Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 146(1): 37-52, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21700325

RESUMEN

Parkinson's disease (PD), an adult neurodegenerative disorder, has been clinically linked to the lysosomal storage disorder Gaucher disease (GD), but the mechanistic connection is not known. Here, we show that functional loss of GD-linked glucocerebrosidase (GCase) in primary cultures or human iPS neurons compromises lysosomal protein degradation, causes accumulation of α-synuclein (α-syn), and results in neurotoxicity through aggregation-dependent mechanisms. Glucosylceramide (GlcCer), the GCase substrate, directly influenced amyloid formation of purified α-syn by stabilizing soluble oligomeric intermediates. We further demonstrate that α-syn inhibits the lysosomal activity of normal GCase in neurons and idiopathic PD brain, suggesting that GCase depletion contributes to the pathogenesis of sporadic synucleinopathies. These findings suggest that the bidirectional effect of α-syn and GCase forms a positive feedback loop that may lead to a self-propagating disease. Therefore, improved targeting of GCase to lysosomes may represent a specific therapeutic approach for PD and other synucleinopathies.


Asunto(s)
Enfermedad de Gaucher/metabolismo , Glucosilceramidasa/metabolismo , alfa-Sinucleína/metabolismo , Animales , Encéfalo/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Retroalimentación Fisiológica , Enfermedad de Gaucher/patología , Glucosilceramidas/metabolismo , Humanos , Lisosomas/metabolismo , Ratones , Neuronas/metabolismo
2.
J Neurosci ; 43(14): 2615-2629, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-36788031

RESUMEN

Macroautophagy is a catabolic process that coordinates with lysosomes to degrade aggregation-prone proteins and damaged organelles. Loss of macroautophagy preferentially affects neuron viability and is associated with age-related neurodegeneration. We previously found that α-synuclein (α-syn) inhibits lysosomal function by blocking ykt6, a farnesyl-regulated soluble NSF attachment protein receptor (SNARE) protein that is essential for hydrolase trafficking in midbrain neurons. Using Parkinson's disease (PD) patient iPSC-derived midbrain cultures, we find that chronic, endogenous accumulation of α-syn directly inhibits autophagosome-lysosome fusion by impairing ykt6-SNAP-29 complexes. In wild-type (WT) cultures, ykt6 depletion caused a near-complete block of autophagic flux, highlighting its critical role for autophagy in human iPSC-derived neurons. In PD, macroautophagy impairment was associated with increased farnesyltransferase (FTase) activity, and FTase inhibitors restored macroautophagic flux through promoting active forms of ykt6 in human cultures, and male and female mice. Our findings indicate that ykt6 mediates cellular clearance by coordinating autophagic-lysosomal fusion and hydrolase trafficking, and that macroautophagy impairment in PD can be rescued by FTase inhibitors.SIGNIFICANCE STATEMENT The pathogenic mechanisms that lead to the death of neurons in Parkinson's disease (PD) and Dementia with Lewy bodies (LBD) are currently unknown. Furthermore, disease modifying treatments for these diseases do not exist. Our study indicates that a cellular clearance pathway termed autophagy is impaired in patient-derived culture models of PD and in vivo We identified a novel druggable target, a soluble NSF attachment protein receptor (SNARE) protein called ykt6, that rescues autophagy in vitro and in vivo upon blocking its farnesylation. Our work suggests that farnesyltransferase (FTase) inhibitors may be useful therapies for PD and DLB through enhancing autophagic-lysosomal clearance of aggregated proteins.


Asunto(s)
Enfermedad de Parkinson , Humanos , Masculino , Ratones , Animales , Femenino , Enfermedad de Parkinson/metabolismo , Farnesiltransferasa/metabolismo , alfa-Sinucleína/metabolismo , Autofagia/fisiología , Mesencéfalo/metabolismo , Neuronas/metabolismo , Lisosomas/metabolismo , Proteínas SNARE/metabolismo , Hidrolasas/metabolismo , Proteínas R-SNARE/metabolismo
3.
Cell ; 137(1): 60-72, 2009 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-19345187

RESUMEN

Huntington's disease (HD) is an incurable neurodegenerative disease caused by neuronal accumulation of the mutant protein huntingtin. Improving clearance of the mutant protein is expected to prevent cellular dysfunction and neurodegeneration in HD. We report here that such clearance can be achieved by posttranslational modification of the mutant Huntingtin (Htt) by acetylation at lysine residue 444 (K444). Increased acetylation at K444 facilitates trafficking of mutant Htt into autophagosomes, significantly improves clearance of the mutant protein by macroautophagy, and reverses the toxic effects of mutant huntingtin in primary striatal and cortical neurons and in a transgenic C. elegans model of HD. In contrast, mutant Htt that is rendered resistant to acetylation dramatically accumulates and leads to neurodegeneration in cultured neurons and in mouse brain. These studies identify acetylation as a mechanism for removing accumulated protein in HD, and more broadly for actively targeting proteins for degradation by autophagy.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Fagosomas/metabolismo , Acetilación , Animales , Animales Modificados Genéticamente , Células COS , Caenorhabditis elegans/metabolismo , Células Cultivadas , Chlorocebus aethiops , Técnicas de Sustitución del Gen , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Ratones , Procesamiento Proteico-Postraduccional , Ratas
4.
Proc Natl Acad Sci U S A ; 118(50)2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34893541

RESUMEN

GBA1 mutations that encode lysosomal ß-glucocerebrosidase (GCase) cause the lysosomal storage disorder Gaucher disease (GD) and are strong risk factors for synucleinopathies, including Parkinson's disease and Lewy body dementia. Only a subset of subjects with GBA1 mutations exhibit neurodegeneration, and the factors that influence neurological phenotypes are unknown. We find that α-synuclein (α-syn) neuropathology induced by GCase depletion depends on neuronal maturity, the physiological state of α-syn, and specific accumulation of long-chain glycosphingolipid (GSL) GCase substrates. Reduced GCase activity does not initiate α-syn aggregation in neonatal mice or immature human midbrain cultures; however, adult mice or mature midbrain cultures that express physiological α-syn oligomers are aggregation prone. Accumulation of long-chain GSLs (≥C22), but not short-chain species, induced α-syn pathology and neurological dysfunction. Selective reduction of long-chain GSLs ameliorated α-syn pathology through lysosomal cathepsins. We identify specific requirements that dictate synuclein pathology in GD models, providing possible explanations for the phenotypic variability in subjects with GCase deficiency.


Asunto(s)
Glicoesfingolípidos/química , Glicoesfingolípidos/metabolismo , alfa-Sinucleína/metabolismo , Secuencia de Aminoácidos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Catepsinas/metabolismo , Diferenciación Celular , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/fisiología , Inositol/análogos & derivados , Inositol/toxicidad , Lisosomas/metabolismo , Ratones , Factores de Tiempo , alfa-Sinucleína/química , alfa-Sinucleína/genética
5.
N Engl J Med ; 381(17): 1644-1652, 2019 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-31597037

RESUMEN

Genome sequencing is often pivotal in the diagnosis of rare diseases, but many of these conditions lack specific treatments. We describe how molecular diagnosis of a rare, fatal neurodegenerative condition led to the rational design, testing, and manufacture of milasen, a splice-modulating antisense oligonucleotide drug tailored to a particular patient. Proof-of-concept experiments in cell lines from the patient served as the basis for launching an "N-of-1" study of milasen within 1 year after first contact with the patient. There were no serious adverse events, and treatment was associated with objective reduction in seizures (determined by electroencephalography and parental reporting). This study offers a possible template for the rapid development of patient-customized treatments. (Funded by Mila's Miracle Foundation and others.).


Asunto(s)
Proteínas de Transporte de Membrana/genética , Mutagénesis Insercional , Lipofuscinosis Ceroideas Neuronales/tratamiento farmacológico , Lipofuscinosis Ceroideas Neuronales/genética , Oligonucleótidos Antisentido/uso terapéutico , Medicina de Precisión , Enfermedades Raras/tratamiento farmacológico , Biopsia , Niño , Desarrollo Infantil , Descubrimiento de Drogas , Drogas en Investigación/uso terapéutico , Electroencefalografía , Femenino , Humanos , Pruebas Neuropsicológicas , ARN Mensajero , Convulsiones/diagnóstico , Convulsiones/tratamiento farmacológico , Piel/patología , Secuenciación Completa del Genoma
6.
J Neurosci ; 39(29): 5760-5772, 2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-31097622

RESUMEN

The accumulation of misfolded proteins is a common pathological feature of many neurodegenerative disorders, including synucleinopathies such as Parkinson's disease (PD), which is characterized by the presence of α-synuclein (α-syn)-containing Lewy bodies. However, although recent studies have investigated α-syn accumulation and propagation in neurons, the molecular mechanisms underlying α-syn transmission have been largely unexplored. Here, we examined a monogenic form of synucleinopathy caused by loss-of-function mutations in lysosomal ATP13A2/PARK9. These studies revealed that lysosomal exocytosis regulates intracellular levels of α-syn in human neurons. Loss of PARK9 function in patient-derived dopaminergic neurons disrupted lysosomal Ca2+ homeostasis, reduced lysosomal Ca2+ storage, increased cytosolic Ca2+, and impaired lysosomal exocytosis. Importantly, this dysfunction in lysosomal exocytosis impaired α-syn secretion from both axons and soma, promoting α-syn accumulation. However, activation of the lysosomal Ca2+ channel transient receptor potential mucolipin 1 (TRPML1) was sufficient to upregulate lysosomal exocytosis, rescue defective α-syn secretion, and prevent α-syn accumulation. Together, these results suggest that intracellular α-syn levels are regulated by lysosomal exocytosis in human dopaminergic neurons and may represent a potential therapeutic target for PD and other synucleinopathies.SIGNIFICANCE STATEMENT Parkinson's disease (PD) is the second most common neurodegenerative disease linked to the accumulation of α-synuclein (α-syn) in patient neurons. However, it is unclear what the mechanism might be. Here, we demonstrate a novel role for lysosomal exocytosis in clearing intracellular α-syn and show that impairment of this pathway by mutations in the PD-linked gene ATP13A2/PARK9 contributes to α-syn accumulation in human dopaminergic neurons. Importantly, upregulating lysosomal exocytosis by increasing lysosomal Ca2+ levels was sufficient to rescue defective α-syn secretion and accumulation in patient neurons. These studies identify lysosomal exocytosis as a potential therapeutic target in diseases characterized by the accumulation of α-syn, including PD.


Asunto(s)
Agonistas de los Canales de Calcio/farmacología , Neuronas Dopaminérgicas/metabolismo , Exocitosis/fisiología , Células Madre Pluripotentes Inducidas/metabolismo , Lisosomas/metabolismo , alfa-Sinucleína/toxicidad , Línea Celular Tumoral , Células Cultivadas , Neuronas Dopaminérgicas/efectos de los fármacos , Exocitosis/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Lisosomas/efectos de los fármacos , Lisosomas/genética , ATPasas de Translocación de Protón/genética , ATPasas de Translocación de Protón/metabolismo
7.
Neurobiol Dis ; 127: 147-162, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30790616

RESUMEN

Lysosomes are organelles involved in the degradation and recycling of macromolecules, and play a critical role in sensing metabolic information in the cell. A class of rare metabolic diseases called lysosomal storage disorders (LSD) are characterized by lysosomal dysfunction and the accumulation of macromolecular substrates. The central nervous system appears to be particularly vulnerable to lysosomal dysfunction, since many LSDs are characterized by severe, widespread neurodegeneration with pediatric onset. Furthermore, variants in lysosomal genes are strongly associated with some common neurodegenerative disorders such as Parkinson's disease (PD). To better understand disease pathology and develop novel treatment strategies, it is critical to study the fundamental molecular disease mechanisms in the affected cell types that harbor endogenously expressed mutations. The discovery of methods for reprogramming of patient-derived somatic cells into induced pluripotent stem cells (iPSCs), and their differentiation into distinct neuronal and glial cell types, have provided novel opportunities to study mechanisms of lysosomal dysfunction within the relevant, vulnerable cell types. These models also expand our ability to develop and test novel therapeutic targets. We discuss recently developed methods for iPSC differentiation into distinct neuronal and glial cell types, while addressing the need for meticulous experimental techniques and parameters that are essential to accurately identify inherent cellular pathologies. iPSC models for neuronopathic LSDs and their relationship to sporadic age-related neurodegeneration are also discussed. These models should facilitate the discovery and development of personalized therapies in the future.


Asunto(s)
Enfermedades por Almacenamiento Lisosomal/patología , Lisosomas/patología , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Células Cultivadas , Humanos
8.
Neurobiol Dis ; 122: 72-82, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29550539

RESUMEN

The finding that mutations in the Gaucher's Disease (GD) gene GBA1 are a strong risk factor for Parkinson's Disease (PD) has allowed for unique insights into pathophysiology centered on disruption of the autophagic-lysosomal pathway. Protein aggregations in the form of Lewy bodies and the effects of canonical PD mutations that converge on the lysosomal degradation system suggest that neurodegeneration in PD is mediated by dysregulation of protein homeostasis. The well-characterized clinical and pathological relationship between PD and the lysosomal storage disorder GD emphasizes the importance of dysregulated protein metabolism in neurodegeneration, and one intriguing piece of this relationship is a shared phenotype of autophagic-lysosomal dysfunction in both diseases. Translational application of these findings may be accelerated by the use of midbrain dopamine neuronal models derived from induced pluripotent stem cells (iPSCs) that recapitulate several pathological features of GD and PD. In this review, we discuss evidence linking autophagic dysfunction to the pathophysiology of GD and GBA1-linked parkinsonism and focus more specifically on studies performed recently in iPSC-derived neurons.


Asunto(s)
Autofagia/fisiología , Enfermedad de Gaucher/fisiopatología , Lisosomas/fisiología , Enfermedad de Parkinson/fisiopatología , Animales , Humanos
9.
Brain ; 141(8): 2255-2262, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29860491

RESUMEN

Common forms of Parkinson's disease have long been described as idiopathic, with no single penetrant genetic factor capable of influencing disease aetiology. Recent genetic studies indicate a clear association of variants within several lysosomal genes as risk factors for idiopathic Parkinson's disease. The emergence of novel variants suggest that the aetiology of idiopathic Parkinson's disease may be explained by the interaction of several partially penetrant mutations that, while seemingly complex, all appear to converge on cellular clearance pathways. These newly evolving data are consistent with mechanistic studies linking α-synuclein toxicity to lysosomal abnormalities, and indicate that idiopathic Parkinson's disease resembles features of Mendelian lysosomal storage disorders at a genetic and biochemical level. These findings offer novel pathways to exploit for the development of disease-altering therapies for idiopathic Parkinson's disease that target specific components of the lysosomal system.


Asunto(s)
Enfermedades por Almacenamiento Lisosomal/fisiopatología , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/fisiopatología , Enfermedad de Gaucher/genética , Enfermedad de Gaucher/fisiopatología , Humanos , Enfermedades por Almacenamiento Lisosomal/genética , Lisosomas/genética , Lisosomas/fisiología , Mitocondrias/patología , Enfermedad de Parkinson/genética , Factores de Riesgo , alfa-Sinucleína/metabolismo
10.
Proc Natl Acad Sci U S A ; 113(7): 1931-6, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26839413

RESUMEN

Parkinson's disease (PD) is an age-related neurodegenerative disorder characterized by the accumulation of protein aggregates comprised of α-synuclein (α-syn). A major barrier in treatment discovery for PD is the lack of identifiable therapeutic pathways capable of reducing aggregates in human neuronal model systems. Mutations in key components of protein trafficking and cellular degradation machinery represent important risk factors for PD; however, their precise role in disease progression and interaction with α-syn remains unclear. Here, we find that α-syn accumulation reduced lysosomal degradation capacity in human midbrain dopamine models of synucleinopathies through disrupting hydrolase trafficking. Accumulation of α-syn at the cell body resulted in aberrant association with cis-Golgi-tethering factor GM130 and disrupted the endoplasmic reticulum-Golgi localization of rab1a, a key mediator of vesicular transport. Overexpression of rab1a restored Golgi structure, improved hydrolase trafficking and activity, and reduced pathological α-syn in patient neurons. Our work suggests that enhancement of lysosomal hydrolase trafficking may prove beneficial in synucleinopathies and indicates that human midbrain disease models may be useful for identifying critical therapeutic pathways in PD and related disorders.


Asunto(s)
Lisosomas/fisiología , Mesencéfalo/patología , Modelos Biológicos , Enfermedad de Parkinson/fisiopatología , alfa-Sinucleína/fisiología , Humanos , Mesencéfalo/metabolismo , Enfermedad de Parkinson/metabolismo , Transporte de Proteínas
11.
Proc Natl Acad Sci U S A ; 113(14): 3791-6, 2016 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-27001828

RESUMEN

The lysosomal integral membrane protein type-2 (LIMP-2) plays a pivotal role in the delivery of ß-glucocerebrosidase (GC) to lysosomes. Mutations in GC result in Gaucher's disease (GD) and are the major genetic risk factor for the development of Parkinson's disease (PD). Variants in the LIMP-2 gene cause action myoclonus-renal failure syndrome and also have been linked to PD. Given the importance of GC and LIMP-2 in disease pathogenesis, we studied their interaction sites in more detail. Our previous data demonstrated that the crystal structure of LIMP-2 displays a hydrophobic three-helix bundle composed of helices 4, 5, and 7, of which helix 5 and 7 are important for ligand binding. Here, we identified a similar helical motif in GC through surface potential analysis. Coimmunoprecipitation and immunofluorescence studies revealed a triple-helical interface region within GC as critical for LIMP-2 binding and lysosomal transport. Based on these findings, we generated a LIMP-2 helix 5-derived peptide that precipitated and activated recombinant wild-type and GD-associated N370S mutant GC in vitro. The helix 5 peptide fused to a cell-penetrating peptide also activated endogenous lysosomal GC and reduced α-synuclein levels, suggesting that LIMP-2-derived peptides can be used to activate endogenous as well as recombinant wild-type or mutant GC efficiently. Our data also provide a structural model of the LIMP-2/GC complex that will facilitate the development of GC chaperones and activators as potential therapeutics for GD, PD, and related synucleinopathies.


Asunto(s)
Glucosilceramidasa/metabolismo , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Lisosomas/metabolismo , Secuencias de Aminoácidos/fisiología , Animales , Sitios de Unión , Células COS , Línea Celular , Chlorocebus aethiops , Cristalografía por Rayos X , Glucosilceramidasa/genética , Humanos , Proteína 2 de la Membrana Asociada a los Lisosomas/genética , Ratones , Unión Proteica , Estructura Terciaria de Proteína
12.
Cell Tissue Res ; 373(1): 51-60, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29064079

RESUMEN

Parkinson's disease (PD) is a neurodegenerative movement disorder characterized pathologically by the presence of Lewy bodies comprised of insoluble alpha (α)-synuclein. Pathological, clinical and genetic studies demonstrate that mutations in the GBA1 gene, which encodes the lysosomal enzyme glucocerebrosidase (GCase) that is deficient in Gaucher's disease, are important risk factors for the development of PD. The molecular mechanism for the association between these two diseases is not completely understood. We discuss several possible mechanisms that may lead to GBA1-related neuronal death and α-synuclein accumulation including disruptions in lipid metabolism, protein trafficking and impaired protein quality control mechanisms. Elucidating the mechanism between GCase and α-synuclein may provide insight into potential therapeutic pathways for PD and related synucleinopathies.


Asunto(s)
Glucosilceramidasa/metabolismo , Enfermedad de Parkinson/genética , alfa-Sinucleína/metabolismo , Animales , Humanos , Lisosomas/metabolismo , Modelos Biológicos , Mutación/genética , Enfermedad de Parkinson/terapia
13.
J Neurosci ; 36(29): 7693-706, 2016 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-27445146

RESUMEN

UNLABELLED: Parkinson's disease (PD) is characterized by the accumulation of α-synuclein (α-syn) within Lewy body inclusions in the nervous system. There are currently no disease-modifying therapies capable of reducing α-syn inclusions in PD. Recent data has indicated that loss-of-function mutations in the GBA1 gene that encodes lysosomal ß-glucocerebrosidase (GCase) represent an important risk factor for PD, and can lead to α-syn accumulation. Here we use a small-molecule modulator of GCase to determine whether GCase activation within lysosomes can reduce α-syn levels and ameliorate downstream toxicity. Using induced pluripotent stem cell (iPSC)-derived human midbrain dopamine (DA) neurons from synucleinopathy patients with different PD-linked mutations, we find that a non-inhibitory small molecule modulator of GCase specifically enhanced activity within lysosomal compartments. This resulted in reduction of GCase substrates and clearance of pathological α-syn, regardless of the disease causing mutations. Importantly, the reduction of α-syn was sufficient to reverse downstream cellular pathologies induced by α-syn, including perturbations in hydrolase maturation and lysosomal dysfunction. These results indicate that enhancement of a single lysosomal hydrolase, GCase, can effectively reduce α-syn and provide therapeutic benefit in human midbrain neurons. This suggests that GCase activators may prove beneficial as treatments for PD and related synucleinopathies. SIGNIFICANCE STATEMENT: The presence of Lewy body inclusions comprised of fibrillar α-syn within affected regions of PD brain has been firmly documented, however no treatments exist that are capable of clearing Lewy bodies. Here, we used a mechanistic-based approach to examine the effect of GCase activation on α-syn clearance in human midbrain DA models that naturally accumulate α-syn through genetic mutations. Small molecule-mediated activation of GCase was effective at reducing α-syn inclusions in neurons, as well as associated downstream toxicity, demonstrating a therapeutic effect. Our work provides an example of how human iPSC-derived midbrain models could be used for testing potential treatments for neurodegenerative disorders, and identifies GCase as a critical therapeutic convergence point for a wide range of synucleinopathies.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Glucosilceramidasa/metabolismo , Lisosomas/metabolismo , Mesencéfalo/patología , Enfermedad de Parkinson/patología , alfa-Sinucleína/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular Tumoral , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/ultraestructura , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Células Madre Pluripotentes Inducidas , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Mutación/genética , Neuroblastoma/patología , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , ATPasas de Translocación de Protón/metabolismo , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/patología , Sinaptofisina/metabolismo
14.
Proc Natl Acad Sci U S A ; 111(43): 15573-8, 2014 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-25316793

RESUMEN

Mutations within the lysosomal enzyme ß-glucocerebrosidase (GC) result in Gaucher disease and represent a major risk factor for developing Parkinson disease (PD). Loss of GC activity leads to accumulation of its substrate glucosylceramide and α-synuclein. Since lysosomal activity of GC is tightly linked to expression of its trafficking receptor, the lysosomal integral membrane protein type-2 (LIMP-2), we studied α-synuclein metabolism in LIMP-2-deficient mice. These mice showed an α-synuclein dosage-dependent phenotype, including severe neurological impairments and premature death. In LIMP-2-deficient brains a significant reduction in GC activity led to lipid storage, disturbed autophagic/lysosomal function, and α-synuclein accumulation mediating neurotoxicity of dopaminergic (DA) neurons, apoptotic cell death, and inflammation. Heterologous expression of LIMP-2 accelerated clearance of overexpressed α-synuclein, possibly through increasing lysosomal GC activity. In surviving DA neurons of human PD midbrain, LIMP-2 levels were increased, probably to compensate for lysosomal GC deficiency. Therefore, we suggest that manipulating LIMP-2 expression to increase lysosomal GC activity is a promising strategy for the treatment of synucleinopathies.


Asunto(s)
Glucosilceramidasa/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/enzimología , Tronco Encefálico/patología , Tronco Encefálico/ultraestructura , Gliosis/complicaciones , Gliosis/patología , Humanos , Lípidos/química , Proteínas de Membrana de los Lisosomas/deficiencia , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lisosomas/patología , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Neuronas/ultraestructura , Neurotoxinas/toxicidad
15.
Anal Chem ; 88(4): 2399-405, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26813311

RESUMEN

Aging and oxidative stress are two prominent pathological mechanisms for Parkinson's disease (PD) that are strongly associated with the degeneration of dopamine (DA) neurons in the midbrain. DA and other catechols readily oxidize into highly reactive o-quinone species that are precursors of neuromelanin (NM) pigment and under pathological conditions can modify and damage macromolecules. The role of DA oxidation in PD pathogenesis remains unclear in part due to the lack of appropriate disease models and the absence of a simple method for the quantification of DA-derived oxidants. Here, we describe a rapid, simple, and reproducible method for the quantification of o-quinones in cells and tissues that relies on the near-infrared fluorescent properties of these species. Importantly, we demonstrate that catechol-derived oxidants can be quantified in human neuroblastoma cells and midbrain dopamine neurons derived from induced pluripotent stem cells, providing a novel model to study the downstream actions of o-quinones. This method should facilitate further study of oxidative stress and DA oxidation in PD and related diseases that affect the dopaminergic system.


Asunto(s)
Neuronas Dopaminérgicas/química , Fluorescencia , Rayos Infrarrojos , Neuroblastoma/química , Quinonas/análisis , Quinonas/química , Catecoles/química , Dopamina/química , Dopamina/metabolismo , Neuronas Dopaminérgicas/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Mesencéfalo/citología , Neuroblastoma/patología , Oxidación-Reducción , Estrés Oxidativo , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología
16.
Hum Mol Genet ; 22(10): 2067-82, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23393156

RESUMEN

Mutations in ATP13A2 (PARK9), encoding a lysosomal P-type ATPase, are associated with both Kufor-Rakeb syndrome (KRS) and neuronal ceroid lipofuscinosis (NCL). KRS has recently been classified as a rare genetic form of Parkinson's disease (PD), whereas NCL is a lysosomal storage disorder. Although the transport activity of ATP13A2 has not been defined, in vitro studies show that its loss compromises lysosomal function, which in turn is thought to cause neuronal degeneration. To understand the role of ATP13A2 dysfunction in disease, we disrupted its gene in mice. Atp13a2(-/-) and Atp13a2(+/+) mice were tested behaviorally to assess sensorimotor and cognitive function at multiple ages. In the brain, lipofuscin accumulation, α-synuclein aggregation and dopaminergic pathology were measured. Behaviorally, Atp13a2(-/-) mice displayed late-onset sensorimotor deficits. Accelerated deposition of autofluorescent storage material (lipofuscin) was observed in the cerebellum and in neurons of the hippocampus and the cortex of Atp13a2(-/-) mice. Immunoblot analysis showed increased insoluble α-synuclein in the hippocampus, but not in the cortex or cerebellum. There was no change in the number of dopaminergic neurons in the substantia nigra or in striatal dopamine levels in aged Atp13a2(-/-) mice. These results show that the loss of Atp13a2 causes sensorimotor impairments, α-synuclein accumulation as occurs in PD and related synucleinopathies, and accumulation of lipofuscin deposits characteristic of NCL, thus providing the first direct demonstration that null mutations in Atp13a2 can cause pathological features of both diseases in the same organism.


Asunto(s)
Adenosina Trifosfatasas , Envejecimiento/metabolismo , Encéfalo/metabolismo , Retroalimentación Sensorial , Proteínas de la Membrana , Lipofuscinosis Ceroideas Neuronales/enzimología , Trastornos Parkinsonianos/enzimología , alfa-Sinucleína/metabolismo , Envejecimiento/genética , Envejecimiento/patología , Animales , Conducta Animal , Encéfalo/patología , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Humanos , Ratones , Ratones Mutantes , Lipofuscinosis Ceroideas Neuronales/genética , Lipofuscinosis Ceroideas Neuronales/patología , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/patología , ATPasas de Translocación de Protón , alfa-Sinucleína/genética
17.
Neuron ; 112(15): 2558-2580.e13, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-38761794

RESUMEN

Neurodegenerative diseases are commonly classified as proteinopathies that are defined by the aggregation of a specific protein. Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are classified as synucleinopathies since α-synuclein (α-syn)-containing inclusions histopathologically define these diseases. Unbiased biochemical analysis of PD and DLB patient material unexpectedly revealed novel pathological inclusions in the nucleus comprising adenosine-to-inosine (A-to-I)-edited mRNAs and NONO and SFPQ proteins. These inclusions showed no colocalization with Lewy bodies and accumulated at levels comparable to α-syn. NONO and SFPQ aggregates reduced the expression of the editing inhibitor ADAR3, increasing A-to-I editing mainly within human-specific, Alu-repeat regions of axon, synaptic, and mitochondrial transcripts. Inosine-containing transcripts aberrantly accumulated in the nucleus, bound tighter to recombinant purified SFPQ in vitro, and potentiated SFPQ aggregation in human dopamine neurons, resulting in a self-propagating pathological state. Our data offer new insight into the inclusion composition and pathophysiology of PD and DLB.


Asunto(s)
Enfermedad por Cuerpos de Lewy , Factor de Empalme Asociado a PTB , Enfermedad de Parkinson , Edición de ARN , Humanos , Enfermedad por Cuerpos de Lewy/metabolismo , Enfermedad por Cuerpos de Lewy/patología , Enfermedad por Cuerpos de Lewy/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Factor de Empalme Asociado a PTB/metabolismo , Factor de Empalme Asociado a PTB/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Inosina/metabolismo , Adenosina/metabolismo , Núcleo Celular/metabolismo , Masculino , Anciano , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Femenino , ARN Mensajero/metabolismo , alfa-Sinucleína/metabolismo , alfa-Sinucleína/genética , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Anciano de 80 o más Años
18.
Neuroscientist ; : 10738584241232963, 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38420922

RESUMEN

Parkinson's disease (PD) is a common age-related neurodegenerative disorder characterized by the loss of dopaminergic neurons in the midbrain. A hallmark of both familial and sporadic PD is the presence of Lewy body inclusions composed mainly of aggregated α-synuclein (α-syn), a presynaptic protein encoded by the SNCA gene. The mechanisms driving the relationship between α-syn accumulation and neurodegeneration are not completely understood, although recent evidence indicates that multiple branches of the proteostasis pathway are simultaneously perturbed when α-syn aberrantly accumulates within neurons. Studies from patient-derived midbrain cultures that develop α-syn pathology through the endogenous expression of PD-causing mutations show that proteostasis disruption occurs at the level of synthesis/folding in the endoplasmic reticulum (ER), downstream ER-Golgi trafficking, and autophagic-lysosomal clearance. Here, we review the fundamentals of protein transport, highlighting the specific steps where α-syn accumulation may intervene and the downstream effects on proteostasis. Current therapeutic efforts are focused on targeting single pathways or proteins, but the multifaceted pathogenic role of α-syn throughout the proteostasis pathway suggests that manipulating several targets simultaneously will provide more effective disease-modifying therapies for PD and other synucleinopathies.

19.
Nat Commun ; 15(1): 5206, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38897986

RESUMEN

Disrupted glucose metabolism and protein misfolding are key characteristics of age-related neurodegenerative disorders including Parkinson's disease, however their mechanistic linkage is largely unexplored. The hexosamine biosynthetic pathway utilizes glucose and uridine-5'-triphosphate to generate N-linked glycans required for protein folding in the endoplasmic reticulum. Here we find that Parkinson's patient midbrain cultures accumulate glucose and uridine-5'-triphosphate, while N-glycan synthesis rates are reduced. Impaired glucose flux occurred by selective reduction of the rate-limiting enzyme, GFPT2, through disrupted signaling between the unfolded protein response and the hexosamine pathway. Failure of the unfolded protein response and reduced N-glycosylation caused immature lysosomal hydrolases to misfold and accumulate, while accelerating glucose flux through the hexosamine pathway rescued hydrolase function and reduced pathological α-synuclein. Our data indicate that the hexosamine pathway integrates glucose metabolism with lysosomal activity, and its failure in Parkinson's disease occurs by uncoupling of the unfolded protein response-hexosamine pathway axis. These findings offer new methods to restore proteostasis by hexosamine pathway enhancement.


Asunto(s)
Vías Biosintéticas , Glucosa , Hexosaminas , Células Madre Pluripotentes Inducidas , Lisosomas , Mesencéfalo , Neuronas , Enfermedad de Parkinson , Respuesta de Proteína Desplegada , Humanos , Hexosaminas/biosíntesis , Hexosaminas/metabolismo , Lisosomas/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Neuronas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Mesencéfalo/metabolismo , Glucosa/metabolismo , Glicosilación , alfa-Sinucleína/metabolismo , Glutamina-Fructosa-6-Fosfato Transaminasa (Isomerizadora)/metabolismo , Glutamina-Fructosa-6-Fosfato Transaminasa (Isomerizadora)/genética
20.
Nat Commun ; 15(1): 264, 2024 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-38238311

RESUMEN

Alzheimer's disease (AD) is characterized by progressive neurodegeneration, but the specific events that cause cell death remain poorly understood. Death Induced by Survival gene Elimination (DISE) is a cell death mechanism mediated by short (s) RNAs acting through the RNA-induced silencing complex (RISC). DISE is thus a form of RNA interference, in which G-rich 6mer seed sequences in the sRNAs (position 2-7) target hundreds of C-rich 6mer seed matches in genes essential for cell survival, resulting in the activation of cell death pathways. Here, using Argonaute precipitation and RNAseq (Ago-RP-Seq), we analyze RISC-bound sRNAs to quantify 6mer seed toxicity in several model systems. In mouse AD models and aging brain, in induced pluripotent stem cell-derived neurons from AD patients, and in cells exposed to Aß42 oligomers, RISC-bound sRNAs show a shift to more toxic 6mer seeds compared to controls. In contrast, in brains of "SuperAgers", humans over age 80 who have superior memory performance, RISC-bound sRNAs are shifted to more nontoxic 6mer seeds. Cells depleted of nontoxic sRNAs are sensitized to Aß42-induced cell death, and reintroducing nontoxic RNAs is protective. Altogether, the correlation between DISE and Aß42 toxicity suggests that increasing the levels of nontoxic miRNAs in the brain or blocking the activity of toxic RISC-bound sRNAs could ameliorate neurodegeneration.


Asunto(s)
Enfermedad de Alzheimer , MicroARNs , Ratones , Animales , Humanos , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , MicroARNs/genética , Complejo Silenciador Inducido por ARN/genética , Interferencia de ARN , Envejecimiento/genética , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA