Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Brain ; 147(2): 427-443, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-37671615

RESUMEN

Mer tyrosine kinase (MerTK) is a receptor tyrosine kinase that mediates non-inflammatory, homeostatic phagocytosis of diverse types of cellular debris. Highly expressed on the surface of microglial cells, MerTK is of importance in brain development, homeostasis, plasticity and disease. Yet, involvement of this receptor in the clearance of protein aggregates that accumulate with ageing and in neurodegenerative diseases has yet to be defined. The current study explored the function of MerTK in the microglial uptake of alpha-synuclein fibrils which play a causative role in the pathobiology of synucleinopathies. Using human primary and induced pluripotent stem cell-derived microglia, the MerTK-dependence of alpha-synuclein fibril internalization was investigated in vitro. Relevance of this pathway in synucleinopathies was assessed through burden analysis of MERTK variants and analysis of MerTK expression in patient-derived cells and tissues. Pharmacological inhibition of MerTK and siRNA-mediated MERTK knockdown both caused a decreased rate of alpha-synuclein fibril internalization by human microglia. Consistent with the non-inflammatory nature of MerTK-mediated phagocytosis, alpha-synuclein fibril internalization was not observed to induce secretion of pro-inflammatory cytokines such as IL-6 or TNF, and downmodulated IL-1ß secretion from microglia. Burden analysis in two independent patient cohorts revealed a significant association between rare functionally deleterious MERTK variants and Parkinson's disease in one of the cohorts (P = 0.002). Despite a small upregulation in MERTK mRNA expression in nigral microglia from Parkinson's disease/Lewy body dementia patients compared to those from non-neurological control donors in a single-nuclei RNA-sequencing dataset (P = 5.08 × 10-21), no significant upregulation in MerTK protein expression was observed in human cortex and substantia nigra lysates from Lewy body dementia patients compared to controls. Taken together, our findings define a novel role for MerTK in mediating the uptake of alpha-synuclein fibrils by human microglia, with possible involvement in limiting alpha-synuclein spread in synucleinopathies such as Parkinson's disease. Upregulation of this pathway in synucleinopathies could have therapeutic values in enhancing alpha-synuclein fibril clearance in the brain.


Asunto(s)
Enfermedad por Cuerpos de Lewy , Enfermedad de Parkinson , Sinucleinopatías , Humanos , alfa-Sinucleína/metabolismo , Tirosina Quinasa c-Mer/metabolismo , Enfermedad por Cuerpos de Lewy/metabolismo , Microglía/metabolismo , Enfermedad de Parkinson/metabolismo , Proteínas Tirosina Quinasas , Sinucleinopatías/metabolismo
2.
Artículo en Inglés | MEDLINE | ID: mdl-37330108

RESUMEN

Fibrillary aggregated α-synuclein represents the neurologic hallmark of Parkinson's disease and is considered to play a causative role in the disease. Although the causes leading to α-synuclein aggregation are not clear, the GM1 ganglioside interaction is recognized to prevent this process. How GM1 exerts these functions is not completely clear, although a primary role of its soluble oligosaccharide (GM1-OS) is emerging. Indeed, we recently identified GM1-OS as the bioactive moiety responsible for GM1 neurotrophic and neuroprotective properties, specifically reverting the parkinsonian phenotype both in in vitro and in vivo models. Here, we report on GM1-OS efficacy against the α-synuclein aggregation and toxicity in vitro. By amyloid seeding aggregation assay and NMR spectroscopy, we demonstrated that GM1-OS was able to prevent both the spontaneous and the prion-like α-synuclein aggregation. Additionally, circular dichroism spectroscopy of recombinant monomeric α-synuclein showed that GM1-OS did not induce any change in α-synuclein secondary structure. Importantly, GM1-OS significantly increased neuronal survival and preserved neurite networks of dopaminergic neurons affected by α-synuclein oligomers, together with a reduction of microglia activation. These data further demonstrate that the ganglioside GM1 acts through its oligosaccharide also in preventing the α-synuclein pathogenic aggregation in Parkinson's disease, opening a perspective window for GM1-OS as drug candidate.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Humanos , alfa-Sinucleína/genética , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/patología , Gangliósido G(M1)/farmacología , Gangliósido G(M1)/química , Oligosacáridos/farmacología
3.
NPJ Parkinsons Dis ; 9(1): 4, 2023 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-36646701

RESUMEN

In Parkinson's disease and other synucleinopathies, the elevation of α-synuclein phosphorylated at Serine129 (pS129) is a widely cited marker of pathology. However, the physiological role for pS129 has remained undefined. Here we use multiple approaches to show for the first time that pS129 functions as a physiological regulator of neuronal activity. Neuronal activity triggers a sustained increase of pS129 in cultured neurons (200% within 4 h). In accord, brain pS129 is elevated in environmentally enriched mice exhibiting enhanced long-term potentiation. Activity-dependent α-synuclein phosphorylation is S129-specific, reversible, confers no cytotoxicity, and accumulates at synapsin-containing presynaptic boutons. Mechanistically, our findings are consistent with a model in which neuronal stimulation enhances Plk2 kinase activity via a calcium/calcineurin pathway to counteract PP2A phosphatase activity for efficient phosphorylation of membrane-bound α-synuclein. Patch clamping of rat SNCA-/- neurons expressing exogenous wild-type or phospho-incompetent (S129A) α-synuclein suggests that pS129 fine-tunes the balance between excitatory and inhibitory neuronal currents. Consistently, our novel S129A knock-in (S129AKI) mice exhibit impaired hippocampal plasticity. The discovery of a key physiological function for pS129 has implications for understanding the role of α-synuclein in neurotransmission and adds nuance to the interpretation of pS129 as a synucleinopathy biomarker.

4.
Acta Neuropathol ; 143(4): 453-469, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35141810

RESUMEN

The protein α-synuclein, a key player in Parkinson's disease (PD) and other synucleinopathies, exists in different physiological conformations: cytosolic unfolded aggregation-prone monomers and helical aggregation-resistant multimers. It has been shown that familial PD-associated missense mutations within the α-synuclein gene destabilize the conformer equilibrium of physiologic α-synuclein in favor of unfolded monomers. Here, we characterized the relative levels of unfolded and helical forms of cytosolic α-synuclein in post-mortem human brain tissue and showed that the equilibrium of α-synuclein conformations is destabilized in sporadic PD and DLB patients. This disturbed equilibrium is decreased in a brain region-specific manner in patient samples pointing toward a possible "prion-like" propagation of the underlying pathology and forms distinct disease-specific patterns in the two different synucleinopathies. We are also able to show that a destabilization of multimers mechanistically leads to increased levels of insoluble, pathological α-synuclein, while pharmacological stabilization of multimers leads to a "prion-like" aggregation resistance. Together, our findings suggest that these disease-specific patterns of α-synuclein multimer destabilization in sporadic PD and DLB are caused by both regional neuronal vulnerability and "prion-like" aggregation transmission enabled by the destabilization of local endogenous α-synuclein protein.


Asunto(s)
Enfermedad por Cuerpos de Lewy , Enfermedad de Parkinson , Priones , Sinucleinopatías , Encéfalo/patología , Humanos , Cuerpos de Lewy/patología , Enfermedad por Cuerpos de Lewy/patología , Enfermedad de Parkinson/patología , Priones/metabolismo , alfa-Sinucleína/metabolismo
5.
Front Neurosci ; 15: 639414, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33613189

RESUMEN

α-Synuclein is a presynaptic protein that regulates synaptic vesicle trafficking under physiological conditions. However, in several neurodegenerative diseases, including Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy, α-synuclein accumulates throughout the neuron, including at synapses, leading to altered synaptic function, neurotoxicity, and motor, cognitive, and autonomic dysfunction. Neurons typically contain both monomeric and multimeric forms of α-synuclein, and it is generally accepted that disrupting the balance between them promotes aggregation and neurotoxicity. However, it remains unclear how distinct molecular species of α-synuclein affect synapses where α-synuclein is normally expressed. Using the lamprey reticulospinal synapse model, we previously showed that acute introduction of excess recombinant monomeric or dimeric α-synuclein impaired distinct stages of clathrin-mediated synaptic vesicle endocytosis, leading to a loss of synaptic vesicles. Here, we expand this knowledge by investigating the effects of native, physiological α-synuclein isolated from the brain of a neuropathologically normal human subject, which comprised predominantly helically folded multimeric α-synuclein with a minor component of monomeric α-synuclein. After acute introduction of excess brain-derived human α-synuclein, there was a moderate reduction in the synaptic vesicle cluster and an increase in the number of large, atypical vesicles called "cisternae." In addition, brain-derived α-synuclein increased synaptic vesicle and cisternae sizes and induced atypical fusion/fission events at the active zone. In contrast to monomeric or dimeric α-synuclein, the brain-derived multimeric α-synuclein did not appear to alter clathrin-mediated synaptic vesicle endocytosis. Taken together, these data suggest that excess brain-derived human α-synuclein impairs intracellular vesicle trafficking and further corroborate the idea that different molecular species of α-synuclein produce distinct trafficking defects at synapses. These findings provide insights into the mechanisms by which excess α-synuclein contributes to synaptic deficits and disease phenotypes.

6.
Science ; 370(6512): 66-69, 2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-33004513

RESUMEN

Dementia is a rapidly rising global health crisis that silently disables families and ends lives and livelihoods around the world. To date, however, no early biomarkers or effective therapies exist. It is now clear that brain microglia are more than mere bystanders or amyloid phagocytes; they can act as governors of neuronal function and homeostasis in the adult brain. Here, we highlight the fundamental role of microglia as tissue-resident macrophages in neuronal health. Then, we suggest how chronic impairment in microglia-neuron cross-talk may secure the permanence of the failure of synaptic and neuronal function and health in Alzheimer's and Parkinson's diseases. Understanding how to assess and modulate microglia-neuron interactions critical for brain health will be key to developing effective therapies for dementia.


Asunto(s)
Enfermedad de Alzheimer/patología , Amiloide/metabolismo , Macrófagos/metabolismo , Microglía/metabolismo , Enfermedad de Parkinson/patología , Sinapsis/patología , Animales , Comunicación Celular , Humanos , Ratones , Neuronas/metabolismo , Sinaptosomas/patología , alfa-Sinucleína/metabolismo
7.
Brain Commun ; 2(1): fcaa010, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32280944

RESUMEN

Since researchers identified α-synuclein as the principal component of Lewy bodies and Lewy neurites, studies have suggested that it plays a causative role in the pathogenesis of dementia with Lewy bodies and other 'synucleinopathies'. While α-synuclein dyshomeostasis likely contributes to the neurodegeneration associated with the synucleinopathies, few direct biochemical analyses of α-synuclein from diseased human brain tissue currently exist. In this study, we analysed sequential protein extracts from a substantial number of patients with neuropathological diagnoses of dementia with Lewy bodies and corresponding controls, detecting a shift of cytosolic and membrane-bound physiological α-synuclein to highly aggregated forms. We then fractionated aqueous extracts (cytosol) from cerebral cortex using non-denaturing methods to search for soluble, disease-associated high molecular weight species potentially associated with toxicity. We applied these fractions and corresponding insoluble fractions containing Lewy-type aggregates to several reporter assays to determine their bioactivity and cytotoxicity. Ultimately, high molecular weight cytosolic fractions enhances phospholipid membrane permeability, while insoluble, Lewy-associated fractions induced morphological changes in the neurites of human stem cell-derived neurons. While the concentrations of soluble, high molecular weight α-synuclein were only slightly elevated in brains of dementia with Lewy bodies patients compared to healthy, age-matched controls, these observations suggest that a small subset of soluble α-synuclein aggregates in the brain may drive early pathogenic effects, while Lewy body-associated α-synuclein can drive neurotoxicity.

8.
Nat Neurosci ; 22(7): 1043-1045, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31235905
9.
J Biol Chem ; 294(25): 9799-9812, 2019 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-31048377

RESUMEN

Parkinson's disease (PD) is one of the most common neurodegenerative disorders, and both genetic and histopathological evidence have implicated the ubiquitous presynaptic protein α-synuclein (αSyn) in its pathogenesis. Recent work has investigated how disrupting αSyn's interaction with membranes triggers trafficking defects, cellular stress, and apoptosis. Special interest has been devoted to a series of mutants exacerbating the effects of the E46K mutation (associated with autosomal dominant PD) through homologous Glu-to-Lys substitutions in αSyn's N-terminal region (i.e. E35K and E61K). Such E46K-like mutants have been shown to cause dopaminergic neuron loss and severe but L-DOPA-responsive motor defects in mouse overexpression models, presenting enormous translational potential for PD and other "synucleinopathies." In this work, using a variety of biophysical techniques, we characterize the molecular pathology of E46K-like αSyn mutants by studying their structure and membrane-binding and remodeling abilities. We find that, although a slight increase in the mutants' avidity for synaptic vesicle-like membranes can be detected, most of their deleterious effects are connected to their complete disruption of αSyn's curvature selectivity. Indiscriminate binding can shift αSyn's subcellular localization away from its physiological interactants at the synaptic bouton toward trafficking vesicles and organelles, as observed in E46K-like cellular and murine models, as well as in human pathology. In conclusion, our findings suggest that a loss of curvature selectivity, rather than increased membrane affinity, could be the critical dyshomeostasis in synucleinopathies.


Asunto(s)
Membrana Celular/patología , Ácido Glutámico/química , Lípidos/análisis , Lisina/química , Proteínas Mutantes/metabolismo , Mutación , alfa-Sinucleína/metabolismo , Membrana Celular/metabolismo , Ácido Glutámico/genética , Humanos , Lípidos/química , Lisina/genética , Proteínas Mutantes/genética , alfa-Sinucleína/genética
10.
PLoS One ; 13(7): e0198715, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29995905

RESUMEN

N-terminal acetylation is one of the most common co- and post-translational modifications of the eukaryotic proteome and regulates numerous aspects of cellular physiology, such as protein folding, localization and turnover. In particular α-synuclein, whose dyshomeostasis has been tied to the pathogenesis of several neurodegenerative disorders, is completely Nα-acetylated in nervous tissue. In this work, building on previous reports, we develop and characterize a bacterial N-terminal acetylation system based on the expression of the yeast N-terminal acetyltransferase B (NatB) complex under the control of the PBAD (L-arabinose-inducible) promoter. We show its functionality and the ability to completely Nα-acetylate our model substrate α-synuclein both upon induction of the construct with L-arabinose and also by only relying on the constitutive expression of the NatB genes.


Asunto(s)
Acetiltransferasas/genética , Escherichia coli/genética , Vectores Genéticos/química , Plásmidos/química , Procesamiento Proteico-Postraduccional , alfa-Sinucleína/genética , Acetilación/efectos de los fármacos , Acetiltransferasas/metabolismo , Secuencia de Aminoácidos , Arabinosa/farmacología , Clonación Molecular , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/metabolismo , Humanos , Fragmentos de Péptidos/análisis , Plásmidos/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos , Proteolisis , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , alfa-Sinucleína/metabolismo
11.
FEBS Lett ; 592(9): 1464-1472, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29633780

RESUMEN

α-Synuclein (αSyn) is a key player in the pathogenesis of Parkinson's disease and other synucleinopathies. Here, we report the existence of a novel soluble α-helical conformer of αSyn, obtained through transient interaction with lipid interfaces, and propose dynamic oligomerization as the mechanism underlying its stability. The conformational space of αSyn appears to be highly context-dependent, and lipid bilayers might thus play crucial roles as molecular chaperones in a cellular environment.


Asunto(s)
Metabolismo de los Lípidos , Replegamiento Proteico , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica en Hélice alfa , Solubilidad
12.
Neurobiol Dis ; 111: 26-35, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29246723

RESUMEN

Missense mutations in the multi-domain kinase LRRK2 cause late onset familial Parkinson's disease. They most commonly with classic proteinopathy in the form of Lewy bodies and Lewy neurites comprised of insoluble α-synuclein, but in rare cases can also manifest tauopathy. The normal function of LRRK2 has remained elusive, as have the cellular consequences of its mutation. Data from LRRK2 null model organisms and LRRK2-inhibitor treated animals support a physiological role for LRRK2 in regulating lysosome function. Since idiopathic and LRRK2-linked PD are associated with the intraneuronal accumulation of protein aggregates, a series of critical questions emerge. First, how do pathogenic mutations that increase LRRK2 kinase activity affect lysosome biology in neurons? Second, are mutation-induced changes in lysosome function sufficient to alter the metabolism of α-synuclein? Lastly, are changes caused by pathogenic mutation sensitive to reversal with LRRK2 kinase inhibitors? Here, we report that mutation of LRRK2 induces modest but significant changes in lysosomal morphology and acidification, and decreased basal autophagic flux when compared to WT neurons. These changes were associated with an accumulation of detergent-insoluble α-synuclein and increased neuronal release of α-synuclein and were reversed by pharmacologic inhibition of LRRK2 kinase activity. These data demonstrate a critical and disease-relevant influence of native neuronal LRRK2 kinase activity on lysosome function and α-synuclein homeostasis. Furthermore, they also suggest that lysosome dysfunction, altered neuronal α-synuclein metabolism, and the insidious accumulation of aggregated protein over decades may contribute to pathogenesis in this late-onset form of familial PD.


Asunto(s)
Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Lisosomas/metabolismo , Mutación , Neuronas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Autofagia , Células Cultivadas , Humanos , Concentración de Iones de Hidrógeno , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/antagonistas & inhibidores , Lisosomas/patología , Ratones Transgénicos , Neuronas/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo
13.
Hum Mol Genet ; 26(18): 3466-3481, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28911198

RESUMEN

α-Synuclein (αS) forms round cytoplasmic inclusions in Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Evidence suggests a physiological function of αS in vesicle trafficking and release. In contrast to earlier tenets, recent work indicates that αS normally exists in cells in a dynamic equilibrium between monomers and tetramers/multimers. We engineered αS mutants incapable of multimerization, leading to excess monomers at vesicle membranes. By EM, such mutants induced prominent vesicle clustering, leading to round cytoplasmic inclusions. Immunogold labeling revealed abundant αS intimately associated with vesicles of varied size. Fluorescence microscopy with marker proteins showed that the αS-associated vesicles were of diverse endocytic and secretory origin. An αS '3K' mutant (E35K + E46K + E61K) that amplifies the PD/DLB-causing E46K mutation induced αS-rich vesicle clusters resembling the vesicle-rich areas of Lewy bodies, supporting pathogenic relevance. Mechanistically, E46K can increase αS vesicle binding via membrane-induced amphipathic helix formation, and '3K' further enhances this effect. Another engineered αS variant added hydrophobicity to the hydrophobic half of αS helices, thereby stabilizing αS-membrane interactions. Importantly, substituting charged for uncharged residues within the hydrophobic half of the stabilized helix not only reversed the strong membrane interaction of the multimer-abolishing αS variant but also restored multimerization and prevented the aberrant vesicle interactions. Thus, reversible αS amphipathic helix formation and dynamic multimerization regulate a normal function of αS at vesicles, and abrogating multimers has pathogenic consequences.


Asunto(s)
Cuerpos de Inclusión/metabolismo , Mutación , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Secuencia de Aminoácidos , Animales , Células Cultivadas , Secuencia Conservada , Humanos , Cuerpos de Inclusión/genética , Cuerpos de Lewy/genética , Cuerpos de Lewy/metabolismo , Enfermedad por Cuerpos de Lewy/genética , Enfermedad por Cuerpos de Lewy/metabolismo , Enfermedad por Cuerpos de Lewy/patología , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente/métodos , Neuronas/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Estructura Secundaria de Proteína
14.
Science ; 357(6354): 891-898, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28860381

RESUMEN

Copy number mutations implicate excess production of α-synuclein as a possibly causative factor in Parkinson's disease (PD). Using an unbiased screen targeting endogenous gene expression, we discovered that the ß2-adrenoreceptor (ß2AR) is a regulator of the α-synuclein gene (SNCA). ß2AR ligands modulate SNCA transcription through histone 3 lysine 27 acetylation of its promoter and enhancers. Over 11 years of follow-up in 4 million Norwegians, the ß2AR agonist salbutamol, a brain-penetrant asthma medication, was associated with reduced risk of developing PD (rate ratio, 0.66; 95% confidence interval, 0.58 to 0.76). Conversely, a ß2AR antagonist correlated with increased risk. ß2AR activation protected model mice and patient-derived cells. Thus, ß2AR is linked to transcription of α-synuclein and risk of PD in a ligand-specific fashion and constitutes a potential target for therapies.


Asunto(s)
Regulación de la Expresión Génica , Enfermedad de Parkinson/etnología , Enfermedad de Parkinson/genética , Receptores Adrenérgicos beta 2/metabolismo , alfa-Sinucleína/genética , Acetilación , Agonistas de Receptores Adrenérgicos beta 1/farmacología , Antagonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/uso terapéutico , Albuterol/farmacología , Albuterol/uso terapéutico , Animales , Línea Celular Tumoral , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica/efectos de los fármacos , Histonas/metabolismo , Humanos , Ligandos , Ratones , Fármacos Neuroprotectores/farmacología , Noruega/etnología , Enfermedad de Parkinson/tratamiento farmacológico , Regiones Promotoras Genéticas , Propranolol/farmacología , Propranolol/uso terapéutico , Receptores Adrenérgicos beta 2/genética , Riesgo , Sustancia Negra/metabolismo , Transcripción Genética/efectos de los fármacos
16.
Curr Opin Neurobiol ; 36: 15-22, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26282834

RESUMEN

α-Synuclein (αSyn) is a highly abundant neuronal protein whose exact structure and function are under debate. Misfolding and aggregation of this normally soluble, 140-residue polypeptide underlies a group of neurodegenerative disorders called synucleinopathies, including Parkinson's disease (PD) and dementia with Lewy bodies (DLB). The αSyn field has focused increasing attention on the hypotheses that certain aggregates of αSyn may be directly toxic to the neurons in which they arise and/or that aggregates can be released from some neurons and diffuse by undefined mechanisms to other neurons to seed αSyn in the recipient cells, thus propagating neuropathology by a non-cell autonomous process ('pathogenic spread'). While intense interest in these hypotheses has led to new approaches and tools to model aspects of the disorders, it is important to analyze which molecular events initiate αSyn aggregation inside neurons in the first place. Here, we review new insights into how neuronal αSyn homeostasis may be maintained under physiological conditions but perturbed by pathological factors.


Asunto(s)
Encéfalo/metabolismo , Enfermedad por Cuerpos de Lewy/metabolismo , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Membrana Celular , Homeostasis , Humanos , Enfermedades Neurodegenerativas/metabolismo , Multimerización de Proteína
17.
Proc Natl Acad Sci U S A ; 112(31): 9596-601, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26153422

RESUMEN

α-Synuclein (αS) is a highly abundant neuronal protein that aggregates into ß-sheet-rich inclusions in Parkinson's disease (PD). αS was long thought to occur as a natively unfolded monomer, but recent work suggests it also occurs normally in α-helix-rich tetramers and related multimers. To elucidate the fundamental relationship between αS multimers and monomers in living neurons, we performed systematic mutagenesis to abolish self-interactions and learn which structural determinants underlie native multimerization. Unexpectedly, tetramers/multimers still formed in cells expressing each of 14 sequential 10-residue deletions across the 140-residue polypeptide. We postulated compensatory effects among the six highly conserved and one to three additional αS repeat motifs (consensus: KTKEGV), consistent with αS and its homologs ß- and γ-synuclein all forming tetramers while sharing only the repeats. Upon inserting in-register missense mutations into six or more αS repeats, certain mutations abolished tetramer formation, shown by intact-cell cross-linking and independently by fluorescent-protein complementation. For example, altered repeat motifs KLKEGV, KTKKGV, KTKEIV, or KTKEGW did not support tetramerization, indicating the importance of charged or small residues. When we expressed numerous different in-register repeat mutants in human neural cells, all multimer-abolishing but no multimer-neutral mutants caused frank neurotoxicity akin to the proapoptotic protein Bax. The multimer-abolishing variants became enriched in buffer-insoluble cell fractions and formed round cytoplasmic inclusions in primary cortical neurons. We conclude that the αS repeat motifs mediate physiological tetramerization, and perturbing them causes PD-like neurotoxicity. Moreover, the mutants we describe are valuable tools for studying normal and pathological properties of αS and screening for tetramer-stabilizing therapeutics.


Asunto(s)
Mutación/genética , Neuronas/patología , Multimerización de Proteína , Secuencias Repetitivas de Aminoácido , alfa-Sinucleína/química , alfa-Sinucleína/toxicidad , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Muerte Celular/efectos de los fármacos , Secuencia Conservada , Reactivos de Enlaces Cruzados/farmacología , Humanos , Cuerpos de Inclusión/efectos de los fármacos , Cuerpos de Inclusión/metabolismo , Microscopía Fluorescente , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas Sprague-Dawley , Eliminación de Secuencia , Relación Estructura-Actividad , alfa-Sinucleína/genética
19.
Nat Commun ; 6: 7314, 2015 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-26076669

RESUMEN

ß-Sheet-rich α-synuclein (αS) aggregates characterize Parkinson's disease (PD). αS was long believed to be a natively unfolded monomer, but recent work suggests it also occurs in α-helix-rich tetramers. Crosslinking traps principally tetrameric αS in intact normal neurons, but not after cell lysis, suggesting a dynamic equilibrium. Here we show that freshly biopsied normal human brain contains abundant αS tetramers. The PD-causing mutation A53T decreases tetramers in mouse brain. Neurons derived from an A53T patient have decreased tetramers. Neurons expressing E46K do also, and adding 1-2 E46K-like mutations into the canonical αS repeat motifs (KTKEGV) further reduces tetramers, decreases αS solubility and induces neurotoxicity and round inclusions. The other three fPD missense mutations likewise decrease tetramer:monomer ratios. The destabilization of physiological tetramers by PD-causing missense mutations and the neurotoxicity and inclusions induced by markedly decreasing tetramers suggest that decreased α-helical tetramers and increased unfolded monomers initiate pathogenesis. Tetramer-stabilizing compounds should prevent this.


Asunto(s)
Encéfalo/metabolismo , Neuronas/metabolismo , Enfermedad de Parkinson/genética , alfa-Sinucleína/genética , Animales , Ensayo de Inmunoadsorción Enzimática , Humanos , Immunoblotting , Inmunohistoquímica , Células Madre Pluripotentes Inducidas , Ratones , Mutación Missense , Enfermedad de Parkinson/metabolismo , Estructura Cuaternaria de Proteína/genética , Ratas , Ratas Sprague-Dawley , alfa-Sinucleína/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...