The Use of Pharmacological Retromer Chaperones in Alzheimer's

Total Page:16

File Type:pdf, Size:1020Kb

The Use of Pharmacological Retromer Chaperones in Alzheimer's Neurotherapeutics (2015) 12:12–18 DOI 10.1007/s13311-014-0321-y REVIEW The Use of Pharmacological Retromer Chaperones in Alzheimer’s Disease and other Endosomal-related Disorders Diego E. Berman & Dagmar Ringe & Greg A. Petsko & Scott A. Small Published online: 4 December 2014 # The American Society for Experimental NeuroTherapeutics, Inc. 2014 Abstract The retromer is an evolutionary conserved Endosomes, Retromer, and Alzheimer’sDisease multiprotein complex involved in the sorting and retrograde trafficking of cargo from endosomal compartments to the A common emerging theme in the research field of age-related Golgi network and to the cell surface. The neuronal retromer neurodegenerative diseases is that of endosomal dysfunction traffics the amyloid precursor protein away from the [1]. Specifically in Alzheimer’s disease (AD), primary evi- endosomes, a site where amyloid precursor protein is enzy- dence pinpointing the endosome as the intracellular site of matically cleaved into pathogenic fragments in Alzheimer’s dysfunction was described in a seminal series of histological disease. In recent years, deficiencies in retromer-mediated studies performed by Cataldo et al. [2]. They showed that transport have been implicated in several neurological and endosomes in the entorhinal cortex and the cornu ammonis non-neurological diseases, including Parkinson’sdisease,sug- subfields of the hippocampal formation are abnormally en- gesting that improving the efficacy of the retromer trafficking larged during the earliest stages of disease. Remarkably, the pathway would result in decreased pathology. We recently distribution of abnormally enlarged endosomes was nearly identified a new family of small molecules that appear to diagnostic of the disease, with very little overlap in the size stabilize the interaction between members of the retromer of endosomes observed in age-matched controls. Additional- complex and enhance its function in neurons: the retromer ly, they showed that carrying the apolipoprotein E4 gene pharmacological chaperones. Here we discuss the role of these variant, the strongest genetic risk factor for late-onset AD, molecules in the improvement of retromer trafficking and further enhanced endosomal enlargement, and that abnormal endosomal dysfunction, as well as their potential as therapeu- enlargement did not occur as part of normal aging. The near tics for neurological and non-neurological disorders. uniformity of the abnormality in patients with late-onset “spo- radic” AD suggests a convergence of pathogenic mechanisms leading to endosomal enlargement. In the case of AD, this is Key Words Retromer . Alzheimer’s disease . particularly important as the endosomal compartment of the neurodegeneration . pharmacological chaperones cell is one of the main sites where amyloid precursor protein β : (APP) interacts with -site APP-cleaving enzyme 1 D. E. Berman (*) S. A. Small (BACE1), initiating a pathogenic process leading to the accu- ’ The Taub Institute for Research on Alzheimer s Disease and the mulation of a number of putatively neurotoxic proteolytic Aging Brain, and Department of Pathology and Cell Biology, β Columbia University College of Physicians and Surgeons, New fragments, including beta-amyloid (A ), the main component York, NY 10032, USA of the amyloid plaques found in AD brains. The fact that e-mail: [email protected] enlarged endosomes represent a cell-biological phenotype of AD has recently been validated in neurons derived from D. Ringe Department of Biochemistry and Chemistry, Rosenstiel Basic patients via induced pluripotent stem cells technology [3]. Medical Sciences Research Center, Brandeis University, Waltham, Endosomes are a hub of membrane trafficking, and mecha- MA 02454, USA nistically it can be assumed that they become enlarged either because of accelerated delivery, via cell-surface endocytosis G. A. Petsko Helen and Robert Appel Alzheimer’s Disease Research Institute, or by reducing the transport of molecules out of endosomes. Weill Cornell Medical College, New York, NY 10065, USA Taken together, these observations establish that endosomal Retromer Chaperones in Alzheimer’sDisease 13 dysfunction is a pathogenic mechanism, and validate takes place; however, many of the studies suggesting a endosomal transport as a “cell biological” target for drug nonendosomal locus of amyloidogenic APP processing have discovery. been performed in experimental conditions that do not resem- As we will discuss below, proteins and mechanisms that ble the normal physiological neuronal state. Nevertheless, one modulate the endosomal residency time of APP, BACE1, and aspect of Aß production is clear: the more time APP and other key molecular players, and hence their physical interac- BACE1 reside and interact with each other in any given tion, could thus regulate the production of Aβ and have cellular compartment under the right conditions (e.g., an acid- implications for AD. One such mechanism is the retromer- ic pH for optimal BACE1 enzymatic activity), the more Aβ is dependent retrieval of transmembrane proteins, such as APP, likely to be produced by the cell. This interplay will be from endosomes to the trans-Golgi network and the plasma determined by the dynamic trafficking routes of each of these membrane. We will begin by looking in some detail at these molecular components separately. While we know that key molecular entities, as their basic biology is important to retromer regulates APP trafficking, less is known about the further understand their roles in neuronal AD-related role of retromer affecting BACE1 transport. BACE1 also retromer-mediated trafficking. shuttles between different membranous subcellular compart- APP is a type I transmembrane protein that can undergo ments [9], hence raising the critical, and still unanswered, proteolytic processing in different ways by different sets of question of how both these important AD molecular players enzymes—in normal healthy conditions, a minor pathway come in closer contact and trigger amyloid-related AD leads to amyloid plaque formation (the amyloidogenic path- pathophysiology. way), while the other major pathway does not (the Another key player in the regulation of APP processing at nonamyloidogenic pathway). In the nonamyloidogenic path- the endosomal level is a sorting protein-related receptor con- way, APP is cleaved first by α-secretase to yield a soluble taining low-density lipoprotein receptor class A repeats N-terminal fragment (sAPPα) and a C-terminal fragment (SorLA). Like APP, SorLA is a type I transmembrane protein (αCTF). The sAPPα fragment may be involved in the that is expressed in neurons and is a critical participant in the enhancement of synaptogenesis, neurite outgrowth, and neu- intracellular transport and processing of APP. Through its ronal survival, and is considered to be neuroprotective [4]. In Vps10-like structural similarity, SorLA binds to the retromer the amyloidogenic pathway, APP is cleaved first by BACE1 complex and serves as an adaptor protein for the trafficking of (a transmembrane aspartic protease), yielding a soluble APP from endosomes to the TGN [10]. It has been shown that N-terminal fragment (sAPPβ) and a membrane-bound SorLA levels are reduced in affected brain areas in patients C-terminal fragment (βCTF). βCTF is then acted upon by with AD [10], and that overexpression of SorLA redistributes γ-secretase yielding a membrane-bound C-terminal fragment APP to the Golgi apparatus thereby reducing the residency and a soluble N-terminal fragment, Aβ.Aβ can then accu- and interaction time of APP and BACE1 in the early mulate in the extracellular space of the brain, where it can endosomes. Consequently, Aβ levels are decreased in SorLA form oligomers, insoluble fibrils, and amyloid plaques if overexpression conditions. These findings became more rele- they are insufficiently cleared, as observed in advanced AD. vant when variants in SORL1 (coding for SorLA) associated The progression to AD is thus determined by the proportion with late-onset AD were identified a few years ago [11]. of APP that is processed by the pathways initiated by The retromer complex was first identified in yeast, medi- BACE1 (i.e., amyloidogenic) and α-secretases (i.e., ating the retrieval of the acid hydrolase receptors from nonamyloidogenic). APP is constitutively actively sorted endosomes (vacuoles in yeast) to the Golgi apparatus [12]. and trafficked through membrane compartments of the cell. Further studies showed that the function and components of After internalization from the cell surface and while it tempo- the retromer complex is evolutionary conserved across spe- rarily resides in endosomes, APP is most likely to interact and cies. Retromers are heteropentameric protein complexes com- be cleaved by BACE1 [5]. Previous studies have shown that posed mainly of 2 components: 1) a structural component retromer-related defects reduced trafficking of APP out of made of a dimer of sorting nexins (SNX), which mediates endosomes and caused an elevation in Aβ levels in vivo the binding to lipids in the endosomal membrane; and 2) a [6, 7]. That said, a recent study performed on HEK293 cells core cargo recognition element, a heterotrimer comprised of using an RNA interference approach suggests that Aβ40 is ‘vacuolar protein sorting’ (VPS) proteins Vps35–Vps29– generated predominantly in the TGN [8]; however, in this case Vps26. Vps35 is the core component of the complex, which no other APP proteolytic fragments were studied. Yet another
Recommended publications
  • The Effects of P75 and Sorcs2 on Neuronal Function and Structure
    The Effects of Zn2+ on the Binding of the BDNF Prodomain with SorCS2 on Neuronal Function and Structure: Implications for Learning and Memory Caroline Pennacchio Briarcliff High School http://www.newscientist.com/data/images/ns/cms/teaser/blog/201211/f0049969_lead.jpg Introduction • Neurons -> Brain • Hippocampus = Control Center • Neural connections -> Brain-Derived Neurotrophic Factor (BDNF) Introduction Review of Literature Research Questions/Hypotheses Methods Bibliography Ligands • Regulate cell proliferation, differentiation • Axon and dendrite growth, synaptogenesis, and synaptic function and plasticity (Reichardt, 2006, Lu, B, Pang, PT, Woo. N.H., 2005, Minichiello L, 2009) http://www.mdpi.com/ijms/ijms-13-13713/article_deploy/html/images/ijms-13-13713f1-1024.png Introduction Review of Literature Research Questions/Hypotheses Methods Bibliography Pro-Neurotrophins • Proteolytically cleaved in trans-Golgi by furin or in secretory granules by pro-protein contervases ] • Extracellular cleavages created in mature domain formation show how to control synaptic functions of neurotrophins (Lu, 2003) • proBDNF regulates hippocampal structure, synaptic transmission, and plasticity (Yang et al., 2014) https://www.researchgate.net/profile/Jay_Pundavela/publication/269520194/figure/fig1/Fig-1-Binding-of-neurotrophins- • Induce apoptotic signaling (Nykjaer et al. 2004; Teng et al. 2005; Jansen et al. and-proneurotrophins-to-Trk-receptors-and-p75NTR-NGF_small.png 2007; Willnow et al. 2008; Yano et al. 2009) Introduction Review of Literature
    [Show full text]
  • Sorcs2 Deletion Leads to Altered Neuronal Lysosome Activity
    bioRxiv preprint doi: https://doi.org/10.1101/2021.04.08.439000; this version posted April 10, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 SorCS2 deletion leads to altered neuronal lysosome activity 2 3 Sérgio Almeida1*, André M. Miranda2, Andrea E. Tóth1, Morten S. Nielsen1 and Tiago Gil Oliveira2 4 1 Department of Biomedicine, Aarhus University, Aarhus, Denmark 5 2 Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal 6 *Corresponding author: Sérgio Almeida, Department of Biomedicine, Aarhus University, Høegh- 7 Guldbergsgade 10, DK-8000C Aarhus, Denmark. Phone: +45 60511406. E-mail: [email protected] 8 9 10 11 12 13 Abstract 14 Vps10p domain receptors are important for regulating intracellular protein sorting within the central 15 nervous system and as such constitute risk factors for different brain pathologies. Here, we show that 16 removal of SorCS2 leads to altered lysosomal activity in mouse primary neurons. SorCS2-/- neurons show 17 elevated lysosomal markers such as LAMP1 and acidic hydrolases including cathepsin B and D. Despite 18 increased levels, SorCS2-/- neurons fail to degrade cathepsin specific substrates in a live context. SorCS2- 19 deficient mice present an increase in lysolipids, which may contribute to membrane permeabilization and 20 increased susceptibility to lysosomal stress. Our findings highlight SorCS2 as an important factor for a 21 balanced neuronal lysosome milieu.
    [Show full text]
  • The Contributions of Microglial Vps35 to Adult Hippocampal Neurogenesis and Neurodegenerative Disorders
    THE CONTRIBUTIONS OF MICROGLIAL VPS35 TO ADULT HIPPOCAMPAL NEUROGENESIS AND NEURODEGENERATIVE DISORDERS By Joanna Ruth Erion Submitted to the Faculty of the Graduate School of Augusta University in partial fulfillment of the Requirements of the Degree of Doctor of Philosophy April 2017 COPYRIGHT© 2017 by Joanna Ruth Erion THE CONTRIBUTIONS OF MICROGLIAL VPS35 TO ADULT HIPPOCAMPAL NEUROGENESIS AND NEURODEGENERATIVE DISORDERS This thesis/dissertation is submitted by Joanna Ruth Erion and has been examined and approved by an appointed committee of the faculty of the Graduate School of Augusta University. The signatures which appear below verify the fact that all required changes have been incorporated and that the thesis/dissertation has received final approval with reference to content, form and accuracy of presentation. This thesis/dissertation is therefore in partial fulfillment of the requirements for the degree of Doctor of Philosophy). ___________________ __________________________________ Date Major Advisor __________________________________ Departmental Chairperson __________________________________ Dean, Graduate School ACKNOWLEDGEMENTS I will be forever grateful to my mentor, Dr. Wen-Cheng Xiong, for welcoming me into her laboratory and providing me with the honor and opportunity to work under her astute leadership. I have learned many valuable lessons under her guidance, all of which have enabled me to grow as both a student and a scientist. It was with her erudite guidance and suggestions that I was able to examine aspects of my investigations that I had yet to consider and ask questions that would not have otherwise occurred to me. I would also like to thank Dr. Lin Mei for contributing not only his laboratory and resources to my endeavors, but also his guidance and input into my investigations, providing me with additional avenues for directing my efforts.
    [Show full text]
  • Recent Efforts to Dissect the Genetic Basis of Alcohol Use and Abuse
    Title: Recent efforts to dissect the genetic basis of alcohol use and abuse Authors: Sandra Sanchez-Roige, PhD1, Abraham A. Palmer, PhD1,2, Toni-Kim Clarke, PhD3 Affiliations: 1 Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA, 2 Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, 92093, USA, 3 Division of Psychiatry, University of Edinburgh, Edinburgh, UK Correspondence: Sandra Sanchez-Roige, Ph.D. Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA. E-mail: [email protected] Short Title: Alcohol genetics Keywords: alcoholism; alcohol consumption; AUDIT; alcohol-metabolizing genes; Genome- wide association studies; Genetics Word count: 3,981 1 Abstract (202 words) Alcohol use disorders (AUD) are defined by several symptom criteria, which can be further dissected at the genetic level. Over the past several years, our understanding of the genetic factors influencing alcohol use and abuse has progressed tremendously; hundreds of loci have now been implicated in different aspects of alcohol use. Previously known associations with alcohol metabolizing enzymes (ADH1B, ALDH2) have been definitively replicated. Additionally, novel associations with loci containing the genes KLB, GCKR, CRHR1 and CADM2 have been reported. Downstream analyses have leveraged these genetic findings to reveal important relationships between alcohol use behaviors and both physical and mental health. AUD and aspects of alcohol misuse have been shown to overlap strongly with psychiatric disorders, whereas aspects of alcohol consumption have shown stronger links to metabolism. These results demonstrate that the genetic architecture of alcohol consumption only partially overlaps with the genetics of clinically defined AUD.
    [Show full text]
  • Parkinson's Disease Causative Mutation in Vps35 Disturbs Tetherin
    cells Article Parkinson’s Disease Causative Mutation in Vps35 Disturbs Tetherin Trafficking to Cell Surfaces and Facilitates Virus Spread Yingzhuo Ding 1,†, Yan Li 1,† , Gaurav Chhetri 1 , Xiaoxin Peng 1, Jing Wu 1, Zejian Wang 1, Bo Zhao 1 , Wenjuan Zhao 1 and Xueyi Li 1,2,* 1 School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; [email protected] (Y.D.); [email protected] (Y.L.); [email protected] (G.C.); [email protected] (X.P.); [email protected] (J.W.); [email protected] (Z.W.); [email protected] (B.Z.); [email protected] (W.Z.) 2 Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA * Correspondence: [email protected] or [email protected] † These authors contributed equally to this work. Abstract: Parkinson’s disease (PD) is the most common neurodegenerative movement disorder, characterized by progressive loss of dopaminergic neurons in the substantia nigra, intraneuronal deposition of misfolded proteins known as Lewy bodies, and chronic neuroinflammation. PD can arise from monogenic mutations, but in most cases, the etiology is unclear. Viral infection is gaining increasing attentions as a trigger of PD. In this study, we investigated whether the PD-causative Citation: Ding, Y.; Li, Y.; Chhetri, G.; 620 aspartate (D) to asparagine (N) mutation in the vacuolar protein sorting 35 ortholog (Vps35) Peng, X.; Wu, J.; Wang, Z.; Zhao, B.; precipitated herpes simplex virus (HSV) infection. We observed that ectopic expression of Vps35 Zhao, W.; Li, X. Parkinson’s Disease significantly reduced the proliferation and release of HSV-1 virions; the D620N mutation rendered Causative Mutation in Vps35 Vps35 a partial loss of such inhibitory effects.
    [Show full text]
  • Retromer Deficiency Observed in Alzheimer's Disease Causes
    Retromer deficiency observed in Alzheimer’s disease causes hippocampal dysfunction, neurodegeneration, and A␤ accumulation Alim Muhammad*, Ingrid Flores*, Hong Zhang*†, Rui Yu*, Agnieszka Staniszewski*†, Emmanuel Planel*†, Mathieu Herman*†, Lingling Ho‡, Robert Kreber‡, Lawrence S. Honig*§, Barry Ganetzky‡¶, Karen Duff*†, Ottavio Arancio*†, and Scott A. Small*§¶ *Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, and Departments of §Neurology and †Pathology, Columbia University College of Physicians and Surgeons, New York, NY 10032; and ‡Laboratory of Genetics, University of Wisconsin, Madison, WI 53706-1580 Contributed by Barry Ganetzky, March 13, 2008 (sent for review December 2, 2007) Although deficiencies in the retromer sorting pathway have been ␥-secretase, it was important to investigate retromer deficiency linked to late-onset Alzheimer’s disease, whether these deficien- on a nonmutated genetic background. To determine whether cies underlie the disease remains unknown. Here we characterized retromer deficiency causes hippocampal dysfunction, a key two genetically modified animal models to test separate but clinical feature of Alzheimer’s disease, we began by investigating related questions about the effects that retromer deficiency has on retromer deficiency in genetically modified mice. Our results the brain. First, testing for cognitive defects, we investigated suggest that retromer deficiency causes hippocampal dysfunction retromer-deficient mice and found that they develop hippocampal- by elevating concentrations of endogenous A␤ peptide. dependent memory and synaptic dysfunction, which was associ- Although APP and BACE are highly homologous across species, ated with elevations in endogenous A␤ peptide. Second, testing important sequence differences do exist, and these differences can for neurodegeneration and amyloid deposits, we investigated affect their intracellular transport, APP processing, and the neu- retromer-deficient flies expressing human wild-type amyloid pre- rotoxicity of APP end products (12).
    [Show full text]
  • Mood Stabilizers in Psychiatric Disorders and Mechanisms Learnt from in Vitro Model Systems
    International Journal of Molecular Sciences Review Mood Stabilizers in Psychiatric Disorders and Mechanisms Learnt from In Vitro Model Systems Ritu Nayak, Idan Rosh, Irina Kustanovich and Shani Stern * Sagol Department of Neurobiology, University of Haifa, Haifa 3498838, Israel; [email protected] (R.N.); [email protected] (I.R.); [email protected] (I.K.) * Correspondence: [email protected] Abstract: Bipolar disorder (BD) and schizophrenia are psychiatric disorders that manifest unusual mental, behavioral, and emotional patterns leading to suffering and disability. These disorders span heterogeneous conditions with variable heredity and elusive pathophysiology. Mood stabilizers such as lithium and valproic acid (VPA) have been shown to be effective in BD and, to some extent in schizophrenia. This review highlights the efficacy of lithium and VPA treatment in several randomized, controlled human trials conducted in patients suffering from BD and schizophrenia. Furthermore, we also address the importance of using induced pluripotent stem cells (iPSCs) as a disease model for mirroring the disease’s phenotypes. In BD, iPSC-derived neurons enabled finding an endophenotype of hyperexcitability with increased hyperpolarizations. Some of the disease phenotypes were significantly alleviated by lithium treatment. VPA studies have also reported rescuing the Wnt/β-catenin pathway and reducing activity. Another significant contribution of iPSC models can be attributed to studying the molecular etiologies of schizophrenia such as abnormal differentiation of patient-derived neural stem cells, decreased neuronal connectivity and neurite Citation: Nayak, R.; Rosh, I.; number, impaired synaptic function, and altered gene expression patterns. Overall, despite significant Kustanovich, I.; Stern, S. Mood advances using these novel models, much more work remains to fully understand the mechanisms Stabilizers in Psychiatric Disorders by which these disorders affect the patients’ brains.
    [Show full text]
  • Parkinson's Disease-Linked D620N VPS35 Knockin Mice Manifest Tau
    Parkinson’s disease-linked D620N VPS35 knockin mice manifest tau neuropathology and dopaminergic neurodegeneration Xi Chena, Jennifer K. Kordicha, Erin T. Williamsa, Nathan Levinea, Allyson Cole-Straussb, Lee Marshalla, Viviane Labriea,c, Jiyan Maa, Jack W. Liptonb, and Darren J. Moorea,1 aCenter for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503; bDepartment of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI 49503; and cDivision of Psychiatry and Behavioral Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503 Edited by Anders Björklund, Lund University, Lund, Sweden, and approved February 13, 2019 (received for review August 30, 2018) Mutations in the vacuolar protein sorting 35 ortholog (VPS35) since only a single D620N mutation carrier has been evaluated at gene represent a cause of late-onset, autosomal dominant familial autopsy but with the notable exception of key PD-relevant brain Parkinson’s disease (PD). A single missense mutation, D620N, is regions (i.e., substantia nigra, locus ceruleus, or any brainstem considered pathogenic based upon its segregation with disease area) (7). Outside of these areas, VPS35 mutation carriers lack in multiple families with PD. At present, the mechanism(s) by extranigral α-synuclein–positive Lewy body pathology (7), a which familial VPS35 mutations precipitate neurodegeneration in characteristic hallmark of PD brains. The mechanism by which PD are poorly understood. Here, we employ a germline D620N dominantly inherited mutations in VPS35 induce neuropathology VPS35 knockin (KI) mouse model of PD to formally establish the and neurodegeneration in PD remains enigmatic. age-related pathogenic effects of the D620N mutation at physiolog- VPS35 encodes a core component of the retromer complex, ical expression levels.
    [Show full text]
  • 14Th Annual OAK Meeting Aarhus 29 May 2015
    14th Annual OAK Meeting Danish Brain Research Laboratories Meeting Aarhus 29 May 2015 Aarhus University Merete Barker Auditorium www.cfin.au.dk/OAK-2015 14th Annual OAK Meeting Danish Brain Research Laboratories Meeting PROGRAM Friday 29 May 2015 10:00 Arrival & registration at Merete Barker Auditorium, Aarhus University 10:15-10:30 Welcome by Arne Møller Session 1: In-vivo Neuroimaging (Chair: Anne M. Landau & Kim Ryun Drasbek) 10:30-10:45 Ali Khalidan Vibholm: Preclinical in-vivo imaging of activated NMDA receptor ion channels with the novel radioligand [18F]-GE179 10:45-11:00 Athanasios Metaxas: PET imaging of the NMDA receptor using [18F]PK209 11:00-11:15 Andreas N. Glud: Parkinson’s disease models of abnormal protein aggregation in the Göttingen minipig CNS 11:15-11:30 Jenny-Ann Phan: Alpha synuclein model of Parkinson’s disease displays early synaptic disruption 11:30-11:45 Janne Vejlby: Endocannabinoid modulation of noradrenaline release 11:45-12:00 Majken Borup Thomsen: Increased receptor density of α2 adrenoceptors and GABAA α5 receptors in limbic brain regions in the domoic acid rat model of epilepsy 12.00-13.00 LUNCH BREAK Session 2: Animal models (Chair: Flemming Fryd Johansen) 13:00-13:15 Charlotte Havelund Nykjær: Stereological estimation of the brain white matter in Multiple System Atrophy 13:15-13:30 Jonas Folke: Deregulation of Wnt pathway in the prefrontal cortex from Alzheimer’s disease brains 13:30-13:45 Anders Malmendal: Insights into Alzheimer’s disease from NMR metabolomics of Aβ-expressing Drosophila 13:45-14:00
    [Show full text]
  • Independent and Epistatic Effects of Variants in VPS10-D Receptors on Alzheimer Disease Risk and Processing of the Amyloid Precursor Protein (APP)
    Citation: Transl Psychiatry (2013) 3, e256; doi:10.1038/tp.2013.13 & 2013 Macmillan Publishers Limited All rights reserved 2158-3188/13 www.nature.com/tp Independent and epistatic effects of variants in VPS10-d receptors on Alzheimer disease risk and processing of the amyloid precursor protein (APP) C Reitz1,2,3, G Tosto2, B Vardarajan4, E Rogaeva5, M Ghani5, RS Rogers2, C Conrad2, JL Haines6, MA Pericak-Vance7, MD Fallin8, T Foroud9, LA Farrer4,10,11,12,13,14, GD Schellenberg15, PS George-Hyslop5,16,17, R Mayeux1,2,3,18,19,20 and the Alzheimer’s Disease Genetics Consortium (ADGC) Genetic variants in the sortilin-related receptor (SORL1) and the sortilin-related vacuolar protein sorting 10 (VPS10) domain- containing receptor 1 (SORCS1) are associated with increased risk of Alzheimer’s disease (AD), declining cognitive function and altered amyloid precursor protein (APP) processing. We explored whether other members of the (VPS10) domain-containing receptor protein family (the sortilin-related VPS10 domain-containing receptors 2 and 3 (SORCS2 and SORCS3) and sortilin (SORT1)) would have similar effects either independently or together. We conducted the analyses in a large Caucasian case control data set (n ¼ 11 840 cases, 10 931 controls) to determine the associations between single nucleotide polymorphisms (SNPs) in all the five homologous genes and AD risk. Evidence for interactions between SNPs in the five VPS10 domain receptor family genes was determined in epistatic statistical models. We also compared expression levels of SORCS2, SORCS3 and SORT1 in AD and control brains using microarray gene expression analyses and assessed the effects of these genes on c-secretase processing of APP.
    [Show full text]
  • Sorcs2-Mediated NR2A Trafficking Regulates Motor Deficits in Huntington’S Disease
    SorCS2-mediated NR2A trafficking regulates motor deficits in Huntington’s disease Qian Ma, … , Lino Tessarollo, Barbara L. Hempstead JCI Insight. 2017;2(9):e88995. https://doi.org/10.1172/jci.insight.88995. Research Article Neuroscience Motor dysfunction is a prominent and disabling feature of Huntington’s disease (HD), but the molecular mechanisms that dictate its onset and progression are unknown. The N-methyl-D-aspartate receptor 2A (NR2A) subunit regulates motor skill development and synaptic plasticity in medium spiny neurons (MSNs) of the striatum, cells that are most severely impacted by HD. Here, we document reduced NR2A receptor subunits on the dendritic membranes and at the synapses of MSNs in zQ175 mice that model HD. We identify that SorCS2, a vacuolar protein sorting 10 protein–domain (VPS10P- domain) receptor, interacts with VPS35, a core component of retromer, thereby regulating surface trafficking of NR2A in MSNs. In the zQ175 striatum, SorCS2 is markedly decreased in an age- and allele-dependent manner. Notably, SorCS2 selectively interacts with mutant huntingtin (mtHTT), but not WT huntingtin (wtHTT), and is mislocalized to perinuclear clusters in striatal neurons of human HD patients and zQ175 mice. Genetic deficiency of SorCS2 accelerates the onset and exacerbates the motor coordination deficit of zQ175 mice. Together, our results identify SorCS2 as an interacting protein of mtHTT and demonstrate that impaired SorCS2-mediated NR2A subunit trafficking to dendritic surface of MSNs is, to our knowledge, a novel mechanism contributing to motor coordination deficits of HD. Find the latest version: https://jci.me/88995/pdf RESEARCH ARTICLE SorCS2-mediated NR2A trafficking regulates motor deficits in Huntington’s disease Qian Ma,1 Jianmin Yang,2,3 Teresa A.
    [Show full text]
  • The Genetic Architecture of Parkinson's Disease
    Review The genetic architecture of Parkinson’s disease Cornelis Blauwendraat, Mike A Nalls, Andrew B Singleton Lancet Neurol 2020; 19: 170–78 Parkinson’s disease is a complex neurodegenerative disorder for which both rare and common genetic variants Published Online contribute to disease risk, onset, and progression. Mutations in more than 20 genes have been associated with the September 11, 2019 disease, most of which are highly penetrant and often cause early onset or atypical symptoms. Although our http://dx.doi.org/10.1016/ understanding of the genetic basis of Parkinson’s disease has advanced considerably, much remains to be done. S1474-4422(19)30287-X Further disease-related common genetic variability remains to be identified and the work in identifying rare risk Laboratory of Neurogenetics, National Institute on Aging, alleles has only just begun. To date, genome-wide association studies have identified 90 independent risk-associated National Institutes of Health, variants. However, most of them have been identified in patients of European ancestry and we know relatively little of Bethesda, MD, USA the genetics of Parkinson’s disease in other populations. We have a limited understanding of the biological functions (C Blauwendraat PhD, of the risk alleles that have been identified, although Parkinson’s disease risk variants appear to be in close proximity M A Nalls PhD, A B Singleton PhD); and Data to known Parkinson’s disease genes and lysosomal-related genes. In the past decade, multiple efforts have been made Tecnica International, to investigate the genetic architecture of Parkinson’s disease, and emerging technologies, such as machine learning, Glen Echo, MD, USA (M A Nalls) single-cell RNA sequencing, and high-throughput screens, will improve our understanding of genetic risk.
    [Show full text]