Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Acta Neuropathol ; 146(2): 211-226, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37351604

RESUMEN

Two siblings with deletion mutation ∆K281 in MAPT developed frontotemporal dementia. At autopsy, numerous inclusions of hyperphosphorylated 3R Tau were present in neurons and glial cells of neocortex and some subcortical regions, including hippocampus, caudate/putamen and globus pallidus. The inclusions were argyrophilic with Bodian silver, but not with Gallyas-Braak silver. They were not labelled by an antibody specific for tau phosphorylated at S262 and/or S356. The inclusions were stained by luminescent conjugated oligothiophene HS-84, but not by bTVBT4. Electron cryo-microscopy revealed that the core of tau filaments was made of residues K254-F378 of 3R Tau and was indistinguishable from that of Pick's disease. We conclude that MAPT mutation ∆K281 causes Pick's disease.


Asunto(s)
Demencia Frontotemporal , Enfermedad de Pick , Humanos , Enfermedad de Pick/genética , Plata , Proteínas tau/genética , Proteínas tau/química , Demencia Frontotemporal/genética , Neuronas , Mutación/genética
2.
Sci Rep ; 12(1): 1163, 2022 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-35064139

RESUMEN

Accumulation of phosphorylated α-synuclein aggregates has been implicated in several diseases, such as Parkinson's disease (PD) and dementia with Lewy bodies (DLB), and is thought to spread in a prion-like manner. Elucidating the mechanisms of prion-like transmission of α-synuclein is important for the development of therapies for these diseases, but little is known about the details. Here, we injected α-synuclein fibrils into the brains of wild-type mice and examined the early phase of the induction of phosphorylated α-synuclein accumulation. We found that phosphorylated α-synuclein appeared within a few days after the intracerebral injection. It was observed initially in presynaptic regions and subsequently extended its localization to axons and cell bodies. These results suggest that extracellular α-synuclein fibrils are taken up into the presynaptic region and seed-dependently convert the endogenous normal α-synuclein that is abundant there to an abnormal phosphorylated form, which is then transported through the axon to the cell body.


Asunto(s)
Hipocampo/patología , Enfermedades Neurodegenerativas/patología , Sinapsis/patología , alfa-Sinucleína/metabolismo , Animales , Axones/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Humanos , Masculino , Ratones , Fosforilación , Cultivo Primario de Células , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Sinapsis/metabolismo , alfa-Sinucleína/administración & dosificación , alfa-Sinucleína/genética , alfa-Sinucleína/aislamiento & purificación
3.
Brain ; 145(1): 349-361, 2022 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-34515757

RESUMEN

The phenomenon of 'prion-like propagation' in which aggregates of abnormal amyloid-fibrilized protein propagate between neurons and spread pathology, is attracting attention as a new mechanism in neurodegenerative diseases. There is a strong correlation between the accumulation or spread of abnormal tau aggregates and the clinical symptoms of tauopathies. Microtubule-associated protein tau (MAPT) contains a microtubule-binding domain that consists of three or four repeats (3R/4R) due to alternative mRNA splicing of transcripts for the MAPT gene. Although a number of models for tau propagation have been reported, most use 4R human tau transgenic mice or adult wild-type mice expressing only endogenous 4R tau and these models have not been able to reproduce the pathology of Alzheimer's disease in which 3R and 4R tau accumulate simultaneously, or that of Pick's disease in which only 3R tau is aggregated. These deficiencies may reflect differences between human and rodent tau isoforms in the brain. To overcome this problem, we used genome editing techniques to generate mice that express an equal ratio of endogenous 3R and 4R tau, even after they become adults. We injected these mice with sarkosyl-insoluble fractions derived from the brains of human tauopathy patients such as those afflicted with Alzheimer's disease (3R and 4R tauopathy), corticobasal degeneration (4R tauopathy) or Pick's disease (3R tauopathy). At 8-9 months following intracerebral injection of mice, histopathological and biochemical analyses revealed that the abnormal accumulation of tau was seed-dependent, with 3R and 4R tau in Alzheimer's disease-injected brains, 4R tau only in corticobasal degeneration-injected brains and 3R tau only in Pick disease-injected brains, all of which contained isoforms related to those found in the injected seeds. The injected abnormal tau was seeded, and accumulated at the site of injection and at neural connections, predominantly within the same site. The abnormal tau newly accumulated was found to be endogenous in these mice and to have crossed the species barrier. Of particular importance, Pick's body-like inclusions were observed in Pick's disease-injected mice, and accumulations characteristic of Pick's disease were reproduced, suggesting that we have developed the first model that recapitulates the pathology of Pick's disease. These models are not only useful for elucidating the mechanism of propagation of tau pathology involving both 3R and 4R isoforms, but can also reproduce the pathology of tauopathies, which should lead to the discovery of new therapeutic agents.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Pick , Tauopatías , Enfermedad de Alzheimer/patología , Animales , Encéfalo/patología , Humanos , Ratones , Ratones Transgénicos , Enfermedad de Pick/patología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Tauopatías/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
4.
Acta Neuropathol Commun ; 9(1): 189, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34819144

RESUMEN

Peripheral administration (oral, intranasal, intraperitoneal, intravenous) of assembled A53T α-synuclein induced synucleinopathy in heterozygous mice transgenic for human mutant A53T α-synuclein (line M83). The same was the case when cerebellar extracts from a case of multiple system atrophy with type II α-synuclein filaments were administered intraperitoneally, intravenously or intramuscularly. We observed abundant immunoreactivity for pS129 α-synuclein in nerve cells and severe motor impairment, resulting in hindlimb paralysis and shortened lifespan. Filaments immunoreactive for pS129 α-synuclein were in evidence. A 70% loss of motor neurons was present five months after an intraperitoneal injection of assembled A53T α-synuclein or cerebellar extract with type II α-synuclein filaments from an individual with a neuropathologically confirmed diagnosis of multiple system atrophy. Microglial cells changed from a predominantly ramified to a dystrophic appearance. Taken together, these findings establish a close relationship between the formation of α-synuclein inclusions in nerve cells and neurodegeneration, accompanied by a shift in microglial cell morphology. Propagation of α-synuclein inclusions depended on the characteristics of both seeds and transgenically expressed protein.


Asunto(s)
Enfermedades Neurodegenerativas/patología , alfa-Sinucleína/metabolismo , alfa-Sinucleína/farmacología , Anciano , Animales , Animales Modificados Genéticamente , Miembro Posterior , Humanos , Inmunohistoquímica , Masculino , Ratones Mutantes Neurológicos , Microglía/patología , Neuronas Motoras/patología , Trastornos del Movimiento/patología , Atrofia de Múltiples Sistemas/patología , Mutación , Enfermedades Neurodegenerativas/inducido químicamente , Neuronas/metabolismo , Parálisis/inducido químicamente , Parálisis/patología , alfa-Sinucleína/administración & dosificación
5.
Methods Mol Biol ; 2322: 131-139, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34043199

RESUMEN

The propagation of assembled α-synuclein (αS) is key to understanding the pathological mechanisms of synucleinopathies such as Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy.Here we describe a nonhuman primate model of αS propagation using common marmosets (Callithrix jacchus) with an intracerebral injection of synthetic preformed αS fibrils. This protocol enables observation of the formation of phosphorylated αS pathology and its propagation three months after the injection.


Asunto(s)
alfa-Sinucleína/metabolismo , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , Encéfalo/patología , Callithrix , Modelos Animales de Enfermedad , Fosforilación/fisiología , Sinucleinopatías/metabolismo , Sinucleinopatías/patología
6.
Brain Pathol ; 31(5): e12952, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33754430

RESUMEN

Prion-like spreading of abnormal proteins is proposed to occur in neurodegenerative diseases, and the progression of α-synuclein (α-syn) deposits has been reported in the brains of animal models injected with synthetic α-syn fibrils or pathological α-syn prepared from patients with Parkinson's disease (PD) and dementia with Lewy bodies (DLB). However, α-syn transmission in nonhuman primates, which are more similar to humans, has not been fully clarified. Here, we injected synthetic human α-syn fibrils into the left striatum of a macaque monkey (Macaca fuscata). At 3 months after the injection, we examined neurodegeneration and α-syn pathology in the brain using α-syn epitope-specific antibodies, antiphosphorylated α-syn antibodies (pSyn#64 and pSer129), anti-ubiquitin antibodies, and anti-p62 antibodies. Immunohistochemical examination with pSyn#64, pSer129, and α-syn epitope-specific antibodies revealed Lewy bodies, massive α-syn-positive neuronal intracytoplasmic inclusions (NCIs), and neurites in the left putamen. These inclusions were also positive for ubiquitin and p62. LB509, a human-specific α-syn antibody targeting amino acid residues 115-122, showed limited immunoreactivity around the injection site. The left substantia nigra (SN) and the bilateral frontal cortex also contained some NCIs and neurites. The left hemisphere, including parietal/temporal cortex presented sparse α-syn pathology, and no immunoreactivity was seen in olfactory nerves, amygdala, hippocampus, or right parietal/temporal cortex. Neuronal loss and gliosis in regions with α-syn pathology were mild, except for the left striatum and SN. Our results indicate that abnormal α-syn fibrils propagate throughout the brain of M. fuscata via projection, association, and commissural fibers, though the progression of α-syn pathology is limited.


Asunto(s)
Encéfalo/patología , Cuerpos de Lewy/patología , Sinucleinopatías/metabolismo , alfa-Sinucleína/metabolismo , Animales , Cuerpos de Inclusión/patología , Macaca fuscata , Masculino , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Putamen/metabolismo , Sustancia Negra/metabolismo , Sustancia Negra/patología , Sinucleinopatías/patología
7.
Brain Commun ; 2(2): fcaa091, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33005889

RESUMEN

Accumulation of assembled tau protein in the central nervous system is characteristic of Alzheimer's disease and several other neurodegenerative diseases, called tauopathies. Recent studies have revealed that propagation of assembled tau is key to understanding the pathological mechanisms of these diseases. Mouse models of tau propagation are established by injecting human-derived tau seeds intracerebrally; nevertheless, these have a limitation in terms of regulation of availability. To date, no study has shown that synthetic assembled tau induce tau propagation in non-transgenic mice. Here we confirm that dextran sulphate, a sulphated glycosaminoglycan, induces the assembly of recombinant tau protein into filaments in vitro. As compared to tau filaments induced by heparin, those induced by dextran sulphate showed higher thioflavin T fluorescence and lower resistance to guanidine hydrochloride, which suggests that the two types of filaments have distinct conformational features. Unlike other synthetic filament seeds, intracerebral injection of dextran sulphate-induced assemblies of recombinant tau caused aggregation of endogenous murine tau in wild-type mice. AT8-positive tau was present at the injection site 1 month after injection, from where it spread to anatomically connected regions. Induced tau assemblies were also stained by anti-tau antibodies AT100, AT180, 12E8, PHF1, anti-pS396 and anti-pS422. They were thioflavin- and Gallyas-Braak silver-positive, indicative of amyloid. In biochemical analyses, accumulated sarkosyl-insoluble and hyperphosphorylated tau was observed in the injected mice. In conclusion, we revealed that intracerebral injection of synthetic full-length wild-type tau seeds prepared in the presence of dextran sulphate caused tau propagation in non-transgenic mice. These findings establish that propagation of tau assemblies does not require tau to be either mutant and/or overexpressed.

8.
J Biol Chem ; 295(19): 6652-6664, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32209651

RESUMEN

Assembled α-synuclein in nerve cells and glial cells is the defining pathological feature of neurodegenerative diseases called synucleinopathies. Seeds of α-synuclein can induce the assembly of monomeric protein. Here, we used sucrose gradient centrifugation and transiently transfected HEK 293T cells to identify the species of α-synuclein from the brains of homozygous, symptomatic mice transgenic for human mutant A53T α-synuclein (line M83) that seed aggregation. The most potent fractions contained Sarkosyl-insoluble assemblies enriched in filaments. We also analyzed six cases of idiopathic Parkinson's disease (PD), one case of familial PD, and six cases of multiple system atrophy (MSA) for their ability to induce α-synuclein aggregation. The MSA samples were more potent than those of idiopathic PD in seeding aggregation. We found that following sucrose gradient centrifugation, the most seed-competent fractions from PD and MSA brains are those that contain Sarkosyl-insoluble α-synuclein. The fractions differed between PD and MSA, consistent with the presence of distinct conformers of assembled α-synuclein in these different samples. We conclude that α-synuclein filaments are the main driving force for amplification and propagation of pathology in synucleinopathies.


Asunto(s)
Encéfalo/metabolismo , Sinucleinopatías/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Animales , Encéfalo/patología , Células HEK293 , Homocigoto , Humanos , Ratones , Ratones Transgénicos , Sinucleinopatías/genética , Sinucleinopatías/patología
9.
Nihon Yakurigaku Zasshi ; 154(6): 301-305, 2019.
Artículo en Japonés | MEDLINE | ID: mdl-31787680

RESUMEN

Accumulation of insoluble alpha-synuclein (αS) is a pathological hallmark of some progressive neurodegenerative diseases including Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy, collectively termed synucleinopathies. In diseased brain, αS forms ß-sheet-rich amyloid fibrils and it is accumulated in neurons or glial cells. A growing body of evidence suggests that spreading of αS pathology occur by prion-like propagation mechanisms. Our study revealed that intracerebral injection of synthetic αS amyloid fibrils into wild-type mice induced prion-like propagation of αS pathology at 1 month post injection, while injection of soluble αS did not induce αS pathology. Furthermore, injection of αS amyloid fibrils into αS knockout mice failed to induce any pathologies. We also have demonstrated that intracerebral injection of αS amyloid fibrils into small primates, adult common marmosets, resulted in spreading of αS pathologies and loss of TH-positive neurons. These in vivo experiments clearly indicate that αS amyloid fibrils has prion-like properties and it propagates through neural networks. The underlying mechanisms of αS propagation are poorly understood, however, αS propagation model animals would be useful in elucidating pathogenetic mechanisms and developing disease-modifying drugs for sporadic synucleinopathies.


Asunto(s)
Modelos Animales , Priones , Sinucleinopatías/patología , alfa-Sinucleína/química , Animales , Encéfalo/fisiopatología , Ratones , Ratones Noqueados
10.
Acta Neuropathol Commun ; 7(1): 148, 2019 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-31522685

RESUMEN

Synucleinopathies [Parkinson's disease (PD), dementia with Lewy bodies (DLB) and multiple system atrophy (MSA)] share filamentous α-synuclein assemblies in nerve cells and glial cells. We compared the abilities of brain extracts from MSA and PD patients to induce neuronal α-synuclein assembly and neurodegeneration following intracerebral injection in heterozygous mice transgenic for human mutant A53T α-synuclein. MSA extracts were more potent than PD extracts in inducing α-synuclein assembly and in causing neurodegeneration. MSA assemblies were Campbell-Switzer- and Gallyas-silver-positive, whereas PD assemblies were only Campbell-Switzer-positive, in confirmation of previous findings. However, induced α-synuclein inclusions were invariably Campbell-Switzer-positive and Gallyas-negative, irrespective of whether MSA or PD brain extracts were injected. The α-synuclein inclusions of non-injected homozygous mice transgenic for A53T α-synuclein were also Campbell-Switzer-positive and Gallyas-negative. These findings demonstrate that transgene expression and its intracellular environment dominated over the silver staining properties of the conformers of assembled α-synuclein.


Asunto(s)
Encéfalo/patología , Atrofia de Múltiples Sistemas/patología , Neuronas/patología , Enfermedad de Parkinson/patología , Tinción con Nitrato de Plata/métodos , alfa-Sinucleína/análisis , Animales , Química Encefálica/genética , Humanos , Ratones , Ratones Transgénicos , Atrofia de Múltiples Sistemas/genética , Neuronas/química , Enfermedad de Parkinson/genética , Técnicas Estereotáxicas , alfa-Sinucleína/administración & dosificación , alfa-Sinucleína/toxicidad
11.
Yakugaku Zasshi ; 139(7): 1007-1013, 2019.
Artículo en Japonés | MEDLINE | ID: mdl-31257247

RESUMEN

α-Synuclein (αS) is the major component of the filamentous inclusions that constitute the defining characteristic of neurodegenerative synucleinopathies, including Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. αS is deposited in a hyperphosphorylated and ubiquitinated form with a ß-sheet-rich fibrillar structure in diseased brains. In 2008, some researchers reported that embryonic neurons transplanted into Parkinson's disease brains had Lewy body-like pathologies, suggesting that pathological αS propagates from diseased neurons to young neurons. Subsequently, a growing body of evidence supported the cell-to-cell spread of αS pathologies. Recent studies have revealed that intracerebral injection of insoluble αS into wild-type mice can induce prion-like propagation of phosphorylated αS pathology even 1 month after injection, while injection into αS-knockout mice failed to induce any pathology. We also showed that intracerebral injection of insoluble αS into adult common marmoset brains results in the spreading of abundant αS pathology. These in vivo experiments clearly indicate that insoluble αS has prion-like properties and that it propagates through neural networks. The underlying mechanisms of αS propagation are still poorly understood, but αS propagation model animals could be helpful in elucidating the pathogenetic mechanisms and developing drugs for synucleinopathies.


Asunto(s)
Enfermedades Neurodegenerativas/metabolismo , Agregación Patológica de Proteínas , alfa-Sinucleína/metabolismo , Animales , Encéfalo/metabolismo , Callithrix , Humanos , Enfermedad por Cuerpos de Lewy , Ratones , Enfermedades Neurodegenerativas/etiología , Enfermedad de Parkinson , Fosforilación , Priones
12.
PLoS One ; 13(7): e0200763, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30021006

RESUMEN

Alpha-synuclein can form beta-sheet filaments, the accumulation of which plays a key role in the development of Parkinson's disease, dementia with Lewy bodies and multiple system atrophy. It has previously been shown that alpha-synuclein is a substrate for the HECT domain-containing ubiquitin ligase Nedd4, and is subject to ubiquitin-mediated endosomal degradation. We show here that alpha-synuclein filaments are much better substrates for ubiquitination in vitro than monomeric alpha-synuclein, and that this increased susceptibility cannot be mimicked by the mere clustering of monomers. Recognition by Nedd4 family enzymes is not through the conventional binding of PPxY-containing sequences to WW domains of the ligase, but it also involves C2 and HECT domains. The disease-causing alpha-synuclein mutant A53T is a much less efficient substrate for Nedd4 ligases than the wild-type protein. We suggest that preferential recognition, ubiquitination and degradation of beta-sheet-containing filaments may help to limit toxicity, and that A53T alpha-synuclein may be more toxic, at least in part because it avoids this fate.


Asunto(s)
Ubiquitina-Proteína Ligasas Nedd4/metabolismo , alfa-Sinucleína/metabolismo , Células HEK293 , Células HeLa , Humanos , Unión Proteica , Ubiquitinación/genética , Ubiquitinación/fisiología
13.
Hum Mol Genet ; 27(15): 2658-2670, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29750243

RESUMEN

Amyotrophic lateral sclerosis and frontotemporal lobar degeneration are neurodegenerative diseases characterized by accumulation of insoluble aggregates of phosphorylated 43 kDa TAR DNA-binding protein (TDP-43) and linked with abnormal expansion of a hexanucleotide repeat in an intron of chromosome 9 open reading frame 72 (C9ORF72). However, the relationship between C9ORF72 mutations and TDP-43 aggregation remains unknown. Non-ATG-dependent translation of C9ORF72 repeats produces dipeptide repeat proteins, which form p62-positive aggregates in cerebral cortex and cerebellum of patients. Here, we show that the formation of poly-GA protein inclusions induced intracellular aggregation of endogenous and exogenous TDP-43 in cultured cells. Poly-GA aggregation preceded accumulation of phosphorylated TDP-43. These inclusions induced intracellular aggregation of phosphorylated TDP-43, but not tau or α-synuclein. Formation of phosphorylated TDP-43 aggregates depends on the number of poly-GA repeats. Detergent-insoluble fraction from cells co-expressing poly-GA and TDP-43 could function as seeds for further TDP-43 aggregation. These findings suggest a novel pathogenic mechanism that poly-GA protein aggregation directly promotes pathogenic changes of TDP-43 without the formation of nuclear RNA foci containing GGGGCC repeat expansion or loss-of-function of the C9ORF72 protein.


Asunto(s)
Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Proteínas de Unión al ADN/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Células Cultivadas , Expansión de las Repeticiones de ADN , Dipéptidos/genética , Dipéptidos/metabolismo , Degeneración Lobar Frontotemporal/genética , Degeneración Lobar Frontotemporal/patología , Humanos , Fosforilación , Ácido Poliglutámico/metabolismo , Secuencias Repetitivas de Aminoácido , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo
14.
Mol Neurobiol ; 55(3): 1847-1860, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28229331

RESUMEN

The cellular prion protein, encoded by the gene Prnp, has been reported to be a receptor of ß-amyloid. Their interaction is mandatory for neurotoxic effects of ß-amyloid oligomers. In this study, we aimed to explore whether the cellular prion protein participates in the spreading of α-synuclein. Results demonstrate that Prnp expression is not mandatory for α-synuclein spreading. However, although the pathological spreading of α-synuclein can take place in the absence of Prnp, α-synuclein expanded faster in PrPC-overexpressing mice. In addition, α-synuclein binds strongly on PrPC-expressing cells, suggesting a role in modulating the effect of α-synuclein fibrils.


Asunto(s)
Neuronas/metabolismo , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Animales , Células Cultivadas , Células HEK293 , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Priónicas/genética , Proteínas Priónicas/metabolismo , Transporte de Proteínas/fisiología
15.
Neuropathology ; 38(1): 64-71, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28948653

RESUMEN

Intracellular inclusions composed of abnormal protein aggregates are one of the neuropathological features of neurodegenerative diseases, and the formation of intracellular aggregates is believed to be associated with neurodegeneration leading to the onset of these diseases. In typical or pure cases, characteristic pathologies with one particular protein, such as tau, alpha-synuclein or trans-activation response DNA protein 43 (TDP-43), can be observed in brains of patients. On the other hand, multiple protein pathologies co-exist in many cases, raising the possibility that they may influence each other reciprocally in the pathogenesis and progression of the diseases. However, the molecular mechanisms through which these proteins interact with each other and through which they are co-deposited in brains of patients remain poorly understood. In this review, we focus on the mechanisms of deposition of multiple pathological proteins, such as tau, alpha-synuclein and/or TDP-43, and on co-deposition models of these proteins in vitro and in vivo intended to recapitulate the multiple pathologies found in diseased brains.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Cuerpos de Inclusión/patología , Enfermedades Neurodegenerativas/patología , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Encéfalo/patología , Humanos , Cuerpos de Inclusión/metabolismo , Enfermedades Neurodegenerativas/metabolismo
17.
Acta Neuropathol Commun ; 5(1): 12, 2017 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-28148299

RESUMEN

α-Synuclein is a defining, key component of Lewy bodies and Lewy neurites in Parkinson's disease (PD) and dementia with Lewy bodies (DLB), as well as glial cytoplasmic inclusions in multiple system atrophy (MSA). The distribution and spreading of these pathologies are closely correlated with disease progression. Recent studies have revealed that intracerebral injection of synthetic α-synuclein fibrils or pathological α-synuclein prepared from DLB or MSA brains into wild-type or transgenic animal brains induced prion-like propagation of phosphorylated α-synuclein pathology. The common marmoset is a very small primate that is expected to be a useful model of human diseases. Here, we show that intracerebral injection of synthetic α-synuclein fibrils into adult wild-type marmoset brains (caudate nucleus and/or putamen) resulted in spreading of abundant α-synuclein pathologies, which were positive for various antibodies to α-synuclein, including phospho Ser129-specific antibody, anti-ubiquitin and anti-p62 antibodies, at three months after injection. Remarkably, robust Lewy body-like inclusions were formed in tyrosine hydroxylase (TH)-positive neurons in these marmosets, strongly suggesting the retrograde spreading of abnormal α-synuclein from striatum to substantia nigra. Moreover, a significant decrease in the numbers of TH-positive neurons was observed in the injection-side of the brain, where α-synuclein inclusions were deposited. Furthermore, most of the α-synuclein inclusions were positive for 1-fluoro-2,5-bis (3-carboxy-4-hydroxystyryl) benzene (FSB) and thioflavin-S, which are dyes widely used to visualize the presence of amyloid. Thus, injection of synthetic α-synuclein fibrils into brains of non-transgenic primates induced PD-like α-synuclein pathologies within only 3 months after injection. Finally, we provide evidence indicating that neurons with abnormal α-synuclein inclusions may be cleared by microglial cells. This is the first marmoset model for α-synuclein propagation. It should be helpful in studies to elucidate mechanisms of disease progression and in development and evaluation of disease-modifying drugs for α-synucleinopathies.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/patología , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología , alfa-Sinucleína/metabolismo , Animales , Benzotiazoles , Callithrix , Femenino , Inmunohistoquímica , Cuerpos de Lewy/metabolismo , Cuerpos de Lewy/patología , Microglía/metabolismo , Microglía/patología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Neuronas/metabolismo , Neuronas/patología , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Tiazoles/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , alfa-Sinucleína/administración & dosificación , alfa-Sinucleína/genética
18.
Pharmacol Ther ; 172: 22-33, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27916654

RESUMEN

Prion-like propagation of abnormal intracytoplasmic proteins, which are the defining features of major neurodegenerative disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD) and amyotrophic lateral sclerosis (ALS), has been proposed. A growing body of evidence strongly suggests that abnormal tau, α-synuclein and TDP-43 have prion-like properties, convert the corresponding normal proteins into abnormal forms, and are transmitted from cell to cell, spreading throughout the brain. This idea is extremely important not only for understanding the pathogenesis and progression of these diseases, but also for the development of molecular therapies. Since the distributions and spreading of the abnormal proteins are closely associated with disease symptoms and progression, gain-of-toxic-function of these proteins may affect the neurons and glial cells either directly or indirectly, or both. It is essential to regulate the aggregation of abnormal intracellular proteins and their cell-to-cell transmission in order to stop, or at least slow, the progression of these diseases.


Asunto(s)
Enfermedades Neurodegenerativas/fisiopatología , Priones/metabolismo , Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/terapia , Esclerosis Amiotrófica Lateral/fisiopatología , Esclerosis Amiotrófica Lateral/terapia , Animales , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Humanos , Enfermedades Neurodegenerativas/terapia , Enfermedad de Parkinson/fisiopatología , Enfermedad de Parkinson/terapia , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo
19.
Brain ; 140(2): 266-278, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27658420

RESUMEN

The abnormal aggregation of a small number of known proteins underlies the most common human neurodegenerative diseases. In tauopathies and synucleinopathies, the normally soluble intracellular proteins tau and α-synuclein become insoluble and filamentous. In recent years, non-cell autonomous mechanisms of aggregate formation have come to the fore, suggesting that nucleation-dependent aggregation may occur in a localized fashion in human tauopathies and synucleinopathies, followed by seed-dependent propagation. There is a long prodromal phase between the formation of protein aggregates and the appearance of the first clinical symptoms, which manifest only after extensive propagation, opening novel therapeutic avenues.


Asunto(s)
Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/fisiopatología , Proteínas Priónicas/metabolismo , Agregación Patológica de Proteínas , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Animales , Humanos
20.
Artículo en Inglés | MEDLINE | ID: mdl-27481772

RESUMEN

α-Synuclein, which is present as a small, soluble, cytosolic protein in healthy subjects, is converted to amyloid-like fibrils in diseases such as Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). Bulk synthesis of purified α-synuclein has made it more convenient to study the nature of the normal protein and the mechanism of its conversion to an abnormal form in vitro and in vivo. Synthetic α-synuclein fibrils and pathological α-synuclein from diseased brains can act as triggers to convert normal α-synuclein to an abnormal form via prion-like mechanisms. In this article, we describe the experimental pathologies of α-synuclein both in vitro and in vivo in human and animal models. Prion-like spreading of abnormal α-synuclein from cell to cell can account for the progression of these α-synucleinopathies.


Asunto(s)
Amiloide/metabolismo , Encéfalo/patología , alfa-Sinucleína/metabolismo , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Enfermedad por Cuerpos de Lewy/patología , Ratones , Atrofia de Múltiples Sistemas/patología , Enfermedad de Parkinson/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...