44 results on '"Milnerwood, Aj"'
Search Results
2. Translation initiator EIF4G1 mutations in familial Parkinson disease
- Author
-
Chartier Harlin, M, Dachsel, Jc, Vilariño Güell, C, Lincoln, Sj, Leprêtre, F, Hulihan, Mm, Kachergus, J, Milnerwood, Aj, Tapia, L, Song, M, Le Rhun, E, Mutez, E, Larvor, L, Duflot, A, Vanbesien Mailliot, C, Kreisler, A, Ross, Oa, Nishioka, K, Soto Ortolaza, Ai, Cobb, Sa, Melrose, Hl, Behrouz, B, Keeling, Bh, Bacon, Ja, Hentati, E, Williams, L, Yanagiya, A, Sonenberg, N, Lockhart, Pj, Zubair, Ac, Uitti, Rj, Aasly, Jo, Krygowska Wajs, A, Opala, G, Wszolek, Zk, Frigerio, R, Maraganore, Dm, Gosal, D, Lynch, T, Hutchinson, M, Bentivoglio, Anna Rita, Valente, Enza Maria, Nichols, Wc, Pankratz, N, Foroud, T, Gibson, Ra, Hentati, F, Dickson, Dw, Destée, A, Farrer, Mj, Bentivoglio, Anna Rita (ORCID:0000-0002-9663-095X), Chartier Harlin, M, Dachsel, Jc, Vilariño Güell, C, Lincoln, Sj, Leprêtre, F, Hulihan, Mm, Kachergus, J, Milnerwood, Aj, Tapia, L, Song, M, Le Rhun, E, Mutez, E, Larvor, L, Duflot, A, Vanbesien Mailliot, C, Kreisler, A, Ross, Oa, Nishioka, K, Soto Ortolaza, Ai, Cobb, Sa, Melrose, Hl, Behrouz, B, Keeling, Bh, Bacon, Ja, Hentati, E, Williams, L, Yanagiya, A, Sonenberg, N, Lockhart, Pj, Zubair, Ac, Uitti, Rj, Aasly, Jo, Krygowska Wajs, A, Opala, G, Wszolek, Zk, Frigerio, R, Maraganore, Dm, Gosal, D, Lynch, T, Hutchinson, M, Bentivoglio, Anna Rita, Valente, Enza Maria, Nichols, Wc, Pankratz, N, Foroud, T, Gibson, Ra, Hentati, F, Dickson, Dw, Destée, A, Farrer, Mj, and Bentivoglio, Anna Rita (ORCID:0000-0002-9663-095X)
- Abstract
Genome-wide analysis of a multi-incident family with autosomal-dominant parkinsonism has implicated a locus on chromosomal region 3q26-q28. Linkage and disease segregation is explained by a missense mutation c.3614G>A (p.Arg1205His) in eukaryotic translation initiation factor 4-gamma (EIF4G1). Subsequent sequence and genotype analysis identified EIF4G1 c.1505C>T (p.Ala502Val), c.2056G>T (p.Gly686Cys), c.3490A>C (p.Ser1164Arg), c.3589C>T (p.Arg1197Trp) and c.3614G>A (p.Arg1205His) substitutions in affected subjects with familial parkinsonism and idiopathic Lewy body disease but not in control subjects. Despite different countries of origin, persons with EIF4G1 c.1505C>T (p.Ala502Val) or c.3614G>A (p.Arg1205His) mutations appear to share haplotypes consistent with ancestral founders. eIF4G1 p.Ala502Val and p.Arg1205His disrupt eIF4E or eIF3e binding, although the wild-type protein does not, and render mutant cells more vulnerable to reactive oxidative species. EIF4G1 mutations implicate mRNA translation initiation in familial parkinsonism and highlight a convergent pathway for monogenic, toxin and perhaps virally-induced Parkinson disease.
- Published
- 2011
3. Akt and AMPK activators rescue hyperexcitability in neurons from patients with bipolar disorder.
- Author
-
Khayachi A, Abuzgaya M, Liu Y, Jiao C, Dejgaard K, Schorova L, Kamesh A, He Q, Cousineau Y, Pietrantonio A, Farhangdoost N, Castonguay CE, Chaumette B, Alda M, Rouleau GA, and Milnerwood AJ
- Subjects
- Humans, Lithium pharmacology, Lithium therapeutic use, Signal Transduction, Gene Expression Profiling, Transcriptome, Bipolar Disorder metabolism, Bipolar Disorder drug therapy, Neurons metabolism, AMP-Activated Protein Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Induced Pluripotent Stem Cells metabolism, Induced Pluripotent Stem Cells cytology
- Abstract
Background: Bipolar disorder (BD) is a multifactorial psychiatric illness affecting ∼1% of the global adult population. Lithium (Li), is the most effective mood stabilizer for BD but works only for a subset of patients and its mechanism of action remains largely elusive., Methods: In the present study, we used iPSC-derived neurons from patients with BD who are responsive (LR) or not (LNR) to lithium. Combined electrophysiology, calcium imaging, biochemistry, transcriptomics, and phosphoproteomics were employed to provide mechanistic insights into neuronal hyperactivity in BD, investigate Li's mode of action, and identify alternative treatment strategies., Findings: We show a selective rescue of the neuronal hyperactivity phenotype by Li in LR neurons, correlated with changes to Na
+ conductance. Whole transcriptome sequencing in BD neurons revealed altered gene expression pathways related to glutamate transmission, alterations in cell signalling and ion transport/channel activity. We found altered Akt signalling as a potential therapeutic effect of Li in LR neurons from patients with BD, and that Akt activation mimics Li effect in LR neurons. Furthermore, the increased neural network activity observed in both LR & LNR neurons from patients with BD were reversed by AMP-activated protein kinase (AMPK) activation., Interpretation: These results suggest potential for new treatment strategies in BD, such as Akt activators in LR cases, and the use of AMPK activators for LNR patients with BD., Funding: Supported by funding from ERA PerMed, Bell Brain Canada Mental Research Program and Brain & Behavior Research Foundation., Competing Interests: Declaration of interests All the authors declare no conflict of interest., (Copyright © 2024 The Author(s). Published by Elsevier B.V. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
4. Maternal gastrointestinal nematode infection alters hippocampal neuroimmunity, promotes synaptic plasticity, and improves resistance to direct infection in offspring.
- Author
-
Noel SC, Madranges JF, Gothié JM, Ewald J, Milnerwood AJ, Kennedy TE, and Scott ME
- Subjects
- Animals, Female, Pregnancy, Mice, Nematode Infections immunology, Nematode Infections parasitology, Long-Term Potentiation, Prenatal Exposure Delayed Effects immunology, Strongylida Infections immunology, Strongylida Infections parasitology, Male, Neuroimmunomodulation, Hippocampus metabolism, Hippocampus parasitology, Neuronal Plasticity
- Abstract
The developing brain is vulnerable to maternal bacterial and viral infections which induce strong inflammatory responses in the mother that are mimicked in the offspring brain, resulting in irreversible neurodevelopmental defects, and associated cognitive and behavioural impairments. In contrast, infection during pregnancy and lactation with the immunoregulatory murine intestinal nematode, Heligmosomoides bakeri, upregulates expression of genes associated with long-term potentiation (LTP) of synaptic networks in the brain of neonatal uninfected offspring, and enhances spatial memory in uninfected juvenile offspring. As the hippocampus is involved in spatial navigation and sensitive to immune events during development, here we assessed hippocampal gene expression, LTP, and neuroimmunity in 3-week-old uninfected offspring born to H. bakeri infected mothers. Further, as maternal immunity shapes the developing immune system, we assessed the impact of maternal H. bakeri infection on the ability of offspring to resist direct infection. In response to maternal infection, we found an enhanced propensity to induce LTP at Schaffer collateral synapses, consistent with RNA-seq data indicating accelerated development of glutamatergic synapses in uninfected offspring, relative to those from uninfected mothers. Hippocampal RNA-seq analysis of offspring of infected mothers revealed increased expression of genes associated with neurogenesis, gliogenesis, and myelination. Furthermore, maternal infection improved resistance to direct infection of H. bakeri in offspring, correlated with transfer of parasite-specific IgG1 to their serum. Hippocampal immunohistochemistry and gene expression suggest Th2/Treg biased neuroimmunity in offspring, recapitulating peripheral immunoregulation of H. bakeri infected mothers. These findings indicate maternal H. bakeri infection during pregnancy and lactation alters peripheral and neural immunity in uninfected offspring, in a manner that accelerates neural maturation to promote hippocampal LTP, and upregulates the expression of genes associated with neurogenesis, gliogenesis, and myelination., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
5. Dendritic Polyglycerol Amine: An Enhanced Substrate to Support Long-Term Neural Cell Culture.
- Author
-
Clément JP, Al-Alwan L, Glasgow SD, Stolow A, Ding Y, Quevedo Melo T, Khayachi A, Liu Y, Hellmund M, Haag R, Milnerwood AJ, Grütter P, and Kennedy TE
- Subjects
- Cell Culture Techniques, Cell Differentiation, Glycerol, Humans, Neurons, Polymers, Amines, Induced Pluripotent Stem Cells
- Abstract
Long-term stable cell culture is a critical tool to better understand cell function. Most adherent cell culture models require a polymer substrate coating of poly-lysine or poly-ornithine for the cells to adhere and survive. However, polypeptide-based substrates are degraded by proteolysis and it remains a challenge to maintain healthy cell cultures for extended periods of time. Here, we report the development of an enhanced cell culture substrate based on a coating of dendritic polyglycerol amine (dPGA), a non-protein macromolecular biomimetic of poly-lysine, to promote the adhesion and survival of neurons in cell culture. We show that this new polymer coating provides enhanced survival, differentiation and long-term stability for cultures of primary neurons or neurons derived from human induced pluripotent stem cells (hiPSCs). Atomic force microscopy analysis provides evidence that greater nanoscale roughness contributes to the enhanced capacity of dPGA-coated surfaces to support cells in culture. We conclude that dPGA is a cytocompatible, functionally superior, easy to use, low cost and highly stable alternative to poly-cationic polymer cell culture substrate coatings such as poly-lysine and poly-ornithine. Summary statement Here, we describe a novel dendritic polyglycerol amine-based substrate coating, demonstrating superior performance compared to current polymer coatings for long-term culture of primary neurons and neurons derived from induced pluripotent stem cells.
- Published
- 2022
- Full Text
- View/download PDF
6. Endosomal traffic and glutamate synapse activity are increased in VPS35 D620N mutant knock-in mouse neurons, and resistant to LRRK2 kinase inhibition.
- Author
-
Kadgien CA, Kamesh A, and Milnerwood AJ
- Subjects
- Animals, Cells, Cultured, Dendrites metabolism, Gain of Function Mutation, Gene Knock-In Techniques, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 physiology, Mice, Mice, Inbred C57BL, Miniature Postsynaptic Potentials physiology, Nerve Tissue Proteins metabolism, Patch-Clamp Techniques, Protein Binding, Protein Interaction Mapping, Receptors, AMPA metabolism, Recombinant Proteins genetics, Recombinant Proteins metabolism, Synapses metabolism, Vesicular Transport Proteins physiology, rab GTP-Binding Proteins metabolism, Endosomes physiology, Glutamic Acid physiology, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 antagonists & inhibitors, Mutation, Missense, Parkinson Disease genetics, Point Mutation, Vesicular Transport Proteins genetics
- Abstract
Vacuolar protein sorting 35 (VPS35) regulates neurotransmitter receptor recycling from endosomes. A missense mutation (D620N) in VPS35 leads to autosomal-dominant, late-onset Parkinson's disease. Here, we study the basic neurobiology of VPS35 and Parkinson's disease mutation effects in the D620N knock-in mouse and the effect of leucine-rich repeat kinase 2 (LRRK2) inhibition on synaptic phenotypes. The study was conducted using a VPS35 D620N knock-in mouse that expresses VPS35 at endogenous levels. Protein levels, phosphorylation states, and binding ratios in brain lysates from knock-in mice and wild-type littermates were assayed by co-immunoprecipitation and western blot. Dendritic protein co-localization, AMPA receptor surface expression, synapse density, and glutamatergic synapse activity in primary cortical cultures from knock-in and wild-type littermates were assayed using immunocytochemistry and whole-cell patch clamp electrophysiology. In brain tissue, we confirm VPS35 forms complexes with LRRK2 and AMPA-type glutamate receptor GluA1 subunits, in addition to NMDA-type glutamate receptor GluN1 subunits and D2-type dopamine receptors. Receptor and LRRK2 binding was unaltered in D620N knock-in mice, but we confirm the mutation results in reduced binding of VPS35 with WASH complex member FAM21, and increases phosphorylation of the LRRK2 kinase substrate Rab10, which is reversed by LRRK2 kinase inhibition in vivo. In cultured cortical neurons from knock-in mice, pRab10 is also increased, and reversed by LRRK2 inhibition. The mutation also results in increased endosomal recycling protein cluster density (VPS35-FAM21 co-clusters and Rab11 clusters), glutamate transmission, and GluA1 surface expression. LRRK2 kinase inhibition, which reversed Rab10 hyper-phosphorylation, did not rescue elevated glutamate release or surface GluA1 expression in knock-in neurons, but did alter AMPAR traffic in wild-type cells. The results improve our understanding of the cell biology of VPS35, and the consequences of the D620N mutation in developing neuronal networks. Together the data support a chronic synaptopathy model for latent neurodegeneration, providing phenotypes and candidate pathophysiological stresses that may drive eventual transition to late-stage parkinsonism in VPS35 PD. The study demonstrates the VPS35 mutation has effects that are independent of ongoing LRRK2 kinase activity, and that LRRK2 kinase inhibition alters basal physiology of glutamate synapses in vitro., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
7. Posttranslational modifications & lithium's therapeutic effect-Potential biomarkers for clinical responses in psychiatric & neurodegenerative disorders.
- Author
-
Khayachi A, Schorova L, Alda M, Rouleau GA, and Milnerwood AJ
- Subjects
- Biomarkers, Humans, Lithium therapeutic use, Protein Processing, Post-Translational, Bipolar Disorder drug therapy, Neurodegenerative Diseases drug therapy
- Abstract
Several neurodegenerative diseases and neuropsychiatric disorders display aberrant posttranslational modifications (PTMs) of one, or many, proteins. Lithium treatment has been used for mood stabilization for many decades, and is highly effective for large subsets of patients with diverse neurological conditions. However, the differential effectiveness and mode of action are not fully understood. In recent years, studies have shown that lithium alters several protein PTMs, altering their function, and consequently neuronal physiology. The impetus for this review is to outline the links between lithium's therapeutic mode of action and PTM homeostasis. We first provide an overview of the principal PTMs affected by lithium. We then describe several neuropsychiatric disorders in which PTMs have been implicated as pathogenic. For each of these conditions, we discuss lithium's clinical use and explore the putative mechanism of how it restores PTM homeostasis, and thereby cellular physiology. Evidence suggests that determining specific PTM patterns could be a promising strategy to develop biomarkers for disease and lithium responsiveness., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
8. Chronic and Acute Manipulation of Cortical Glutamate Transmission Induces Structural and Synaptic Changes in Co-cultured Striatal Neurons.
- Author
-
Kuhlmann N, Wagner Valladolid M, Quesada-Ramírez L, Farrer MJ, and Milnerwood AJ
- Abstract
In contrast to the prenatal topographic development of sensory cortices, striatal circuit organization is slow and requires the functional maturation of cortical and thalamic excitatory inputs throughout the first postnatal month. While mechanisms regulating synapse development and plasticity are quite well described at excitatory synapses of glutamatergic neurons in the neocortex, comparatively little is known of how this translates to glutamate synapses onto GABAergic neurons in the striatum. Here we investigate excitatory striatal synapse plasticity in an in vitro system, where glutamate can be studied in isolation from dopamine and other neuromodulators. We examined pre-and post-synaptic structural and functional plasticity in GABAergic striatal spiny projection neurons (SPNs), co-cultured with glutamatergic cortical neurons. After synapse formation, medium-term (24 h) TTX silencing increased the density of filopodia, and modestly decreased dendritic spine density, when assayed at 21 days in vitro (DIV). Spine reductions appeared to require residual spontaneous activation of ionotropic glutamate receptors. Conversely, chronic (14 days) TTX silencing markedly reduced spine density without any observed increase in filopodia density. Time-dependent, biphasic changes to the presynaptic marker Synapsin-1 were also observed, independent of residual spontaneous activity. Acute silencing (3 h) did not affect presynaptic markers or postsynaptic structures. To induce rapid, activity-dependent plasticity in striatal neurons, a chemical NMDA receptor-dependent "long-term potentiation (LTP)" paradigm was employed. Within 30 min, this increased spine and GluA1 cluster densities, and the percentage of spines containing GluA1 clusters, without altering the presynaptic signal. The results demonstrate that the growth and pruning of dendritic protrusions is an active process, requiring glutamate receptor activity in striatal projection neurons. Furthermore, NMDA receptor activation is sufficient to drive glutamatergic structural plasticity in SPNs, in the absence of dopamine or other neuromodulators., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Kuhlmann, Wagner Valladolid, Quesada-Ramírez, Farrer and Milnerwood.)
- Published
- 2021
- Full Text
- View/download PDF
9. A Critical LRRK at the Synapse? The Neurobiological Function and Pathophysiological Dysfunction of LRRK2.
- Author
-
Kuhlmann N and Milnerwood AJ
- Abstract
Since the discovery of LRRK2 mutations causal to Parkinson's disease (PD) in the early 2000s, the LRRK2 protein has been implicated in a plethora of cellular processes in which pathogenesis could occur, yet its physiological function remains elusive. The development of genetic models of LRRK2 PD has helped identify the etiological and pathophysiological underpinnings of the disease, and may identify early points of intervention. An important role for LRRK2 in synaptic function has emerged in recent years, which links LRRK2 to other genetic forms of PD, most notably those caused by mutations in the synaptic protein α-synuclein. This point of convergence may provide useful clues as to what drives dysfunction in the basal ganglia circuitry and eventual death of substantia nigra (SN) neurons. Here, we discuss the evolution and current state of the literature placing LRRK2 at the synapse, through the lens of knock-out, overexpression, and knock-in animal models. We hope that a deeper understanding of LRRK2 neurobiology, at the synapse and beyond, will aid the eventual development of neuroprotective interventions for PD, and the advancement of useful treatments in the interim., (Copyright © 2020 Kuhlmann and Milnerwood.)
- Published
- 2020
- Full Text
- View/download PDF
10. Neuron-autonomous susceptibility to induced synuclein aggregation is exacerbated by endogenous Lrrk2 mutations and ameliorated by Lrrk2 genetic knock-out.
- Author
-
MacIsaac S, Quevedo Melo T, Zhang Y, Volta M, Farrer MJ, and Milnerwood AJ
- Abstract
Neuronal aggregates containing α-synuclein are a pathological hallmark of several degenerative diseases; including Parkinson's disease, Parkinson's disease with dementia and dementia with Lewy bodies. Understanding the process of α-synuclein aggregation, and discovering means of preventing it, may help guide therapeutic strategy and drug design. Recent advances provide tools to induce α-synuclein aggregation in neuronal cultures. Application of exogenous pre-formed fibrillar α-synuclein induces pathological phosphorylation and accumulation of endogenous α-synuclein, typical of that seen in disease. Genomic variability and mutations in α-synuclein and leucine-rich repeat kinase 2 proteins are the major genetic risk factors for Parkinson's disease. Reports demonstrate fibril-induced α-synuclein aggregation is increased in cells from leucine-rich repeat kinase 2 pathogenic mutant (G2019S) overexpressing mice, and variously decreased by leucine-rich repeat kinase 2 inhibitors. Elsewhere in vivo antisense knock-down of leucine-rich repeat kinase 2 protein has been shown to protect mice from fibril-induced α-synuclein aggregation, whereas kinase inhibition did not. To help bring clarity to this issue, we took a purely genetic approach in a standardized neuron-enriched culture, lacking glia. We compared fibril treatment of leucine-rich repeat kinase 2 germ-line knock-out, and G2019S germ-line knock-in, mouse cortical neuron cultures with those from littermates. We found leucine-rich repeat kinase 2 knock-out neurons are resistant to α-synuclein aggregation, which predominantly forms within axons, and may cause axonal fragmentation. Conversely, leucine-rich repeat kinase 2 knock-in neurons are more vulnerable to fibril-induced α-synuclein accumulation. Protection and resistance correlated with basal increases in a lysosome marker in knock-out, and an autophagy marker in knock-in cultures. The data add to a growing number of studies that argue leucine-rich repeat kinase 2 silencing, and potentially kinase inhibition, may be a useful therapeutic strategy against synucleinopathy., (© The Author(s) (2020). Published by Oxford University Press on behalf of the Guarantors of Brain.)
- Published
- 2020
- Full Text
- View/download PDF
11. Quantitative Profiling of Synuclein Species: Application to Transgenic Mouse Models of Parkinson's Disease.
- Author
-
Singh S, Khayachi A, Milnerwood AJ, and DeMarco ML
- Subjects
- Animals, Chromatography, Liquid, Disease Models, Animal, Mice, Transgenic, Peptide Hydrolases, Proof of Concept Study, Species Specificity, Tandem Mass Spectrometry, Biological Assay methods, Brain metabolism, Parkinson Disease metabolism, alpha-Synuclein metabolism, beta-Synuclein metabolism
- Abstract
Introduction: Improved analytical tools for detailed characterization of synucleins in pre-clinical models of Parkinson's disease (PD) and related synucleinopathies are needed., Objective: Develop a multiple reaction monitoring (MRM) liquid chromatography tandem mass spectrometry (LC-MS/MS) assay to quantify species-specific sequences and structural heterogeneity in soluble α- and β-synucleins in brain tissue., Methods: Using a proteolytic digestion workflow, the MRM LC-MS/MS method assayed six proteotypic peptides from the α-synuclein sequence; three unique to mouse or human α-synuclein and three conserved in α- and β-synuclein. For quantification, we used labeled α-synuclein as the internal standard and an external calibration curve. As proof of concept, the synuclein LC-MS/MS method was applied to brain tissue specimens from M83 transgenic PD mice, which overexpresses human α-synuclein, relative to wild-type littermate controls., Results: The synuclein MRM assay was linear over a wide concentration range (at least one order of magnitude). The assay had several advantages over ligand binding analytical methods, such as western blotting and enzyme-linked immunosorbent assays. These advantages included the ability to: quantify 1) total α-synuclein, 2) combined α- and β-synucleins, 3) species-specific contributions to total α-synuclein (e.g., in mice expressing both mouse and human α-synuclein), and 4) identify peptide-specific profile differences that may reflect post-translational modifications, all within a single analysis., Conclusion: With improved and expanded analytical characteristics coupled with a streamlined sample preparation workflow, the quantitative synuclein profiling LC-MS/MS assay provides a versatile and efficient platform to characterize synuclein biology in pre-clinical models and the potential for application to human tissues and fluids.
- Published
- 2020
- Full Text
- View/download PDF
12. The X-Linked Intellectual Disability Gene Zdhhc9 Is Essential for Dendrite Outgrowth and Inhibitory Synapse Formation.
- Author
-
Shimell JJ, Shah BS, Cain SM, Thouta S, Kuhlmann N, Tatarnikov I, Jovellar DB, Brigidi GS, Kass J, Milnerwood AJ, Snutch TP, and Bamji SX
- Subjects
- Acyltransferases genetics, Animals, Cells, Cultured, Epilepsy genetics, Epilepsy metabolism, Genes, X-Linked genetics, Hippocampus metabolism, Humans, Intellectual Disability genetics, Lipoylation genetics, Lipoylation physiology, Mice, Mice, Knockout, Synapses genetics, ras Proteins metabolism, rho GTP-Binding Proteins genetics, rho GTP-Binding Proteins metabolism, Acyltransferases metabolism, Dendrites metabolism, Genes, X-Linked physiology, Intellectual Disability metabolism, Synapses metabolism
- Abstract
Palmitoylation is a reversible post-translational lipid modification that facilitates vesicular transport and subcellular localization of modified proteins. This process is catalyzed by ZDHHC enzymes that are implicated in several neurological and neurodevelopmental disorders. Loss-of-function mutations in ZDHHC9 have been identified in patients with X-linked intellectual disability (XLID) and associated with increased epilepsy risk. Loss of Zdhhc9 function in hippocampal cultures leads to shorter dendritic arbors and fewer inhibitory synapses, altering the ratio of excitatory-to-inhibitory inputs formed onto Zdhhc9-deficient cells. While Zdhhc9 promotes dendrite outgrowth through the palmitoylation of the GTPase Ras, it promotes inhibitory synapse formation through the palmitoylation of another GTPase, TC10. Zdhhc9 knockout mice exhibit seizure-like activity together with increased frequency and amplitude of both spontaneous and miniature excitatory and inhibitory postsynaptic currents. These findings present a plausible mechanism for how the loss of ZDHHC9 function may contribute to XLID and epilepsy., (Crown Copyright © 2019. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
13. DNAJC13 p.Asn855Ser, implicated in familial parkinsonism, alters membrane dynamics of sorting nexin 1.
- Author
-
Follett J, Fox JD, Gustavsson EK, Kadgien C, Munsie LN, Cao LP, Tatarnikov I, Milnerwood AJ, and Farrer MJ
- Subjects
- Alleles, Animals, Cells, Cultured, Endosomes metabolism, Mice, Mice, Transgenic, Molecular Chaperones metabolism, Neurons metabolism, Parkinsonian Disorders metabolism, Protein Transport, Sorting Nexins metabolism, Vesicular Transport Proteins metabolism, Cell Membrane metabolism, Molecular Chaperones genetics, Parkinsonian Disorders genetics, Sorting Nexins genetics, Vesicular Transport Proteins genetics
- Abstract
DNAJC13 (RME-8) is a core co-chaperone that facilitates membrane recycling and cargo sorting of endocytosed proteins. DNAJ/Hsp40 (heat shock protein 40) proteins are highly conserved throughout evolution and mediate the folding of nascent proteins, and the unfolding, refolding or degradation of misfolded proteins while assisting in associated-membrane translocation. DNAJC13 is one of five DNAJ 'C' class chaperone variants implicated in monogenic parkinsonism. Here we examine the effect of the DNAJC13 disease-linked mutation (p.Asn855Ser) on its interacting partners, focusing on sorting nexin 1 (SNX1) membrane dynamics in primary cortical neurons derived from a novel Dnajc13 p.Asn855Ser knock-in (DKI) mouse model. Dnajc13 p.Asn855Ser mutant and wild type protein expression were equivalent in mature heterozygous cultures (DIV21). While SNX1-positive puncta density, area, and WASH-retromer assembly were comparable between cultures derived from DKI and wild type littermates, the formation of SNX1-enriched tubules in DKI neuronal cultures was significantly increased. Thus, Dnajc13 p.Asn855Ser disrupts SNX1 membrane-tubulation and trafficking, analogous to results from RME-8 depletion studies. The data suggest the mutation confers a dominant-negative gain-of-function in RME-8. Implications for the pathogenesis of Parkinson's disease are discussed., (Copyright © 2019 Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
14. Altered dopamine release and monoamine transporters in Vps35 p.D620N knock-in mice.
- Author
-
Cataldi S, Follett J, Fox JD, Tatarnikov I, Kadgien C, Gustavsson EK, Khinda J, Milnerwood AJ, and Farrer MJ
- Abstract
Vacuolar protein sorting 35 (VPS35) is a core component of the retromer trimer required for endosomal membrane-associated protein trafficking. The discovery of a missense mutation, Vps35 p.D620N implicates retromer dysfunction in the pathogenesis of Parkinson's disease (PD). We have characterized a knock-in mouse with a Vps35 p.D620N substitution (hereafter referred to as VKI) at 3 months of age. Standardized behavioral testing did not observe overt movement disorder. Tyrosine hydroxylase (TH)-positive nigral neuron counts and terminal expression in striata were comparable across genotypes. Fast scan cyclic voltammetry revealed increased dopamine release in VKI striatal slices. While extracellular dopamine collected via striatal microdialysis of freely moving animals was comparable across genotypes, the ratio of dopamine metabolites to dopamine suggests increased dopamine turnover in VKI homozygous mice. Western blot of striatal proteins revealed a genotype-dependent decrease in dopamine transporter (DAT) along with an increase in vesicular monoamine transporter 2 (VMAT2), albeit independent of changes in other synaptic markers. The reduction in DAT was further supported by immunohistochemical analysis. The data show that the dopaminergic system of VKI mice is profoundly altered relative to wild-type littermates. We conclude early synaptic dysfunction contributes to age-related pathophysiology in the nigrostriatal system that may lead to parkinsonism in man., Competing Interests: The authors declare no competing interests.
- Published
- 2018
- Full Text
- View/download PDF
15. Initial elevations in glutamate and dopamine neurotransmission decline with age, as does exploratory behavior, in LRRK2 G2019S knock-in mice.
- Author
-
Volta M, Beccano-Kelly DA, Paschall SA, Cataldi S, MacIsaac SE, Kuhlmann N, Kadgien CA, Tatarnikov I, Fox J, Khinda J, Mitchell E, Bergeron S, Melrose H, Farrer MJ, and Milnerwood AJ
- Subjects
- Animals, Gene Knock-In Techniques, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 genetics, Mutant Proteins genetics, Mutant Proteins metabolism, Aging, Dopamine metabolism, Exploratory Behavior, Glutamic Acid metabolism, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 metabolism, Neurons metabolism, Synaptic Transmission
- Abstract
LRRK2 mutations produce end-stage Parkinson's disease (PD) with reduced nigrostriatal dopamine, whereas, asymptomatic carriers have increased dopamine turnover and altered brain connectivity. LRRK2 pathophysiology remains unclear, but reduced dopamine and mitochondrial abnormalities occur in aged G2019S mutant knock-in (GKI) mice. Conversely, cultured GKI neurons exhibit increased synaptic transmission. We assessed behavior and synaptic glutamate and dopamine function across a range of ages. Young GKI mice exhibit more vertical exploration, elevated glutamate and dopamine transmission, and aberrant D2-receptor responses. These phenomena decline with age, but are stable in littermates. In young GKI mice, dopamine transients are slower, independent of dopamine transporter (DAT), increasing the lifetime of extracellular dopamine. Slowing of dopamine transients is observed with age in littermates, suggesting premature ageing of dopamine synapses in GKI mice. Thus, GKI mice exhibit early, but declining, synaptic and behavioral phenotypes, making them amenable to investigation of early pathophysiological, and later parkinsonian-like, alterations. This model will prove valuable in efforts to develop neuroprotection for PD.
- Published
- 2017
- Full Text
- View/download PDF
16. G2019S-LRRK2 Expression Augments α-Synuclein Sequestration into Inclusions in Neurons.
- Author
-
Volpicelli-Daley LA, Abdelmotilib H, Liu Z, Stoyka L, Daher JP, Milnerwood AJ, Unni VK, Hirst WD, Yue Z, Zhao HT, Fraser K, Kennedy RE, and West AB
- Subjects
- Animals, Gene Expression Regulation genetics, Humans, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Oligoribonucleotides, Antisense pharmacology, Photobleaching, Rats, Synucleins metabolism, Transcytosis genetics, Tubulin metabolism, Voltage-Dependent Anion Channels genetics, Voltage-Dependent Anion Channels metabolism, Inclusion Bodies pathology, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 genetics, Mutation genetics, Neurons metabolism, Transcytosis physiology, alpha-Synuclein metabolism
- Abstract
Unlabelled: Pathologic inclusions define α-synucleinopathies that include Parkinson's disease (PD). The most common genetic cause of PD is the G2019S LRRK2 mutation that upregulates LRRK2 kinase activity. However, the interaction between α-synuclein, LRRK2, and the formation of α-synuclein inclusions remains unclear. Here, we show that G2019S-LRRK2 expression, in both cultured neurons and dopaminergic neurons in the rat substantia nigra pars compact, increases the recruitment of endogenous α-synuclein into inclusions in response to α-synuclein fibril exposure. This results from the expression of mutant G2019S-LRRK2, as overexpression of WT-LRRK2 not only does not increase formation of inclusions but reduces their abundance. In addition, treatment of primary mouse neurons with LRRK2 kinase inhibitors, PF-06447475 and MLi-2, blocks G2019S-LRRK2 effects, suggesting that the G2019S-LRRK2 potentiation of inclusion formation depends on its kinase activity. Overexpression of G2019S-LRRK2 slightly increases, whereas WT-LRRK2 decreases, total levels of α-synuclein. Knockdown of total α-synuclein with potent antisense oligonucleotides substantially reduces inclusion formation in G2019S-LRRK2-expressing neurons, suggesting that LRRK2 influences α-synuclein inclusion formation by altering α-synuclein levels. These findings support the hypothesis that G2019S-LRRK2 may increase the progression of pathological α-synuclein inclusions after the initial formation of α-synuclein pathology by increasing a pool of α-synuclein that is more susceptible to forming inclusions., Significance Statement: α-Synuclein inclusions are found in the brains of patients with many different neurodegenerative diseases. Point mutation, duplication, or triplication of the α-synuclein gene can all cause Parkinson's disease (PD). The G2019S mutation in LRRK2 is the most common known genetic cause of PD. The interaction between G2019S-LRRK2 and α-synuclein may uncover new mechanisms and targets for neuroprotection. Here, we show that expression of G2019S-LRRK2 increases α-synuclein mobility and enhances aggregation of α-synuclein in primary cultured neurons and in dopaminergic neurons of the substantia nigra pars compacta, a susceptible brain region in PD. Potent LRRK2 kinase inhibitors, which are being developed for clinical use, block the increased α-synuclein aggregation in G2019S-LRRK2-expressing neurons. These results demonstrate that α-synuclein inclusion formation in neurons can be blocked and that novel therapeutic compounds targeting this process by inhibiting LRRK2 kinase activity may slow progression of PD-associated pathology., (Copyright © 2016 the authors 0270-6474/16/367416-13$15.00/0.)
- Published
- 2016
- Full Text
- View/download PDF
17. Changes in Dopamine Signalling Do Not Underlie Aberrant Hippocampal Plasticity in a Mouse Model of Huntington's Disease.
- Author
-
Dallérac GM, Cummings DM, Hirst MC, Milnerwood AJ, and Murphy KP
- Subjects
- Animals, Disease Models, Animal, Female, Gene Expression Regulation, Humans, Huntingtin Protein genetics, Huntington Disease metabolism, Long-Term Synaptic Depression, Male, Mice, Mice, Transgenic, Receptors, Dopamine D1 metabolism, Receptors, Dopamine D2 metabolism, Synaptic Transmission, Dopamine physiology, Hippocampus physiopathology, Huntington Disease physiopathology, Neuronal Plasticity
- Abstract
Altered dopamine receptor labelling has been demonstrated in presymptomatic and symptomatic Huntington's disease (HD) gene carriers, indicating that alterations in dopaminergic signalling are an early event in HD. We have previously described early alterations in synaptic transmission and plasticity in both the cortex and hippocampus of the R6/1 mouse model of Huntington's disease. Deficits in cortical synaptic plasticity were associated with altered dopaminergic signalling and could be reversed by D1- or D2-like dopamine receptor activation. In light of these findings we here investigated whether defects in dopamine signalling could also contribute to the marked alteration in hippocampal synaptic function. To this end we performed dopamine receptor labelling and pharmacology in the R6/1 hippocampus and report a marked, age-dependent elevation of hippocampal D1 and D2 receptor labelling in R6/1 hippocampal subfields. Yet, pharmacological inhibition or activation of D1- or D2-like receptors did not modify the aberrant synaptic plasticity observed in R6/1 mice. These findings demonstrate that global perturbations to dopamine receptor expression do occur in HD transgenic mice, similarly in HD gene carriers and patients. However, the direction of change and the lack of effect of dopaminergic pharmacological agents on synaptic function demonstrate that the perturbations are heterogeneous and region-specific, a finding that may explain the mixed results of dopamine therapy in HD.
- Published
- 2016
- Full Text
- View/download PDF
18. Insights from late-onset familial parkinsonism on the pathogenesis of idiopathic Parkinson's disease.
- Author
-
Volta M, Milnerwood AJ, and Farrer MJ
- Subjects
- Animals, Humans, Parkinson Disease genetics, Parkinson Disease metabolism, Parkinson Disease therapy
- Abstract
Disease-modifying therapies that slow or halt the progression of Parkinson's disease are an unmet clinical need. Many hypotheses have been put forward to explain the pathogenesis of the disease, but none has led to the development of disease-modifying drugs. Here we focus on familial forms of late-onset parkinsonism that most closely resemble idiopathic Parkinson's disease and present a synthesis of emerging molecular advances. Genetic discoveries and mechanistic investigations have highlighted early alterations to synaptic function, endosomal maturation, and protein sorting that might lead to an intracellular proteinopathy. We propose that these cellular processes constitute one pathway to pathogenesis and suggest that neuroprotection, as an adjunct to current symptomatic treatments, need not remain an elusive goal., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
19. Chronic and acute LRRK2 silencing has no long-term behavioral effects, whereas wild-type and mutant LRRK2 overexpression induce motor and cognitive deficits and altered regulation of dopamine release.
- Author
-
Volta M, Cataldi S, Beccano-Kelly D, Munsie L, Tatarnikov I, Chou P, Bergeron S, Mitchell E, Lim R, Khinda J, Lloret A, Bennett CF, Paradiso C, Morari M, Farrer MJ, and Milnerwood AJ
- Subjects
- Animals, Blotting, Western, Chromosomes, Artificial, Bacterial, Corpus Striatum metabolism, Disease Models, Animal, Humans, Immunohistochemistry, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Parkinson Disease metabolism, Cognition Disorders genetics, Dopamine metabolism, Motor Activity physiology, Parkinson Disease genetics, Protein Serine-Threonine Kinases genetics
- Abstract
Introduction: Germline silencing of the PD-related protein LRRK2 does not alter glutamate or dopamine release in adult mice, but some exploratory abnormalities have been reported with ageing. Contrastingly, high levels of human LRRK2 cause locomotor alterations and cognitive deficits accompanied by reduced striatal dopamine levels, with the latter also observed in G2019S mutant mice. Comparative cognitive and motor behavioral testing of LRRK2 KO, overexpressor and mutant overexpressor mice has not previously been reported., Methods: Parallel, comparative behavioral characterization was performed assessing motor and cognitive abilities. Striatal antisense oligonucleotide injections were conducted to investigate the effects of acute LRRK2 silencing on behavior and dopamine fiber density. Striatal synaptosomes prepared from hG2019S mice assessed vesicular release of dopamine and its sensitivity to D2 autoreceptor stimulation., Results: Genetic ablation of LRRK2 has no long-term consequences on motor or cognitive function. Consistently, no effects on behavior or dopaminergic fiber density were observed following acute striatal silencing. Conversely, 12-month OE mice show persistent locomotor deficits and worsening of cognitive abilities; whereas, hG2019S mice display early hyperactivity and effective learning and memory that progress to decreased motor and cognitive deficits at older ages. The G2019S mutation does not affect vesicular dopamine release, but decreases its sensitivity to D2-mediated inhibition., Conclusion: LRRK2 silencing is well tolerated in mouse, arguing PD does not result from LRRK2 loss of function. High levels of WT and G2019S LRRK2 produce similar but temporally distinct phenotypes, potentially modeling different stages of disease progression. The data implicate gain of LRRK2 function in the pathogenesis of PD., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
20. Progressive dopaminergic alterations and mitochondrial abnormalities in LRRK2 G2019S knock-in mice.
- Author
-
Yue M, Hinkle KM, Davies P, Trushina E, Fiesel FC, Christenson TA, Schroeder AS, Zhang L, Bowles E, Behrouz B, Lincoln SJ, Beevers JE, Milnerwood AJ, Kurti A, McLean PJ, Fryer JD, Springer W, Dickson DW, Farrer MJ, and Melrose HL
- Subjects
- Animals, Autophagy genetics, Brain metabolism, Brain ultrastructure, Dopaminergic Neurons metabolism, Female, Gene Knock-In Techniques, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Mitochondria ultrastructure, Motor Activity genetics, Rotarod Performance Test, tau Proteins metabolism, Brain enzymology, Dopamine metabolism, Mitochondria metabolism, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases physiology
- Abstract
Mutations in the LRRK2 gene represent the most common genetic cause of late onset Parkinson's disease. The physiological and pathological roles of LRRK2 are yet to be fully determined but evidence points towards LRRK2 mutations causing a gain in kinase function, impacting on neuronal maintenance, vesicular dynamics and neurotransmitter release. To explore the role of physiological levels of mutant LRRK2, we created knock-in (KI) mice harboring the most common LRRK2 mutation G2019S in their own genome. We have performed comprehensive dopaminergic, behavioral and neuropathological analyses in this model up to 24months of age. We find elevated kinase activity in the brain of both heterozygous and homozygous mice. Although normal at 6months, by 12months of age, basal and pharmacologically induced extracellular release of dopamine is impaired in both heterozygous and homozygous mice, corroborating previous findings in transgenic models over-expressing mutant LRRK2. Via in vivo microdialysis measurement of basal and drug-evoked extracellular release of dopamine and its metabolites, our findings indicate that exocytotic release from the vesicular pool is impaired. Furthermore, profound mitochondrial abnormalities are evident in the striatum of older homozygous G2019S KI mice, which are consistent with mitochondrial fission arrest. We anticipate that this G2019S mouse line will be a useful pre-clinical model for further evaluation of early mechanistic events in LRRK2 pathogenesis and for second-hit approaches to model disease progression., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
21. Retromer-dependent neurotransmitter receptor trafficking to synapses is altered by the Parkinson's disease VPS35 mutation p.D620N.
- Author
-
Munsie LN, Milnerwood AJ, Seibler P, Beccano-Kelly DA, Tatarnikov I, Khinda J, Volta M, Kadgien C, Cao LP, Tapia L, Klein C, and Farrer MJ
- Subjects
- Animals, Dendritic Spines metabolism, Humans, Mice, Protein Transport, Synapses metabolism, Mutation, Missense, Neurons metabolism, Parkinson Disease genetics, Receptors, Glutamate metabolism, Vesicular Transport Proteins genetics
- Abstract
Vacuolar protein sorting 35 (VPS35) is a core component of the retromer complex, crucial to endosomal protein sorting and intracellular trafficking. We recently linked a mutation in VPS35 (p.D620N) to familial parkinsonism. Here, we characterize human VPS35 and retromer function in mature murine neuronal cultures and investigate neuron-specific consequences of the p.D620N mutation. We find VPS35 localizes to dendritic spines and is involved in the trafficking of excitatory AMPA-type glutamate receptors (AMPARs). Fundamental neuronal processes, including excitatory synaptic transmission, AMPAR surface expression and synaptic recycling are altered by VPS35 overexpression. VPS35 p.D620N acts as a loss-of-function mutation with respect to VPS35 activity regulating synaptic transmission and AMPAR recycling in mouse cortical neurons and dopamine neuron-like cells produced from induced pluripotent stem cells of human p.D620N carriers. Such perturbations to synaptic function likely produce chronic pathophysiological stress upon neuronal circuits that may contribute to neurodegeneration in this, and other, forms of parkinsonism., (© The Author 2014. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
22. LRRK2 overexpression alters glutamatergic presynaptic plasticity, striatal dopamine tone, postsynaptic signal transduction, motor activity and memory.
- Author
-
Beccano-Kelly DA, Volta M, Munsie LN, Paschall SA, Tatarnikov I, Co K, Chou P, Cao LP, Bergeron S, Mitchell E, Han H, Melrose HL, Tapia L, Raymond LA, Farrer MJ, and Milnerwood AJ
- Subjects
- Animals, Glutamates, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2, Male, Mice, Mice, Transgenic, Neuronal Plasticity, Neurons metabolism, Parkinson Disease genetics, Phosphoproteins genetics, Phosphoproteins metabolism, Protein Serine-Threonine Kinases genetics, Receptors, Dopamine D2 genetics, Receptors, Dopamine D2 metabolism, Synaptic Transmission, Dopamine metabolism, Memory, Motor Activity, Neostriatum metabolism, Protein Serine-Threonine Kinases metabolism, Signal Transduction
- Abstract
Mutations in leucine-rich repeat kinase 2 (Lrrk2) are the most common genetic cause of Parkinson's disease (PD), a neurodegenerative disorder affecting 1-2% of those >65 years old. The neurophysiology of LRRK2 remains largely elusive, although protein loss suggests a role in glutamatergic synapse transmission and overexpression studies show altered dopamine release in aged mice. We show that glutamate transmission is unaltered onto striatal projection neurons (SPNs) of adult LRRK2 knockout mice and that adult animals exhibit no detectable cognitive or motor deficits. Basal synaptic transmission is also unaltered in SPNs of LRRK2 overexpressing mice, but they do exhibit clear alterations to D2-receptor-mediated short-term synaptic plasticity, behavioral hypoactivity and impaired recognition memory. These phenomena are associated with decreased striatal dopamine tone and abnormal dopamine- and cAMP-regulated phosphoprotein 32 kDa signal integration. The data suggest that LRRK2 acts at the nexus of dopamine and glutamate signaling in the adult striatum, where it regulates dopamine levels, presynaptic glutamate release via D2-dependent synaptic plasticity and dopamine-receptor signal transduction., (© The Author 2014. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
23. Dysfunctional Dopaminergic Neurones in Mouse Models of Huntington's Disease: A Role for SK3 Channels.
- Author
-
Dallérac GM, Levasseur G, Vatsavayai SC, Milnerwood AJ, Cummings DM, Kraev I, Huetz C, Evans KA, Walters SW, Rezaie P, Cho Y, Hirst MC, and Murphy KP
- Subjects
- Animals, Biophysical Phenomena genetics, Disease Models, Animal, Dopamine metabolism, Electric Stimulation, Female, Gene Expression Regulation genetics, Humans, Huntingtin Protein, Huntington Disease genetics, In Vitro Techniques, Male, Membrane Potentials genetics, Mice, Mice, Transgenic, Nerve Tissue Proteins genetics, Trinucleotide Repeat Expansion genetics, Tyrosine 3-Monooxygenase metabolism, Brain pathology, Dopaminergic Neurons physiology, Huntington Disease pathology, Small-Conductance Calcium-Activated Potassium Channels metabolism
- Abstract
Background: Huntington's disease (HD) is a late-onset fatal neurodegenerative disorder caused by a CAG trinucleotide repeat expansion in the gene coding for the protein huntingtin and is characterised by progressive motor, psychiatric and cognitive decline. We previously demonstrated that normal synaptic function in HD could be restored by application of dopamine receptor agonists, suggesting that changes in the release or bioavailability of dopamine may be a contributing factor to the disease process., Objective: In the present study, we examined the properties of midbrain dopaminergic neurones and dopamine release in presymptomatic and symptomatic transgenic HD mice., Methods and Results: Using intracellular sharp recordings and immunohistochemistry, we found that neuronal excitability was increased due to a loss of slow afterhyperpolarisation and that these changes were related to an apparent functional loss and abnormal distribution of SK3 channels (KCa2.3 encoded by the KCNN3 gene), a class of small-conductance calcium-activated potassium channels. Electrochemical detection of dopamine showed that this observation was associated with an enhanced dopamine release in presymptomatic transgenic mice and a drastic reduction in symptomatic animals. These changes occurred in the context of a progressive expansion in the CAG repeat number and nuclear localisation of mutant protein within the substantia nigra pars compacta., Conclusions: Dopaminergic neuronal dysfunction is a key early event in HD disease progression. The initial increase in dopamine release appears to be related to a loss of SK3 channel function, a protein containing a polyglutamine tract. Implications for polyglutamine-mediated sequestration of SK3 channels, dopamine-associated DNA damage and CAG expansion are discussed in the context of HD., (© 2015 S. Karger AG, Basel.)
- Published
- 2015
- Full Text
- View/download PDF
24. Synaptic function is modulated by LRRK2 and glutamate release is increased in cortical neurons of G2019S LRRK2 knock-in mice.
- Author
-
Beccano-Kelly DA, Kuhlmann N, Tatarnikov I, Volta M, Munsie LN, Chou P, Cao LP, Han H, Tapia L, Farrer MJ, and Milnerwood AJ
- Abstract
Mutations in Leucine-Rich Repeat Kinase-2 (LRRK2) result in familial Parkinson's disease and the G2019S mutation alone accounts for up to 30% in some ethnicities. Despite this, the function of LRRK2 is largely undetermined although evidence suggests roles in phosphorylation, protein interactions, autophagy and endocytosis. Emerging reports link loss of LRRK2 to altered synaptic transmission, but the effects of the G2019S mutation upon synaptic release in mammalian neurons are unknown. To assess wild type and mutant LRRK2 in established neuronal networks, we conducted immunocytochemical, electrophysiological and biochemical characterization of >3 week old cortical cultures of LRRK2 knock-out, wild-type overexpressing and G2019S knock-in mice. Synaptic release and synapse numbers were grossly normal in LRRK2 knock-out cells, but discretely reduced glutamatergic activity and reduced synaptic protein levels were observed. Conversely, synapse density was modestly but significantly increased in wild-type LRRK2 overexpressing cultures although event frequency was not. In knock-in cultures, glutamate release was markedly elevated, in the absence of any change to synapse density, indicating that physiological levels of G2019S LRRK2 elevate probability of release. Several pre-synaptic regulatory proteins shown by others to interact with LRRK2 were expressed at normal levels in knock-in cultures; however, synapsin 1 phosphorylation was significantly reduced. Thus, perturbations to the pre-synaptic release machinery and elevated synaptic transmission are early neuronal effects of LRRK2 G2019S. Furthermore, the comparison of knock-in and overexpressing cultures suggests that one copy of the G2019S mutation has a more pronounced effect than an ~3-fold increase in LRRK2 protein. Mutant-induced increases in transmission may convey additional stressors to neuronal physiology that may eventually contribute to the pathogenesis of Parkinson's disease.
- Published
- 2014
- Full Text
- View/download PDF
25. A microfluidic based in vitro model of synaptic competition.
- Author
-
Coquinco A, Kojic L, Wen W, Wang YT, Jeon NL, Milnerwood AJ, and Cynader M
- Subjects
- Action Potentials, Animals, Cells, Cultured, Cerebral Cortex cytology, Cerebral Cortex growth & development, Cerebral Cortex physiology, GABA-A Receptor Agonists pharmacology, Muscimol pharmacology, Neurogenesis, Neurons cytology, Neurons drug effects, Neurons physiology, Rats, Synaptic Potentials, Microfluidics, Models, Neurological, Neuronal Plasticity, Synapses physiology
- Abstract
Synaptic competition is widely believed to be central to the formation and function of neuronal networks, yet the underlying mechanisms are poorly described. To investigate synaptic competition in vitro, we have developed a novel two input pathway competition model using a 3-compartment microfluidic device. Axons from cultured rat cortical neurons from two different lateral compartments (inputs) innervate a common neuronal population in a separate central compartment. Inhibiting one input's activity, using the GABAAR agonist muscimol, resulted in increased synapse numbers and axon elongation of the opposing untreated (uninhibited) inputs in the central compartment. Time lapse imaging revealed that uninhibited inputs outgrew and outconnected their inhibited counterparts. This form of competition occurs during a sensitive period ending prior to 21 DIV and is NMDAR and CamKII dependent. Surprisingly, this form of plasticity was dependent on the age of the center compartment neurons but not of the competing inputs., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
26. DNAJC13 mutations in Parkinson disease.
- Author
-
Vilariño-Güell C, Rajput A, Milnerwood AJ, Shah B, Szu-Tu C, Trinh J, Yu I, Encarnacion M, Munsie LN, Tapia L, Gustavsson EK, Chou P, Tatarnikov I, Evans DM, Pishotta FT, Volta M, Beccano-Kelly D, Thompson C, Lin MK, Sherman HE, Han HJ, Guenther BL, Wasserman WW, Bernard V, Ross CJ, Appel-Cresswell S, Stoessl AJ, Robinson CA, Dickson DW, Ross OA, Wszolek ZK, Aasly JO, Wu RM, Hentati F, Gibson RA, McPherson PS, Girard M, Rajput M, Rajput AH, and Farrer MJ
- Subjects
- Adult, Age of Onset, Aged, Base Sequence, Case-Control Studies, Cells, Cultured, Endocytosis genetics, Endosomes genetics, Family, Female, Genetic Predisposition to Disease, Haplotypes, Humans, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2, Lewy Body Disease genetics, Male, Middle Aged, Molecular Chaperones immunology, Pedigree, Protein Serine-Threonine Kinases genetics, Sequence Alignment, Sequence Analysis, DNA, Vesicular Transport Proteins genetics, Lewy Bodies genetics, Molecular Chaperones genetics, Mutation genetics, Parkinson Disease genetics
- Abstract
A Saskatchewan multi-incident family was clinically characterized with Parkinson disease (PD) and Lewy body pathology. PD segregates as an autosomal-dominant trait, which could not be ascribed to any known mutation. DNA from three affected members was subjected to exome sequencing. Genome alignment, variant annotation and comparative analyses were used to identify shared coding mutations. Sanger sequencing was performed within the extended family and ethnically matched controls. Subsequent genotyping was performed in a multi-ethnic case-control series consisting of 2928 patients and 2676 control subjects from Canada, Norway, Taiwan, Tunisia, and the USA. A novel mutation in receptor-mediated endocytosis 8/RME-8 (DNAJC13 p.Asn855Ser) was found to segregate with disease. Screening of cases and controls identified four additional patients with the mutation, of which two had familial parkinsonism. All carriers shared an ancestral DNAJC13 p.Asn855Ser haplotype and claimed Dutch-German-Russian Mennonite heritage. DNAJC13 regulates the dynamics of clathrin coats on early endosomes. Cellular analysis shows that the mutation confers a toxic gain-of-function and impairs endosomal transport. DNAJC13 immunoreactivity was also noted within Lewy body inclusions. In late-onset disease which is most reminiscent of idiopathic PD subtle deficits in endosomal receptor-sorting/recycling are highlighted by the discovery of pathogenic mutations VPS35, LRRK2 and now DNAJC13. With this latest discovery, and from a neuronal perspective, a temporal and functional ecology is emerging that connects synaptic exo- and endocytosis, vesicular trafficking, endosomal recycling and the endo-lysosomal degradative pathway. Molecular deficits in these processes are genetically linked to the phenotypic spectrum of parkinsonism associated with Lewy body pathology.
- Published
- 2014
- Full Text
- View/download PDF
27. Palmitoylation of δ-catenin by DHHC5 mediates activity-induced synapse plasticity.
- Author
-
Brigidi GS, Sun Y, Beccano-Kelly D, Pitman K, Mobasser M, Borgland SL, Milnerwood AJ, and Bamji SX
- Subjects
- Acyltransferases, Animals, Catenins metabolism, Female, Hippocampus cytology, Hippocampus metabolism, Male, Membrane Proteins metabolism, Memory physiology, Mice, Mice, Inbred C57BL, Neurons cytology, Neurons metabolism, Neurons physiology, Rats, Rats, Sprague-Dawley, Synapses metabolism, Synaptic Membranes metabolism, Synaptic Membranes physiology, Delta Catenin, Catenins physiology, Lipoylation physiology, Neuronal Plasticity physiology, Synapses physiology
- Abstract
Synaptic cadherin adhesion complexes are known to be key regulators of synapse plasticity. However, the molecular mechanisms that coordinate activity-induced modifications in cadherin localization and adhesion and the subsequent changes in synapse morphology and efficacy remain unknown. We demonstrate that the intracellular cadherin binding protein δ-catenin is transiently palmitoylated by DHHC5 after enhanced synaptic activity and that palmitoylation increases δ-catenin-cadherin interactions at synapses. Both the palmitoylation of δ-catenin and its binding to cadherin are required for activity-induced stabilization of N-cadherin at synapses and the enlargement of postsynaptic spines, as well as the insertion of GluA1 and GluA2 subunits into the synaptic membrane and the concomitant increase in miniature excitatory postsynaptic current amplitude. Notably, context-dependent fear conditioning in mice resulted in increased δ-catenin palmitoylation, as well as increased δ-catenin-cadherin associations at hippocampal synapses. Together these findings suggest a role for palmitoylated δ-catenin in coordinating activity-dependent changes in synaptic adhesion molecules, synapse structure and receptor localization that are involved in memory formation.
- Published
- 2014
- Full Text
- View/download PDF
28. Behavioral deficits and striatal DA signaling in LRRK2 p.G2019S transgenic rats: a multimodal investigation including PET neuroimaging.
- Author
-
Walker MD, Volta M, Cataldi S, Dinelle K, Beccano-Kelly D, Munsie L, Kornelsen R, Mah C, Chou P, Co K, Khinda J, Mroczek M, Bergeron S, Yu K, Cao LP, Funk N, Ott T, Galter D, Riess O, Biskup S, Milnerwood AJ, Stoessl AJ, Farrer MJ, and Sossi V
- Subjects
- Animals, Brain diagnostic imaging, Brain metabolism, Dopamine Plasma Membrane Transport Proteins metabolism, Dopamine and cAMP-Regulated Phosphoprotein 32 metabolism, Humans, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2, Male, Neostriatum diagnostic imaging, Phosphorylation, Positron-Emission Tomography, Rats, Rats, Sprague-Dawley, Rats, Transgenic, Receptors, Dopamine D2 metabolism, Rotarod Performance Test, Tyrosine 3-Monooxygenase metabolism, Vesicular Monoamine Transport Proteins metabolism, Disease Models, Animal, Dopamine metabolism, Motor Activity genetics, Neostriatum metabolism, Parkinson Disease genetics, Protein Serine-Threonine Kinases genetics
- Abstract
Background: A major risk-factor for developing Parkinson's disease (PD) is genetic variability in leucine-rich repeat kinase 2 (LRRK2), most notably the p.G2019S mutation. Examination of the effects of this mutation is necessary to determine the etiology of PD and to guide therapeutic development., Objective: Assess the behavioral consequences of LRRK2 p.G2019S overexpression in transgenic rats as they age and test the functional integrity of the nigro-striatal dopamine system. Conduct positron emission tomography (PET) neuroimaging to compare transgenic rats with previous data from human LRRK2 mutation carriers., Methods: Rats overexpressing human LRRK2 p.G2019S were generated by BAC transgenesis and compared to non-transgenic (NT) littermates. Motor skill tests were performed at 3, 6 and 12 months-of-age. PET, performed at 12 months, assessed the density of dopamine and vesicular monoamine transporters (DAT and VMAT2, respectively) and measured dopamine synthesis, storage and availability. Brain tissue was assayed for D2, DAT, dopamine and cAMP-regulated phosphoprotein (DARPP32) and tyrosine hydroxylase (TH) expression by Western blot, and TH by immunohistochemistry., Results: Transgenic rats had no abnormalities in measures of striatal dopamine function at 12 months. A behavioral phenotype was present, with LRRK2 p.G2019S rats performing significantly worse on the rotarod than non-transgenic littermates (26% reduction in average running duration at 6 months), but with normal performance in other motor tests., Conclusions: Neuroimaging using dopaminergic PET did not recapitulate prior studies in human LRRK2 mutation carriers. Consistently, LRRK2 p.G2019S rats do not develop overt neurodegeneration; however, they do exhibit behavioral abnormalities.
- Published
- 2014
- Full Text
- View/download PDF
29. Memory and synaptic deficits in Hip14/DHHC17 knockout mice.
- Author
-
Milnerwood AJ, Parsons MP, Young FB, Singaraja RR, Franciosi S, Volta M, Bergeron S, Hayden MR, and Raymond LA
- Subjects
- Acyltransferases metabolism, Analysis of Variance, Animals, Cell Count, Dendrites ultrastructure, Hippocampus cytology, Hippocampus physiology, Lipoylation, Mice, Mice, Knockout, Neuronal Plasticity physiology, Patch-Clamp Techniques, Synapses physiology, Acyltransferases genetics, Long-Term Potentiation genetics, Long-Term Potentiation physiology, Memory Disorders genetics, Neuronal Plasticity genetics, Synapses genetics
- Abstract
Palmitoylation of neurotransmitter receptors and associated scaffold proteins regulates their membrane association in a rapid, reversible, and activity-dependent fashion. This makes palmitoylation an attractive candidate as a key regulator of the fast, reversible, and activity-dependent insertion of synaptic proteins required during the induction and expression of long-term plasticity. Here we describe that the constitutive loss of huntingtin interacting protein 14 (Hip14, also known as DHHC17), a single member of the broad palmitoyl acyltransferase (PAT) family, produces marked alterations in synaptic function in varied brain regions and significantly impairs hippocampal memory and synaptic plasticity. The data presented suggest that, even though the substrate pool is overlapping for the 23 known PAT family members, the function of a single PAT has marked effects upon physiology and cognition. Moreover, an improved understanding of the role of PATs in synaptic modification and maintenance highlights a potential strategy for intervention against early cognitive impairments in neurodegenerative disease.
- Published
- 2013
- Full Text
- View/download PDF
30. Mitigation of augmented extrasynaptic NMDAR signaling and apoptosis in cortico-striatal co-cultures from Huntington's disease mice.
- Author
-
Milnerwood AJ, Kaufman AM, Sepers MD, Gladding CM, Zhang L, Wang L, Fan J, Coquinco A, Qiao JY, Lee H, Wang YT, Cynader M, and Raymond LA
- Subjects
- Animals, Apoptosis drug effects, Cerebral Cortex drug effects, Cerebral Cortex pathology, Coculture Techniques, Corpus Striatum drug effects, Corpus Striatum pathology, Disease Models, Animal, Excitatory Amino Acid Antagonists pharmacology, Huntington Disease genetics, Huntington Disease pathology, Memantine pharmacology, Mice, Mice, Transgenic, Neurons drug effects, Neurons metabolism, Neurons pathology, Piperidines pharmacology, Receptors, N-Methyl-D-Aspartate antagonists & inhibitors, Signal Transduction drug effects, Apoptosis physiology, Cerebral Cortex metabolism, Corpus Striatum metabolism, Huntington Disease metabolism, Receptors, N-Methyl-D-Aspartate metabolism, Signal Transduction physiology
- Abstract
We recently reported evidence for disturbed synaptic versus extrasynaptic NMDAR transmission in the early pathogenesis of Huntington's disease (HD), a late-onset neurodegenerative disorder caused by CAG repeat expansion in the gene encoding huntingtin. Studies in glutamatergic cells indicate that synaptic NMDAR transmission increases phosphorylated cyclic-AMP response element binding protein (pCREB) levels and drives neuroprotective gene transcription, whereas extrasynaptic NMDAR activation reduces pCREB and promotes cell death. By generating striatal and cortical neuronal co-cultures to investigate the glutamatergic innervation of striatal neurons, we demonstrate that dichotomous synaptic and extrasynaptic NMDAR signaling also occurs in GABAergic striatal medium-sized spiny neurons (MSNs), which are acutely vulnerable in HD. Further, we show that wild-type (WT) and HD transgenic YAC128 MSNs co-cultured with cortical cells have similar levels of glutamatergic synapses, synaptic NMDAR currents and synaptic GluN2B and GluN2A subunit-containing NMDARs. However, NMDAR whole-cell, and especially extrasynaptic, current is elevated in YAC128 MSNs. Moreover, GluN2B subunit-containing NMDAR surface expression is markedly increased, irrespective of whether or not the co-cultured cortical cells express mutant huntingtin. The data suggest that MSN cell-autonomous increases in extrasynaptic NMDARs are driven by the HD mutation. Consistent with these results, we find that extrasynaptic NMDAR-induced pCREB reductions and apoptosis are also augmented in YAC128 MSNs. Moreover, both NMDAR-mediated apoptosis and CREB-off signaling are blocked by co-application of either memantine or the GluN2B subunit-selective antagonist ifenprodil in YAC128 MSNs. GluN2A-subunit-selective concentrations of the antagonist NVP-AAM077 did not reduce cell death in either genotype. Cortico-striatal co-cultures provide an in vitro model system in which to better investigate striatal neuronal dysfunction in disease than mono-cultured striatal cells. Results from the use of this system, which partially recapitulates the cortico-striatal circuit and is amenable to acute genetic and pharmacological manipulations, suggest that pathophysiological NMDAR signaling is an intrinsic frailty in HD MSNs that can be successfully targeted by pharmacological interventions., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
31. Calpain and STriatal-Enriched protein tyrosine phosphatase (STEP) activation contribute to extrasynaptic NMDA receptor localization in a Huntington's disease mouse model.
- Author
-
Gladding CM, Sepers MD, Xu J, Zhang LY, Milnerwood AJ, Lombroso PJ, and Raymond LA
- Subjects
- Animals, Calpain antagonists & inhibitors, Calpain genetics, Coculture Techniques, Disease Models, Animal, Enzyme Activation drug effects, Enzyme Inhibitors pharmacology, Huntington Disease pathology, Ion Channel Gating drug effects, Mice, Models, Biological, Neostriatum drug effects, Neostriatum enzymology, Neostriatum pathology, Neurons drug effects, Neurons enzymology, Phosphorylation drug effects, Phosphotyrosine metabolism, Protein Transport drug effects, Synapses drug effects, Calpain metabolism, Huntington Disease enzymology, Protein Tyrosine Phosphatases, Non-Receptor metabolism, Receptors, N-Methyl-D-Aspartate metabolism, Synapses enzymology
- Abstract
In Huntington's disease (HD), the mutant huntingtin (mhtt) protein is associated with striatal dysfunction and degeneration. Excitotoxicity and early synaptic defects are attributed, in part, to altered NMDA receptor (NMDAR) trafficking and function. Deleterious extrasynaptic NMDAR localization and signalling are increased early in yeast artificial chromosome mice expressing full-length mhtt with 128 polyglutamine repeats (YAC128 mice). NMDAR trafficking at the plasma membrane is regulated by dephosphorylation of the NMDAR subunit GluN2B tyrosine 1472 (Y1472) residue by STriatal-Enriched protein tyrosine Phosphatase (STEP). NMDAR function is also regulated by calpain cleavage of the GluN2B C-terminus. Activation of both STEP and calpain is calcium-dependent, and disruption of calcium homeostasis occurs early in the HD striatum. Here, we show increased calpain cleavage of GluN2B at both synaptic and extrasynaptic sites, and elevated extrasynaptic total GluN2B expression in the YAC128 striatum. Calpain inhibition significantly reduced extrasynaptic GluN2B expression in the YAC128 but not wild-type striatum. Furthermore, calpain inhibition reduced whole-cell NMDAR current and the surface/internal GluN2B ratio in co-cultured striatal neurons, without affecting synaptic GluN2B localization. Synaptic STEP activity was also significantly higher in the YAC128 striatum, correlating with decreased GluN2B Y1472 phosphorylation. A substrate-trapping STEP protein (TAT-STEP C-S) significantly increased VGLUT1-GluN2B colocalization, as well as increasing synaptic GluN2B expression and Y1472 phosphorylation. Moreover, combined calpain inhibition and STEP inactivation reduced extrasynaptic, while increasing synaptic GluN2B expression in the YAC128 striatum. These results indicate that increased STEP and calpain activation contribute to altered NMDAR localization in an HD mouse model, suggesting new therapeutic targets for HD.
- Published
- 2012
- Full Text
- View/download PDF
32. Opposing roles of synaptic and extrasynaptic NMDA receptor signaling in cocultured striatal and cortical neurons.
- Author
-
Kaufman AM, Milnerwood AJ, Sepers MD, Coquinco A, She K, Wang L, Lee H, Craig AM, Cynader M, and Raymond LA
- Subjects
- 4-Aminopyridine pharmacology, Analysis of Variance, Animals, Bicuculline pharmacology, CREB-Binding Protein metabolism, Calcium Channel Blockers pharmacology, Cells, Cultured, Coculture Techniques, Electric Stimulation, Embryo, Mammalian, Excitatory Amino Acid Agents pharmacology, Female, GABA-A Receptor Antagonists pharmacology, Glutamate Decarboxylase metabolism, Glycine Agents pharmacology, Luminescent Proteins genetics, Luminescent Proteins metabolism, Microfluidic Analytical Techniques methods, N-Methylaspartate pharmacology, Nerve Tissue Proteins metabolism, Neurons physiology, Nifedipine pharmacology, Patch-Clamp Techniques, Potassium Channel Blockers pharmacology, Pregnancy, Rats, Rats, Wistar, Sodium Channel Blockers pharmacology, Strychnine pharmacology, Tetrodotoxin pharmacology, Transfection methods, Vesicular Glutamate Transport Protein 1 metabolism, Cerebral Cortex cytology, Corpus Striatum cytology, Neurons cytology, Receptors, N-Methyl-D-Aspartate metabolism, Signal Transduction physiology, Synapses physiology
- Abstract
The NMDAR plays a unique and vital role in subcellular signaling. Calcium influx initiates signaling cascades important for both synaptic plasticity and survival; however, overactivation of the receptor leads to toxicity and cell death. This dichotomy is partially explained by the subcellular location of the receptor. NMDARs located at the synapse stimulate cell survival pathways, while extrasynaptic receptors signal for cell death. Thus far, this interplay between synaptic and extrasynaptic NMDARs has been studied exclusively in cortical (CTX) and hippocampal neurons. It was unknown whether other cell types, such as GABAergic medium-sized spiny projection neurons of the striatum (MSNs), which bear the brunt of neurodegeneration in Huntington's disease, follow the same pattern. Here we report synaptic versus extrasynaptic NMDAR signaling in striatal MSNs and resultant activation of cAMP response element binding protein (CREB), in rat primary corticostriatal cocultures. Similarly to CTX, we found in striatal MSNs that synaptic NMDARs activate CREB, whereas extrasynaptic NMDARs dominantly oppose CREB activation. However, MSNs are much less susceptible to NMDA-mediated toxicity than CTX cells and show differences in subcellular GluN2B distribution. Blocking NMDARs with memantine (30 μm) or GluN2B-containing receptors with ifenprodil (3 μm) prevents CREB shutoff effectively in CTX and MSNs, and also rescues both neuronal types from NMDA-mediated toxicity. This work may provide cell and NMDAR subtype-specific targets for treatment of diseases with putative NMDAR involvement, including neurodegenerative disorders and ischemia.
- Published
- 2012
- Full Text
- View/download PDF
33. P38 MAPK is involved in enhanced NMDA receptor-dependent excitotoxicity in YAC transgenic mouse model of Huntington disease.
- Author
-
Fan J, Gladding CM, Wang L, Zhang LY, Kaufman AM, Milnerwood AJ, and Raymond LA
- Subjects
- Analysis of Variance, Animals, Animals, Newborn, Apoptosis drug effects, Apoptosis genetics, Bacterial Proteins genetics, Cerebral Cortex cytology, Chromosomes, Artificial, Yeast genetics, Coculture Techniques, Disease Models, Animal, Disks Large Homolog 4 Protein, Embryo, Mammalian, Enzyme Inhibitors pharmacology, Gene Expression Regulation drug effects, Gene Expression Regulation genetics, Guanylate Kinases metabolism, Humans, Huntingtin Protein, Huntington Disease genetics, Immunoprecipitation methods, In Situ Nick-End Labeling, Luminescent Proteins genetics, Membrane Proteins metabolism, Mice, Mice, Transgenic, N-Methylaspartate pharmacology, Nerve Tissue Proteins genetics, Neurons drug effects, Nuclear Proteins genetics, Peptides genetics, Peptides pharmacology, Receptors, N-Methyl-D-Aspartate chemistry, Subcellular Fractions drug effects, Subcellular Fractions metabolism, Corpus Striatum pathology, Huntington Disease pathology, Neurons metabolism, Receptors, N-Methyl-D-Aspartate metabolism, p38 Mitogen-Activated Protein Kinases metabolism
- Abstract
Huntington disease (HD) is a dominantly inherited neurodegenerative disease caused by a polyglutamine (polyQ) expansion in the protein huntingtin (htt). Previous studies have shown enhanced N-methyl-d-aspartate (NMDA)-induced excitotoxicity in neuronal models of HD, mediated in part by increased NMDA receptor (NMDAR) GluN2B subunit binding with the postsynaptic density protein-95 (PSD-95). In cultured hippocampal neurons, the NMDAR-activated p38 Mitogen-activated Protein Kinase (MAPK) death pathway is disrupted by a peptide (Tat-NR2B9c) that uncouples GluN2B from PSD-95, whereas NMDAR-mediated activation of c-Jun N-terminal Kinase (JNK) MAPK is PSD-95-independent. To investigate the mechanism by which Tat-NR2B9c protects striatal medium spiny neurons (MSNs) from mutant htt (mhtt)-enhanced NMDAR toxicity, we compared striatal tissue and cultured MSNs from presymptomatic yeast artificial chromosome (YAC) mice expressing htt with 128 polyQ (YAC128) to those from YAC18 and/or WT mice as controls. Similar to the previously published shift of GluN2B-containing NMDARs to extrasynaptic sites, we found increased PSD-95 localization as well as elevated PSD-95-GluN2B interactions in the striatal non-PSD (extrasynaptic) fraction from YAC128 mice. Notably, basal levels of both activated p38 and JNK MAPKs were elevated in the YAC128 striatum. NMDA stimulation of acute slices increased activation of p38 and JNK in WT and YAC128 striatum, but Tat-NR2B9c pretreatment reduced only the p38 activation in YAC128. In cultured MSNs, p38 MAPK inhibition reduced YAC128 NMDAR-mediated cell death to WT levels, and occluded the Tat-NR2B9c peptide protective effect; in contrast, inhibition of JNK had a similar protective effect in cultured MSNs from both WT and YAC128 mice. Our results suggest that altered activation of p38 MAPK contributes to mhtt enhancement of GluN2B/PSD-95 toxic signaling., (Copyright © 2011. Published by Elsevier Inc.)
- Published
- 2012
- Full Text
- View/download PDF
34. Pathophysiology of Huntington's disease: time-dependent alterations in synaptic and receptor function.
- Author
-
Raymond LA, André VM, Cepeda C, Gladding CM, Milnerwood AJ, and Levine MS
- Subjects
- Animals, Disease Models, Animal, Humans, Mice, Time Factors, Corpus Striatum pathology, Huntington Disease pathology, Huntington Disease physiopathology, Receptors, N-Methyl-D-Aspartate metabolism, Synapses physiology
- Abstract
Huntington's disease (HD) is a progressive, fatal neurological condition caused by an expansion of CAG (glutamine) repeats in the coding region of the Huntington gene. To date, there is no cure but great strides have been made to understand pathophysiological mechanisms. In particular, genetic animal models of HD have been instrumental in elucidating the progression of behavioral and physiological alterations, which had not been possible using classic neurotoxin models. Our groups have pioneered the use of transgenic HD mice to examine the excitotoxicity hypothesis of striatal neuronal dysfunction and degeneration, as well as alterations in excitation and inhibition in striatum and cerebral cortex. In this review, we focus on synaptic and receptor alterations of striatal medium-sized spiny (MSNs) and cortical pyramidal neurons in genetic HD mouse models. We demonstrate a complex series of alterations that are region-specific and time-dependent. In particular, many changes are bidirectional depending on the degree of disease progression, that is, early vs. late, and also on the region examined. Early synaptic dysfunction is manifested by dysregulated glutamate release in striatum followed by progressive disconnection between cortex and striatum. The differential effects of altered glutamate release on MSNs originating the direct and indirect pathways is also elucidated, with the unexpected finding that cells of the direct striatal pathway are involved early in the course of the disease. In addition, we review evidence for early N-methyl-D-aspartate receptor (NMDAR) dysfunction leading to enhanced sensitivity of extrasynaptic receptors and a critical role of GluN2B subunits. Some of the alterations in late HD could be compensatory mechanisms designed to cope with early synaptic and receptor dysfunctions. The main findings indicate that HD treatments need to be designed according to the stage of disease progression and should consider regional differences., (Copyright © 2011 IBRO. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
35. Altered palmitoylation and neuropathological deficits in mice lacking HIP14.
- Author
-
Singaraja RR, Huang K, Sanders SS, Milnerwood AJ, Hines R, Lerch JP, Franciosi S, Drisdel RC, Vaid K, Young FB, Doty C, Wan J, Bissada N, Henkelman RM, Green WN, Davis NG, Raymond LA, and Hayden MR
- Subjects
- Acyltransferases genetics, Acyltransferases metabolism, Animals, Cell Death genetics, Corpus Striatum metabolism, Corpus Striatum pathology, Disease Models, Animal, Dopamine and cAMP-Regulated Phosphoprotein 32 metabolism, Enkephalins metabolism, Huntington Disease genetics, Huntington Disease pathology, Mice, Mice, Knockout, Motor Activity genetics, Mutant Proteins metabolism, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Neurons metabolism, Neurons pathology, Serotonin Plasma Membrane Transport Proteins genetics, Serotonin Plasma Membrane Transport Proteins metabolism, Synapses metabolism, Acyltransferases deficiency, Huntington Disease etiology, Lipoylation genetics, Nerve Tissue Proteins deficiency
- Abstract
Huntingtin interacting protein 14 (HIP14, ZDHHC17) is a huntingtin (HTT) interacting protein with palmitoyl transferase activity. In order to interrogate the function of Hip14, we generated mice with disruption in their Hip14 gene. Hip14-/- mice displayed behavioral, biochemical and neuropathological defects that are reminiscent of Huntington disease (HD). Palmitoylation of other HIP14 substrates, but not Htt, was reduced in the Hip14-/- mice. Hip14 is dysfunctional in the presence of mutant htt in the YAC128 mouse model of HD, suggesting that altered palmitoylation mediated by HIP14 may contribute to HD.
- Published
- 2011
- Full Text
- View/download PDF
36. Translation initiator EIF4G1 mutations in familial Parkinson disease.
- Author
-
Chartier-Harlin MC, Dachsel JC, Vilariño-Güell C, Lincoln SJ, Leprêtre F, Hulihan MM, Kachergus J, Milnerwood AJ, Tapia L, Song MS, Le Rhun E, Mutez E, Larvor L, Duflot A, Vanbesien-Mailliot C, Kreisler A, Ross OA, Nishioka K, Soto-Ortolaza AI, Cobb SA, Melrose HL, Behrouz B, Keeling BH, Bacon JA, Hentati E, Williams L, Yanagiya A, Sonenberg N, Lockhart PJ, Zubair AC, Uitti RJ, Aasly JO, Krygowska-Wajs A, Opala G, Wszolek ZK, Frigerio R, Maraganore DM, Gosal D, Lynch T, Hutchinson M, Bentivoglio AR, Valente EM, Nichols WC, Pankratz N, Foroud T, Gibson RA, Hentati F, Dickson DW, Destée A, and Farrer MJ
- Subjects
- Base Sequence, Cloning, Molecular, DNA Copy Number Variations, DNA Mutational Analysis, Flow Cytometry, Genetic Linkage, Genotype, Humans, Immunoprecipitation, Mitochondria physiology, Molecular Sequence Data, Mutation, Missense genetics, Pedigree, Chromosomes, Human, Pair 3 genetics, Eukaryotic Initiation Factor-4G genetics, Parkinson Disease genetics, Protein Biosynthesis genetics
- Abstract
Genome-wide analysis of a multi-incident family with autosomal-dominant parkinsonism has implicated a locus on chromosomal region 3q26-q28. Linkage and disease segregation is explained by a missense mutation c.3614G>A (p.Arg1205His) in eukaryotic translation initiation factor 4-gamma (EIF4G1). Subsequent sequence and genotype analysis identified EIF4G1 c.1505C>T (p.Ala502Val), c.2056G>T (p.Gly686Cys), c.3490A>C (p.Ser1164Arg), c.3589C>T (p.Arg1197Trp) and c.3614G>A (p.Arg1205His) substitutions in affected subjects with familial parkinsonism and idiopathic Lewy body disease but not in control subjects. Despite different countries of origin, persons with EIF4G1 c.1505C>T (p.Ala502Val) or c.3614G>A (p.Arg1205His) mutations appear to share haplotypes consistent with ancestral founders. eIF4G1 p.Ala502Val and p.Arg1205His disrupt eIF4E or eIF3e binding, although the wild-type protein does not, and render mutant cells more vulnerable to reactive oxidative species. EIF4G1 mutations implicate mRNA translation initiation in familial parkinsonism and highlight a convergent pathway for monogenic, toxin and perhaps virally-induced Parkinson disease., (Copyright © 2011 The American Society of Human Genetics. Published by Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
37. Impaired long-term potentiation in the prefrontal cortex of Huntington's disease mouse models: rescue by D1 dopamine receptor activation.
- Author
-
Dallérac GM, Vatsavayai SC, Cummings DM, Milnerwood AJ, Peddie CJ, Evans KA, Walters SW, Rezaie P, Hirst MC, and Murphy KP
- Subjects
- Animals, Disease Models, Animal, Dopamine Agonists pharmacology, Electrophysiology, Female, Immunohistochemistry, Long-Term Potentiation drug effects, Male, Mice, Mice, Transgenic, Organ Culture Techniques, Prefrontal Cortex drug effects, Synaptic Transmission physiology, Huntington Disease physiopathology, Long-Term Potentiation physiology, Prefrontal Cortex physiopathology, Receptors, Dopamine D1 metabolism
- Abstract
Background: The introduction of gene testing for Huntington's disease (HD) has enabled the neuropsychiatric and cognitive profiling of human gene carriers prior to the onset of overt motor and cognitive symptoms. Such studies reveal an early decline in working memory and executive function, altered EEG and a loss of striatal dopamine receptors. Working memory is processed in the prefrontal cortex and modulated by extrinsic dopaminergic inputs., Objective: We sought to study excitatory synaptic function and plasticity in the medial prefrontal cortex of mouse models of HD., Methods: We have used 2 mouse models of HD, carrying 89 and 116 CAG repeats (corresponding to a preclinical and symptomatic state, respectively) and performed electrophysiological field recording in coronal slices of the medial prefrontal cortex., Results: We report that short-term synaptic plasticity and long-term potentiation (LTP) are impaired and that the severity of impairment is correlated with the size of the CAG repeat. Remarkably, the deficits in LTP and short-term plasticity are reversed in the presence of a D(1) dopamine receptor agonist (SKF38393)., Conclusion: In a previous study, we demonstrated that a deficit in long-term depression (LTD) in the perirhinal cortex could also be reversed by a dopamine agonist. These and our current data indicate that inadequate dopaminergic modulation of cortical synaptic function is an early event in HD and may provide a route for the alleviation of cognitive dysfunction., (Copyright © 2011 S. Karger AG, Basel.)
- Published
- 2011
- Full Text
- View/download PDF
38. Early synaptic pathophysiology in neurodegeneration: insights from Huntington's disease.
- Author
-
Milnerwood AJ and Raymond LA
- Subjects
- Animals, Humans, Mice, Neuronal Plasticity physiology, Synaptic Transmission physiology, Huntington Disease physiopathology, Nerve Degeneration physiopathology, Synapses metabolism, Synapses pathology
- Abstract
Investigations of synaptic transmission and plasticity in mouse models of Huntington's disease (HD) demonstrate neuronal dysfunction long before the onset of classical disease indicators. Similarly, recent human studies reveal synaptic dysfunction decades before predicted clinical diagnosis in HD gene carriers. These studies guide premanifest tracking of disease and the development of treatment assessment tools. New discoveries of mechanisms underlying early neuronal dysfunction, including elevated pathogenic extrasynaptic NMDA receptor signaling, reduced synaptic connectivity and loss of brain-derived neurotrophic factor (BDNF) support have led to pharmacological interventions that can reverse or delay phenotype onset and disease progression in HD mice. Further understanding the primary effects of gene mutations associated with late-onset neurodegeneration should translate to novel treatments for HD families and guide therapeutic strategies for other neurodegenerative diseases., (Copyright © 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
39. Early increase in extrasynaptic NMDA receptor signaling and expression contributes to phenotype onset in Huntington's disease mice.
- Author
-
Milnerwood AJ, Gladding CM, Pouladi MA, Kaufman AM, Hines RM, Boyd JD, Ko RW, Vasuta OC, Graham RK, Hayden MR, Murphy TH, and Raymond LA
- Subjects
- Action Potentials genetics, Animals, Huntington Disease genetics, Mice, Mice, Transgenic, Receptors, N-Methyl-D-Aspartate genetics, Signal Transduction genetics, Synapses chemistry, Synapses genetics, Synaptic Transmission genetics, Time Factors, Disease Models, Animal, Gene Expression Regulation, Huntington Disease metabolism, Phenotype, Receptors, N-Methyl-D-Aspartate biosynthesis, Signal Transduction physiology, Synapses metabolism
- Abstract
N-methyl-D-aspartate receptor (NMDAR) excitotoxicity is implicated in the pathogenesis of Huntington's disease (HD), a late-onset neurodegenerative disorder. However, NMDARs are poor therapeutic targets, due to their essential physiological role. Recent studies demonstrate that synaptic NMDAR transmission drives neuroprotective gene transcription, whereas extrasynaptic NMDAR activation promotes cell death. We report specifically increased extrasynaptic NMDAR expression, current, and associated reductions in nuclear CREB activation in HD mouse striatum. The changes are observed in the absence of dendritic morphological alterations, before and after phenotype onset, correlate with mutation severity, and require caspase-6 cleavage of mutant huntingtin. Moreover, pharmacological block of extrasynaptic NMDARs with memantine reversed signaling and motor learning deficits. Our data demonstrate elevated extrasynaptic NMDAR activity in an animal model of neurodegenerative disease. We provide a candidate mechanism linking several pathways previously implicated in HD pathogenesis and demonstrate successful early therapeutic intervention in mice., (Copyright 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
40. Corticostriatal synaptic function in mouse models of Huntington's disease: early effects of huntingtin repeat length and protein load.
- Author
-
Milnerwood AJ and Raymond LA
- Subjects
- Animals, Excitatory Postsynaptic Potentials physiology, Genetic Load, Genotype, Huntingtin Protein, Mice, Mice, Transgenic, Nerve Tissue Proteins physiology, Nuclear Proteins physiology, Receptors, AMPA genetics, Receptors, AMPA physiology, Receptors, N-Methyl-D-Aspartate genetics, Receptors, N-Methyl-D-Aspartate physiology, Receptors, Presynaptic physiology, Synaptic Transmission genetics, Synaptic Transmission physiology, Cerebral Cortex physiopathology, Huntington Disease physiopathology, Neostriatum physiopathology, Nerve Tissue Proteins genetics, Nuclear Proteins genetics, Synapses physiology
- Abstract
Huntington's disease (HD) is an autosomal dominant, late onset, neurodegenerative disease characterized by motor deficits and dementia that is caused by expansion of a CAG repeat in the HD gene. Clinical manifestations result from selective neuronal degeneration of predominantly GABAergic striatal medium-sized spiny neurons (MSNs). A growing number of studies demonstrate that personality, mood and cognitive disturbances are some of the earliest signs of HD and may reflect synaptic dysfunction prior to neuronal loss. Previous studies in striatal MSNs demonstrated early alterations in NMDA-type glutamate receptor currents in several HD mouse models, as well as evidence for presynaptic dysfunction prior to disease manifestations in the R6/2 HD fragment mouse model. We have compared corticostriatal synaptic function in full-length, human HD gene-carrying YAC transgenic mice expressing a non-pathogenic CAG repeat (YAC18; control) with three increasingly severe variants of pathogenic HD gene-expressing mice (YAC72 and two different lines of YAC128), at ages that precede any detectable disease phenotype. We report presynaptic dysfunction and a propensity towards synaptic depression in YAC72 and YAC128 compared to YAC18 mice, and, in the most severe model, we also observed altered AMPA receptor function. When normalized to evoked AMPAR currents, postsynaptic NMDAR currents are augmented in all three pathogenic HD YAC variants. These findings demonstrate multiple perturbations to corticostriatal synaptic function in HD mice, furthering our understanding of the early effects of the HD mutation that may contribute to cognitive dysfunction, mood disorders and later development of more serious dysfunction. Furthermore, this study provides a set of neurophysiological sequelae against which to test and compare other mouse models and potential therapies in HD.
- Published
- 2007
- Full Text
- View/download PDF
41. Progressive CAG expansion in the brain of a novel R6/1-89Q mouse model of Huntington's disease with delayed phenotypic onset.
- Author
-
Vatsavayai SC, Dallérac GM, Milnerwood AJ, Cummings DM, Rezaie P, Murphy KP, and Hirst MC
- Subjects
- Animals, Brain pathology, Genotype, Huntingtin Protein, Immunohistochemistry, Mice, Transgenic, Nerve Tissue Proteins genetics, Nuclear Proteins genetics, Phenotype, Polymerase Chain Reaction, Trinucleotide Repeat Expansion, Brain metabolism, Disease Models, Animal, Huntington Disease genetics, Mice
- Abstract
Transgenic models representing Huntington's disease (HD) have proved useful for understanding the cascade of molecular events leading to the disease. We report an initial characterisation of a novel transgenic mouse model derived from a spontaneous truncation event within the R6/1 transgene. The transgene is widely expressed, carries 89 CAG repeats and the animals exhibit a significantly milder neurological phenotype with delayed onset compared to R6/1. Moreover, we report evidence of progressive somatic CAG expansions in the brain starting at an early age before an overt phenotype has developed. This novel line shares a common genetic ancestry with R6/1, differing only in CAG repeat number, and therefore, provides an additional tool with which to examine early molecular and neurophysiological changes in HD.
- Published
- 2007
- Full Text
- View/download PDF
42. Abnormal cortical synaptic plasticity in a mouse model of Huntington's disease.
- Author
-
Cummings DM, Milnerwood AJ, Dallérac GM, Vatsavayai SC, Hirst MC, and Murphy KP
- Subjects
- Animals, Disease Models, Animal, Huntington Disease complications, Memory Disorders etiology, Mice, Microelectrodes, Organ Culture Techniques, Synapses pathology, Cerebral Cortex physiopathology, Excitatory Postsynaptic Potentials physiology, Huntington Disease physiopathology, Long-Term Synaptic Depression physiology
- Abstract
Huntington's disease is a fatal neurodegenerative disorder characterised by a progressive motor, psychiatric and cognitive decline and associated with a marked loss of neurons in the cortex and striatum of affected individuals. The disease is inherited in an autosomal dominant fashion and is caused by a trinucleotide (CAG) repeat expansion in the gene encoding the protein huntingtin. Predictive genetic testing has revealed early cognitive deficits in asymptomatic gene carriers such as altered working memory, executive function and recognition memory. The perirhinal cortex is believed to process aspects of recognition memory. Evidence from primate studies suggests that decrements in neuronal firing within this cortical region encode recognition memory and that the underlying mechanism is an activity-dependent long-term depression (LTD) of excitatory neurotransmission, the converse of long-term potentiation (LTP). We have used the R6/1 mouse model of HD to assess synaptic plasticity in the perirhinal cortex. This mouse model provides an ideal tool for investigating early and progressive changes in synaptic function in HD. We report here that LTD at perirhinal synapses is markedly reduced in R6/1 mice. We also provide evidence to suggest that a reduction in dopamine D2 receptor signalling may be implicated.
- Published
- 2007
- Full Text
- View/download PDF
43. Aberrant cortical synaptic plasticity and dopaminergic dysfunction in a mouse model of Huntington's disease.
- Author
-
Cummings DM, Milnerwood AJ, Dallérac GM, Waights V, Brown JY, Vatsavayai SC, Hirst MC, and Murphy KP
- Subjects
- Animals, Cerebral Cortex physiopathology, Disease Models, Animal, Female, Humans, Huntingtin Protein, Huntington Disease genetics, Male, Mice, Mice, Inbred C57BL, Mice, Inbred CBA, Mice, Transgenic, Nerve Tissue Proteins genetics, Nuclear Proteins genetics, Receptors, Dopamine metabolism, Synaptic Transmission, Dopamine physiology, Huntington Disease physiopathology, Neuronal Plasticity physiology
- Abstract
Predictive genetic testing for Huntington's disease (HD) has revealed early cognitive deficits in asymptomatic gene carriers, such as altered working memory, executive function and impaired recognition memory. The perirhinal cortex processes aspects of recognition memory and the underlying mechanism is believed to be long-term depression (LTD) of excitatory neurotransmission, the converse of long-term potentiation (LTP). We have used the R6/1 mouse model of HD to assess synaptic plasticity in the perirhinal cortex. We report here a progressive derailment of both LTD and short-term plasticity at perirhinal synapses. Layer II/III neurones gradually lose their ability to support LTD, show early nuclear localization of mutant huntingtin and display a progressive loss of membrane integrity (depolarization and loss of cell capacitance) accompanied by a reduction in the expression of D1 and D2 dopamine receptors visualized in layer I of the perirhinal cortex. Importantly, abnormalities in both short-term and long-term plasticity can be reversed by the introduction of a D2 dopamine receptor agonist (Quinpirole), suggesting that alterations in dopaminergic signalling may underlie early cognitive dysfunction in HD.
- Published
- 2006
- Full Text
- View/download PDF
44. Early development of aberrant synaptic plasticity in a mouse model of Huntington's disease.
- Author
-
Milnerwood AJ, Cummings DM, Dallérac GM, Brown JY, Vatsavayai SC, Hirst MC, Rezaie P, and Murphy KP
- Subjects
- Animals, Cell Nucleus pathology, Disease Models, Animal, Hippocampus metabolism, Hippocampus pathology, Huntingtin Protein, Huntington Disease genetics, Huntington Disease pathology, Mice, Mice, Transgenic, Mutation, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Nuclear Proteins genetics, Nuclear Proteins metabolism, Phenotype, Receptors, N-Methyl-D-Aspartate physiology, Synaptic Transmission, Aging physiology, Huntington Disease physiopathology, Long-Term Synaptic Depression, Synapses pathology
- Abstract
Huntington's disease (HD) is a fatal neurodegenerative disorder characterized by progressive motor, psychiatric and cognitive decline. Marked neuronal loss occurs in the cortex and striatum. HD is inherited in an autosomal dominant fashion and caused by a trinucleotide repeat expansion (CAG) in the gene encoding the protein huntingtin. Predictive genetic testing has revealed early cognitive deficits in asymptomatic gene carriers at a time when there is little evidence for cell death, suggesting that impaired cognition results from a cellular or synaptic deficit, such as aberrant synaptic plasticity. Altered hippocampal long-term potentiation has been reported in mouse models of HD; however, the relationship between synaptic dysfunction and phenotype progression has not previously been characterized. We examined the age-dependency of aberrant hippocampal synaptic plasticity in the R6/1 mouse model of HD. Long-term depression (LTD) is a developmentally regulated form of plasticity, which normally declines by early adulthood. Young R6/1 mice follow the same pattern of LTD expression as controls, in that they express LTD in the first weeks of life, and then lose the ability with age. Unlike controls, R6/1 synapses later regain the ability to support LTD. This is associated with nuclear localization of mutant huntingtin, but occurs months prior to the formation of nuclear aggregates. We present the first detailed description of a progressive derailment of a functional neural correlate of cognitive processing in HD.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.