Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
J Neurosci ; 43(21): 3970-3984, 2023 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-37019623

RESUMO

Endolysosomal defects in neurons are central to the pathogenesis of prion and other neurodegenerative disorders. In prion disease, prion oligomers traffic through the multivesicular body (MVB) and are routed for degradation in lysosomes or for release in exosomes, yet how prions impact proteostatic pathways is unclear. We found that prion-affected human and mouse brain showed a marked reduction in Hrs and STAM1 (ESCRT-0), which route ubiquitinated membrane proteins from early endosomes into MVBs. To determine how the reduction in ESCRT-0 impacts prion conversion and cellular toxicity in vivo, we prion-challenged conditional knockout mice (male and female) having Hrs deleted from neurons, astrocytes, or microglia. The neuronal, but not astrocytic or microglial, Hrs-depleted mice showed a shortened survival and an acceleration in synaptic derangements, including an accumulation of ubiquitinated proteins, deregulation of phosphorylated AMPA and metabotropic glutamate receptors, and profoundly altered synaptic structure, all of which occurred later in the prion-infected control mice. Finally, we found that neuronal Hrs (nHrs) depletion increased surface levels of the cellular prion protein, PrPC, which may contribute to the rapidly advancing disease through neurotoxic signaling. Taken together, the reduced Hrs in the prion-affected brain hampers ubiquitinated protein clearance at the synapse, exacerbates postsynaptic glutamate receptor deregulation, and accelerates neurodegeneration.SIGNIFICANCE STATEMENT Prion diseases are rapidly progressive neurodegenerative disorders characterized by prion aggregate spread through the central nervous system. Early disease features include ubiquitinated protein accumulation and synapse loss. Here, we investigate how prion aggregates alter ubiquitinated protein clearance pathways (ESCRT) in mouse and human prion-infected brain, discovering a marked reduction in Hrs. Using a prion-infection mouse model with neuronal Hrs (nHrs) depleted, we show that low neuronal Hrs is detrimental and markedly shortens survival time while accelerating synaptic derangements, including ubiquitinated protein accumulation, indicating that Hrs loss exacerbates prion disease progression. Additionally, Hrs depletion increases the surface distribution of prion protein (PrPC), linked to aggregate-induced neurotoxic signaling, suggesting that Hrs loss in prion disease accelerates disease through enhancing PrPC-mediated neurotoxic signaling.


Assuntos
Doenças Neurodegenerativas , Doenças Priônicas , Príons , Masculino , Feminino , Camundongos , Humanos , Animais , Príons/metabolismo , Proteínas Priônicas/metabolismo , Receptores de AMPA/metabolismo , Neurônios/metabolismo , Doenças Priônicas/metabolismo , Doenças Priônicas/patologia , Doenças Neurodegenerativas/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo
2.
Brain ; 145(3): 879-886, 2022 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-35258081

RESUMO

Loss of midbrain dopamine neurons causes the cardinal symptoms of Parkinson's disease. However, not all dopamine neurons are equally vulnerable and a better understanding of the cell-type specific properties relating to selective dopamine neuron degeneration is needed. Most midbrain dopamine neurons express the vesicular glutamate transporter VGLUT2 during development and a subset continue to express low levels of VGLUT2 in adulthood, enabling the co-release of glutamate. Moreover, VGLUT2 expression in dopamine neurons can be neuroprotective since its genetic disruption was shown to sensitize dopamine neurons to neurotoxins. Here, we show that in response to toxic insult, and in two distinct models of alpha-synuclein stress, VGLUT2 dopamine neurons were resilient to degeneration. Dopamine neurons expressing VGLUT2 were enriched whether or not insult induced dopamine neuron loss, suggesting that while VGLUT2 dopamine neurons are more resilient, VGLUT2 expression can also be transcriptionally upregulated by injury. Finally, we observed that VGLUT2 expression was enhanced in surviving dopamine neurons from post-mortem Parkinson's disease individuals. These data indicate that emergence of a glutamatergic identity in dopamine neurons may be part of a neuroprotective response in Parkinson's disease.


Assuntos
Neurônios Dopaminérgicos , Doença de Parkinson , Adulto , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Humanos , Mesencéfalo , Degeneração Neural/metabolismo , Doença de Parkinson/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/genética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
3.
Nat Rev Neurosci ; 18(12): 713-726, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29097785

RESUMO

The corporeal beauty of the neuronal cytoskeleton has captured the imagination of generations of scientists. One of the easiest cellular structures to visualize by light microscopy, its existence has been known for well over 100 years, yet we have only recently begun to fully appreciate its intricacy and diversity. Recent studies combining new probes with super-resolution microscopy and live imaging have revealed surprising details about the axonal cytoskeleton and, in particular, have discovered previously unknown actin-based structures. Along with traditional electron microscopy, these newer techniques offer a nanoscale view of the axonal cytoskeleton, which is important for our understanding of neuronal form and function, and lay the foundation for future studies. In this Review, we summarize existing concepts in the field and highlight contemporary discoveries that have fundamentally altered our perception of the axonal cytoskeleton.


Assuntos
Transporte Axonal/fisiologia , Axônios/metabolismo , Citoesqueleto/metabolismo , Neurônios/metabolismo , Animais , Arquitetura/métodos , Axônios/ultraestrutura , Citoesqueleto/ultraestrutura , Humanos , Microscopia Eletrônica/métodos , Neurônios/ultraestrutura
4.
Proc Natl Acad Sci U S A ; 116(23): 11116-11118, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-31110014

RESUMO

The normal function of α-synuclein (α-syn) remains elusive. Although recent studies suggest α-syn as a physiologic attenuator of synaptic vesicle (SV) recycling, mechanisms are unclear. Here, we show that synapsin-a cytosolic protein with known roles in SV mobilization and clustering-is required for presynaptic functions of α-syn. Our data offer a critical missing link and advocate a model where α-syn and synapsin cooperate to cluster SVs and attenuate recycling.


Assuntos
Sinapsinas/metabolismo , Vesículas Sinápticas/metabolismo , alfa-Sinucleína/metabolismo , Animais , Linhagem Celular , Camundongos , Neurônios/metabolismo , Sinapses/metabolismo
5.
Proc Natl Acad Sci U S A ; 116(23): 11113-11115, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-31110017

RESUMO

The function of α-synuclein (α-syn) has been long debated, and two seemingly divergent views have emerged. In one, α-syn binds to VAMP2, acting as a SNARE chaperone-but with no effect on neurotransmission-while another posits that α-syn attenuates neurotransmitter release by restricting synaptic vesicle mobilization and recycling. Here, we show that α-syn-VAMP2 interactions are necessary for α-syn-induced synaptic attenuation. Our data connect divergent views and suggest a unified model of α-syn function.


Assuntos
Vesículas Sinápticas/metabolismo , Proteína 2 Associada à Membrana da Vesícula/metabolismo , alfa-Sinucleína/metabolismo , Transporte Biológico/fisiologia , Humanos , Neurônios/metabolismo , Proteínas SNARE/metabolismo , Transmissão Sináptica/imunologia
6.
J Proteome Res ; 18(3): 1380-1391, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30735395

RESUMO

The brain represents one of the most divergent and critical organs in the human body. Yet, it can be afflicted by a variety of neurodegenerative diseases specifically linked to aging, about which we lack a full biomolecular understanding of onset and progression, such as Alzheimer's disease (AD). Here we provide a proteomic resource comprising nine anatomically distinct sections from three aged individuals, across a spectrum of disease progression, categorized by quantity of neurofibrillary tangles. Using state-of-the-art mass spectrometry, we identify a core brain proteome that exhibits only small variance in expression, accompanied by a group of proteins that are highly differentially expressed in individual sections and broader regions. AD affected tissue exhibited slightly elevated levels of tau protein with similar relative expression to factors associated with the AD pathology. Substantial differences were identified between previous proteomic studies of mature adult brains and our aged cohort. Our findings suggest considerable value in examining specifically the brain proteome of aged human populations from a multiregional perspective. This resource can serve as a guide, as well as a point of reference for how specific regions of the brain are affected by aging and neurodegeneration.


Assuntos
Doença de Alzheimer/genética , Encéfalo/metabolismo , Proteoma/isolamento & purificação , Proteínas tau/isolamento & purificação , Adulto , Idoso , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/patologia , Encéfalo/patologia , Mapeamento Encefálico/métodos , Feminino , Humanos , Masculino , Espectrometria de Massas , Proteoma/genética , Proteômica/métodos , Proteínas tau/genética
7.
J Neurosci ; 37(7): 1675-1684, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28039370

RESUMO

Despite considerable research to uncover them, the anatomic and neuropathologic correlates of memory impairment in dementia with Lewy bodies (DLB) remain unclear. While some studies have implicated Lewy bodies in the neocortex, others have pointed to α-synuclein pathology in the hippocampus. We systematically examined hippocampal Lewy pathology and its distribution in hippocampal subfields in 95 clinically and neuropathologically characterized human cases of DLB, finding that α-synuclein pathology was highest in two hippocampal-related subregions: the CA2 subfield and the entorhinal cortex (EC). While the EC had numerous classic somatic Lewy bodies, CA2 contained mainly Lewy neurites in presumed axon terminals, suggesting the involvement of the EC → CA2 circuitry in the pathogenesis of DLB symptoms. Clinicopathological correlations with measures of verbal and visual memory supported a role for EC Lewy pathology, but not CA2, in causing these memory deficits. Lewy pathology in CA1-the main output region for CA2-correlated best with results from memory testing despite a milder pathology. This result indicates that CA1 may be more functionally relevant than CA2 in the context of memory impairment in DLB. These correlations remained significant after controlling for several factors, including concurrent Alzheimer's pathology (neuritic plaques and neurofibrillary tangles) and the interval between time of testing and time of death. Our data suggest that although hippocampal Lewy pathology in DLB is predominant in CA2 and EC, memory performance correlates most strongly with CA1 burden.SIGNIFICANCE STATEMENT This study provides a detailed neuropathologic analysis of hippocampal Lewy pathology in human patients with autopsy-confirmed dementia with Lewy bodies. The approach-informed by regional molecular markers, concurrent Alzheimer's pathology analysis, and relevant clinical data-helps tease out the relative contribution of Lewy pathology to memory dysfunction in the disease. Levels of Lewy pathology were found to be highest in the hippocampal CA2 subregion and entorhinal cortex, implicating a potentially overlooked circuit in disease pathogenesis. However, correlation with memory performance was strongest with CA1. This unexpected finding suggests that Lewy pathology must reach a critical burden across hippocampal circuitry to contribute to memory dysfunction beyond that related to other factors, notably coexisting Alzheimer's disease tau pathology.


Assuntos
Hipocampo/metabolismo , Doença por Corpos de Lewy/complicações , Transtornos da Memória/etiologia , Transtornos da Memória/patologia , alfa-Sinucleína/metabolismo , Idoso , Idoso de 80 Anos ou mais , Autopsia , Proteínas de Transporte de Cátions/metabolismo , Feminino , Humanos , Masculino , Testes Neuropsicológicos , Escalas de Graduação Psiquiátrica , Análise de Regressão , Sinucleínas/metabolismo
8.
J Neurosci ; 37(29): 6926-6937, 2017 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-28626017

RESUMO

High levels of amyloid-ß peptide (Aß) have been related to Alzheimer's disease pathogenesis. However, in the healthy brain, low physiologically relevant concentrations of Aß are necessary for long-term potentiation (LTP) and memory. Because cGMP plays a key role in these processes, here we investigated whether the cyclic nucleotide cGMP influences Aß levels and function during LTP and memory. We demonstrate that the increase of cGMP levels by the phosphodiesterase-5 inhibitors sildenafil and vardenafil induces a parallel release of Aß due to a change in the approximation of amyloid precursor protein (APP) and the ß-site APP cleaving enzyme 1. Moreover, electrophysiological and behavioral studies performed on animals of both sexes showed that blocking Aß function, by using anti-murine Aß antibodies or APP knock-out mice, prevents the cGMP-dependent enhancement of LTP and memory. Our data suggest that cGMP positively regulates Aß levels in the healthy brain which, in turn, boosts synaptic plasticity and memory.SIGNIFICANCE STATEMENT Amyloid-ß (Aß) is a key pathogenetic factor in Alzheimer's disease. However, low concentrations of endogenous Aß, mimicking levels of the peptide in the healthy brain, enhance hippocampal long-term potentiation (LTP) and memory. Because the second messenger cGMP exerts a central role in LTP mechanisms, here we studied whether cGMP affects Aß levels and function during LTP. We show that cGMP enhances Aß production by increasing the APP/BACE-1 convergence in endolysosomal compartments. Moreover, the cGMP-induced enhancement of LTP and memory was disrupted by blockade of Aß, suggesting that the physiological effect of the cyclic nucleotide on LTP and memory is dependent upon Aß.


Assuntos
Peptídeos beta-Amiloides/metabolismo , GMP Cíclico/metabolismo , Hipocampo/fisiologia , Potenciação de Longa Duração/fisiologia , Memória/fisiologia , Rememoração Mental/fisiologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos Sprague-Dawley , Análise e Desempenho de Tarefas
9.
N Engl J Med ; 370(13): 1209-1219, 2014 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-24670167

RESUMO

BACKGROUND: Autism involves early brain overgrowth and dysfunction, which is most strongly evident in the prefrontal cortex. As assessed on pathological analysis, an excess of neurons in the prefrontal cortex among children with autism signals a disturbance in prenatal development and may be concomitant with abnormal cell type and laminar development. METHODS: To systematically examine neocortical architecture during the early years after the onset of autism, we used RNA in situ hybridization with a panel of layer- and cell-type-specific molecular markers to phenotype cortical microstructure. We assayed markers for neurons and glia, along with genes that have been implicated in the risk of autism, in prefrontal, temporal, and occipital neocortical tissue from postmortem samples obtained from children with autism and unaffected children between the ages of 2 and 15 years. RESULTS: We observed focal patches of abnormal laminar cytoarchitecture and cortical disorganization of neurons, but not glia, in prefrontal and temporal cortical tissue from 10 of 11 children with autism and from 1 of 11 unaffected children. We observed heterogeneity between cases with respect to cell types that were most abnormal in the patches and the layers that were most affected by the pathological features. No cortical layer was uniformly spared, with the clearest signs of abnormal expression in layers 4 and 5. Three-dimensional reconstruction of layer markers confirmed the focal geometry and size of patches. CONCLUSIONS: In this small, explorative study, we found focal disruption of cortical laminar architecture in the cortexes of a majority of young children with autism. Our data support a probable dysregulation of layer formation and layer-specific neuronal differentiation at prenatal developmental stages. (Funded by the Simons Foundation and others.).


Assuntos
Transtorno Autístico/patologia , Neocórtex/ultraestrutura , Adolescente , Transtorno Autístico/genética , Biomarcadores/análise , Biomarcadores/metabolismo , Calbindina 1/genética , Contagem de Células , Criança , Pré-Escolar , Crioultramicrotomia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 1/genética , Expressão Gênica , Humanos , Imageamento Tridimensional , Hibridização In Situ , Neocórtex/crescimento & desenvolvimento , Proteínas do Tecido Nervoso/genética , Proteínas de Neurofilamentos/genética , Neurogênese , Neurônios/patologia , Membro 2 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , RNA/genética
10.
Traffic ; 13(5): 681-93, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22309053

RESUMO

The downstream targets of amyloid ß (Aß)-oligomers remain elusive. One hypothesis is that Aß-oligomers interrupt axonal transport. Although previous studies have demonstrated Aß-induced transport blockade, early effects of low-n soluble Aß-oligomers on axonal transport remain unclear. Furthermore, the cargo selectivity for such deficits (if any) or the specific effects of Aß on the motility kinetics of transported cargoes are also unknown. Toward this, we visualized axonal transport of vesicles in cultured hippocampal neurons treated with picomolar (pm) levels of cell-derived soluble Aß-oligomers. We examined select cargoes thought to move as distinct organelles and established imaging parameters that allow organelle tracking with consistency and high fidelity - analyzing all data in a blinded fashion. Aß-oligomers induced early and selective diminutions in velocities of synaptic cargoes but had no effect on mitochondrial motility, contrary to previous reports. These changes were N-methyl D-aspartate receptor/glycogen synthase kinase-3ß dependent and reversible upon washout of the oligomers. Cluster-mode analyses reveal selective attenuations in faster-moving synaptic vesicles, suggesting possible decreases in cargo/motor associations, and biochemical experiments implicate tau phosphorylation in the process. Collectively, the data provide a biological basis for Aß-induced axonal transport deficits.


Assuntos
Peptídeos beta-Amiloides/química , Axônios/metabolismo , Sinapses/metabolismo , Animais , Transporte Biológico , Células CHO , Análise por Conglomerados , Cricetinae , Hipocampo/citologia , Hipocampo/metabolismo , Cinética , Camundongos , Mitocôndrias/metabolismo , Neurônios/metabolismo , Distribuição Normal , Fosforilação , Solubilidade
11.
J Neurosci ; 33(39): 15362-75, 2013 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-24068803

RESUMO

Although it is known that cytosolic/soluble proteins synthesized in cell bodies are transported at much lower overall velocities than vesicles in fast axonal transport, the fundamental basis for this slow movement is unknown. Recently, we found that cytosolic proteins in axons of mouse cultured neurons are conveyed in a manner that superficially resembles diffusion, but with a slow anterograde bias that is energy- and motor-dependent (Scott et al., 2011). Here we show that slow axonal transport of synapsin, a prototypical member of this rate class, is dependent upon fast vesicle transport. Despite the distinct overall dynamics of slow and fast transport, experimentally induced and intrinsic variations in vesicle transport have analogous effects on slow transport of synapsin as well. Dynamic cotransport of vesicles and synapsin particles is also seen in axons, consistent with a model where higher-order assemblies of synapsin are conveyed by transient and probabilistic associations with vesicles moving in fast axonal transport. We posit that such dynamic associations generate the slow overall anterogradely biased flow of the population ("dynamic-recruitment model"). Our studies uncover the underlying kinetic basis for a classic cytosolic/soluble protein moving in slow axonal transport and reveal previously unknown links between slow and fast transport, offering a clearer conceptual picture of this curious phenomenon.


Assuntos
Transporte Axonal , Sinapsinas/metabolismo , Vesículas Transportadoras/metabolismo , Animais , Células Cultivadas , Cinética , Camundongos , Neurônios/metabolismo , Transporte Proteico
12.
Elife ; 122024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38713200

RESUMO

The cytosolic proteins synucleins and synapsins are thought to play cooperative roles in regulating synaptic vesicle (SV) recycling, but mechanistic insight is lacking. Here, we identify the synapsin E-domain as an essential functional binding-partner of α-synuclein (α-syn). Synapsin E-domain allows α-syn functionality, binds to α-syn, and is necessary and sufficient for enabling effects of α-syn at synapses of cultured mouse hippocampal neurons. Together with previous studies implicating the E-domain in clustering SVs, our experiments advocate a cooperative role for these two proteins in maintaining physiologic SV clusters.


Assuntos
Hipocampo , Neurônios , Sinapsinas , alfa-Sinucleína , Animais , Humanos , Camundongos , alfa-Sinucleína/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/química , Células Cultivadas , Hipocampo/metabolismo , Neurônios/metabolismo , Ligação Proteica , Domínios Proteicos , Sinapses/metabolismo , Sinapsinas/metabolismo , Sinapsinas/genética , Vesículas Sinápticas/metabolismo
13.
bioRxiv ; 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38895400

RESUMO

Coordinated assembly of individual components into higher-order structures is a defining theme in biology, but underlying principles are not well-understood. In neurons, α/ß spectrins, adducin, and actinfilaments assemble into a lattice wrapping underneath the axonal plasma membrane, but mechanistic events leading to this periodic axonal structure (PAS) are unclear. Visualizing PAS components in axons as they develop, we found focal patches in distal axons containing spectrins and adducin (but sparse actin filaments) with biophysical properties reminiscent of biomolecular condensation. Overexpressing spectrin-repeats - constituents of α/ß-spectrins - in heterologous cells triggered condensate formation, and preventing association of ßII-spectrin with actin-filaments/membranes also facilitated condensation. Finally, overexpressing condensate-triggering spectrin repeats in neurons before PAS establishment disrupted the lattice, presumably by competing with innate assembly, supporting a functional role for biomolecular condensation. We propose a condensation-assembly model where PAS components form focal phase-separated condensates that eventually unfurl into a stable lattice-structure by associating with subplasmalemmal actin. By providing local 'depots' of assembly parts, biomolecular condensation may play a wider role in the construction of intricate cytoskeletal structures.

14.
bioRxiv ; 2024 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-38895278

RESUMO

Gene-editing technologies promise to create a new class of therapeutics that can achieve permanent correction with a single intervention. Besides eliminating mutant alleles in familial disease, gene-editing can also be used to favorably manipulate upstream pathophysiologic events and alter disease-course in wider patient populations, but few such feasible therapeutic avenues have been reported. Here we use CRISPR-Cas9 to edit the last exon of amyloid precursor protein (App), relevant for Alzheimer's disease (AD). Our strategy effectively eliminates an endocytic (YENPTY) motif at APP C-terminus, while preserving the N-terminus and compensatory APP-homologues. This manipulation favorably alters events along the amyloid-pathway - inhibiting toxic APP-ß-cleavage fragments (including Aß) and upregulating neuroprotective APP-α-cleavage products. AAV-driven editing ameliorates neuropathologic, electrophysiologic, and behavioral deficits in an AD knockin mouse model. Effects persist for many months, and no abnormalities are seen in WT mice even after germline App-editing; underlining overall efficacy and safety. Pathologic alterations in the glial-transcriptome of App-KI mice, as seen by single nuclei RNA-sequencing (sNuc-Seq), are also normalized by App C-terminus editing. Our strategy takes advantage of innate transcriptional rules that render terminal exons insensitive to nonsense-decay, and the upstream manipulation is expected to be effective for all forms of AD. These studies offer a path for a one-time disease-modifying treatment for AD.

15.
J Neurosci ; 32(30): 10129-35, 2012 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-22836248

RESUMO

Although the presynaptic protein α-synuclein is a recognized player in neurodegeneration, its precise physiologic function(s) and/or role in human disease remains unclear. An emerging consensus from previous studies in lower-order systems is that α-synuclein interferes with vesicle-trafficking pathways; however putative neuronal correlates are unknown. Here we explore consequences of α-synuclein modulation in cultured mouse hippocampal neurons; coupling α-synuclein overexpression and knock-out model-systems with contemporary imaging paradigms. Our studies reveal an unexpected role of α-synuclein in attenuating the mobility of recycling pool (RP) vesicles between presynaptic boutons--called "superpool" trafficking--and also in maintaining the overall size of RPs at synapses. While an excess of α-synuclein led to smaller RPs and inhibited intersynaptic trafficking, an absence of α-synuclein triggered converse changes with larger RPs and enhanced intersynaptic trafficking. The data collectively suggest a model where α-synuclein maintains RP homeostasis by modulating intersynaptic vesicular dynamics, and provide a putative neuronal correlate of α-synuclein-induced impairments in vesicle-trafficking previously reported in lower-order systems.


Assuntos
Homeostase/fisiologia , Neurônios/metabolismo , Terminações Pré-Sinápticas/metabolismo , Vesículas Sinápticas/metabolismo , alfa-Sinucleína/metabolismo , Animais , Células Cultivadas , Hipocampo/metabolismo , Camundongos , Camundongos Knockout , Vesículas Sinápticas/genética , alfa-Sinucleína/genética
16.
bioRxiv ; 2023 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-37425805

RESUMO

The cytosolic proteins synucleins and synapsins are thought to play cooperative roles in regulating synaptic vesicle (SV) recycling, but mechanistic insight is lacking. Here we identify the synapsin E-domain as an essential functional binding-partner of α-synuclein (α-syn). Synapsin E-domain allows α-syn functionality, binds to α-syn, and is necessary and sufficient for enabling effects of α-syn at the synapse. Together with previous studies implicating the E-domain in clustering SVs, our experiments advocate a cooperative role for these two proteins in maintaining physiologic SV clusters.

17.
Biomaterials ; 293: 121959, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36527789

RESUMO

Genome editing of somatic cells via clustered regularly interspaced short palindromic repeats (CRISPR) offers promise for new therapeutics to treat a variety of genetic disorders, including neurological diseases. However, the dense and complex parenchyma of the brain and the post-mitotic state of neurons make efficient genome editing challenging. In vivo delivery systems for CRISPR-Cas proteins and single guide RNA (sgRNA) include both viral vectors and non-viral strategies, each presenting different advantages and disadvantages for clinical application. We developed non-viral and biodegradable PEGylated nanocapsules (NCs) that deliver preassembled Cas9-sgRNA ribonucleoproteins (RNPs). Here, we show that the RNP NCs led to robust genome editing in neurons following intracerebral injection into the healthy mouse striatum. Genome editing was predominantly observed in medium spiny neurons (>80%), with occasional editing in cholinergic, calretinin, and parvalbumin interneurons. Glial activation was minimal and was localized along the needle tract. Our results demonstrate that the RNP NCs are capable of safe and efficient neuronal genome editing in vivo.


Assuntos
Edição de Genes , Nanocápsulas , Animais , Camundongos , Edição de Genes/métodos , Sistemas CRISPR-Cas/genética , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Neurônios/metabolismo , Encéfalo/metabolismo
18.
Methods Mol Biol ; 2431: 163-179, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35412276

RESUMO

The polarized morphology of neurons necessitates the delivery of proteins synthesized in the soma along the length of the axon to distal synapses; critical for sustaining communication between neurons. This constitutive and dynamic process of protein transport along axons termed "axonal transport" was initially characterized by classic pulse-chase radiolabeling studies which identified two major rate components: a fast component and a slow component. Early radiolabeling studies indicated "cohesive co-transport" of slow transport cargos. However, this approach could not be used to visualize or provide mechanistic insights on this highly dynamic process. The advent of fluorescent and photoactivatable imaging probes have now enabled real-time imaging of axonal transport. Conventional fluorescent probes have helped visualize and characterize the molecular mechanisms of transport of vesicular proteins. These proteins typically move in the fast component of axonal transport and appear as "punctate structures" along axons. However, a large majority of transported proteins that move in the slow component of transport, typically show a "uniform diffusive glow" along axons when tagged to conventional fluorescent probes. This makes it challenging to unequivocally track them in real time. Our lab has used photoactivatable fluorescent probes to tag three individual cytosolic proteins moving in the slow component of axonal transport, and identified three distinct modes of transport along axons. Our data from these experiments argue against the prevailing hypothesis based on classic radiolabeling studies, which suggested that all slow-transport proteins may move along the axon as one large macromolecular protein complex. Although other labs have started using photoactivation to study axonal transport of cytosolic proteins, this technique remains largely under-utilized. Here, we describe the detailed protocols to image and analyze axonal transport of three typical slow-component cargoes along axons of cultured hippocampal neurons.


Assuntos
Transporte Axonal , Corantes Fluorescentes , Transporte Axonal/fisiologia , Axônios/metabolismo , Corantes Fluorescentes/metabolismo , Neurônios , Transporte Proteico/fisiologia
19.
J Neurosci ; 30(24): 8083-95, 2010 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-20554859

RESUMO

Several neurodegenerative diseases are typified by intraneuronal alpha-synuclein deposits, synaptic dysfunction, and dementia. While even modest alpha-synuclein elevations can be pathologic, the precise cascade of events induced by excessive alpha-synuclein and eventually culminating in synaptotoxicity is unclear. To elucidate this, we developed a quantitative model system to evaluate evolving alpha-synuclein-induced pathologic events with high spatial and temporal resolution, using cultured neurons from brains of transgenic mice overexpressing fluorescent-human-alpha-synuclein. Transgenic alpha-synuclein was pathologically altered over time and overexpressing neurons showed striking neurotransmitter release deficits and enlarged synaptic vesicles; a phenotype reminiscent of previous animal models lacking critical presynaptic proteins. Indeed, several endogenous presynaptic proteins involved in exocytosis and endocytosis were undetectable in a subset of transgenic boutons ("vacant synapses") with diminished levels in the remainder, suggesting that such diminutions were triggering the overall synaptic pathology. Similar synaptic protein alterations were also retrospectively seen in human pathologic brains, highlighting potential relevance to human disease. Collectively the data suggest a previously unknown cascade of events where pathologic alpha-synuclein leads to a loss of a number of critical presynaptic proteins, thereby inducing functional synaptic deficits.


Assuntos
Proteínas de Filamentos Intermediários/efeitos adversos , Proteínas de Filamentos Intermediários/metabolismo , Degeneração Neural/induzido quimicamente , Degeneração Neural/patologia , Neurônios/efeitos dos fármacos , Sinapses/patologia , Idoso , Idoso de 80 Anos ou mais , Animais , Animais Recém-Nascidos , Células Cultivadas , Demência/metabolismo , Demência/patologia , Proteína 4 Homóloga a Disks-Large , Relação Dose-Resposta a Droga , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , Proteínas de Fluorescência Verde/genética , Guanilato Quinases , Hipocampo/citologia , Humanos , Proteínas de Filamentos Intermediários/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão/métodos , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/ultraestrutura , Técnicas de Patch-Clamp , Peptídeo Hidrolases/farmacologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Compostos de Piridínio/metabolismo , Compostos de Amônio Quaternário/metabolismo , Sinapses/genética , Sinapses/ultraestrutura
20.
Blood ; 114(12): 2542-52, 2009 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-19506297

RESUMO

Cystinosis is an autosomal recessive metabolic disease that belongs to the family of lysosomal storage disorders. The defective gene is CTNS encoding the lysosomal cystine transporter, cystinosin. Cystine accumulates in every organ in the body and leads to organ damage and dysfunction, including renal defects. Using the murine model for cystinosis, Ctns(-/-) mice, we performed syngeneic bone marrow cell (BMC), hematopoietic stem cell (HSC), and mesenchymal stem cell transplantation. Organ-specific cystine content was reduced by 57% to 94% in all organs tested in the BMC-treated mice. Confocal microscopy and quantitative polymerase chain reaction revealed a large quantity of transplanted BMC in all organs tested, from 5% to 19% of the total cells. Most of these cells were not from the lymphoid lineage but part of the intrinsic structure of the organ. The natural progression of renal dysfunction was prevented, and deposition of corneal cystine crystals was significantly improved in the BMC-treated mice. HSC had the same therapeutic effect as whole BMC. In contrast, mesenchymal stem cell did not integrate efficiently in any organ. This work is a proof of concept for using HSC transplantation as a therapy for cystinosis and highlights the efficiency of this strategy for a chronic, progressive degenerative disease.


Assuntos
Transplante de Medula Óssea , Cistinose/cirurgia , Modelos Animais de Doenças , Animais , Western Blotting , Cistinose/sangue , Cistinose/urina , Citometria de Fluxo , Imunofluorescência , Transplante de Células-Tronco Hematopoéticas , Técnicas Imunoenzimáticas , Luciferases/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa