons. In humans, neuromelanin accumulates with age, the latter being the main risk factor for PD. The contribution of neuromelanin to PD pathogenesis remains unknown because, unlike humans, common laboratory animals lack neuromelanin. Synthesis of peripheral melanins is mediated by tyrosinase, an enzyme also present at low levels in the brain. Here we report that overexpression of human tyrosinase in rat substantia nigra results in age-dependent production of human-like neuromelanin within nigral dopaminergic neurons, up to levels reached in elderly humans. In these animals, intracellular neuromelanin accumulation above a specific threshold is associated to an age-dependent PD phenotype, including hypokinesia, Lewy body-like formation and nigrostriatal neurodegeneration. Enhancing lysosomal proteostasis reduces intracellular neuromelanin and prevents neurodegeneration in tyrosinase-overexpressing animals. Our results suggest that intracellular neuromelanin levels may set the threshold for the initiation of PD.


Asunto(s)
Encéfalo/metabolismo , Melaninas/biosíntesis , Monofenol Monooxigenasa/metabolismo , Enfermedad de Parkinson/metabolismo , Envejecimiento/metabolismo , Animales , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Humanos , Cuerpos de Lewy/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Monofenol Monooxigenasa/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sustancia Negra/metabolismo , alfa-Sinucleína/deficiencia , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
7.
J Neurosci ; 27(45): 12413-8, 2007 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-17989306

RESUMEN

Degeneration of dopaminergic neurons in the substantia nigra is characteristic for Parkinson's disease (PD), the second most common neurodegenerative disorder. Mitochondrial dysfunction is believed to contribute to the etiology of PD. Although most cases are sporadic, recent evidence points to a number of genes involved in familial variants of PD. Among them, a loss-of-function of phosphatase and tensin homolog-induced kinase 1 (PINK1; PARK6) is associated with rare cases of autosomal recessive parkinsonism. In HeLa cells, RNA interference-mediated downregulation of PINK1 results in abnormal mitochondrial morphology and altered membrane potential. Morphological changes of mitochondria can be rescued by expression of wild-type PINK1 but not by PD-associated PINK1 mutants. Moreover, primary cells derived from patients with two different PINK1 mutants showed a similar defect in mitochondrial morphology. Human parkin but not PD-associated mutants could rescue mitochondrial pathology in human cells like wild-type PINK1. Our results may therefore suggest that PINK1 deficiency in humans results in mitochondrial abnormalities associated with cellular stress, a pathological phenotype, which can be ameliorated by enhanced expression of parkin.


Asunto(s)
Mitocondrias/enzimología , Mitocondrias/patología , Proteínas Quinasas/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Células Cultivadas , Células HeLa , Humanos , Mitocondrias/genética , Membranas Mitocondriales/enzimología , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Fenotipo , Proteínas Quinasas/deficiencia , Proteínas Quinasas/genética
8.
Acta Neuropathol Commun ; 6(1): 8, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29422109

RESUMEN

In Parkinson's disease (PD) there is widespread accumulation in the brain of abnormal α-synuclein aggregates forming intraneuronal Lewy bodies (LB). It is now well established that LB-type α-synuclein aggregates also occur in the peripheral autonomic nervous system in PD, from where it has been speculated they may progressively spread to the central nervous system through synaptically-connected brain networks and reach the substantia nigra to trigger herein dopaminergic dysfunction/degeneration and subsequent parkinsonism. Supporting a pathogenic role for α-synuclein aggregates we have previously shown that LB purified from postmortem PD brains promote α-synuclein pathology and dopaminergic neurodegeneration when intracerebrally inoculated into wild-type mice. However, the pathogenic capacity of PD-derived peripheral α-synuclein aggregates remains unknown. Here we addressed this question using purified LB-type α-synuclein aggregates from postmortem PD stellate ganglia (SG), a paravertebral sympathetic ganglion that exhibits consistent and conspicuous Lewy pathology in all PD patients. In contrast to our previous findings using nigral LB extracts, intracerebral inoculation of SG-derived LB into mice did not trigger long-term nigrostriatal neurodegeneration nor α-synuclein pathology. The differential pathogenic capacities of central- and peripheral-derived α-synuclein aggregates appear independent of the absolute amount and basic biochemical properties of α-synuclein within these aggregates and may rely instead on differences in α-synuclein conformation and/or yet unrecognized brain region-specific intrinsic factors. Our results argue against a putative pathogenic capacity of peripheral α-synuclein aggregates to promote α-synuclein pathology in the brain, propagate between neuronal networks or induce neurodegeneration.


Asunto(s)
Encéfalo/metabolismo , Cuerpos de Lewy/metabolismo , Enfermedad de Parkinson/metabolismo , Agregación Patológica de Proteínas/metabolismo , Ganglio Estrellado/metabolismo , alfa-Sinucleína/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/patología , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , alfa-Sinucleína/administración & dosificación , alfa-Sinucleína/química , alfa-Sinucleína/aislamiento & purificación
9.
Front Neuroanat ; 12: 113, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30618654

RESUMEN

When James Parkinson described the classical symptoms of the disease he could hardly foresee the evolution of our understanding over the next two hundred years. Nowadays, Parkinson's disease is considered a complex multifactorial disease in which genetic factors, either causative or susceptibility variants, unknown environmental cues, and the potential interaction of both could ultimately trigger the pathology. Noteworthy advances have been made in different fields from the clinical phenotype to the decoding of some potential neuropathological features, among which are the fields of genetics, drug discovery or biomaterials for drug delivery, which, though recent in origin, have evolved swiftly to become the basis of research into the disease today. In this review, we highlight some of the key advances in the field over the past two centuries and discuss the current challenges focusing on exciting new research developments likely to come in the next few years. Also, the importance of pre-motor symptoms and early diagnosis in the search for more effective therapeutic options is discussed.

10.
Cell Rep ; 25(9): 2484-2496.e9, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30485814

RESUMEN

Although accumulating data indicate that increased α-synuclein expression is crucial for Parkinson disease (PD), mechanisms regulating the transcription of its gene, SNCA, are largely unknown. Here, we describe a pathway regulating α-synuclein expression. Our data show that ZSCAN21 stimulates SNCA transcription in neuronal cells and that TRIM41 is an E3 ubiquitin ligase for ZSCAN21. In contrast, TRIM17 decreases the TRIM41-mediated degradation of ZSCAN21. Silencing of ZSCAN21 and TRIM17 consistently reduces SNCA expression, whereas TRIM41 knockdown increases it. The mRNA levels of TRIM17, ZSCAN21, and SNCA are simultaneously increased in the midbrains of mice following MPTP treatment. In addition, rare genetic variants in ZSCAN21, TRIM17, and TRIM41 genes occur in patients with familial forms of PD. Expression of variants in ZSCAN21 and TRIM41 genes results in the stabilization of the ZSCAN21 protein. Our data thus suggest that deregulation of the TRIM17/TRIM41/ZSCAN21 pathway may be involved in the pathogenesis of PD.


Asunto(s)
Proteínas Portadoras/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , alfa-Sinucleína/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Femenino , Regulación de la Expresión Génica , Humanos , Factores de Transcripción de Tipo Kruppel/química , Masculino , Ratones Endogámicos C57BL , Mutación/genética , Proteínas Nucleares/química , Linaje , Unión Proteica , Proteolisis , Transcripción Genética , Proteínas de Motivos Tripartitos , Ubiquitinación , alfa-Sinucleína/genética
12.
Mol Neurobiol ; 55(9): 7533-7552, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29429047

RESUMEN

Parkinson's disease is associated with intracellular α-synuclein accumulation and ventral midbrain dopaminergic neuronal death in the Substantia Nigra of brain patients. The Rho GTPase pathway, mainly linking surface receptors to the organization of the actin and microtubule cytoskeletons, has been suggested to participate to Parkinson's disease pathogenesis. Nevertheless, its exact contribution remains obscure. To unveil the participation of the Rho GTPase family to the molecular pathogenesis of Parkinson's disease, we first used C elegans to demonstrate the role of the small GTPase RAC1 (ced-10 in the worm) in maintaining dopaminergic function and survival in the presence of alpha-synuclein. In addition, ced-10 mutant worms determined an increase of alpha-synuclein inclusions in comparison to control worms as well as an increase in autophagic vesicles. We then used a human neuroblastoma cells (M17) stably over-expressing alpha-synuclein and found that RAC1 function decreased the amount of amyloidogenic alpha-synuclein. Further, by using dopaminergic neurons derived from patients of familial LRRK2-Parkinson's disease we report that human RAC1 activity is essential in the regulation of dopaminergic cell death, alpha-synuclein accumulation, participates in neurite arborization and modulates autophagy. Thus, we determined for the first time that RAC1/ced-10 participates in Parkinson's disease associated pathogenesis and established RAC1/ced-10 as a new candidate for further investigation of Parkinson's disease associated mechanisms, mainly focused on dopaminergic function and survival against α-synuclein-induced toxicity.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/fisiología , Neuronas Dopaminérgicas/enzimología , alfa-Sinucleína/toxicidad , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Amiloide/metabolismo , Animales , Autofagia/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Biomarcadores/metabolismo , Caenorhabditis elegans/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dopamina/metabolismo , Humanos , Cuerpos de Inclusión/metabolismo , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Mesencéfalo/patología , Mutación/genética , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Neuroprotección/efectos de los fármacos , Enfermedad de Parkinson/patología
13.
EMBO Mol Med ; 7(12): 1529-46, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26516212

RESUMEN

The epigenomic landscape of Parkinson's disease (PD) remains unknown. We performed a genomewide DNA methylation and a transcriptome studies in induced pluripotent stem cell (iPSC)-derived dopaminergic neurons (DAn) generated by cell reprogramming of somatic skin cells from patients with monogenic LRRK2-associated PD (L2PD) or sporadic PD (sPD), and healthy subjects. We observed extensive DNA methylation changes in PD DAn, and of RNA expression, which were common in L2PD and sPD. No significant methylation differences were present in parental skin cells, undifferentiated iPSCs nor iPSC-derived neural cultures not-enriched-in-DAn. These findings suggest the presence of molecular defects in PD somatic cells which manifest only upon differentiation into the DAn cells targeted in PD. The methylation profile from PD DAn, but not from controls, resembled that of neural cultures not-enriched-in-DAn indicating a failure to fully acquire the epigenetic identity own to healthy DAn in PD. The PD-associated hypermethylation was prominent in gene regulatory regions such as enhancers and was related to the RNA and/or protein downregulation of a network of transcription factors relevant to PD (FOXA1, NR3C1, HNF4A, and FOSL2). Using a patient-specific iPSC-based DAn model, our study provides the first evidence that epigenetic deregulation is associated with monogenic and sporadic PD.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Enfermedad de Parkinson/genética , Reprogramación Celular , Metilación de ADN , Epigenómica , Perfilación de la Expresión Génica , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Enfermedad de Parkinson/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo
14.
EMBO Mol Med ; 4(5): 380-95, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22407749

RESUMEN

Induced pluripotent stem cells (iPSC) offer an unprecedented opportunity to model human disease in relevant cell types, but it is unclear whether they could successfully model age-related diseases such as Parkinson's disease (PD). Here, we generated iPSC lines from seven patients with idiopathic PD (ID-PD), four patients with familial PD associated to the G2019S mutation in the Leucine-Rich Repeat Kinase 2 (LRRK2) gene (LRRK2-PD) and four age- and sex-matched healthy individuals (Ctrl). Over long-time culture, dopaminergic neurons (DAn) differentiated from either ID-PD- or LRRK2-PD-iPSC showed morphological alterations, including reduced numbers of neurites and neurite arborization, as well as accumulation of autophagic vacuoles, which were not evident in DAn differentiated from Ctrl-iPSC. Further induction of autophagy and/or inhibition of lysosomal proteolysis greatly exacerbated the DAn morphological alterations, indicating autophagic compromise in DAn from ID-PD- and LRRK2-PD-iPSC, which we demonstrate occurs at the level of autophagosome clearance. Our study provides an iPSC-based in vitro model that captures the patients' genetic complexity and allows investigation of the pathogenesis of both sporadic and familial PD cases in a disease-relevant cell type.


Asunto(s)
Dopamina/metabolismo , Neuronas/patología , Neuronas/fisiología , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología , Células Madre Pluripotentes/patología , Células Madre Pluripotentes/fisiología , Autofagia , Células Cultivadas , Humanos , Receptores Dopaminérgicos/metabolismo , Vacuolas/metabolismo , Vacuolas/ultraestructura
15.
Cell Signal ; 22(5): 821-7, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20074637

RESUMEN

Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most frequent cause of autosomal-dominant Parkinson's disease (PD). The second known autosomal-dominant PD gene (SNCA) encodes alpha-synuclein, which is deposited in Lewy bodies, the neuropathological hallmark of PD. LRRK2 contains a kinase domain with homology to mitogen-activated protein kinase kinase kinases (MAPKKKs) and its activity has been suggested to be a key factor in LRRK2-associated PD. Here we investigated the role of LRRK2 in signal transduction pathways to identify putative PD-relevant downstream targets. Over-expression of wild-type [wt]LRRK2 in human embryonic kidney HEK293 cells selectively activated the extracellular signal-regulated kinase (ERK) module. PD-associated mutants G2019S and R1441C, but not kinase-dead LRRK2, induced ERK phosphorylation to the same extent as [wt]LRRK2, indicating that this effect is kinase-dependent. However, ERK activation by mutant R1441C and G2019S was significantly slower than that for [wt]LRRK2, despite similar levels of expression. Furthermore, induction of the ERK module by LRRK2 was associated to a small but significant induction of SNCA, which was suppressed by treatment with the selective MAPK/ERK kinase inhibitor U0126. This pathway linking the two dominant PD genes LRRK2 and SNCA may offer an interesting target for drug therapy in both familial and sporadic disease.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Serina-Treonina Quinasas/metabolismo , alfa-Sinucleína/genética , Butadienos/farmacología , Línea Celular , Activación Enzimática/efectos de los fármacos , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , MAP Quinasa Quinasa 2/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Mutantes/metabolismo , Mutación/genética , Nitrilos/farmacología , Enfermedad de Parkinson/enzimología , Enfermedad de Parkinson/patología , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , alfa-Sinucleína/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA