Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 34
1.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119720, 2024 Apr 04.
Article En | MEDLINE | ID: mdl-38582237

Nitric oxide can covalently modify cysteine thiols on target proteins to alter that protein's function in a process called S-nitrosylation (SNO). S-nitrosylation of synaptic proteins plays an integral part in neurotransmission. Here we review the function of the SNO-proteome at the synapse and whether clusters of SNO-modification may predict synaptic dysfunction associated with disease. We used a systematic search strategy to concatenate SNO-proteomic datasets from normal human or murine brain samples. Identified SNO-modified proteins were then filtered against proteins reported in the Synaptome Database, which provides a detailed and experimentally verified annotation of all known synaptic proteins. Subsequently, we performed an unbiased network analysis of all known SNO-synaptic proteins to identify clusters of SNO proteins commonly involved in biological processes or with known disease associations. The resulting SNO networks were significantly enriched in biological processes related to metabolism, whereas significant gene-disease associations were related to Schizophrenia, Alzheimer's, Parkinson's and Huntington's disease. Guided by an unbiased network analysis, the current review presents a thorough discussion of how clustered changes to the SNO-proteome influence health and disease.

2.
Cell Death Dis ; 15(4): 246, 2024 Apr 04.
Article En | MEDLINE | ID: mdl-38575601

Parkinson's disease (PD) is a debilitating neurodegenerative disease characterized by the loss of midbrain dopaminergic neurons (DaNs) and the abnormal accumulation of α-Synuclein (α-Syn) protein. Currently, no treatment can slow nor halt the progression of PD. Multiplications and mutations of the α-Syn gene (SNCA) cause PD-associated syndromes and animal models that overexpress α-Syn replicate several features of PD. Decreasing total α-Syn levels, therefore, is an attractive approach to slow down neurodegeneration in patients with synucleinopathy. We previously performed a genetic screen for modifiers of α-Syn levels and identified CDK14, a kinase of largely unknown function as a regulator of α-Syn. To test the potential therapeutic effects of CDK14 reduction in PD, we ablated Cdk14 in the α-Syn preformed fibrils (PFF)-induced PD mouse model. We found that loss of Cdk14 mitigates the grip strength deficit of PFF-treated mice and ameliorates PFF-induced cortical α-Syn pathology, indicated by reduced numbers of pS129 α-Syn-containing cells. In primary neurons, we found that Cdk14 depletion protects against the propagation of toxic α-Syn species. We further validated these findings on pS129 α-Syn levels in PD patient neurons. Finally, we leveraged the recent discovery of a covalent inhibitor of CDK14 to determine whether this target is pharmacologically tractable in vitro and in vivo. We found that CDK14 inhibition decreases total and pathologically aggregated α-Syn in human neurons, in PFF-challenged rat neurons and in the brains of α-Syn-humanized mice. In summary, we suggest that CDK14 represents a novel therapeutic target for PD-associated synucleinopathy.


Neurodegenerative Diseases , Parkinson Disease , Synucleinopathies , Animals , Humans , Mice , Rats , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Dopaminergic Neurons/metabolism , Mesencephalon/metabolism , Neurodegenerative Diseases/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Parkinson Disease/metabolism , Synucleinopathies/metabolism , Synucleinopathies/pathology
3.
Child Abuse Negl ; 146: 106502, 2023 12.
Article En | MEDLINE | ID: mdl-37844460

BACKGROUND: A growing body of research has focused on the relationships of policies and other macro factors and child welfare outcomes. However, to date, few studies have examined state child welfare policies and reunification, despite reunification being the priority case goal among children in foster care. OBJECTIVE: This study examined the relationship between state child welfare policies and other macro factors and reunification, while controlling for child factors. PARTICIPANTS AND SETTING: Data came from the 2016-2019 Adoption and Foster Care Analysis Reporting System (AFCARS), which contains case-level information on all children in foster care during each fiscal year. METHODS: We conducted a series of multi-level survival analyses to observe the fixed effects of state-level factors and child-level factors on time to reunification among children who entered foster care in 2016. RESULTS: In the unadjusted model, children in states with Title IV-E stipend programs for caseworkers had higher hazards of reunification (HR = 1.21, 95 % CI = 1.01-1.44) as did children in states with statewide in-home post-reunification services (HR = 1.63, 95 % CI = 1.16-2.28). Children in states that required a master's degree for caseworkers had lower hazards of reunification (HR = 0.72, 95 % CI = 0.59-0.95). CONCLUSIONS: Findings from this study highlight the importance of considering state policies and their impact on reunification. Implications for policy, practice, and research are explored.


Child Welfare , Foster Home Care , Humans , Child , Survival Analysis , Policy , Motivation
4.
J Neurosci ; 42(50): 9473-9487, 2022 12 14.
Article En | MEDLINE | ID: mdl-36414406

Cortical synucleinopathies, including dementia with Lewy bodies and Parkinson's disease dementia, collectively known as Lewy body dementia, are characterized by the aberrant aggregation of misfolded α-synuclein (α-syn) protein into large inclusions in cortical tissue, leading to impairments in proteostasis and synaptic connectivity and eventually resulting in neurodegeneration. Here, we show that male and female rat cortical neurons exposed to exogenous α-syn preformed fibrils accumulate large, detergent-insoluble, PS129-labeled deposits at synaptic terminals. Live-cell imaging of calcium dynamics coupled with assessment of network activity reveals that aberrant intracellular accumulation of α-syn inhibits synaptic response to glutamate through NMDARs, although deficits manifest slowly over a 7 d period. Impairments in NMDAR activity temporally correlated with increased nitric oxide synthesis and S-nitrosylation of the dendritic scaffold protein, microtubule-associated protein 1A. Inhibition of nitric oxide synthesis via the nitric oxide synthase inhibitor l-NG-nitroarginine methyl ester blocked microtubule-associated protein 1A S-nitrosylation and normalized NMDAR-dependent inward calcium transients and overall network activity. Collectively, these data suggest that loss of synaptic function in Lewy body dementia may result from synucleinopathy-evoked nitrosative stress and subsequent NMDAR dysfunction.SIGNIFICANCE STATEMENT This work shows the importance of the redox state of microtubule-associated protein 1A in the maintenance of synaptic function through regulation of NMDAR. We show that α-syn preformed fibrils promote nitric oxide synthesis, which triggers S-nitrosylation of microtubule-associated protein 1A, leading to impairment of NMDAR-dependent glutamate responses. This offers insight into the mechanism of synaptic dysfunction in Lewy body dementia.


Dementia , Lewy Body Disease , Parkinson Disease , Synucleinopathies , Male , Female , Animals , Rats , alpha-Synuclein/metabolism , Lewy Body Disease/metabolism , Calcium/metabolism , Nitric Oxide/metabolism , Parkinson Disease/metabolism , Synucleinopathies/metabolism , Receptors, N-Methyl-D-Aspartate , Glutamates , Microtubule-Associated Proteins/metabolism
6.
NPJ Parkinsons Dis ; 8(1): 104, 2022 Aug 11.
Article En | MEDLINE | ID: mdl-35953517

Parkinson's Disease (PD) is a neurodegenerative disorder characterized, in part, by the loss of dopaminergic neurons within the nigral-striatal pathway. Multiple lines of evidence support a role for reactive nitrogen species (RNS) in degeneration of this pathway, specifically nitric oxide (NO). This review will focus on how RNS leads to loss of dopaminergic neurons in PD and whether RNS accumulation represents a central signal in the degenerative cascade. Herein, we provide an overview of how RNS accumulates in PD by considering the various cellular sources of RNS including nNOS, iNOS, nitrate, and nitrite reduction and describe evidence that these sources are upregulating RNS in PD. We document that over 1/3 of the proteins that deposit in Lewy Bodies, are post-translationally modified (S-nitrosylated) by RNS and provide a broad description of how this elicits deleterious effects in neurons. In doing so, we identify specific proteins that are modified by RNS in neurons which are implicated in PD pathogenesis, with an emphasis on exacerbation of synucleinopathy. How nitration of alpha-synuclein (aSyn) leads to aSyn misfolding and toxicity in PD models is outlined. Furthermore, we delineate how RNS modulates known PD-related phenotypes including axo-dendritic-, mitochondrial-, and dopamine-dysfunctions. Finally, we discuss successful outcomes of therapeutics that target S-nitrosylation of proteins in Parkinson's Disease related clinical trials. In conclusion, we argue that targeting RNS may be of therapeutic benefit for people in early clinical stages of PD.

7.
Cell Rep ; 35(6): 109099, 2021 05 11.
Article En | MEDLINE | ID: mdl-33979611

Neuronal loss in Parkinson's disease (PD) is associated with impaired proteostasis and accumulation of α-syn microaggregates in dopaminergic neurons. These microaggregates promote seeding of α-synuclein (α-syn) pathology between synaptically linked neurons. However, the mechanism by which seeding is initiated is not clear. Using human pluripotent stem cell (hPSC) models of PD that allow comparison of SNCA mutant cells with isogenic controls, we find that SNCA mutant neurons accumulate α-syn deposits that cluster to multiple endomembrane compartments, specifically multivesicular bodies (MVBs) and lysosomes. We demonstrate that A53T and E46K α-syn variants bind and sequester LC3B monomers into detergent-insoluble microaggregates on the surface of late endosomes, increasing α-syn excretion via exosomes and promoting seeding of α-syn from SNCA mutant neurons to wild-type (WT) isogenic controls. Finally, we show that constitutive inactivation of LC3B promotes α-syn accumulation and seeding, while LC3B activation inhibits these events, offering mechanistic insight into the spread of synucleinopathy in PD.


Exocytosis/genetics , Exosomes/metabolism , Parkinson Disease/genetics , alpha-Synuclein/metabolism , Cell Differentiation , Humans , Mutation , Parkinson Disease/pathology , Transfection
8.
Biomol NMR Assign ; 15(2): 297-303, 2021 10.
Article En | MEDLINE | ID: mdl-33797711

Alpha-synuclein (α-syn) is a small presynaptic protein that is believed to play an important role in the pathogenesis of Parkinson's disease (PD). It localizes to presynaptic terminals where it partitions between a cytosolic soluble and a lipid-bound state. Recent evidence suggests that α-syn can also associate with mitochondrial membranes where it interacts with a unique anionic phospholipid cardiolipin (CL). Here, we examine the conformation of the flexible fragments of a monomeric α-syn bound to lipid vesicles composed of anionic 1,2-dioleoyl-sn-glycero-3-phosphate (DOPA) and 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) lipids, of tetraoleoyl CL (TOCL) and DOPC, and of fibrils. The dynamic properties of α-syn associated with DOPA:DOPC vesicles were the most favorable for conducting three-dimensional NMR experiments, and the 13C, 15N and amide 1H chemical shifts of the flexible and disordered C-terminus of α-syn could be assigned using three-dimensional through-bond magic angle spinning NMR spectroscopy. Although the C-terminus is more dynamically constrained in fibrils and in α-syn bound to TOCL:DOPC vesicles, a direct comparison of carbon chemical shifts detected using through bond two-dimensional spectroscopy indicates that the C-terminus is flexible and unstructured in all the three samples.


alpha-Synuclein
9.
Mol Biol Cell ; 32(7): 511-520, 2021 04 01.
Article En | MEDLINE | ID: mdl-33502893

Evidence suggests that n-3 polyunsaturated fatty acids may act as activators of the Nrf2 antioxidant pathway. The antioxidant response, in turn, promotes neuronal differentiation and neurite outgrowth. Nrf2 has recently been suggested to be a cell intrinsic mediator of docosohexanoic acid (DHA) signaling. In the current study, we assessed whether DHA-mediated axodendritic development was dependent on activation of the Nrf2 pathway and whether Nrf2 protected from agrochemical-induced neuritic retraction. Expression profiling of the DHA-enriched Fat-1 mouse brain relative to wild type showed a significant enrichment of genes associated with neuronal development and neuronal projection and genes associated with the Nrf2-transcriptional pathway. Moreover, we found that primary cortical neurons treated with DHA showed a dose-dependent increase in Nrf2 transcriptional activity and Nrf2-target gene expression. DHA-mediated activation of Nrf2 promoted neurite outgrowth and inhibited oxidative stress-induced neuritic retraction evoked by exposure to agrochemicals. Finally, we provide evidence that this effect is largely dependent on induction of the Nrf2-target gene NAD(P)H: (quinone acceptor) oxidoreductase 1 (NQO1), and that silencing of either Nrf2 or Nqo1 blocks the effects of DHA on the axodendritic compartment. Collectively, these data support a role for the Nrf2-NQO1 pathway in DHA-mediated axodendritic development and protection from agrochemical exposure.


NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Neuronal Outgrowth/physiology , Animals , Antioxidants/pharmacology , Cell Line, Tumor , Dendrites/metabolism , Docosahexaenoic Acids/pharmacology , Humans , Mice , NF-E2-Related Factor 2/genetics , Neuronal Outgrowth/genetics , Neurons/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , Signal Transduction/drug effects , Transcription, Genetic/drug effects
10.
Proc Natl Acad Sci U S A ; 116(28): 14280-14289, 2019 07 09.
Article En | MEDLINE | ID: mdl-31235589

While mutations in the SNCA gene (α-synuclein [α-syn]) are causal in rare familial forms of Parkinson's disease (PD), the prevalence of α-syn aggregates in the cortices of sporadic disease cases emphasizes the need to understand the link between α-syn accumulation and disease pathogenesis. By employing a combination of human pluripotent stem cells (hPSCs) that harbor the SNCA-A53T mutation contrasted against isogenic controls, we evaluated the consequences of α-syn accumulation in human A9-type dopaminergic (DA) neurons (hNs). We show that the early accumulation of α-syn in SNCA-A53T hNs results in changes in gene expression consistent with the expression profile of the substantia nigra (SN) from PD patients, analyzed post mortem. Differentially expressed genes from both PD patient SN and SNCA-A53T hNs were associated with regulatory motifs transcriptionally activated by the antioxidant response pathway, particularly Nrf2 gene targets. Differentially expressed gene targets were also enriched for gene ontologies related to microtubule binding processes. We thus assessed the relationship between Nrf2-mediated gene expression and neuritic pathology in SNCA-A53T hNs. We show that SNCA-mutant hNs have deficits in neuritic length and complexity relative to isogenic controls as well as contorted axons with Tau-positive varicosities. Furthermore, we show that mutant α-syn fails to complex with protein kinase C (PKC), which, in turn, results in impaired activation of Nrf2. These neuritic defects result from impaired Nrf2 activity on antioxidant response elements (AREs) localized to a microtubule-associated protein (Map1b) gene enhancer and are rescued by forced expression of Map1b as well as by both Nrf2 overexpression and pharmaceutical activation in PD neurons.


Microtubule-Associated Proteins/genetics , NF-E2-Related Factor 2/genetics , Parkinson Disease/genetics , alpha-Synuclein/genetics , Animals , Antioxidant Response Elements/genetics , Axons/drug effects , Axons/pathology , Cell Differentiation/genetics , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Enhancer Elements, Genetic , Humans , Mitochondria/metabolism , Mitochondria/pathology , Mutation , Neurites/metabolism , Neurites/pathology , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , Protein Kinase C/genetics , Substantia Nigra/metabolism , Substantia Nigra/pathology
11.
FASEB J ; 32(10): 5350-5364, 2018 10.
Article En | MEDLINE | ID: mdl-29688812

Neuronal loss in Parkinson's disease (PD) is associated with aberrant mitochondrial function in dopaminergic (DA) neurons of the substantia nigra pars compacta. An association has been reported between PD onset and exposure to mitochondrial toxins, including the agrochemicals paraquat (PQ), maneb (MB), and rotenone (Rot). Here, with the use of a patient-derived stem cell model of PD, allowing comparison of DA neurons harboring a mutation in the α-synuclein (α-syn) gene ( SNCA-A53T) against isogenic, mutation-corrected controls, we describe a novel mechanism whereby NO, generated from SNCA-A53T mutant neurons exposed to Rot or PQ/MB, inhibits anterograde mitochondrial transport through nitration of α-tubulin (α-Tub). Nitration of α-Tub inhibited the association of both α-syn and the mitochondrial motor protein kinesin 5B with the microtubules, arresting anterograde transport. This was, in part, a result of nitration of α-Tub in the C-terminal domain. These effects were rescued by inhibiting NO synthesis with the NOS inhibitor Nω-nitro-L-arginine methyl ester. Collectively, our results are the first to demonstrate a gene by environment interaction in PD, whereby agrochemical exposure selectively triggers a deficit in mitochondrial transport by nitrating the microtubules in neurons harboring the SNCA-A53T mutation.-Stykel, M. G., Humphries, K., Kirby, M. P., Czaniecki, C., Wang, T., Ryan, T., Bamm, V., Ryan, S. D. Nitration of microtubules blocks axonal mitochondrial transport in a human pluripotent stem cell model of Parkinson's disease.


Axonal Transport , Axons/metabolism , Induced Pluripotent Stem Cells/metabolism , Microtubules/metabolism , Models, Biological , Parkinson Disease/metabolism , Amino Acid Substitution , Axons/pathology , Cell Line , Humans , Induced Pluripotent Stem Cells/pathology , Microtubules/genetics , Microtubules/pathology , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mutation, Missense , Nitric Oxide/genetics , Nitric Oxide/metabolism , Parkinson Disease/genetics , Protein Transport/genetics , Tubulin/genetics , Tubulin/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
12.
Nat Commun ; 9(1): 817, 2018 02 26.
Article En | MEDLINE | ID: mdl-29483518

Neuronal loss in Parkinson's disease (PD) is associated with aberrant mitochondrial function and impaired proteostasis. Identifying the mechanisms that link these pathologies is critical to furthering our understanding of PD pathogenesis. Using human pluripotent stem cells (hPSCs) that allow comparison of cells expressing mutant SNCA (encoding α-synuclein (α-syn)) with isogenic controls, or SNCA-transgenic mice, we show that SNCA-mutant neurons display fragmented mitochondria and accumulate α-syn deposits that cluster to mitochondrial membranes in response to exposure of cardiolipin on the mitochondrial surface. Whereas exposed cardiolipin specifically binds to and facilitates refolding of α-syn fibrils, prolonged cardiolipin exposure in SNCA-mutants initiates recruitment of LC3 to the mitochondria and mitophagy. Moreover, we find that co-culture of SNCA-mutant neurons with their isogenic controls results in transmission of α-syn pathology coincident with mitochondrial pathology in control neurons. Transmission of pathology is effectively blocked using an anti-α-syn monoclonal antibody (mAb), consistent with cell-to-cell seeding of α-syn.


Cardiolipins/pharmacology , Mitochondria/drug effects , Mitochondrial Membranes/drug effects , Neurons/metabolism , Parkinson Disease, Secondary/genetics , alpha-Synuclein/genetics , Animals , Antibodies, Monoclonal/pharmacology , Cell Communication , Cell Differentiation , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Gene Expression , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/ultrastructure , Mitophagy/drug effects , Mutation , Neurons/drug effects , Neurons/pathology , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Protein Folding/drug effects , Protein Transport/drug effects , alpha-Synuclein/metabolism
13.
Brain Plast ; 4(2): 151-167, 2018 Dec 26.
Article En | MEDLINE | ID: mdl-30598867

Alzheimer's disease (AD) and Parkinson's disease (PD) share many commonalities ranging from signaling deficits such as altered cholinergic activity, neurotrophin and insulin signaling to cell stress cascades that result in proteinopathy, mitochondrial dysfunction and neuronal cell death. These pathological processes are not unidirectional, but are intertwined, resulting in a series of feed-forward loops that worsen symptoms and advance disease progression. At the center of these loops is glycogen synthase kinase-3 (GSK-3), a keystone protein involved in many of the multidirectional biological processes that contribute to AD and PD neuropathology. Here, a unified overview of the involvement of GSK-3 in the major processes involved in these diseases will be presented. The mechanisms by which these processes are linked will be discussed and the feed-forward pathways identified. In this regard, this review will put forth the notion that combination therapy, targeting these multiple facets of AD or PD neuropathology is a necessary next step in the search for effective therapies.

14.
Nat Commun ; 8(1): 1488, 2017 11 14.
Article En | MEDLINE | ID: mdl-29133852

Transcription factor MEF2C regulates multiple genes linked to autism spectrum disorder (ASD), and human MEF2C haploinsufficiency results in ASD, intellectual disability, and epilepsy. However, molecular mechanisms underlying MEF2C haploinsufficiency syndrome remain poorly understood. Here we report that Mef2c +/-(Mef2c-het) mice exhibit behavioral deficits resembling those of human patients. Gene expression analyses on brains from these mice show changes in genes associated with neurogenesis, synapse formation, and neuronal cell death. Accordingly, Mef2c-het mice exhibit decreased neurogenesis, enhanced neuronal apoptosis, and an increased ratio of excitatory to inhibitory (E/I) neurotransmission. Importantly, neurobehavioral deficits, E/I imbalance, and histological damage are all ameliorated by treatment with NitroSynapsin, a new dual-action compound related to the FDA-approved drug memantine, representing an uncompetitive/fast off-rate antagonist of NMDA-type glutamate receptors. These results suggest that MEF2C haploinsufficiency leads to abnormal brain development, E/I imbalance, and neurobehavioral dysfunction, which may be mitigated by pharmacological intervention.


Autistic Disorder/genetics , Brain/growth & development , Excitatory Amino Acid Antagonists/therapeutic use , Haploinsufficiency , Memantine/analogs & derivatives , Memantine/therapeutic use , Animals , Autistic Disorder/pathology , Autistic Disorder/physiopathology , Behavior, Animal , Biomarkers/metabolism , Brain/pathology , Brain/physiopathology , Cell Death , Disease Models, Animal , Down-Regulation , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Profiling , Humans , Long-Term Potentiation/genetics , MEF2 Transcription Factors/genetics , Memantine/pharmacology , Mice, Inbred C57BL , Neurogenesis/genetics , Neurons/pathology , Phenotype , Receptors, N-Methyl-D-Aspartate/drug effects , Synapses/pathology , Synaptic Transmission/genetics
15.
Hum Mutat ; 35(9): 1101-13, 2014 Sep.
Article En | MEDLINE | ID: mdl-24930953

Early-onset dystonia is associated with the deletion of one of a pair of glutamic acid residues (c.904_906delGAG/c.907_909delGAG; p.Glu302del/Glu303del; ΔE 302/303) near the carboxyl-terminus of torsinA, a member of the AAA(+) protein family that localizes to the endoplasmic reticulum lumen and nuclear envelope. This deletion commonly underlies early-onset DYT1 dystonia. While the role of the disease-causing mutation, torsinAΔE, has been established through genetic association studies, it is much less clear whether other rare human variants of torsinA are pathogenic. Two missense variations have been described in single patients: R288Q (c.863G>A; p.Arg288Gln; R288Q) identified in a patient with onset of severe generalized dystonia and myoclonus since infancy and F205I (c.613T>A, p.Phe205Ile; F205I) in a psychiatric patient with late-onset focal dystonia. In this study, we have undertaken a series of analyses comparing the biochemical and cellular effects of these rare variants to torsinAΔE and wild-type (wt) torsinA to reveal whether there are common dysfunctional features. The results revealed that the variants, R288Q and F205I, are more similar in their properties to torsinAΔE protein than to torsinAwt. These findings provide functional evidence for the potential pathogenic nature of these rare sequence variants in the TOR1A gene, thus implicating these pathologies in the development of dystonia.


Dystonia Musculorum Deformans/genetics , Genetic Variation , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , Gene Knockdown Techniques , Genetic Association Studies , Humans , Models, Molecular , Molecular Chaperones/metabolism , Molecular Dynamics Simulation , Mutation , Phenotype , Protein Conformation , Protein Multimerization , Protein Transport , Viral Envelope Proteins/metabolism
16.
Cell ; 155(6): 1351-64, 2013 Dec 05.
Article En | MEDLINE | ID: mdl-24290359

Parkinson's disease (PD) is characterized by loss of A9 dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc). An association has been reported between PD and exposure to mitochondrial toxins, including environmental pesticides paraquat, maneb, and rotenone. Here, using a robust, patient-derived stem cell model of PD allowing comparison of A53T α-synuclein (α-syn) mutant cells and isogenic mutation-corrected controls, we identify mitochondrial toxin-induced perturbations in A53T α-syn A9 DA neurons (hNs). We report a pathway whereby basal and toxin-induced nitrosative/oxidative stress results in S-nitrosylation of transcription factor MEF2C in A53T hNs compared to corrected controls. This redox reaction inhibits the MEF2C-PGC1α transcriptional network, contributing to mitochondrial dysfunction and apoptotic cell death. Our data provide mechanistic insight into gene-environmental interaction (GxE) in the pathogenesis of PD. Furthermore, using small-molecule high-throughput screening, we identify the MEF2C-PGC1α pathway as a therapeutic target to combat PD.


Gene-Environment Interaction , Mitochondria/drug effects , Paraquat/toxicity , Parkinson Disease/genetics , Parkinson Disease/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , MEF2 Transcription Factors , Mutation/drug effects , Neurons/metabolism , Oxidative Stress , Parkinson Disease/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Reactive Nitrogen Species/metabolism , Substantia Nigra/metabolism , Transcription Factors/metabolism , Transcription, Genetic , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
17.
J Neurosci ; 33(35): 14170-83, 2013 Aug 28.
Article En | MEDLINE | ID: mdl-23986251

The synaptic toxicity of soluble amyloid-ß (Aß) oligomers plays a critical role in the pathophysiology of Alzheimer's disease (AD). Here we report that overexpressed α1-takusan, which we previously identified as a protein that enhances synaptic activity via interaction with PSD-95, mitigates oligomeric Aß-induced synaptic loss. In contrast, takusan knockdown results in enhanced synaptic damage. α1-Takusan interacts with tau either directly or indirectly, and prevents Aß-induced tau hyperphosphorylation and mitochondrial fragmentation. Deletion analysis identified the second domain (D2) within the takusan protein that is required for PSD-95 clustering and synaptic protection from Aß. A 51 aa sequence linking D2 to the PDZ-binding C terminus was found to be as effective as full-length takusan in protecting synapses from Aß-induced damage. Moreover, a sequence containing the D2 from the human protein discs large homolog 5, when linked to a C-terminal PDZ-binding motif, can also increase the clustering of PSD-95 in cortical dendrites. In summary, α1-takusan protects synapses from Aß-induced insult via interaction with PSD-95 and tau. Thus, takusan-based protein sequences from either mouse or human may be of potential therapeutic benefit in AD.


Amyloid beta-Peptides/toxicity , Membrane Proteins/metabolism , Neurons/metabolism , Synapses/metabolism , tau Proteins/metabolism , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Disks Large Homolog 4 Protein , Hippocampus/cytology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mitochondria/metabolism , Neurons/physiology , PDZ Domains , Rats , Synaptic Potentials , Tumor Suppressor Proteins/metabolism
18.
J Neurosci ; 32(45): 15837-42, 2012 Nov 07.
Article En | MEDLINE | ID: mdl-23136422

After transplantation, individual stem cell-derived neurons can functionally integrate into the host CNS; however, evidence that neurons derived from transplanted human embryonic stem cells (hESCs) can drive endogenous neuronal network activity in CNS tissue is still lacking. Here, using multielectrode array recordings, we report activation of high-frequency oscillations in the ß and γ ranges (10-100 Hz) in the host hippocampal network via targeted optogenetic stimulation of transplanted hESC-derived neurons.


Embryonic Stem Cells/transplantation , Hippocampus/physiology , Neural Stem Cells/transplantation , Neurons/transplantation , Action Potentials/physiology , Animals , Embryonic Stem Cells/cytology , Female , Hippocampus/cytology , Humans , Male , Neural Stem Cells/cytology , Neurogenesis/physiology , Neurons/cytology , Optogenetics , Rats , Rats, Sprague-Dawley
19.
Bioarchitecture ; 2(1): 2-5, 2012 Jan 01.
Article En | MEDLINE | ID: mdl-22754620

Crosslinking proteins maintain organelle structure and facilitate their function through the crosslinking of cytoskeletal elements. We recently found an interaction between the giant crosslinking protein dystonin-a2 and the microtubule-associated protein-1B (MAP1B), occurring in the centrosomal region of the cell. In addition, we showed that this interaction is necessary to maintain microtubule acetylation. Loss of dystonin-a2 disrupts MT stability, Golgi organization, and flux through the secretory pathway. This, coupled to our recent finding that dystonin-a2 is critical in maintaining endoplasmic reticulum (ER) structure and function, provides novel insight into the importance of dystonin in maintenance of organelle structure and in facilitating intracellular transport. These results highlight the importance of cytoskeletal dynamics in communicating signals between organelle membranes and the cytoskeleton. Importantly, they demonstrate how defects in cytoskeletal dynamics can translate into a failure of vesicular trafficking associated with neurodegenerative disease.

20.
Int J Cell Biol ; 2012: 634214, 2012.
Article En | MEDLINE | ID: mdl-22611399

Most cases of early onset DYT1 dystonia in humans are caused by a GAG deletion in the TOR1A gene leading to loss of a glutamic acid (ΔE) in the torsinA protein, which underlies a movement disorder associated with neuronal dysfunction without apparent neurodegeneration. Mutation/deletion of the gene (Dst) encoding dystonin in mice results in a dystonic movement disorder termed dystonia musculorum, which resembles aspects of dystonia in humans. While torsinA and dystonin proteins do not share modular domain architecture, they participate in a similar function by modulating a structural link between the nuclear envelope and the cytoskeleton in neuronal cells. We suggest that through a shared interaction with the nuclear envelope protein nesprin-3α, torsinA and the neuronal dystonin-a2 isoform comprise a bridge complex between the outer nuclear membrane and the cytoskeleton, which is critical for some aspects of neuronal development and function. Elucidation of the overlapping roles of torsinA and dystonin-a2 in nuclear/endoplasmic reticulum dynamics should provide insights into the cellular mechanisms underlying the dystonic phenotype.

...