Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Cell ; 154(1): 103-17, 2013 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-23827677

RESUMEN

Many neurodegenerative diseases are characterized by the accumulation of insoluble protein aggregates, including neurofibrillary tangles comprised of tau in Alzheimer's disease and Lewy bodies composed of α-synuclein in Parkinson's disease. Moreover, different pathological proteins frequently codeposit in disease brains. To test whether aggregated α-synuclein can directly cross-seed tau fibrillization, we administered preformed α-synuclein fibrils assembled from recombinant protein to primary neurons and transgenic mice. Remarkably, we discovered two distinct strains of synthetic α-synuclein fibrils that demonstrated striking differences in the efficiency of cross-seeding tau aggregation, both in neuron cultures and in vivo. Proteinase K digestion revealed conformational differences between the two synthetic α-synuclein strains and also between sarkosyl-insoluble α-synuclein extracted from two subgroups of Parkinson's disease brains. We speculate that distinct strains of pathological α-synuclein likely exist in neurodegenerative disease brains and may underlie the tremendous heterogeneity of synucleinopathies.


Asunto(s)
Neuronas/metabolismo , Enfermedad de Parkinson/patología , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Amiloide/química , Amiloide/metabolismo , Animales , Células Cultivadas , Embrión de Mamíferos/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , Enfermedad de Parkinson/metabolismo , Proteínas Recombinantes/metabolismo , alfa-Sinucleína/química
2.
Acta Neuropathol ; 141(2): 193-215, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33385254

RESUMEN

The microtubule-associated protein tau (tau) forms hyperphosphorylated aggregates in the brains of tauopathy patients that can be pathologically and biochemically defined as distinct tau strains. Recent studies show that these tau strains exhibit strain-specific biological activities, also referred to as pathogenicities, in the tau spreading models. Currently, the specific pathogenicity of human-derived tau strains cannot be fully recapitulated by synthetic tau preformed fibrils (pffs), which are generated from recombinant tau protein. Reproducing disease-relevant tau pathology in cell and animal models necessitates the use of human brain-derived tau seeds. However, the availability of human-derived tau is extremely limited. Generation of tau variants that can mimic the pathogenicity of human-derived tau seeds would significantly extend the scale of experimental design within the field of tauopathy research. Previous studies have demonstrated that in vitro seeding reactions can amplify the beta-sheet structure of tau protein from a minute quantity of human-derived tau. However, whether the strain-specific pathogenicities of the original, human-derived tau seeds are conserved in the amplified tau strains has yet to be experimentally validated. Here, we used biochemically enriched brain-derived tau seeds from Alzheimer's disease (AD), corticobasal degeneration (CBD) and progressive supranuclear palsy (PSP) patient brains with a modified seeding protocol to template the recruitment of recombinant 2N4R (T40) tau in vitro. We quantitatively interrogated efficacy of the amplification reactions and the pathogenic fidelity of the amplified material to the original tau seeds using recently developed sporadic tau spreading models. Our data suggest that different tau strains can be faithfully amplified in vitro from tau isolated from different tauopathy brains and that the amplified tau variants retain their strain-dependent pathogenic characteristics.


Asunto(s)
Tauopatías/patología , Proteínas tau/genética , Enfermedad de Alzheimer/patología , Animales , Encéfalo/patología , Células Cultivadas , Secuencia Conservada , Amplificación de Genes , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Enfermedades Neurodegenerativas/patología , Ovillos Neurofibrilares/patología , Cultivo Primario de Células , Parálisis Supranuclear Progresiva/patología
3.
J Neurosci ; 39(26): 5080-5094, 2019 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-31036761

RESUMEN

Synucleinopathies are characterized by the accumulation of insoluble α-synuclein (αSyn). To test whether αSyn aggregates modulate synaptic activity, we used a recently developed model in primary neurons for inducing αSyn pathology. We demonstrated that preformed fibrils (PFFs) generated with recombinant human αSyn compromised synaptic activity in a time- and dose-dependent manner and that the magnitude of these deficits correlated with the formation of αSyn pathology in cultured excitatory hippocampal neurons from both sexes of mice. Remarkably, acute passive infusion of αSyn PFFs from whole-cell patch-clamp pipette decreased mEPSC frequency within 10 min followed by induction of αSyn pathology within 1 d. Moreover, by direct addition of αSyn PFFs into culture medium, the formation of misfolded αSyn inclusions dramatically compromised the colocalization of synaptic markers and altered dynamic changes of dendritic spines, but the viability of neurons was not affected up to 7 d post-treatment with αSyn PFFs. Our data indicate that intraneuronal αSyn fibrils impaired the initiation of synaptogenesis and their physiological functions, thereby suggesting that targeting synaptic dysfunction in synucleinopathies may provide a promising therapeutic direction.SIGNIFICANCE STATEMENT Under pathological conditions, the presynaptic protein α-synuclein (αSyn) aggregates to form intraneuronal inclusions. To understand how and to what extent αSyn aggregates modulate synaptic activity before neuron loss, we demonstrate that αSyn preformed fibrils (PFFs) reduced synaptic activity in a dose- and time-dependent manner. The magnitude of these deficits correlated with the deposition of αSyn pathology, which dramatically compromised the colocalization of synaptic markers and altered the dendritic spine dynamics. The present work further highlights the impact of αSyn PFFs on synaptogenesis and physiological function, which may be applicable to other types of synucleinopathies.


Asunto(s)
Hipocampo/metabolismo , Neuronas/metabolismo , Agregado de Proteínas/fisiología , Sinapsis/metabolismo , alfa-Sinucleína/metabolismo , Animales , Supervivencia Celular , Hipocampo/patología , Ratones , Ratones Noqueados , Neuronas/patología , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología , Sinapsis/patología
4.
Neurobiol Dis ; 136: 104712, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31837422

RESUMEN

Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are progressive neurodegenerative diseases for which there is no disease-modifying treatment. PD and DLB are characterized by aggregation of the synaptic protein α-synuclein, and there is compelling evidence to suggest that progression of these diseases is associated with the trans-cellular spread of pathogenic α-synuclein through the brains of afflicted individuals. Therapies targeting extracellular, pathogenic α-synuclein may therefore hold promise for slowing or halting disease progression. In this regard, it has been suggested that highly-selective antibodies can be administered as therapeutic agents targeting pathogenic proteins. In the current study, we screened a series of antibodies using multiple selection criterion to identify those that selectively bind pathogenic α-synuclein and show potent inhibition of pathology seeding in a neuronal model of α-synucleinopathy. A lead antibody was tested in a mouse model of PD, and it was able to reduce the spread of α-synuclein pathology in the brain and attenuate dopamine reductions in the striatum. This study highlights the therapeutic potential of α-synuclein immunotherapy for the treatment of PD and DLB, and provides a framework for screening of α-synuclein antibodies to identify those with preferred properties.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Inmunoterapia/métodos , Enfermedad por Cuerpos de Lewy/inmunología , Enfermedad por Cuerpos de Lewy/terapia , Enfermedad de Parkinson/inmunología , Enfermedad de Parkinson/terapia , alfa-Sinucleína/administración & dosificación , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Células Cultivadas , Relación Dosis-Respuesta Inmunológica , Femenino , Humanos , Enfermedad por Cuerpos de Lewy/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Enfermedad de Parkinson/genética , alfa-Sinucleína/química , alfa-Sinucleína/genética
5.
J Neurosci ; 37(24): 5870-5884, 2017 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-28522732

RESUMEN

Parkinson's disease (PD) patients progressively accumulate intracytoplasmic inclusions formed by misfolded α-synuclein known as Lewy bodies (LBs). LBs also contain other proteins that may or may not be relevant in the disease process. To identify proteins involved early in LB formation, we performed proteomic analysis of insoluble proteins in a primary neuron culture model of α-synuclein pathology. We identified proteins previously found in authentic LBs in PD as well as several novel proteins, including the microtubule affinity-regulating kinase 1 (MARK1), one of the most enriched proteins in this model of LB formation. Activated MARK proteins (MARKs) accumulated in LB-like inclusions in this cell-based model as well as in a mouse model of LB disease and in LBs of postmortem synucleinopathy brains. Inhibition of MARKs dramatically exacerbated α-synuclein pathology. These findings implicate MARKs early in synucleinopathy pathogenesis and as potential therapeutic drug targets.SIGNIFICANCE STATEMENT Neurodegenerative diseases are diagnosed definitively only in postmortem brains by the presence of key misfolded and aggregated disease proteins, but cellular processes leading to accumulation of these proteins have not been well elucidated. Parkinson's disease (PD) patients accumulate misfolded α-synuclein in LBs, the diagnostic signatures of PD. Here, unbiased mass spectrometry was used to identify the microtubule affinity-regulating kinase family (MARKs) as activated and insoluble in a neuronal culture PD model. Aberrant activation of MARKs was also found in a PD mouse model and in postmortem PD brains. Further, inhibition of MARKs led to increased pathological α-synuclein burden. We conclude that MARKs play a role in PD pathogenesis.


Asunto(s)
Cuerpos de Lewy/enzimología , Proteínas del Tejido Nervioso/metabolismo , Enfermedad de Parkinson/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteoma/metabolismo , alfa-Sinucleína/metabolismo , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
6.
J Neurosci ; 37(47): 11485-11494, 2017 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-28986461

RESUMEN

Neurodegenerative proteinopathies characterized by intracellular aggregates of tau proteins, termed tauopathies, include Alzheimer's disease (AD), frontotemporal lobar degeneration (FTLD) with tau pathology (FTLD-tau), and related disorders. Pathological tau proteins derived from human AD brains (AD-tau) act as proteopathic seeds that initiate the templated aggregation of soluble tau upon intracerebral injection into tau transgenic (Tg) and wild-type mice, thereby modeling human tau pathology. In this study, we found that aged Tg mice of both sexes expressing human tau proteins harboring a pathogenic P301L MAPT mutation labeled with green fluorescent protein (T40PL-GFP Tg mouse line) exhibited hyperphosphorylated tau mislocalized to the somatodentritic domain of neurons, but these mice did not develop de novo insoluble tau aggregates, which are characteristic of human AD and related tauopathies. However, intracerebral injections of either T40PL preformed fibrils (PFFs) or AD-tau seeds into T40PL-GFP mice induced abundant intraneuronal pathological inclusions of hyperphosphorylated T40PL-GFP. These injections of pathological tau resulted in the propagation of tau pathology from the injection site to neuroanatomically connected brain regions, and these tau inclusions consisted of both T40PL-GFP and WT endogenous mouse tau. Primary neurons cultured from the brains of neonatal T40PL-GFP mice provided an informative in vitro model for examining the uptake and localization of tau PFFs. These findings demonstrate the seeded aggregation of T40PL-GFP in vivo by synthetic PFFs and human AD-tau and the utility of this system to study the neuropathological spread of tau aggregates.SIGNIFICANCE STATEMENT The stereotypical spread of pathological tau protein aggregates have recently been attributed to the transmission of proteopathic seeds. Despite the extensive use of transgenic mouse models to investigate the propagation of tau pathology in vivo, details of the aggregation process such as the early seeding events leading to new tau pathology have remained elusive. This study validates the use of GFP-labeled tau expressed by neurons in vivo and in vitro as models for investigating mechanisms underlying the seeded transmission of tau pathology as well as tau-focused drug discovery to identify disease-modifying therapies for AD and related tauopathies.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas tau/toxicidad , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inyecciones Intraventriculares , Masculino , Ratones , Mutación , Neuronas/metabolismo , Neuronas/patología , Proteínas Recombinantes , Proteínas tau/administración & dosificación , Proteínas tau/genética , Proteínas tau/metabolismo
7.
Acta Neuropathol ; 135(6): 855-875, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29502200

RESUMEN

The accumulation of misfolded α-synuclein (aSyn) and neuron loss define several neurodegenerative disorders including Parkinson's disease (PD) and dementia with Lewy bodies (DLB). However, the precise relationship between pathology and neurotoxicity and why these processes disproportionately affect certain neuron subpopulations are poorly understood. We show here that Math2-expressing neurons in the hippocampal Cornu ammonis (CA), a region significantly affected by aSyn pathology in advanced PD and DLB, are highly susceptible to pathological seeding with pre-formed fibrils (PFFs), in contrast to dentate gyrus neurons, which are relatively spared. Math2+ neurons also exhibited more rapid and severe cell loss in both in vitro and in vivo models of synucleinopathy. Toxicity resulting from PFF exposure was dependent on endogenous aSyn and could be attenuated by N-acetyl-cysteine through a glutathione-dependent process. Moreover, aSyn expression levels strongly correlate with relative vulnerability among hippocampal neuron subtypes of which Math2+ neurons contained the highest amount. Consistent with this, antisense oligonucleotide (ASO)-mediated knockdown of aSyn reduced the neuronal pathology in a time-dependent manner. However, significant neuroprotection was observed only with early ASO intervention and a substantial reduction of aSyn pathology, indicating toxicity occurs after a critical threshold of pathological burden is exceeded in vulnerable neurons. Together, our findings reveal considerable heterogeneity in endogenous aSyn levels among hippocampal neurons and suggest that this may contribute to the selective vulnerability observed in the context of synucleinopathies.


Asunto(s)
Hipocampo/metabolismo , Neuronas/metabolismo , Agregación Patológica de Proteínas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Muerte Celular/fisiología , Células Cultivadas , Femenino , Técnicas de Silenciamiento del Gen , Hipocampo/patología , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Neuronas/patología , Cultivo Primario de Células , Agregación Patológica de Proteínas/patología , Deficiencias en la Proteostasis/metabolismo , Deficiencias en la Proteostasis/patología , alfa-Sinucleína/genética
8.
Acta Neuropathol Commun ; 10(1): 30, 2022 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-35246269

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder that exhibits pathological changes in both tau and synaptic function. AD patients display increases in hyperphosphorylated tau and synaptic activity. Previous studies have individually identified the role of NR2B subunit-containing NMDA receptors in AD related synaptic dysfunction and aggregated tau without reconciling the conflicting differences and implications of NR2B expression. Inhibition of extrasynaptically located NR2B mitigates tau pathology in AD models, whereas the inhibition of synaptic NR2B replicates tau-associated hyperactivity. This suggests that a simultaneous increase in extrasynaptic NR2B and decrease in synaptic NR2B may be responsible for tau pathology and synaptic dysfunction, respectively. The synaptic location of NR2B is regulated by casein kinase 2 (CK2), which is highly expressed in AD patients. Here, we used patient brains diagnosed with AD, corticobasal degeneration, progressive supranuclear palsy or Pick's disease to characterize CK2 expression across these diverse tauopathies. Human derived material was also utilized in conjunction with cultured hippocampal neurons in order to investigate AD-induced changes in NR2B location. We further assessed the therapeutic effect of CK2 inhibition on NR2B synaptic distribution and tau pathology. We found that aberrant expression of CK2, and synaptically translocated NR2B, is unique to AD patients compared to other tauopathies. Increased CK2 was also observed in AD-tau treated neurons in addition to the mislocalization of NR2B receptors. Tau burden was alleviated in vitro by correcting synaptic:extrasynaptic NR2B function. Restoring NR2B physiological expression patterns with CK2 inhibition and inhibiting the function of excessive extrasynaptic NR2B with Memantine both mitigated tau accumulation in vitro. However, the combined pharmacological treatment promoted the aggregation of tau. Our data suggests that the synaptic:extrasynaptic balance of NR2B function regulates AD-tau pathogenesis, and that the inhibition of CK2, and concomitant prevention of NR2B mislocalization, may be a useful therapeutic tool for AD patients.


Asunto(s)
Enfermedad de Alzheimer , Quinasa de la Caseína II , Receptores de N-Metil-D-Aspartato , Tauopatías , Enfermedad de Alzheimer/patología , Quinasa de la Caseína II/antagonistas & inhibidores , Quinasa de la Caseína II/metabolismo , Humanos , Enfermedad de Pick/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Tauopatías/patología , Proteínas tau/metabolismo
9.
Psychol Trauma ; 12(4): 431-435, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-31328939

RESUMEN

OBJECTIVE: The purpose of this study was to pilot the iCOVER curriculum-a training product designed to teach service members how to recognize and respond to acute stress reactions (ASRs) using a simple, 6-step procedure, iCOVER. Three goals guided the pilot: (a) assess training acceptability, (b) assess impact of training on knowledge and performance of the iCOVER procedure, and (c) explore the effects of in-person and computer-simulated practical exercises. METHOD: Six military squads (N = 66) were randomly assigned to 3 conditions: iCOVER Standard (iCOVER instruction with an in-person practical exercise), iCOVER Tech (iCOVER instruction with a computer-simulated practical exercise), or Control (no iCOVER instruction). Squads in the iCOVER conditions received iCOVER instruction, completed a knowledge test and practical exercise to which they were assigned (i.e., Standard or Tech), demonstrated their iCOVER skills in live-action scenarios, and reported their perceptions of the training. RESULTS: iCOVER training was acceptable to most participants and associated with improved knowledge about iCOVER (Mpre = 3.33 vs. Mpost = 5.15; t[42] = -7.61, p < .001, d = 1.41); iCOVER Standard resulted in more iCOVER behaviors during a live-action scenario compared with the other conditions, F(2, 35) = 13.36, p < .001, η2 = 0.43. Compared with iCOVER Tech, iCOVER Standard had greater acceptability and resulted in better performance of iCOVER. CONCLUSIONS: This is the first U.S. demonstration of a training program designed to address ASRs during high-risk operations and offers a potential way ahead for preparing military teams to manage ASRs. (PsycInfo Database Record (c) 2020 APA, all rights reserved).


Asunto(s)
Personal Militar/educación , Estrés Psicológico/diagnóstico , Adulto , Curriculum , Humanos , Masculino , Proyectos Piloto
10.
Neuron ; 105(2): 260-275.e6, 2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31759806

RESUMEN

Studies have shown an overlap of Aß plaques, tau tangles, and α-synuclein (α-syn) pathologies in the brains of Alzheimer's disease (AD) and Parkinson's disease (PD) with dementia (PDD) patients, with increased pathological burden correlating with severity of cognitive and motor symptoms. Despite the observed co-pathology and concomitance of motor and cognitive phenotypes, the consequences of the primary amyloidogenic protein on the secondary pathologies remain poorly understood. To better define the relationship between α-syn and Aß plaques, we injected α-syn preformed fibrils (α-syn mpffs) into mice with abundant Aß plaques. Aß deposits dramatically accelerated α-syn pathogenesis and spread throughout the brain. Remarkably, hyperphosphorylated tau (p-tau) was induced in α-syn mpff-injected 5xFAD mice. Finally, α-syn mpff-injected 5xFAD mice showed neuron loss that correlated with the progressive decline of cognitive and motor performance. Our findings suggest a "feed-forward" mechanism whereby Aß plaques enhance endogenous α-syn seeding and spreading over time post-injection with mpffs.


Asunto(s)
Enfermedad por Cuerpos de Lewy/metabolismo , Neuronas/patología , Placa Amiloide/metabolismo , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Animales , Encéfalo/metabolismo , Recuento de Células , Disfunción Cognitiva/patología , Humanos , Enfermedad por Cuerpos de Lewy/patología , Ratones , Actividad Motora , Fosforilación , alfa-Sinucleína/administración & dosificación
11.
J Exp Med ; 217(2)2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31826239

RESUMEN

Tauopathies are characterized by abnormal accumulation of tau protein in neurons and glia. In Alzheimer's disease (AD), tau aggregates in neurons, while in corticobasal degeneration (CBD) and progressive supranuclear palsy (PSP), tau also aggregates in astrocytes and oligodendrocytes. We previously demonstrated that human CBD and PSP tauopathy lysates (CBD-tau and PSP-tau) contain distinct tau strains that propagate neuronal and glial tau aggregates in nontransgenic (nonTg) mouse brain. Yet the mechanism of glial tau transmission is unknown. Here, we developed a novel mouse model to knock down tau in neurons to test for glial tau transmission. While oligodendroglial tau pathology propagated across the mouse brain in the absence of neuronal tau pathology, astrocytic tau pathology did not. Oligodendroglial tau aggregates propagated along white matter tracts independently of neuronal axons, and resulted in oligodendrocyte cell loss. Thus, glial tau pathology has significant functional consequences independent of neuronal tau pathology.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Agregación Patológica de Proteínas/metabolismo , Parálisis Supranuclear Progresiva/metabolismo , Proteínas tau/metabolismo , Anciano , Enfermedad de Alzheimer/patología , Animales , Astrocitos/metabolismo , Encéfalo/patología , Células Cultivadas , Técnicas de Cocultivo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Modelos Animales , Neuroglía/patología , Oligodendroglía/metabolismo , Ratas , Parálisis Supranuclear Progresiva/patología , Proteínas tau/genética , Proteínas tau/aislamiento & purificación
12.
Mol Neurodegener ; 15(1): 64, 2020 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-33148293

RESUMEN

BACKGROUND: The spread of tau pathology in Alzheimer's disease (AD) is mediated by cell-to-cell transmission of pathological tau seeds released from neurons that, upon internalization by recipient neurons, template the misfolding of naïve cellular tau, thereby propagating fibrillization. We hypothesize that anti-tau monoclonal antibodies (mAbs) that selectively bind to pathological tau seeds will inhibit propagation of tau aggregates and reduce the spread of tau pathology in vivo. METHODS: We inoculated mice with human AD brain-derived extracts containing tau paired helical filaments (AD-tau) and identified two novel mAbs, DMR7 and SKT82, that selectively bind to a misfolded pathological conformation of tau relative to recombinant tau monomer. To evaluate the effects of these mAbs on the spread of pathological tau in vivo, 5xFAD mice harboring significant brain Aß plaque burden were unilaterally injected with AD-tau in the hippocampus, to initiate the formation of neuritic plaque (NP) tau pathology, and were treated weekly with intraperitoneal (i.p.) injections of DMR7, SKT82, or IgG isotype control mAbs. RESULTS: DMR7 and SKT82 bind epitopes comprised of the proline-rich domain and c-terminal region of tau and binding is reduced upon disruption of the pathological conformation of AD-tau by chemical and thermal denaturation. We found that both DMR7 and SKT82 immunoprecipitate pathological tau and significantly reduce the seeding of cellular tau aggregates induced by AD-tau in primary neurons by 60.5 + 13.8% and 82.2 + 8.3%, respectively, compared to IgG control. To investigate the mechanism of mAb inhibition, we generated pH-sensitive fluorophore-labeled recombinant tau fibrils seeded by AD-tau to track internalization of tau seeds and demonstrate that the conformation-selective tau mAbs inhibit the internalization of tau seeds. DMR7 and SKT82 treatment reduced hyperphosphorylated NP tau as measured with AT8 immunohistochemistry (IHC) staining, but did not achieve statistical significance in the contralateral cortex and SKT82 significantly reduced tau pathology in the ipsilateral hippocampus by 24.2%; p = 0.044. CONCLUSIONS: These findings demonstrate that conformation-selective tau mAbs, DMR7 and SKT82, inhibit tau pathology in primary neurons by preventing the uptake of tau seeds and reduce tau pathology in vivo, providing potential novel therapeutic candidates for the treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Anticuerpos Monoclonales/farmacología , Neuronas/patología , Proteínas tau/antagonistas & inhibidores , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Neuronas/efectos de los fármacos , Proteínas tau/efectos de los fármacos
13.
Neuron ; 105(5): 822-836.e7, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-31899072

RESUMEN

Mutations in the GBA1 gene are the most common genetic risk factor for Parkinson's disease (PD) and dementia with Lewy bodies (DLB). GBA1 encodes the lysosomal lipid hydrolase glucocerebrosidase (GCase), and its activity has been linked to accumulation of α-synuclein. The current study systematically examines the relationship between GCase activity and both pathogenic and non-pathogenic forms of α-synuclein in primary hippocampal, cortical, and midbrain neuron and astrocyte cultures, as well as in transgenic mice and a non-transgenic mouse model of PD. We find that reduced GCase activity does not result in aggregation of α-synuclein. However, in the context of extant misfolded α-synuclein, GCase activity modulates neuronal susceptibility to pathology. Furthermore, this modulation does not depend on neuron type but rather is driven by the level of pathological α-synuclein seeds. This study has implications for understanding how GBA1 mutations influence PD pathogenesis and provides a platform for testing novel therapeutics.


Asunto(s)
Astrocitos/metabolismo , Glucosilceramidasa/genética , Neuronas/metabolismo , Trastornos Parkinsonianos/genética , Agregación Patológica de Proteínas/genética , alfa-Sinucleína/metabolismo , Animales , Astrocitos/citología , Astrocitos/patología , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Susceptibilidad a Enfermedades , Predisposición Genética a la Enfermedad , Glucosilceramidasa/metabolismo , Células HEK293 , Hipocampo/citología , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Enfermedad por Cuerpos de Lewy/genética , Enfermedad por Cuerpos de Lewy/metabolismo , Enfermedad por Cuerpos de Lewy/patología , Mesencéfalo/citología , Mesencéfalo/metabolismo , Mesencéfalo/patología , Ratones , Ratones Transgénicos , Neuronas/citología , Neuronas/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Cultivo Primario de Células , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología , Sinucleinopatías/genética , Sinucleinopatías/metabolismo , Sinucleinopatías/patología
14.
Front Psychol ; 10: 1480, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31293490

RESUMEN

Recent advances in the science of teams have provided much insight into the important attitudes (e.g., team cohesion and efficacy), cognitions (e.g., shared team cognition), and behaviors (e.g., teamwork communications) of high performing teams and how these competencies emerge as team members interact, and appropriate measurement methods for tracking development. Numerous training interventions have been found to effectively improve these competencies, and more recently have begun addressing the problem of team dynamics. Team science researchers have increasingly called for more field studies to better understand training and team development processes in the wild and to advance the theory of team development. In addition to the difficulty of gaining access to teams that operate in isolated, confined, and extreme environments (ICE), a major practical challenge for trainers of ICE teams whose schedules are already strained is the need to prioritize the most effective strategies to optimize the time available for implementation. To address these challenges, we describe an applied research experiment that developed and evaluated an integrated team training approach to improve Tactical Combat Casualty (TC3) skills in U.S. Army squads. Findings showed that employing effective team training best practices improved learning, team cognition, emergent team processes, and performance. We recommend future research should focus on understanding the types of training strategies needed to enable teams and team leaders to develop from novices to experts. Effectively modifying training to scale it to team expertise requires more research. More laboratory and field research is needed to further develop measures of team knowledge emergence for complex task domains, and include other potential emergent factors such as team leadership and resilience. Practical implications for research include developing automated tools and technologies needed to implement training and collect team data, and employ more sensitive indicators (e.g., behavioral markers) of team attitudes, cognitions and behaviors to model the dynamics of how they naturally change over time. These tools are critical to understanding the dynamics of team development and to implement interventions that more effectively support teams as they develop over time.

15.
J Neuropathol Exp Neurol ; 77(3): 216-228, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29415231

RESUMEN

Aggregation of tau into fibrillar structures within the CNS is a pathological hallmark of a clinically heterogeneous set of neurodegenerative diseases termed tauopathies. Unique misfolded conformations of tau, referred to as strains, are hypothesized to underlie the distinct neuroanatomical and cellular distribution of pathological tau aggregates. Here, we report the identification of novel tau monoclonal antibodies (mAbs) that selectively bind to an Alzheimer disease (AD)-specific conformation of pathological tau. Immunohistochemical analysis of tissue from various AD and nonAD tauopathies demonstrate selective binding of mAbs GT-7 and GT-38 to AD tau pathologies and absence of immunoreactivity for tau aggregates that are diagnostic of corticobasal degenerations (CBD), progressive supranuclear palsy (PSP), and Pick's disease (PiD). In cases with co-occurring AD tauopathy, GT-7 and GT-38 distinguish comorbid AD tau from pathological tau in frontotemporal lobar degeneration characterized by tau inclusions (FTLD-Tau), as confirmed by the presence of both 3 versus 4 microtubule-binding repeat isoforms (3R and 4R tau isoforms, respectively), in AD neurofibrillary tangles but not in the tau aggregates of CBD, PSP, or PiD. These findings support the concept of an AD-specific tau strain. The mAbs described here enable the selective detection of AD tau pathology in nonAD tauopathies.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico , Anticuerpos Monoclonales/metabolismo , Conformación Molecular , Tauopatías/metabolismo , Proteínas tau/inmunología , Proteínas tau/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Anticuerpos Monoclonales/química , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Fosforilación , Enfermedad de Pick/metabolismo , Enfermedad de Pick/patología , Conformación Proteica , Isoformas de Proteínas/metabolismo , Parálisis Supranuclear Progresiva/metabolismo , Parálisis Supranuclear Progresiva/patología , Tauopatías/patología
17.
Patient Educ Couns ; 50(1): 39-42, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12767583

RESUMEN

Communication between patients and physicians likely mediates traditional patient and physician predispositions in determining patient outcomes, including perceptions and decision making. However, the extent to which a mediating effect occurs is unclear. The purpose of this essay is to outline the need for conceptualizing more holistic models of consumer-provider interaction that demonstrate the role of the therapeutic relationship in treatment outcomes. We focus on an important communicative context for exploring this question: the situation where patients, with the help of oncologists, are faced with making treatment choices, particularly whether to enroll in a clinical trial in response to their life-threatening cancer diagnosis. We explore the question from the perspectives of the medical provider, the patient, and the accompanying family member, in order to better frame the complex interactional dynamics occurring during the interaction.


Asunto(s)
Ensayos Clínicos como Asunto , Comunicación , Neoplasias/psicología , Participación del Paciente , Selección de Paciente , Relaciones Médico-Paciente , Toma de Decisiones , Familia/psicología , Humanos , Oncología Médica , Modelos Psicológicos , Evaluación de Necesidades , Neoplasias/terapia , Rol del Médico
18.
Mol Biol Cell ; 25(25): 4010-23, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25298402

RESUMEN

Aggregates of α-synuclein (α-syn) accumulate in neurons in Parkinson's disease and other synucleinopathies. These inclusions predominantly localize to axons even in the early stages of the disease, but their affect on axon function has remained unknown. Previously we established a model in which the addition of preformed α-syn fibrils to primary neurons seeds formation of insoluble α-syn inclusions built from endogenously expressed α-syn that closely recapitulate the neuropathological phenotypes of Lewy neurites found in human diseased brains. Here we show, using live-cell imaging, that immobile α-syn inclusions accumulate in axons from the recruitment of α-syn located on mobile α-syn-positive vesicles. Ultrastructural analyses and live imaging demonstrate that α-syn accumulations do not cause a generalized defect in axonal transport; the inclusions do not fill the axonal cytoplasm, disrupt the microtubule cytoskeleton, or affect the transport of synaptophysin or mitochondria. However, the α-syn aggregates impair the transport of Rab7 and TrkB receptor-containing endosomes, as well as autophagosomes. In addition, the TrkB receptor-associated signaling molecule pERK5 accumulates in α-syn aggregate-bearing neurons. Thus α-syn pathology impairs axonal transport of signaling and degradative organelles. These early effects of α-syn accumulations may predict points of intervention in the neurodegenerative process.


Asunto(s)
Endosomas/metabolismo , alfa-Sinucleína/metabolismo , Amiloide/metabolismo , Animales , Transporte Axonal , Células Cultivadas , Endosomas/patología , Cuerpos de Lewy , Ratones , Enfermedad de Parkinson/metabolismo , Fagosomas/metabolismo , Agregado de Proteínas , Transporte de Proteínas , Receptor trkB/metabolismo
19.
Acta Neuropathol Commun ; 2: 33, 2014 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-24690345

RESUMEN

The RNA/DNA-binding protein, TDP-43, is the key component of ubiquitinated inclusions characteristic of amyotrophic lateral sclerosis (ALS) and the majority of frontotemporal lobar degeneration (FTLD-TDP) referred to collectively as TDP-43 proteinopathies. To further elucidate mechanisms of pathological TDP-43 processing and identify TDP-43 epitopes that could be useful as potential biomarkers of TDP-43 proteinopathies, we developed a panel of novel monoclonal antibodies (MAbs) directed at regions extending across the length of TDP-43. Here, we confirm previous observations that there is no or minimal accumulation of TDP-43 N-terminal domains in neocortical inclusions in human TDP-43 proteinopathy tissues and we identify a subset of these MAbs that are specific for human versus mouse TDP-43. Notably, one of these MAbs recognized an epitope that preferentially detected pathological TDP-43 inclusions with negligible reactivity for normal nuclear TDP-43 resembling anti-phospho-TDP-43 specific antibodies that only bind pathological TDP-43. Hence, we infer that this new MAb recognizes a phosphorylation independent but disease-specific pathologic conformation in abnormal TDP-43. These data suggest that the novel MAbs reported here will be useful for patient-oriented research as well as for studies of animal and cell-based models of TDP-43 proteinopathies including ALS and FTLD-TDP.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Anticuerpos Monoclonales , Proteínas de Unión al ADN/inmunología , Demencia Frontotemporal/metabolismo , Esclerosis Amiotrófica Lateral/diagnóstico , Animales , Especificidad de Anticuerpos , Encéfalo/metabolismo , Proteínas de Unión al ADN/metabolismo , Ensayo de Inmunoadsorción Enzimática , Mapeo Epitopo , Demencia Frontotemporal/diagnóstico , Humanos , Ratones
20.
Neuron ; 72(1): 57-71, 2011 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-21982369

RESUMEN

Inclusions composed of α-synuclein (α-syn), i.e., Lewy bodies (LBs) and Lewy neurites (LNs), define synucleinopathies including Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Here, we demonstrate that preformed fibrils generated from full-length and truncated recombinant α-syn enter primary neurons, probably by adsorptive-mediated endocytosis, and promote recruitment of soluble endogenous α-syn into insoluble PD-like LBs and LNs. Remarkably, endogenous α-syn was sufficient for formation of these aggregates, and overexpression of wild-type or mutant α-syn was not required. LN-like pathology first developed in axons and propagated to form LB-like inclusions in perikarya. Accumulation of pathologic α-syn led to selective decreases in synaptic proteins, progressive impairments in neuronal excitability and connectivity, and, eventually, neuron death. Thus, our data contribute important insights into the etiology and pathogenesis of PD-like α-syn inclusions and their impact on neuronal functions, and they provide a model for discovering therapeutics targeting pathologic α-syn-mediated neurodegeneration.


Asunto(s)
Muerte Celular/fisiología , Cuerpos de Lewy/patología , Neuronas/patología , Sinapsis/patología , alfa-Sinucleína/efectos adversos , alfa-Sinucleína/metabolismo , Animales , Transporte Axonal/fisiología , Endocitosis/fisiología , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/fisiología , Hipocampo/ultraestructura , Humanos , Cuerpos de Lewy/metabolismo , Ratones , Ratones Endogámicos C57BL , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Proteínas del Tejido Nervioso/metabolismo , Neuritas/metabolismo , Neuritas/patología , Neuronas/metabolismo , Neuronas/fisiología , Neuronas/ultraestructura , Cultivo Primario de Células , Sinapsis/metabolismo , Imagen de Colorante Sensible al Voltaje/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA