Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Neurobiol Dis ; 199: 106571, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38901781

RESUMEN

Leucine-rich repeat kinase 2 (LRRK2) is the most common gene responsible for familial Parkinson's disease (PD). The gene product of LRRK2 contains multiple protein domains, including armadillo repeat, ankyrin repeat, leucine-rich repeat (LRR), Ras-of-complex (ROC), C-terminal of ROC (COR), kinase, and WD40 domains. In this study, we performed genetic screening of LRRK2 in our PD cohort, detecting sixteen LRRK2 rare variants. Among them, we selected seven variants that are likely to be familial and characterized them in terms of LRRK2 protein function, along with clinical information and one pathological analysis. The seven variants were S1120P and N1221K in the LRR domain; I1339M, S1403R, and V1447M in the ROC domain; and I1658F and D1873H in the COR domain. The kinase activity of the LRRK2 variants N1221K, S1403R, V1447M, and I1658F toward Rab10, a well-known phosphorylation substrate, was higher than that of wild-type LRRK2. LRRK2 D1873H showed enhanced self-association activity, whereas LRRK2 S1403R and D1873H showed reduced microtubule-binding activity. Pathological analysis of a patient with the LRRK2 V1447M variant was also performed, which revealed Lewy pathology in the brainstem. No functional alterations in terms of kinase activity, self-association activity, and microtubule-binding activity were detected in LRRK2 S1120P and I1339M variants. However, the patient with PD carrying LRRK2 S1120P variant also had a heterozygous Glucosylceramidase beta 1 (GBA1) L444P variant. In conclusion, we characterized seven LRRK2 variants potentially associated with PD. Five of the seven variants in different LRRK2 domains exhibited altered properties in kinase activity, self-association, and microtubule-binding activity, suggesting that each domain variant may contribute to disease progression in different ways.

2.
Hum Mol Genet ; 31(15): 2623-2638, 2022 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-35313349

RESUMEN

The mitochondrial kinase PTEN-induced kinase 1 (PINK1) and cytosolic ubiquitin ligase (E3) Parkin/PRKN are involved in mitochondrial quality control responses. PINK1 phosphorylates ubiquitin and the Parkin ubiquitin-like (Ubl) domain at serine 65 and promotes Parkin activation and translocation to damaged mitochondria. Upon Parkin activation, the Ubl domain is ubiquitinated at lysine (K) 27 and K48 residues. However, the contribution of K27/K48 ubiquitination toward Parkin activity remains unclear. In this study, ubiquitination of K56 (corresponding to K27 in the human), K77 (K48 in the human) or both was blocked by generating Drosophila Parkin (dParkin) mutants to examine the effects of Parkin Ubl domain ubiquitination on Parkin activation in Drosophila. The dParkin, in which K56 was replaced with arginine (dParkin K56R), rescued pupal lethality in flies by co-expression with PINK1, whereas dParkin K77R could not. The dParkin K56R exhibited reduced abilities of mitochondrial fragmentation and motility arrest, which are mediated by degrading Parkin E3 substrates Mitofusin and Miro, respectively. Pathogenic dParkin K56N, unlike dParkin K56R, destabilized the protein, suggesting that not only was dParkin K56N non-ubiquitin-modified at K56, but also the structure of the Ubl domain for activation was largely affected. Ubiquitin attached to K27 of the Ubl domain during PINK1-mediated Parkin activation was likely to be phosphorylated because human Parkin K27R weakened Parkin self-binding and activation in trans. Therefore, our findings suggest a new mechanism of Parkin activation, where an activation complex is formed through phospho-ubiquitin attachment on the K27 residue of the Parkin Ubl domain.


Asunto(s)
Proteínas de Drosophila , Ubiquitina , Animales , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Humanos , Lisina , Fosforilación , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas , Ubiquitina/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
3.
J Neural Transm (Vienna) ; 131(6): 709-719, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38478097

RESUMEN

Twenty-five years have passed since the causative gene for familial Parkinson's disease (PD), Parkin (now PRKN), was identified in 1998; PRKN is the most common causative gene in young-onset PD. Parkin encodes a ubiquitin-protein ligase, and Parkin is involved in mitophagy, a type of macroautophagy, in concert with PTEN-induced kinase 1 (PINK1). Both gene products are also involved in mitochondrial quality control. Among the many genetic PD-causing genes discovered, discovering PRKN as a cause of juvenile-onset PD has significantly impacted other neurodegenerative disorders. This is because the involvement of proteolytic systems has been suggested as a common mechanism in neurodegenerative diseases in which inclusion body formation is observed. The discovery of the participation of PRKN in PD has brought attention to the involvement of the proteolytic system in neurodegenerative diseases. Our research group has successfully isolated and identified CHCHD2, which is involved in the mitochondrial electron transfer system, and prosaposin (PSAP), which is involved in the lysosomal system, in this Parkin mechanism. Hereditary PD is undoubtedly an essential clue to solitary PD, and at least 25 or so genes and loci have been reported so far. This number of genes indicates that PD is a very diverse group of diseases. Currently, the diagnosis of PD is based on clinical symptoms and imaging studies. Although highly accurate diagnostic criteria have been published, early diagnosis is becoming increasingly important in treatment strategies for neurodegenerative diseases. Here, we also describe biomarkers that our group is working on.


Asunto(s)
Biomarcadores , Enfermedad de Parkinson , Ubiquitina-Proteína Ligasas , Humanos , Enfermedad de Parkinson/genética , Ubiquitina-Proteína Ligasas/genética , Biomarcadores/metabolismo
4.
Hum Mol Genet ; 30(18): 1693-1710, 2021 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-33890983

RESUMEN

Type 2 diabetes mellitus (T2DM) has long been considered a risk factor for Alzheimer's disease (AD). However, the molecular links between T2DM and AD remain obscure. Here, we reported that serum-/glucocorticoid-regulated kinase 1 (SGK1) is activated by administering a chronic high-fat diet (HFD), which increases the risk of T2DM, and thus promotes Tau pathology via the phosphorylation of tau at Ser214 and the activation of a key tau kinase, namely, GSK-3ß, forming SGK1-GSK-3ß-tau complex. SGK1 was activated under conditions of elevated glucocorticoid and hyperglycemia associated with HFD, but not of fatty acid-mediated insulin resistance. Elevated expression of SGK1 in the mouse hippocampus led to neurodegeneration and impairments in learning and memory. Upregulation and activation of SGK1, SGK1-GSK-3ß-tau complex were also observed in the hippocampi of AD cases. Our results suggest that SGK1 is a key modifier of tau pathology in AD, linking AD to corticosteroid effects and T2DM.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Dieta Alta en Grasa/efectos adversos , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Activación Enzimática/genética , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Proteínas Inmediatas-Precoces/genética , Ratones , Ratones Transgénicos , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas tau/genética
5.
J Cell Sci ; 134(22)2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34704591

RESUMEN

Mammalian syntaxin 17 (Stx17) has several roles in processes other than membrane fusion, including in mitochondrial division, autophagosome formation and lipid droplet expansion. In contrast to conventional syntaxins, Stx17 has a long C-terminal hydrophobic region with a hairpin-like structure flanked by a basic amino acid-enriched C-terminal tail. Although Stx17 is one of the six ancient SNAREs and is present in diverse eukaryotic organisms, it has been lost in multiple lineages during evolution. In the present study, we compared the localization and function of fly and nematode Stx17s expressed in HeLa cells with those of human Stx17. We found that fly Stx17 predominantly localizes to the cytosol and mediates autophagy, but not mitochondrial division. Nematode Stx17, on the other hand, is predominantly present in mitochondria and facilitates mitochondrial division, but is irrelevant to autophagy. These differences are likely due to different structures in the C-terminal tail. Non-participation of fly Stx17 and nematode Stx17 in mitochondrial division and autophagy, respectively, was demonstrated in individual organisms. Our results provide an insight into the evolution of Stx17 in metazoa. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Fusión de Membrana , Proteínas SNARE , Animales , Autofagia , Células HeLa , Humanos , Proteínas Qa-SNARE/genética
6.
EMBO J ; 37(21)2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30237312

RESUMEN

PGAM5, a mitochondrial protein phosphatase that is genetically and biochemically linked to PINK1, facilitates mitochondrial division by dephosphorylating the mitochondrial fission factor Drp1. At the onset of mitophagy, PGAM5 is cleaved by PARL, a rhomboid protease that degrades PINK1 in healthy cells, and the cleaved form facilitates the engulfment of damaged mitochondria by autophagosomes by dephosphorylating the mitophagy receptor FUNDC1. Here, we show that the function and localization of PGAM5 are regulated by syntaxin 17 (Stx17), a mitochondria-associated membrane/mitochondria protein implicated in mitochondrial dynamics in fed cells and autophagy in starved cells. In healthy cells, loss of Stx17 causes PGAM5 aggregation within mitochondria and thereby failure of the dephosphorylation of Drp1, leading to mitochondrial elongation. In Parkin-mediated mitophagy, Stx17 is prerequisite for PGAM5 to interact with FUNDC1. Our results reveal that the Stx17-PGAM5 axis plays pivotal roles in mitochondrial division and PINK1/Parkin-mediated mitophagy.


Asunto(s)
Dinámicas Mitocondriales , Proteínas Mitocondriales/metabolismo , Mitofagia , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Qa-SNARE/metabolismo , Transducción de Señal , Autofagosomas/metabolismo , Dinaminas , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Células HeLa , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Metaloproteasas/genética , Metaloproteasas/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Mitocondriales/genética , Fosfoproteínas Fosfatasas/genética , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteolisis , Proteínas Qa-SNARE/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
7.
Mov Disord ; 37(10): 2075-2085, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35894540

RESUMEN

BACKGROUND: The α-Synuclein (α-Syn) V15A variant has been found in two Caucasian families with Parkinson's disease (PD). However, the significance of this missense variant remained unclear. OBJECTIVE: We sought to elucidate whether V15A could increase aggregation or change phospholipid affinity. METHODS: A sequencing analysis for the SNCA encoding α-Syn from 875 patients with PD and 324 control subjects was performed. Comparing with known pathogenic missense variants of α-Syn, A30P, and A53T, we analyzed the effects of V15A on binding to phospholipid membrane, self-aggregation, and seed-dependent aggregation in cultured cells. RESULTS: Genetic screening identified SNCA c.44 T>C (p.V15A) from two Japanese PD families. The missense variant V15A was extremely rare in several public databases and predicted as pathogenic using in silico tools. The amplification activity of α-Syn V15A fibrils was stronger than that of wild-type α-Syn fibrils. CONCLUSIONS: The discovery of the V15A variant from Japanese families reinforces the possibility that the V15A variant may be a causative variant for developing PD. V15A had a reduced affinity for phospholipids and increased propagation activity compared with wild-type. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Humanos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Línea Celular , Mutación Missense , Enfermedad de Parkinson/metabolismo , Fosfolípidos
8.
Proc Natl Acad Sci U S A ; 116(41): 20689-20699, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31548400

RESUMEN

Mutations in the iPLA2-VIA/PLA2G6 gene are responsible for PARK14-linked Parkinson's disease (PD) with α-synucleinopathy. However, it is unclear how iPLA2-VIA mutations lead to α-synuclein (α-Syn) aggregation and dopaminergic (DA) neurodegeneration. Here, we report that iPLA2-VIA-deficient Drosophila exhibits defects in neurotransmission during early developmental stages and progressive cell loss throughout the brain, including degeneration of the DA neurons. Lipid analysis of brain tissues reveals that the acyl-chain length of phospholipids is shortened by iPLA2-VIA loss, which causes endoplasmic reticulum (ER) stress through membrane lipid disequilibrium. The introduction of wild-type human iPLA2-VIA or the mitochondria-ER contact site-resident protein C19orf12 in iPLA2-VIA-deficient flies rescues the phenotypes associated with altered lipid composition, ER stress, and DA neurodegeneration, whereas the introduction of a disease-associated missense mutant, iPLA2-VIA A80T, fails to suppress these phenotypes. The acceleration of α-Syn aggregation by iPLA2-VIA loss is suppressed by the administration of linoleic acid, correcting the brain lipid composition. Our findings suggest that membrane remodeling by iPLA2-VIA is required for the survival of DA neurons and α-Syn stability.


Asunto(s)
Encéfalo/patología , Membrana Celular/patología , Neuronas Dopaminérgicas/patología , Proteínas de Drosophila/metabolismo , Fosfolipasas A2 Grupo X/metabolismo , Degeneración Nerviosa/patología , Enfermedad de Parkinson/patología , alfa-Sinucleína/química , Animales , Animales Modificados Genéticamente , Encéfalo/metabolismo , Membrana Celular/metabolismo , Neuronas Dopaminérgicas/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster , Estrés del Retículo Endoplásmico , Femenino , Fosfolipasas A2 Grupo VI/genética , Fosfolipasas A2 Grupo VI/metabolismo , Fosfolipasas A2 Grupo X/genética , Humanos , Masculino , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Degeneración Nerviosa/metabolismo , Enfermedad de Parkinson/metabolismo , Fosfolípidos/metabolismo , Transmisión Sináptica , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
9.
Hum Mol Genet ; 28(23): 3895-3911, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31600778

RESUMEN

Mutations in CHCHD2 are linked to a familial, autosomal dominant form of Parkinson's disease (PD). The gene product may regulate mitochondrial respiratory function. However, whether mitochondrial dysfunction induced by CHCHD2 mutations further yields α-synuclein pathology is unclear. Here, we provide compelling genetic evidence that mitochondrial dysfunction induced by PD-linked CHCHD2 T61I mutation promotes α-synuclein aggregation using brain autopsy, induced pluripotent stem cells (iPSCs) and Drosophila genetics. An autopsy of an individual with CHCHD2 T61I revealed widespread Lewy pathology with both amyloid plaques and neurofibrillary tangles that appeared in the brain stem, limbic regions and neocortex. A prominent accumulation of sarkosyl-insoluble α-synuclein aggregates, the extent of which was comparable to that of a case with α-synuclein (SNCA) duplication, was observed in CHCHD2 T61I brain tissue. The prion-like activity and morphology of α-synuclein fibrils from the CHCHD2 T61I brain tissue were similar to those of fibrils from SNCA duplication and sporadic PD brain tissues. α-Synuclein insolubilization was reproduced in dopaminergic neuron cultures from CHCHD2 T61I iPSCs and Drosophila lacking the CHCHD2 ortholog or expressing the human CHCHD2 T61I. Moreover, the combination of ectopic α-synuclein expression and CHCHD2 null or T61I enhanced the toxicity in Drosophila dopaminergic neurons, altering the proteolysis pathways. Furthermore, CHCHD2 T61I lost its mitochondrial localization by α-synuclein in Drosophila. The mislocalization of CHCHD2 T61I was also observed in the patient brain. Our study suggests that CHCHD2 is a significant mitochondrial factor that determines α-synuclein stability in the etiology of PD.


Asunto(s)
Proteínas de Unión al ADN/genética , Mutación con Pérdida de Función , Enfermedad de Parkinson/genética , Factores de Transcripción/genética , alfa-Sinucleína/química , Anciano , Animales , Autopsia , Encéfalo/metabolismo , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Drosophila , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mitocondrias/metabolismo , Neuronas/citología , Enfermedad de Parkinson/metabolismo , Linaje , Agregado de Proteínas , Estabilidad Proteica , Factores de Transcripción/metabolismo
10.
Genes Dev ; 27(2): 157-62, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23348839

RESUMEN

Eukaryotes employ elaborate mitochondrial quality control (MQC) to maintain the function of the power-generating organelle. Parkinson's disease-associated PINK1 and Parkin actively participate in MQC. However, the signaling events involved are largely unknown. Here we show that mechanistic target of rapamycin 2 (mTORC2) and Tricornered (Trc) kinases act downstream from PINK1 to regulate MQC. Trc is phosphorylated in mTORC2-dependent and mTORC2-independent manners and is specifically localized to mitochondria in response to PINK1, which regulates mTORC2 through mitochondrial complex-I activity. Genetically, mTORC2 and Trc act upstream of Parkin. Thus, multiplex kinase signaling is acting between PINK1 and Parkin to regulate MQC, a process highly conserved in mammals.


Asunto(s)
Proteínas de Drosophila/metabolismo , Mitocondrias/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Animales , Proteínas de Drosophila/genética , Humanos , Masculino , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética
11.
Int J Mol Sci ; 22(7)2021 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-33918221

RESUMEN

Leucine-rich repeat kinase 2 (LRRK2) is a major causative gene of late-onset familial Parkinson's disease (PD). The suppression of kinase activity is believed to confer neuroprotection, as most pathogenic variants of LRRK2 associated with PD exhibit increased kinase activity. We herein report a novel LRRK2 variant-p.G2294R-located in the WD40 domain, detected through targeted gene-panel screening in a patient with familial PD. The proband showed late-onset Parkinsonism with dysautonomia and a good response to levodopa, without cognitive decline or psychosis. Cultured cell experiments revealed that p.G2294R is highly destabilized at the protein level. The LRRK2 p.G2294R protein expression was upregulated in the patient's peripheral blood lymphocytes. However, macrophages differentiated from the same peripheral blood showed decreased LRRK2 protein levels. Moreover, our experiment indicated reduced phagocytic activity in the pathogenic yeasts and α-synuclein fibrils. This PD case presents an example wherein the decrease in LRRK2 activity did not act in a neuroprotective manner. Further investigations are needed in order to elucidate the relationship between LRRK2 expression in the central nervous system and the pathogenesis caused by altered LRRK2 activity.


Asunto(s)
Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Trastornos Parkinsonianos/genética , Anciano , Estudios de Casos y Controles , Femenino , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Trastornos Parkinsonianos/metabolismo
13.
Int J Mol Sci ; 21(9)2020 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-32392751

RESUMEN

Parkinson's disease (PD) is the second most common neurodegenerative disease; it is characterized by the loss of dopaminergic neurons in the midbrain and the accumulation of neuronal inclusions, mainly consisting of α-synuclein (α-syn) fibrils in the affected regions. The prion-like property of the pathological forms of α-syn transmitted via neuronal circuits has been considered inherent in the nature of PD. Thus, one of the potential targets in terms of PD prevention is the suppression of α-syn conversion from the functional form to pathological forms. Recent studies suggested that α-syn interacts with synaptic vesicle membranes and modulate the synaptic functions. A series of studies suggest that transient interaction of α-syn as multimers with synaptic vesicle membranes composed of phospholipids and other lipids is required for its physiological function, while an α-syn-lipid interaction imbalance is believed to cause α-syn aggregation and the resultant pathological α-syn conversion. Altered lipid metabolisms have also been implicated in the modulation of PD pathogenesis. This review focuses on the current literature reporting the role of lipids, especially phospholipids, and lipid metabolism in α-syn dynamics and aggregation processes.


Asunto(s)
Metabolismo de los Lípidos , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Animales , Humanos , Fosfolípidos/metabolismo , Vesículas Sinápticas/química , Vesículas Sinápticas/metabolismo , alfa-Sinucleína/química
14.
Hum Mol Genet ; 26(15): 2933-2948, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28482024

RESUMEN

Mutations of the retromer component Vps35 and endosomal kinase LRRK2 are linked to autosomal dominant forms of familial Parkinson's disease (PD). However, the physiological and pathological roles of Vps35 and LRRK2 in neuronal functions are poorly understood. Here, we demonstrated that the loss of Drosophila Vps35 (dVps35) affects synaptic vesicle recycling, dopaminergic synaptic release and sleep behavior associated with dopaminergic activity, which is rescued by the expression of wild-type dVps35 but not the PD-associated mutant dVps35 D647N. Drosophila LRRK2 dLRRK together with Rab5 and Rab11 is also implicated in synaptic vesicle recycling, and the manipulation of these activities improves the Vps35 synaptic phenotypes. These findings indicate that defects of synaptic vesicle recycling in which two late-onset PD genes, Vps35 and LRRK2, are involved could be key aspects of PD etiology.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animales , Animales Modificados Genéticamente , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Endocitosis/genética , Endocitosis/fisiología , Endosomas/metabolismo , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Mutación , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Transmisión Sináptica , Vesículas Sinápticas/genética , Vesículas Sinápticas/metabolismo , Proteínas de Transporte Vesicular/genética
15.
Hum Mol Genet ; 26(16): 3172-3185, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28541509

RESUMEN

The ubiquitin (Ub) kinase PINK1 and the E3 Ub ligase Parkin, two gene products associated with young-onset Parkinson's disease (PD), participate in mitochondrial quality control. The phosphorylation of mitochondrial polyUb by PINK1, which is activated in a mitochondrial membrane potential (ΔΨm)-dependent manner, facilitates the mitochondrial translocation and concomitant enzymatic activation of Parkin, leading to the clearance of phospho-polyUb-tagged mitochondria via mitophagy. Thus, Ub phosphorylation is a key event in PINK1-Parkin-mediated mitophagy. Here, we examined the role of phospho-Ub signaling in the pathogenesis of PD using fly PD models, human brain tissue and dopaminergic neurons derived from induced pluripotent stem cells (iPSCs) containing Parkin or PINK1 mutations, as well as normal controls. We report that phospho-Ub signaling is highly conserved between humans and Drosophila, and that phospho-Ub signaling and the relocation of axonal mitochondria upon ΔΨm reduction are indeed compromised in human dopaminergic neurons containing Parkin or PINK1 mutations. Moreover, phospho-Ub signaling is prominent in tyrosine hydroxylase-positive neurons compared with tyrosine hydroxylase-negative neurons, suggesting that PINK1-Parkin signaling is more required for dopaminergic neurons. These results shed light on the particular vulnerability of dopaminergic neurons to mitochondrial stress.


Asunto(s)
Enfermedad de Parkinson/genética , Proteínas Quinasas/genética , Ubiquitina/metabolismo , Animales , Encéfalo/metabolismo , Neuronas Dopaminérgicas/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Activación Enzimática , Células HeLa , Humanos , Potencial de la Membrana Mitocondrial , Mitocondrias/metabolismo , Enfermedad de Parkinson/etiología , Fosforilación , Proteínas Quinasas/metabolismo , Transporte de Proteínas , Transducción de Señal , Ubiquitina/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
16.
Int J Mol Sci ; 20(4)2019 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-30791515

RESUMEN

Mutations of coiled-coil-helix-coiled-coil-helix domain containing 2 (CHCHD2) and 10 (CHCHD10) have been found to be linked to Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and/or frontotemporal lobe dementia (FTD). CHCHD2 and CHCHD10 proteins, which are homologous proteins with 54% identity in amino acid sequence, belong to the mitochondrial coiled-coil-helix-coiled-coil-helix (CHCH) domain protein family. A series of studies reveals that these twin proteins form a multimodal complex, producing a variety of pathophysiology by the disease-causing variants of these proteins. In this review, we summarize the present knowledge about the physiological and pathological roles of twin proteins, CHCHD2 and CHCHD10, in neurodegenerative diseases.


Asunto(s)
Esclerosis Amiotrófica Lateral/etiología , Demencia Frontotemporal/etiología , Proteínas Mitocondriales/genética , Enfermedad de Parkinson/etiología , Factores de Transcripción/genética , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Proteínas de Unión al ADN , Susceptibilidad a Enfermedades , Demencia Frontotemporal/metabolismo , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/química , Proteínas Mitocondriales/metabolismo , Mutación , Enfermedad de Parkinson/metabolismo , Unión Proteica , Transporte de Proteínas , Transducción de Señal , Relación Estructura-Actividad , Factores de Transcripción/química , Factores de Transcripción/metabolismo
17.
PLoS Genet ; 11(9): e1005503, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26355680

RESUMEN

Leucine-rich repeat kinase 2 (LRRK2) is a key molecule in the pathogenesis of familial and idiopathic Parkinson's disease (PD). We have identified two novel LRRK2-associated proteins, a HECT-type ubiquitin ligase, HERC2, and an adaptor-like protein with six repeated Neuralized domains, NEURL4. LRRK2 binds to NEURL4 and HERC2 via the LRRK2 Ras of complex proteins (ROC) domain and NEURL4, respectively. HERC2 and NEURL4 link LRRK2 to the cellular vesicle transport pathway and Notch signaling, through which the LRRK2 complex promotes the recycling of the Notch ligand Delta-like 1 (Dll1)/Delta (Dl) through the modulation of endosomal trafficking. This process negatively regulates Notch signaling through cis-inhibition by stabilizing Dll1/Dl, which accelerates neural stem cell differentiation and modulates the function and survival of differentiated dopaminergic neurons. These effects are strengthened by the R1441G ROC domain-mutant of LRRK2. These findings suggest that the alteration of Notch signaling in mature neurons is a component of PD etiology linked to LRRK2.


Asunto(s)
Endosomas/metabolismo , Enfermedad de Parkinson/enzimología , Proteínas Serina-Treonina Quinasas/fisiología , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Animales , Proteínas Portadoras/metabolismo , Dopamina/metabolismo , Drosophila , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HEK293 , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Ubiquitina-Proteína Ligasas
18.
Adv Exp Med Biol ; 997: 157-169, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28815529

RESUMEN

Parkinson's disease (PD) is a common neurodegenerative disorder, with ageing being a major risk factor. Accordingly, estimates predict an increasing number of PD patients due to our expanding life span. Consequently, developing a true disease-modifying therapy is necessary. In this regard, monogenic PD offers a suitable means for determining pathogenesis. Among monogenic forms of PD, mitochondrial dysfunction may be a major cause and is also likely to be involved in sporadic PD. Thus, mitochondrial impairment may be a common pathway. Recently, mitochondria-associated membranes (MAM) were identified as dynamic sites between mitochondria and endoplasmic reticulum. Indeed, the gene product of α-synuclein is a major component of MAM, with other gene products also involved. This review focuses on the possibility of using MAM as novel therapeutic targets.


Asunto(s)
Encéfalo/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/metabolismo , Enfermedad de Parkinson/metabolismo , Transducción de Señal , Animales , Transporte Biológico , Encéfalo/patología , Retículo Endoplásmico/patología , Metabolismo Energético , Humanos , Microdominios de Membrana/metabolismo , Microdominios de Membrana/patología , Proteínas de Transporte de Membrana/genética , Mitocondrias/patología , Dinámicas Mitocondriales , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Membranas Mitocondriales/patología , Proteínas Mitocondriales/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología
19.
PLoS Genet ; 10(6): e1004391, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24901221

RESUMEN

Two genes linked to early onset Parkinson's disease, PINK1 and Parkin, encode a protein kinase and a ubiquitin-ligase, respectively. Both enzymes have been suggested to support mitochondrial quality control. We have reported that Parkin is phosphorylated at Ser65 within the ubiquitin-like domain by PINK1 in mammalian cultured cells. However, it remains unclear whether Parkin phosphorylation is involved in mitochondrial maintenance and activity of dopaminergic neurons in vivo. Here, we examined the effects of Parkin phosphorylation in Drosophila, in which the phosphorylation residue is conserved at Ser94. Morphological changes of mitochondria caused by the ectopic expression of wild-type Parkin in muscle tissue and brain dopaminergic neurons disappeared in the absence of PINK1. In contrast, phosphomimetic Parkin accelerated mitochondrial fragmentation or aggregation and the degradation of mitochondrial proteins regardless of PINK1 activity, suggesting that the phosphorylation of Parkin boosts its ubiquitin-ligase activity. A non-phosphorylated form of Parkin fully rescued the muscular mitochondrial degeneration due to the loss of PINK1 activity, whereas the introduction of the non-phosphorylated Parkin mutant in Parkin-null flies led to the emergence of abnormally fused mitochondria in the muscle tissue. Manipulating the Parkin phosphorylation status affected spontaneous dopamine release in the nerve terminals of dopaminergic neurons, the survivability of dopaminergic neurons and flight activity. Our data reveal that Parkin phosphorylation regulates not only mitochondrial function but also the neuronal activity of dopaminergic neurons in vivo, suggesting that the appropriate regulation of Parkin phosphorylation is important for muscular and dopaminergic functions.


Asunto(s)
Proteínas de Drosophila/metabolismo , Mitocondrias/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/genética , Drosophila melanogaster , Complejo I de Transporte de Electrón/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias/genética , Músculo Esquelético/inervación , Músculo Esquelético/patología , Enfermedad de Parkinson/genética , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Ubiquitina-Proteína Ligasas/biosíntesis , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Alas de Animales/inervación , Alas de Animales/patología , Proteínas de Unión al GTP rho/metabolismo
20.
PLoS Genet ; 10(12): e1004861, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25474007

RESUMEN

The kinase PINK1 and the E3 ubiquitin (Ub) ligase Parkin participate in mitochondrial quality control. The phosphorylation of Ser65 in Parkin's ubiquitin-like (UBl) domain by PINK1 stimulates Parkin activation and translocation to damaged mitochondria, which induces mitophagy generating polyUb chain. However, Parkin Ser65 phosphorylation is insufficient for Parkin mitochondrial translocation. Here we report that Ser65 in polyUb chain is also phosphorylated by PINK1, and that phosphorylated polyUb chain on mitochondria tethers Parkin at mitochondria. The expression of Tom70MTS-4xUb SE, which mimics phospho-Ser65 polyUb chains on the mitochondria, activated Parkin E3 activity and its mitochondrial translocation. An E3-dead form of Parkin translocated to mitochondria with reduced membrane potential in the presence of Tom70(MTS)-4xUb SE, whereas non-phospho-polyUb mutant Tom70(MTS)-4xUb SA abrogated Parkin translocation. Parkin binds to the phospho-polyUb chain through its RING1-In-Between-RING (IBR) domains, but its RING0-linker is also required for mitochondrial translocation. Moreover, the expression of Tom70(MTS)-4xUb SE improved mitochondrial degeneration in PINK1-deficient, but not Parkin-deficient, Drosophila. Our study suggests that the phosphorylation of mitochondrial polyUb by PINK1 is implicated in both Parkin activation and mitochondrial translocation, predicting a chain reaction mechanism of mitochondrial phospho-polyUb production by which rapid translocation of Parkin is achieved.


Asunto(s)
Mitocondrias/metabolismo , Poliubiquitina/metabolismo , Proteínas Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Animales Modificados Genéticamente , Células Cultivadas , Drosophila , Células HEK293 , Células HeLa , Humanos , Ratones , Fosforilación , Unión Proteica , Transporte de Proteínas , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA