551
|
Dragatsis I, Dietrich P, Ren H, Deng YP, Del Mar N, Wang HB, Johnson IM, Jones KR, Reiner A. Effect of early embryonic deletion of huntingtin from pyramidal neurons on the development and long-term survival of neurons in cerebral cortex and striatum. Neurobiol Dis 2017; 111:102-117. [PMID: 29274742 PMCID: PMC5821111 DOI: 10.1016/j.nbd.2017.12.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 11/07/2017] [Accepted: 12/19/2017] [Indexed: 12/12/2022] Open
Abstract
We evaluated the impact of early embryonic deletion of huntingtin (htt) from pyramidal neurons on cortical development, cortical neuron survival and motor behavior, using a cre-loxP strategy to inactivate the mouse htt gene (Hdh) in emx1-expressing cell lineages. Western blot confirmed substantial htt reduction in cerebral cortex of these Emx-httKO mice, with residual cortical htt in all likelihood restricted to cortical interneurons of the subpallial lineage and/or vascular endothelial cells. Despite the loss of htt early in development, cortical lamination was normal, as revealed by layer-specific markers. Cortical volume and neuron abundance were, however, significantly less than normal, and cortical neurons showed reduced brain-derived neurotrophic factor (BDNF) expression and reduced activation of BDNF signaling pathways. Nonetheless, cortical volume and neuron abundance did not show progressive age-related decline in Emx-httKO mice out to 24 months. Although striatal neurochemistry was normal, reductions in striatal volume and neuron abundance were seen in Emx-httKO mice, which were again not progressive. Weight maintenance was normal in Emx-httKO mice, but a slight rotarod deficit and persistent hyperactivity were observed throughout the lifespan. Our results show that embryonic deletion of htt from developing pallium does not substantially alter migration of cortical neurons to their correct laminar destinations, but does yield reduced cortical and striatal size and neuron numbers. The Emx-httKO mice were persistently hyperactive, possibly due to defects in corticostriatal development. Importantly, deletion of htt from cortical pyramidal neurons did not yield age-related progressive cortical or striatal pathology.
Collapse
Affiliation(s)
- I Dragatsis
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - P Dietrich
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - H Ren
- Department of Anatomy & Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Y P Deng
- Department of Anatomy & Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - N Del Mar
- Department of Anatomy & Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - H B Wang
- Department of Anatomy & Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - I M Johnson
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - K R Jones
- Department of Molecular, Cellular, & Developmental Biology, 347 UCB, University of Colorado, Boulder, CO 80309, United States
| | - A Reiner
- Department of Anatomy & Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States; Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States.
| |
Collapse
|
552
|
Emborg ME. Nonhuman Primate Models of Neurodegenerative Disorders. ILAR J 2017; 58:190-201. [PMID: 28985333 PMCID: PMC5886328 DOI: 10.1093/ilar/ilx021] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 05/18/2017] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's (AD), Huntington's (HD), and Parkinson's (PD) disease are age-related neurodegenerative disorders characterized by progressive neuronal cell death. Although each disease has particular pathologies and symptoms, accumulated evidence points to similar mechanisms of neurodegeneration, including inflammation, oxidative stress, and protein aggregation. A significant body of research is ongoing to understand how these pathways affect each other and what ultimately triggers the onset of the disease. Experiments in nonhuman primates (NHPs) account for only 5% of all research in animals. Yet the impact of NHP studies for clinical translation is much greater, especially for neurodegenerative disorders, as NHPs have a complex cognitive and motor functions and highly developed neuroanatomy. New NHP models are emerging to better understand pathology and improve the platform in which to test novel therapies. The goal of this report is to review NHP models of AD, HD, and PD in the context of the current understanding of these diseases and their contribution to the development of novel therapies.
Collapse
Affiliation(s)
- Marina E Emborg
- Marina E. Emborg, MD, PhD, is the director of the Preclinical Parkinson’s Research Program at the Wisconsin National Primate Research Center and an associate professor in the department of Medical Physics at the University of Wisconsin in Madison, Wisconsin.
| |
Collapse
|
553
|
Croese T, Furlan R. Extracellular vesicles in neurodegenerative diseases. Mol Aspects Med 2017; 60:52-61. [PMID: 29137922 DOI: 10.1016/j.mam.2017.11.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/31/2017] [Accepted: 11/10/2017] [Indexed: 12/22/2022]
Abstract
Extracellular vesicles (EVs) are released by all neural cells, including neurons, oligodendrocytes, astrocytes, and microglia. The lack of adequate technology has not halted neuroscientists from investigating EVs as a mean to decipher neurodegenerative disorders, still in search of comprehensible pathogenic mechanisms and efficient treatment. EVs are thought to be one of ways neurodegenerative pathologies spread in the brain, but also one of the ways the brain tries to displace toxic proteins, making their meaning in pathogenesis uncertain. EVs, however do reach biological fluids where they can be analyzed, and might therefore constitute clinically decisive biomarkers for neurodegenerative diseases in the future. Finally, if they constitute a physiological inter-cell communication system, they may represent also a very specific drug delivery tool for a difficult target such as the brain. We try to resume here available information on the role of EVs in neurodegeneration, with a special focus on Alzheimer's disease, progressive multiple sclerosis, amyotrophic lateral sclerosis, and Huntington's disease.
Collapse
Affiliation(s)
- Tommaso Croese
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Roberto Furlan
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
554
|
Veldman MB, Yang XW. Molecular insights into cortico-striatal miscommunications in Huntington's disease. Curr Opin Neurobiol 2017; 48:79-89. [PMID: 29125980 DOI: 10.1016/j.conb.2017.10.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 10/18/2017] [Indexed: 12/12/2022]
Abstract
Huntington's disease (HD), a dominantly inherited neurodegenerative disease, is defined by its genetic cause, a CAG-repeat expansion in the HTT gene, its motor and psychiatric symptomology and primary loss of striatal medium spiny neurons (MSNs). However, the molecular mechanisms from genetic lesion to disease phenotype remain largely unclear. Mouse models of HD have been created that exhibit phenotypes partially recapitulating those in the patient, and specifically, cortico-striatal disconnectivity appears to be a shared pathogenic event shared by HD mouse models and patients. Molecular studies have begun to unveil converging molecular and cellular pathogenic mechanisms that may account for cortico-striatal miscommunication in various HD mouse models. Systems biological approaches help to illuminate synaptic molecular networks as a nexus for HD cortio-striatal pathogenesis, and may offer new candidate targets to modify the disease.
Collapse
Affiliation(s)
- Matthew B Veldman
- Center for Neurobehavioral Genetics and Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
| | - X William Yang
- Center for Neurobehavioral Genetics and Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States.
| |
Collapse
|
555
|
Abstract
A hallmark of Huntington's disease is the presence of intracellular aggregates of mutant huntingtin, the pathological significance of which has long been debated. Using cryo-electron tomography, Bauerlein et al. reveal the fibrillary structure of huntingtin aggregates in situ and show that huntingtin fibrils interact with the endoplasmic reticulum, distorting its morphology and dynamics.
Collapse
Affiliation(s)
- Pedro Guedes-Dias
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA.
| |
Collapse
|
556
|
Wild EJ, Tabrizi SJ. Therapies targeting DNA and RNA in Huntington's disease. Lancet Neurol 2017; 16:837-847. [PMID: 28920889 PMCID: PMC5604739 DOI: 10.1016/s1474-4422(17)30280-6] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/23/2017] [Accepted: 07/12/2017] [Indexed: 01/12/2023]
Abstract
No disease-slowing treatment exists for Huntington's disease, but its monogenic inheritance makes it an appealing candidate for the development of therapies targeting processes close to its genetic cause. Huntington's disease is caused by CAG repeat expansions in the HTT gene, which encodes the huntingtin protein; development of therapies to target HTT transcription and the translation of its mRNA is therefore an area of intense investigation. Huntingtin-lowering strategies include antisense oligonucleotides and RNA interference targeting mRNA, and zinc finger transcriptional repressors and CRISPR-Cas9 methods aiming to reduce transcription by targeting DNA. An intrathecally delivered antisense oligonucleotide that aims to lower huntingtin is now well into its first human clinical trial, with other antisense oligonucleotides expected to enter trials in the next 1-2 years and virally delivered RNA interference and zinc finger transcriptional repressors in advanced testing in animal models. Recent advances in the design and delivery of therapies to target HTT RNA and DNA are expected to improve their efficacy, safety, tolerability, and duration of effect in future studies.
Collapse
Affiliation(s)
- Edward J Wild
- Huntington's Disease Centre, University College London Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK.
| | - Sarah J Tabrizi
- Huntington's Disease Centre, University College London Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| |
Collapse
|
557
|
Whittaker DS, Wang H, Loh DH, Cachope R, Colwell CS. Possible use of a H3R antagonist for the management of nonmotor symptoms in the Q175 mouse model of Huntington's disease. Pharmacol Res Perspect 2017; 5:e00344. [PMID: 28971617 PMCID: PMC5625154 DOI: 10.1002/prp2.344] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 07/11/2017] [Accepted: 07/14/2017] [Indexed: 12/12/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant, neurodegenerative disorder characterized by motor as well as nonmotor symptoms for which there is currently no cure. The Q175 mouse model of HD recapitulates many of the symptoms identified in HD patients including disruptions of the sleep/wake cycle. In this study, we sought to determine if the daily administration of the histamine-3 receptor (H3R) antagonist/inverse agonist 6-[(3-cyclobutyl-2,3,4,5-tetrahydro-1H-3-benzazepin-7-yl)oxy]-N-methyl-3-pyridinecarboxamide hydrochloride (GSK189254) would improve nonmotor symptoms in the Q175 line. This class of drugs acts on autoreceptors found at histaminergic synapses and results in increased levels of histamine (HA). HA is a neuromodulator whose levels vary with a daily rhythm with peak release during the active cycle and relatively lower levels during sleep. H3Rs are widely expressed in brain regions involved in cognitive processes and activation of these receptors promotes wakefulness. We administered GSK189254 nightly to homozygote and heterozygote Q175 mice for 4 weeks and confirmed that the plasma levels of the drug were elevated to a therapeutic range. We demonstrate that daily treatment with GSK189254 improved several behavioral measures in the Q175 mice including strengthening activity rhythms, cognitive performance and mood as measured by the tail suspension test. The treatment also reduced inappropriate activity during the normal sleep time. The drug treatment did not alter motor performance and coordination as measured by the challenging beam test. Our findings suggest that drugs targeting the H3R system may show benefits as cognitive enhancers in the management of HD.
Collapse
Affiliation(s)
- Daniel S. Whittaker
- Department of Psychiatry & Biobehavioral SciencesUniversity of CaliforniaLos AngelesCalifornia90095‐1751
| | - Huei‐Bin Wang
- Department of Psychiatry & Biobehavioral SciencesUniversity of CaliforniaLos AngelesCalifornia90095‐1751
| | - Dawn H. Loh
- Department of Psychiatry & Biobehavioral SciencesUniversity of CaliforniaLos AngelesCalifornia90095‐1751
| | - Roger Cachope
- CHDI Foundation6080 Center DriveSuite 100Los AngelesCalifornia90045
| | - Christopher S. Colwell
- Department of Psychiatry & Biobehavioral SciencesUniversity of CaliforniaLos AngelesCalifornia90095‐1751
| |
Collapse
|
558
|
Brandi V, Di Lella V, Marino M, Ascenzi P, Polticelli F. A comprehensive in silico analysis of huntingtin and its interactome. J Biomol Struct Dyn 2017; 36:3155-3171. [PMID: 28920551 DOI: 10.1080/07391102.2017.1381646] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
A polyglutamine expansion of the N-terminal region of huntingtin (Htt) causes Huntington's disease, a severe neurodegenerative disorder. Htt huge multidomain structure, the presence of disordered regions, and the lack of sequence homologs of known structure, so far prevented structural studies of Htt, making the study of its structure-function relationships very difficult. In this work, the presence and location of five Htt ordered domains (named from Hunt1 to Hunt5) has been detected and the structure of these domains has been predicted for the first time using a combined threading/ab initio modeling approach. This work has led to the identification of a previously undetected HEAT repeats region in the Hunt3 domain. Furthermore, a putative function has been assigned to four out of the five domains. Hunt1 and Hunt5, displaying structural similarity with the regulatory subunit A of protein phosphatase 2A, are predicted to play a role in regulating the phosphorylation status of cellular proteins. Hunt2 and Hunt3 are predicted to be homologs of two yeast importins and to mediate vescicles transport and protein trafficking. Finally, a comprehensive analysis of the Htt interactome has been carried out and is discussed to provide a global picture of the Htt's structure-function relationships.
Collapse
Affiliation(s)
- Valentina Brandi
- a Department of Sciences , Roma Tre University, Viale Guglielmo Marconi 446 , Roma I-00146 , Italy
| | - Valentina Di Lella
- a Department of Sciences , Roma Tre University, Viale Guglielmo Marconi 446 , Roma I-00146 , Italy
| | - Maria Marino
- a Department of Sciences , Roma Tre University, Viale Guglielmo Marconi 446 , Roma I-00146 , Italy
| | - Paolo Ascenzi
- a Department of Sciences , Roma Tre University, Viale Guglielmo Marconi 446 , Roma I-00146 , Italy.,b Interdepartmental Laboratory for Electron Microscopy , Roma Tre University , Roma I-00146 , Italy
| | - Fabio Polticelli
- a Department of Sciences , Roma Tre University, Viale Guglielmo Marconi 446 , Roma I-00146 , Italy.,c National Institute of Nuclear Physics , Roma Tre University, Roma Tre Section , Roma I-00146 , Italy
| |
Collapse
|
559
|
Arbez N, Ratovitski T, Roby E, Chighladze E, Stewart JC, Ren M, Wang X, Lavery DJ, Ross CA. Post-translational modifications clustering within proteolytic domains decrease mutant huntingtin toxicity. J Biol Chem 2017; 292:19238-19249. [PMID: 28972180 DOI: 10.1074/jbc.m117.782300] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 09/18/2017] [Indexed: 01/09/2023] Open
Abstract
Huntington's disease (HD) is caused in large part by a polyglutamine expansion within the huntingtin (Htt) protein. Post-translational modifications (PTMs) control and regulate many protein functions and cellular pathways, and PTMs of mutant Htt are likely important modulators of HD pathogenesis. Alterations of selected numbers of PTMs of Htt fragments have been shown to modulate Htt cellular localization and toxicity. In this study, we systematically introduced site-directed alterations in individual phosphorylation and acetylation sites in full-length Htt constructs. The effects of each of these PTM alteration constructs were tested on cell toxicity using our nuclear condensation assay and on mitochondrial viability by measuring mitochondrial potential and size. Using these functional assays in primary neurons, we identified several PTMs whose alteration can block neuronal toxicity and prevent potential loss and swelling of the mitochondria caused by mutant Htt. These PTMs included previously described sites such as serine 116 and newly found sites such as serine 2652 throughout the protein. We found that these functionally relevant sites are clustered in protease-sensitive domains throughout full-length Htt. These findings advance our understanding of the Htt PTM code and its role in HD pathogenesis. Because PTMs are catalyzed by enzymes, the toxicity-modulating Htt PTMs identified here may be promising therapeutic targets for managing HD.
Collapse
Affiliation(s)
- Nicolas Arbez
- From the Division of Neurobiology, Department of Psychiatry and Behavioral Sciences,
| | - Tamara Ratovitski
- From the Division of Neurobiology, Department of Psychiatry and Behavioral Sciences
| | - Elaine Roby
- From the Division of Neurobiology, Department of Psychiatry and Behavioral Sciences
| | - Ekaterine Chighladze
- From the Division of Neurobiology, Department of Psychiatry and Behavioral Sciences
| | - Jacqueline C Stewart
- From the Division of Neurobiology, Department of Psychiatry and Behavioral Sciences
| | - Mark Ren
- the Department of Neurobiology and Behavior, Cornell University, Ithaca, New York 14853, and
| | - Xiaofang Wang
- From the Division of Neurobiology, Department of Psychiatry and Behavioral Sciences
| | - Daniel J Lavery
- the CHDI Foundation/CHDI Management Inc., Princeton, New Jersey 08540
| | - Christopher A Ross
- From the Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, .,the Department of Neurology and Program in Cellular and Molecular Medicine, and.,the Departments of Pharmacology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| |
Collapse
|
560
|
Lee H, Shin EA, Lee JH, Ahn D, Kim CG, Kim JH, Kim SH. Caspase inhibitors: a review of recently patented compounds (2013-2015). Expert Opin Ther Pat 2017; 28:47-59. [DOI: 10.1080/13543776.2017.1378426] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Hyemin Lee
- Cancer Molecular Targeted Herbal Research Center, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Eun Ah Shin
- Cancer Molecular Targeted Herbal Research Center, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Jae Hee Lee
- Department of East West Medical Science, Graduate School of East West Medical Science Kyung Hee University, Yongin, South Korea
| | - Deoksoo Ahn
- Cancer Molecular Targeted Herbal Research Center, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Chang Geun Kim
- Cancer Molecular Targeted Herbal Research Center, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Ju-Ha Kim
- Cancer Molecular Targeted Herbal Research Center, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Sung-Hoon Kim
- Cancer Molecular Targeted Herbal Research Center, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
561
|
Mutant Huntingtin Inhibits αB-Crystallin Expression and Impairs Exosome Secretion from Astrocytes. J Neurosci 2017; 37:9550-9563. [PMID: 28893927 DOI: 10.1523/jneurosci.1418-17.2017] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 08/23/2017] [Accepted: 08/25/2017] [Indexed: 12/11/2022] Open
Abstract
In the brain, astrocytes secrete diverse substances that regulate neuronal function and viability. Exosomes, which are vesicles produced through the formation of multivesicular bodies and their subsequent fusion with the plasma membrane, are also released from astrocytes via exocytotic secretion. Astrocytic exosomes carry heat shock proteins that can reduce the cellular toxicity of misfolded proteins and prevent neurodegeneration. Although mutant huntingtin (mHtt) affects multiple functions of astrocytes, it remains unknown whether mHtt impairs the production of exosomes from astrocytes. We found that mHtt is not present in astrocytic exosomes, but can decrease exosome secretion from astrocytes in HD140Q knock-in (KI) mice. N-terminal mHtt accumulates in the nuclei and forms aggregates, causing decreased secretion of exosomes from cultured astrocytes. Consistently, there is a significant decrease in secreted exosomes in both female and male HD KI mouse striatum in which abundant nuclear mHtt aggregates are present. Conversely, injection of astrocytic exosomes into the striatum of HD140Q KI mice reduces the density of mHtt aggregates. Further, mHtt in astrocytes decreased the expression of αB-crystallin, a small heat shock protein that is enriched in astrocytes and mediates exosome secretion, by reducing the association of Sp1 with the enhancer of the αB-crystallin gene. Importantly, overexpression of αB-crystallin rescues defective exosome release from HD astrocytes as well as mHtt aggregates in the striatum of HD140Q KI mice. Our results demonstrate that mHtt reduces the expression of αB-crystallin in astrocytes to decrease exosome secretion in the HD brains, contributing to non-cell-autonomous neurotoxicity in HD.SIGNIFICANCE STATEMENT Huntington's disease (HD) is characterized by selective neurodegeneration that preferentially occurs in the striatal medium spiny neurons. Recent studies in different HD mouse models demonstrated that dysfunction of astrocytes, a major type of glial cell, leads to neuronal vulnerability. Emerging evidence shows that exosomes secreted from astrocytes contain neuroprotective cargoes that could support the survival of neighboring neurons. We found that mHtt in astrocytes impairs exosome secretion by decreasing αB-crystallin, a protein that is expressed mainly in glial cells and mediates exosome secretion. Overexpression of αB-crystallin could alleviate the deficient exosome release and neuropathology in HD mice. Our results revealed a new pathological pathway that affects the critical support of glial cells to neurons in the HD brain.
Collapse
|
562
|
Merienne N, Vachey G, de Longprez L, Meunier C, Zimmer V, Perriard G, Canales M, Mathias A, Herrgott L, Beltraminelli T, Maulet A, Dequesne T, Pythoud C, Rey M, Pellerin L, Brouillet E, Perrier AL, du Pasquier R, Déglon N. The Self-Inactivating KamiCas9 System for the Editing of CNS Disease Genes. Cell Rep 2017; 20:2980-2991. [DOI: 10.1016/j.celrep.2017.08.075] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 07/14/2017] [Accepted: 08/23/2017] [Indexed: 10/18/2022] Open
|
563
|
Ratovitski T, O’Meally RN, Jiang M, Chaerkady R, Chighladze E, Stewart JC, Wang X, Arbez N, Roby E, Alexandris A, Duan W, Vijayvargia R, Seong IS, Lavery DJ, Cole RN, Ross CA. Post-Translational Modifications (PTMs), Identified on Endogenous Huntingtin, Cluster within Proteolytic Domains between HEAT Repeats. J Proteome Res 2017; 16:2692-2708. [PMID: 28653853 PMCID: PMC5560079 DOI: 10.1021/acs.jproteome.6b00991] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Post-translational modifications (PTMs) of proteins regulate various cellular processes. PTMs of polyglutamine-expanded huntingtin (Htt) protein, which causes Huntington's disease (HD), are likely modulators of HD pathogenesis. Previous studies have identified and characterized several PTMs on exogenously expressed Htt fragments, but none of them were designed to systematically characterize PTMs on the endogenous full-length Htt protein. We found that full-length endogenous Htt, which was immunoprecipitated from HD knock-in mouse and human post-mortem brain, is suitable for detection of PTMs by mass spectrometry. Using label-free and mass tag labeling-based approaches, we identified near 40 PTMs, of which half are novel (data are available via ProteomeXchange with identifier PXD005753). Most PTMs were located in clusters within predicted unstructured domains rather than within the predicted α-helical structured HEAT repeats. Using quantitative mass spectrometry, we detected significant differences in the stoichiometry of several PTMs between HD and WT mouse brain. The mass-spectrometry identification and quantitation were verified using phospho-specific antibodies for selected PTMs. To further validate our findings, we introduced individual PTM alterations within full-length Htt and identified several PTMs that can modulate its subcellular localization in striatal cells. These findings will be instrumental in further assembling the Htt PTM framework and highlight several PTMs as potential therapeutic targets for HD.
Collapse
Affiliation(s)
- Tamara Ratovitski
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 8-121, Baltimore, Maryland 21287, United States
| | - Robert N. O’Meally
- Mass Spectrometry and Proteomics Facility, Department of Biological Chemistry, Johns Hopkins University School of Medicine, 733 North Broadway Street, Suite 371 BRB, Baltimore, Maryland 21287, United States
| | - Mali Jiang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 8-121, Baltimore, Maryland 21287, United States
| | - Raghothama Chaerkady
- Mass Spectrometry and Proteomics Facility, Department of Biological Chemistry, Johns Hopkins University School of Medicine, 733 North Broadway Street, Suite 371 BRB, Baltimore, Maryland 21287, United States
| | - Ekaterine Chighladze
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 8-121, Baltimore, Maryland 21287, United States
| | - Jacqueline C. Stewart
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 8-121, Baltimore, Maryland 21287, United States
| | - Xiaofang Wang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 8-121, Baltimore, Maryland 21287, United States
| | - Nicolas Arbez
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 8-121, Baltimore, Maryland 21287, United States
| | - Elaine Roby
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 8-121, Baltimore, Maryland 21287, United States
| | - Athanasios Alexandris
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 8-121, Baltimore, Maryland 21287, United States
| | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 8-121, Baltimore, Maryland 21287, United States
- Department of Neurology and Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
- Departments of Pharmacology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Ravi Vijayvargia
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Department of Neurology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Ihn Sik Seong
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Department of Neurology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Daniel J. Lavery
- CHDI Foundation/CHDI Management, Inc., Princeton, New Jersey 08540, United States
| | - Robert N. Cole
- Mass Spectrometry and Proteomics Facility, Department of Biological Chemistry, Johns Hopkins University School of Medicine, 733 North Broadway Street, Suite 371 BRB, Baltimore, Maryland 21287, United States
| | - Christopher A. Ross
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 8-121, Baltimore, Maryland 21287, United States
- Department of Neurology and Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
- Departments of Pharmacology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| |
Collapse
|
564
|
Translating protein phosphatase research into treatments for neurodegenerative diseases. Biochem Soc Trans 2017; 45:101-112. [PMID: 28202663 DOI: 10.1042/bst20160157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/21/2016] [Accepted: 11/23/2016] [Indexed: 12/11/2022]
Abstract
Many of the major neurodegenerative disorders are characterized by the accumulation of intracellular protein aggregates in neurons and other cells in brain, suggesting that errors in protein quality control mechanisms associated with the aging process play a critical role in the onset and progression of disease. The increased understanding of the unfolded protein response (UPR) signaling network and, more specifically, the structure and function of eIF2α phosphatases has enabled the development or discovery of small molecule inhibitors that show great promise in restoring protein homeostasis and ameliorating neuronal damage and death. While this review focuses attention on one or more eIF2α phosphatases, the wide range of UPR proteins that are currently being explored as potential drug targets bodes well for the successful future development of therapies to preserve neuronal function and treat neurodegenerative disease.
Collapse
|
565
|
Interaction of misfolded proteins and mitochondria in neurodegenerative disorders. Biochem Soc Trans 2017; 45:1025-1033. [PMID: 28733489 DOI: 10.1042/bst20170024] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 06/20/2017] [Accepted: 06/23/2017] [Indexed: 12/17/2022]
Abstract
The number of the people affected by neurodegenerative disorders is growing dramatically due to the ageing of population. The major neurodegenerative diseases share some common pathological features including the involvement of mitochondria in the mechanism of pathology and misfolding and the accumulation of abnormally aggregated proteins. Neurotoxicity of aggregated β-amyloid, tau, α-synuclein and huntingtin is linked to the effects of these proteins on mitochondria. All these misfolded aggregates affect mitochondrial energy metabolism by inhibiting diverse mitochondrial complexes and limit ATP availability in neurones. β-Amyloid, tau, α-synuclein and huntingtin are shown to be involved in increased production of reactive oxygen species, which can be generated in mitochondria or can target this organelle. Most of these aggregated proteins are capable of deregulating mitochondrial calcium handling that, in combination with oxidative stress, lead to opening of the mitochondrial permeability transition pore. Despite some of the common features, aggregated β-amyloid, tau, α-synuclein and huntingtin have diverse targets in mitochondria that can partially explain neurotoxic effect of these proteins in different brain regions.
Collapse
|
566
|
Proteostasis of Huntingtin in Health and Disease. Int J Mol Sci 2017; 18:ijms18071568. [PMID: 28753941 PMCID: PMC5536056 DOI: 10.3390/ijms18071568] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/15/2017] [Accepted: 07/18/2017] [Indexed: 12/28/2022] Open
Abstract
Huntington's disease (HD) is a fatal neurodegenerative disorder characterized by motor dysfunction, cognitive deficits and psychosis. HD is caused by mutations in the Huntingtin (HTT) gene, resulting in the expansion of polyglutamine (polyQ) repeats in the HTT protein. Mutant HTT is prone to aggregation, and the accumulation of polyQ-expanded fibrils as well as intermediate oligomers formed during the aggregation process contribute to neurodegeneration. Distinct protein homeostasis (proteostasis) nodes such as chaperone-mediated folding and proteolytic systems regulate the aggregation and degradation of HTT. Moreover, polyQ-expanded HTT fibrils and oligomers can lead to a global collapse in neuronal proteostasis, a process that contributes to neurodegeneration. The ability to maintain proteostasis of HTT declines during the aging process. Conversely, mechanisms that preserve proteostasis delay the onset of HD. Here we will review the link between proteostasis, aging and HD-related changes.
Collapse
|
567
|
Dietrich P, Johnson IM, Alli S, Dragatsis I. Elimination of huntingtin in the adult mouse leads to progressive behavioral deficits, bilateral thalamic calcification, and altered brain iron homeostasis. PLoS Genet 2017; 13:e1006846. [PMID: 28715425 PMCID: PMC5536499 DOI: 10.1371/journal.pgen.1006846] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 07/31/2017] [Accepted: 06/01/2017] [Indexed: 02/07/2023] Open
Abstract
Huntington's Disease (HD) is an autosomal dominant progressive neurodegenerative disorder characterized by cognitive, behavioral and motor dysfunctions. HD is caused by a CAG repeat expansion in exon 1 of the HD gene that is translated into an expanded polyglutamine tract in the encoded protein, huntingtin (HTT). While the most significant neuropathology of HD occurs in the striatum, other brain regions are also affected and play an important role in HD pathology. To date there is no cure for HD, and recently strategies aiming at silencing HTT expression have been initiated as possible therapeutics for HD. However, the essential functions of HTT in the adult brain are currently unknown and hence the consequence of sustained suppression of HTT expression is unpredictable and can potentially be deleterious. Using the Cre-loxP system of recombination, we conditionally inactivated the mouse HD gene homologue at 3, 6 and 9 months of age. Here we show that elimination of Htt expression in the adult mouse results in behavioral deficits, progressive neuropathological changes including bilateral thalamic calcification, and altered brain iron homeostasis.
Collapse
Affiliation(s)
- Paula Dietrich
- Department of Physiology, The University of Tennessee, Health Science Center, Memphis, Tennessee, United States of America
| | - Irudayam Maria Johnson
- Department of Physiology, The University of Tennessee, Health Science Center, Memphis, Tennessee, United States of America
| | - Shanta Alli
- Department of Physiology, The University of Tennessee, Health Science Center, Memphis, Tennessee, United States of America
| | - Ioannis Dragatsis
- Department of Physiology, The University of Tennessee, Health Science Center, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
568
|
Fiocchetti M, Cipolletti M, Brandi V, Polticelli F, Ascenzi P. Neuroglobin and friends. J Mol Recognit 2017; 30. [DOI: 10.1002/jmr.2654] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 06/05/2017] [Accepted: 06/14/2017] [Indexed: 01/02/2023]
Affiliation(s)
| | | | | | - Fabio Polticelli
- Dipartimento di Scienze; Università Roma Tre; Rome Italy
- Istituto Nazionale di Fisica Nucleare; Sezione dell'Università Roma Tre; Rome Italy
| | - Paolo Ascenzi
- Laboratorio Interdipartimentale di Microscopia Elettronica; Università Roma Tre; Rome Italy
| |
Collapse
|
569
|
Defective Sphingosine-1-phosphate metabolism is a druggable target in Huntington's disease. Sci Rep 2017; 7:5280. [PMID: 28706199 PMCID: PMC5509685 DOI: 10.1038/s41598-017-05709-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 06/01/2017] [Indexed: 12/22/2022] Open
Abstract
Huntington’s disease is characterized by a complex and heterogeneous pathogenic profile. Studies have shown that disturbance in lipid homeostasis may represent a critical determinant in the progression of several neurodegenerative disorders. The recognition of perturbed lipid metabolism is only recently becoming evident in HD. In order to provide more insight into the nature of such a perturbation and into the effect its modulation may have in HD pathology, we investigated the metabolism of Sphingosine-1-phosphate (S1P), one of the most important bioactive lipids, in both animal models and patient samples. Here, we demonstrated that S1P metabolism is significantly disrupted in HD even at early stage of the disease and importantly, we revealed that such a dysfunction represents a common denominator among multiple disease models ranging from cells to humans through mouse models. Interestingly, the in vitro anti-apoptotic and the pro-survival actions seen after modulation of S1P-metabolizing enzymes allows this axis to emerge as a new druggable target and unfolds its promising therapeutic potential for the development of more effective and targeted interventions against this incurable condition.
Collapse
|
570
|
Shrivastava AN, Aperia A, Melki R, Triller A. Physico-Pathologic Mechanisms Involved in Neurodegeneration: Misfolded Protein-Plasma Membrane Interactions. Neuron 2017; 95:33-50. [DOI: 10.1016/j.neuron.2017.05.026] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 05/12/2017] [Accepted: 05/19/2017] [Indexed: 12/12/2022]
|
571
|
Pepeu G, Grazia Giovannini M. The fate of the brain cholinergic neurons in neurodegenerative diseases. Brain Res 2017; 1670:173-184. [PMID: 28652219 DOI: 10.1016/j.brainres.2017.06.023] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 06/21/2017] [Accepted: 06/22/2017] [Indexed: 01/03/2023]
Abstract
The aims of this review are: 1) to describe which cholinergic neurons are affected in brain neurodegenerative diseases leading to dementia; 2) to discuss the possible causes of the degeneration of the cholinergic neurons, 3) to summarize the functional consequences of the cholinergic deficit. The brain cholinergic system is basically constituted by three populations of phenotypically similar neurons forming a series of basal forebrain nuclei, the midpontine nuclei and a large population of striatal interneurons. In Alzheimer's disease there is an extensive loss of forebrain cholinergic neurons accompanied by a reduction of the cholinergic fiber network of the cortical mantel and hippocampus. The midpontine cholinergic nuclei are spared. The same situation occurs in the corticobasal syndrome and dementia following alcohol abuse and traumatic brain injury. Conversely, in Parkinson's disease, the midpontine nuclei degenerate, together with the dopaminergic nuclei, reducing the cholinergic input to thalamus and forebrain whereas the forebrain cholinergic neurons are spared. In Parkinson's disease with dementia, Lewis Body Dementia and Parkinsonian syndromes both groups of forebrain and midpontine cholinergic nuclei degenerate. In Huntington's disease a dysfunction of the striatal cholinergic interneurons without cell loss takes place. The formation and accumulation of misfolded proteins such as β-amyloid oligomers and plaques, tau protein tangles and α-synuclein clumps, and aggregated mutated huntingtin play a crucial role in the neuronal degeneration by direct cellular toxicity of the misfolded proteins and through the toxic compounds resulting from an extensive inflammatory reaction. Evidences indicate that β-amyloid disrupts NGF metabolism causing the degeneration of the cholinergic neurons which depend on NGF for their survival, namely the forebrain cholinergic neurons, sparing the midpontine and striatal neurons which express no specific NGF receptors. It is feasible that the latter cholinergic neurons may be damaged by direct toxicity of tau, α-synuclein and inflammations products through mechanisms not fully understood. Attention and learning and memory impairment are the functional consequences of the forebrain cholinergic neuron dysfunction, whereas the loss of midpontine cholinergic neurons results primarily in motor and sleep disturbances.
Collapse
Affiliation(s)
- Giancarlo Pepeu
- Department of Health Sciences, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy.
| | - Maria Grazia Giovannini
- Department of Health Sciences, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy.
| |
Collapse
|
572
|
Cambon K, Zimmer V, Martineau S, Gaillard MC, Jarrige M, Bugi A, Miniarikova J, Rey M, Hassig R, Dufour N, Auregan G, Hantraye P, Perrier AL, Déglon N. Preclinical Evaluation of a Lentiviral Vector for Huntingtin Silencing. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 5:259-276. [PMID: 28603746 PMCID: PMC5453866 DOI: 10.1016/j.omtm.2017.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 05/07/2017] [Indexed: 01/12/2023]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder resulting from a polyglutamine expansion in the huntingtin (HTT) protein. There is currently no cure for this disease, but recent studies suggest that RNAi to downregulate the expression of both normal and mutant HTT is a promising therapeutic approach. We previously developed a small hairpin RNA (shRNA), vectorized in an HIV-1-derived lentiviral vector (LV), that reduced pathology in an HD rodent model. Here, we modified this vector for preclinical development by using a tat-independent third-generation LV (pCCL) backbone and removing the original reporter genes. We demonstrate that this novel vector efficiently downregulated HTT expression in vitro in striatal neurons derived from induced pluripotent stem cells (iPSCs) of HD patients. It reduced two major pathological HD hallmarks while triggering a minimal inflammatory response, up to 6 weeks after injection, when administered by stereotaxic surgery in the striatum of an in vivo rodent HD model. Further assessment of this shRNA vector in vitro showed proper processing by the endogenous silencing machinery, and we analyzed gene expression changes to identify potential off-targets. These preclinical data suggest that this new shRNA vector fulfills primary biosafety and efficiency requirements for further development in the clinic as a cure for HD.
Collapse
Affiliation(s)
- Karine Cambon
- CEA, DRF, Institute of Biology Francois Jacob, Molecular Imaging Research Center, F-92265 Fontenay-aux-Roses, France
- CNRS, CEA, Paris-Sud University, University Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265 Fontenay-aux-Roses, France
| | - Virginie Zimmer
- Department of Clinical Neurosciences, Laboratory of Cellular and Molecular Neurotherapies, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
- Neuroscience Research Center, Laboratory of Cellular and Molecular Neurotherapies, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Sylvain Martineau
- CEA, DRF, Institute of Biology Francois Jacob, Molecular Imaging Research Center, F-92265 Fontenay-aux-Roses, France
- CNRS, CEA, Paris-Sud University, University Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265 Fontenay-aux-Roses, France
| | - Marie-Claude Gaillard
- CEA, DRF, Institute of Biology Francois Jacob, Molecular Imaging Research Center, F-92265 Fontenay-aux-Roses, France
- CNRS, CEA, Paris-Sud University, University Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265 Fontenay-aux-Roses, France
| | - Margot Jarrige
- Institut National de la Santé et de la Recherche Médicale UMR861, I-Stem, AFM, 91100 Corbeil-Essonnes, France
- UEVE UMR861, I-STEM, AFM, 91100 Corbeil-Essonnes, France
- CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France
| | - Aurore Bugi
- CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France
| | - Jana Miniarikova
- Department of Research & Development, uniQure, 1105 Amsterdam, the Netherlands
| | - Maria Rey
- Department of Clinical Neurosciences, Laboratory of Cellular and Molecular Neurotherapies, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
- Neuroscience Research Center, Laboratory of Cellular and Molecular Neurotherapies, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Raymonde Hassig
- CEA, DRF, Institute of Biology Francois Jacob, Molecular Imaging Research Center, F-92265 Fontenay-aux-Roses, France
- CNRS, CEA, Paris-Sud University, University Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265 Fontenay-aux-Roses, France
| | - Noelle Dufour
- CEA, DRF, Institute of Biology Francois Jacob, Molecular Imaging Research Center, F-92265 Fontenay-aux-Roses, France
- CNRS, CEA, Paris-Sud University, University Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265 Fontenay-aux-Roses, France
| | - Gwenaelle Auregan
- CEA, DRF, Institute of Biology Francois Jacob, Molecular Imaging Research Center, F-92265 Fontenay-aux-Roses, France
- CNRS, CEA, Paris-Sud University, University Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265 Fontenay-aux-Roses, France
| | - Philippe Hantraye
- CEA, DRF, Institute of Biology Francois Jacob, Molecular Imaging Research Center, F-92265 Fontenay-aux-Roses, France
- CNRS, CEA, Paris-Sud University, University Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265 Fontenay-aux-Roses, France
| | - Anselme L. Perrier
- Institut National de la Santé et de la Recherche Médicale UMR861, I-Stem, AFM, 91100 Corbeil-Essonnes, France
- UEVE UMR861, I-STEM, AFM, 91100 Corbeil-Essonnes, France
| | - Nicole Déglon
- Department of Clinical Neurosciences, Laboratory of Cellular and Molecular Neurotherapies, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
- Neuroscience Research Center, Laboratory of Cellular and Molecular Neurotherapies, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
- Corresponding author: Nicole Déglon, Lausanne University Hospital (CHUV), Laboratory of Cellular and Molecular Neurotherapies (LNCM), Pavillon 3, Avenue de Beaumont, 1011 Lausanne, Switzerland.
| |
Collapse
|
573
|
Kim KH, Lee D, Lee HL, Kim CE, Jung K, Kang KS. Beneficial effects of Panax ginseng for the treatment and prevention of neurodegenerative diseases: past findings and future directions. J Ginseng Res 2017; 42:239-247. [PMID: 29989012 PMCID: PMC6035378 DOI: 10.1016/j.jgr.2017.03.011] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 03/15/2017] [Indexed: 01/20/2023] Open
Abstract
In recent years, several therapeutic drugs have been rationally designed and synthesized based on the novel knowledge gained from investigating the actions of biologically active chemicals derived from foods, plants, and medicinal herbs. One of the major advantages of these naturalistic chemicals is their ability to interact with multiple targets in the body resulting in a combined beneficial effect. Ginseng is a perennial herb (Araliaceae family), a species within the genus Panax, and a highly valued and popular medicinal plant. Evidence for the medicinal and health benefits of Panax ginseng and its components in preventing neurodegeneration has increased significantly in the past decade. The beneficial effects of P. ginseng on neurodegenerative diseases have been attributed primarily to the antioxidative and immunomodulatory activities of its ginsenoside components. Mechanistic studies on the neuroprotective effects of ginsenosides revealed that they act not only as antioxidants but also as modulators of intracellular neuronal signaling and metabolism, cell survival/death genes, and mitochondrial function. The goal of the present paper is to provide a brief review of recent knowledge and developments concerning the beneficial effects as well as the mechanism of action of P. ginseng and its components in the treatment and prevention of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Dahae Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hye Lim Lee
- College of Korean Medicine, Gachon University, Seongnam, Republic of Korea
| | - Chang-Eop Kim
- College of Korean Medicine, Gachon University, Seongnam, Republic of Korea
| | - Kiwon Jung
- Institute of Pharmaceutical Sciences, College of Pharmacy, CHA University, Sungnam, Republic of Korea
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam, Republic of Korea
| |
Collapse
|
574
|
Aggregation landscapes of Huntingtin exon 1 protein fragments and the critical repeat length for the onset of Huntington's disease. Proc Natl Acad Sci U S A 2017; 114:4406-4411. [PMID: 28400517 DOI: 10.1073/pnas.1702237114] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disease caused by an abnormal expansion in the polyglutamine (polyQ) track of the Huntingtin (HTT) protein. The severity of the disease depends on the polyQ repeat length, arising only in patients with proteins having 36 repeats or more. Previous studies have shown that the aggregation of N-terminal fragments (encoded by HTT exon 1) underlies the disease pathology in mouse models and that the HTT exon 1 gene product can self-assemble into amyloid structures. Here, we provide detailed structural mechanisms for aggregation of several protein fragments encoded by HTT exon 1 by using the associative memory, water-mediated, structure and energy model (AWSEM) to construct their free energy landscapes. We find that the addition of the N-terminal 17-residue sequence ([Formula: see text]) facilitates polyQ aggregation by encouraging the formation of prefibrillar oligomers, whereas adding the C-terminal polyproline sequence ([Formula: see text]) inhibits aggregation. The combination of both terminal additions in HTT exon 1 fragment leads to a complex aggregation mechanism with a basic core that resembles that found for the aggregation of pure polyQ repeats using AWSEM. At the extrapolated physiological concentration, although the grand canonical free energy profiles are uphill for HTT exon 1 fragments having 20 or 30 glutamines, the aggregation landscape for fragments with 40 repeats has become downhill. This computational prediction agrees with the critical length found for the onset of HD and suggests potential therapies based on blocking early binding events involving the terminal additions to the polyQ repeats.
Collapse
|
575
|
Plaza-Zabala A, Sierra-Torre V, Sierra A. Autophagy and Microglia: Novel Partners in Neurodegeneration and Aging. Int J Mol Sci 2017; 18:E598. [PMID: 28282924 PMCID: PMC5372614 DOI: 10.3390/ijms18030598] [Citation(s) in RCA: 217] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 02/28/2017] [Accepted: 03/05/2017] [Indexed: 02/07/2023] Open
Abstract
Autophagy is emerging as a core regulator of Central Nervous System (CNS) aging and neurodegeneration. In the brain, it has mostly been studied in neurons, where the delivery of toxic molecules and organelles to the lysosome by autophagy is crucial for neuronal health and survival. However, we propose that the (dys)regulation of autophagy in microglia also affects innate immune functions such as phagocytosis and inflammation, which in turn contribute to the pathophysiology of aging and neurodegenerative diseases. Herein, we first describe the basic concepts of autophagy and its regulation, discuss key aspects for its accurate monitoring at the experimental level, and summarize the evidence linking autophagy impairment to CNS senescence and disease. We focus on acute, chronic, and autoimmunity-mediated neurodegeneration, including ischemia/stroke, Alzheimer's, Parkinson's, and Huntington's diseases, and multiple sclerosis. Next, we describe the actual and potential impact of autophagy on microglial phagocytic and inflammatory function. Thus, we provide evidence of how autophagy may affect microglial phagocytosis of apoptotic cells, amyloid-β, synaptic material, and myelin debris, and regulate the progression of age-associated neurodegenerative diseases. We also discuss data linking autophagy to the regulation of the microglial inflammatory phenotype, which is known to contribute to age-related brain dysfunction. Overall, we update the current knowledge of autophagy and microglia, and highlight as yet unexplored mechanisms whereby autophagy in microglia may contribute to CNS disease and senescence.
Collapse
Affiliation(s)
| | | | - Amanda Sierra
- Achucarro Basque Center for Neuroscience, 48170 Zamudio, Spain.
- Department of Neurosciences, University of the Basque Country EHU/UPV, 48940 Leioa, Spain.
- Ikerbasque Foundation, 48013 Bilbao, Spain.
| |
Collapse
|
576
|
Stopschinski BE, Diamond MI. The prion model for progression and diversity of neurodegenerative diseases. Lancet Neurol 2017; 16:323-332. [PMID: 28238712 DOI: 10.1016/s1474-4422(17)30037-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 01/12/2017] [Accepted: 01/26/2017] [Indexed: 12/12/2022]
Abstract
The neuropathology of different neurodegenerative diseases begins in different brain regions, and involves distinct brain networks. Evidence indicates that transcellular propagation of protein aggregation, which is the basis of prion disease, might underlie the progression of pathology in neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. The prion model predicts specific patterns of neuronal vulnerability and network involvement on the basis of the conformation of pathological proteins. Indeed, evidence indicates that self-propagating aggregate conformers, or so-called strains, are associated with distinct neuropathological syndromes. The extension of this hypothesis to our understanding of common neurodegenerative disorders can suggest new therapeutic approaches, such as immunotherapy and small molecules, to block transcellular propagation, and new diagnostic tools to detect early evidence of disease.
Collapse
Affiliation(s)
- Barbara E Stopschinski
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr Brain Institute, University of Texas, Southwestern Medical Center, Dallas, TX 75225, USA
| | - Marc I Diamond
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr Brain Institute, University of Texas, Southwestern Medical Center, Dallas, TX 75225, USA.
| |
Collapse
|
577
|
Yu MS, Tanese N. Huntingtin Is Required for Neural But Not Cardiac/Pancreatic Progenitor Differentiation of Mouse Embryonic Stem Cells In vitro. Front Cell Neurosci 2017; 11:33. [PMID: 28270748 PMCID: PMC5318384 DOI: 10.3389/fncel.2017.00033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 02/06/2017] [Indexed: 12/25/2022] Open
Abstract
Mutation in the huntingtin (HTT) gene causes Huntington's disease (HD). It is an autosomal dominant trinucleotide-repeat expansion disease in which CAG repeat sequence expands to >35. This results in the production of mutant HTT protein with an increased stretch of glutamines near the N-terminus. The wild type HTT gene encodes a 350 kD protein whose function remains elusive. Mutant HTT protein has been implicated in transcription, axonal transport, cytoskeletal structure/function, signal transduction, and autophagy. HD is characterized by the appearance of nuclear inclusions and degeneration of the striatum. Although HTT protein is expressed early in embryos, most patients develop symptoms in mid-life. It is also unclear why the ubiquitously expressed mutant HTT specifically causes striatal atrophy. Wild type Htt is essential for development as Htt knockout mice die at day E7.5. Increasing evidence suggests mutant Htt may alter neurogenesis and development of striatal neurons resulting in neuronal loss. Using a mouse embryonic stem cell model, we examined the role of Htt in neural differentiation. We found cells lacking Htt inefficient in generating neural stem cells. In contrast differentiation into progenitors of mesoderm and endoderm lineages was not affected. The data suggests Htt is essential for neural but not cardiac/pancreatic progenitor differentiation of embryonic stem cells in vitro.
Collapse
Affiliation(s)
- Man Shan Yu
- Department of Microbiology, New York University School of Medicine, New York NY, USA
| | - Naoko Tanese
- Department of Microbiology, New York University School of Medicine, New York NY, USA
| |
Collapse
|
578
|
Adegbuyiro A, Sedighi F, Pilkington AW, Groover S, Legleiter J. Proteins Containing Expanded Polyglutamine Tracts and Neurodegenerative Disease. Biochemistry 2017; 56:1199-1217. [PMID: 28170216 DOI: 10.1021/acs.biochem.6b00936] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Several hereditary neurological and neuromuscular diseases are caused by an abnormal expansion of trinucleotide repeats. To date, there have been 10 of these trinucleotide repeat disorders associated with an expansion of the codon CAG encoding glutamine (Q). For these polyglutamine (polyQ) diseases, there is a critical threshold length of the CAG repeat required for disease, and further expansion beyond this threshold is correlated with age of onset and symptom severity. PolyQ expansion in the translated proteins promotes their self-assembly into a variety of oligomeric and fibrillar aggregate species that accumulate into the hallmark proteinaceous inclusion bodies associated with each disease. Here, we review aggregation mechanisms of proteins with expanded polyQ-tracts, structural consequences of expanded polyQ ranging from monomers to fibrillar aggregates, the impact of protein context and post-translational modifications on aggregation, and a potential role for lipid membranes in aggregation. As the pathogenic mechanisms that underlie these disorders are often classified as either a gain of toxic function or loss of normal protein function, some toxic mechanisms associated with mutant polyQ tracts will also be discussed.
Collapse
Affiliation(s)
- Adewale Adegbuyiro
- The C. Eugene Bennett Department of Chemistry, 217 Clark Hall, West Virginia University , Morgantown, West Virginia 26506, United States
| | - Faezeh Sedighi
- The C. Eugene Bennett Department of Chemistry, 217 Clark Hall, West Virginia University , Morgantown, West Virginia 26506, United States
| | - Albert W Pilkington
- The C. Eugene Bennett Department of Chemistry, 217 Clark Hall, West Virginia University , Morgantown, West Virginia 26506, United States
| | - Sharon Groover
- The C. Eugene Bennett Department of Chemistry, 217 Clark Hall, West Virginia University , Morgantown, West Virginia 26506, United States
| | - Justin Legleiter
- The C. Eugene Bennett Department of Chemistry, 217 Clark Hall, West Virginia University , Morgantown, West Virginia 26506, United States.,Blanchette Rockefeller Neurosciences Institute, Robert C. Byrd Health Sciences Center, P.O. Box 9304, West Virginia University , Morgantown, West Virginia 26506, United States.,NanoSAFE, P.O. Box 6223, West Virginia University , Morgantown, West Virginia 26506, United States
| |
Collapse
|
579
|
Coarelli G, Diallo A, Thion MS, Rinaldi D, Calvas F, Boukbiza OL, Tataru A, Charles P, Tranchant C, Marelli C, Ewenczyk C, Tchikviladzé M, Monin ML, Carlander B, Anheim M, Brice A, Mochel F, Tezenas du Montcel S, Humbert S, Durr A. Low cancer prevalence in polyglutamine expansion diseases. Neurology 2017; 88:1114-1119. [DOI: 10.1212/wnl.0000000000003725] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 11/10/2016] [Indexed: 12/24/2022] Open
Abstract
Objective:Polyglutamine (PolyQ) diseases are dominantly transmitted neurologic disorders, caused by coding and expanded CAG trinucleotide repeats. Cancer was reported retrospectively to be rare in patients with PolyQ diseases and we aimed to investigate its prevalence in France.Methods:Consecutive patients with Huntington disease (HD) and spinocerebellar ataxia (SCA) were questioned about cancer, cardiovascular diseases, and related risk factors in 4 university hospitals in Paris, Toulouse, Strasbourg, and Montpellier. Standardized incidence ratios (SIR), based on age- and sex-adjusted rate of the French population, were assessed for different types of cancer.Results:We questioned 372 patients with HD and 134 patients with SCA. SIR showed significantly reduced risk of cancer in HD: 23 observed cases vs 111.05 expected ones (SIR 0.21, 95% confidence interval [CI] 0.13–0.31), as well as in SCA: 7 observed cases vs 34.73 expected (SIR 0.23, 95% CI 0.08–0.42). This was surprising since risk behavior for cancer was increased in these patients, with significantly greater tobacco and alcohol consumption in patients with HD vs patients with SCA (p < 0.0056). There was no association between CAG repeat size and cancer or cardiovascular disease. However, in patients with HD, skin cancers were more frequent than expected (5 vs 0.98, SIR 5.11, 95% CI 1.65–11.95).Conclusions:There was a decreased cancer rate in PolyQ diseases despite high incidence of risk factors. Intriguingly, skin cancer incidence was higher, suggesting a crosstalk between neurodegeneration and skin tumorigenesis.
Collapse
|
580
|
Gomez-Pastor R, Burchfiel ET, Neef DW, Jaeger AM, Cabiscol E, McKinstry SU, Doss A, Aballay A, Lo DC, Akimov SS, Ross CA, Eroglu C, Thiele DJ. Abnormal degradation of the neuronal stress-protective transcription factor HSF1 in Huntington's disease. Nat Commun 2017; 8:14405. [PMID: 28194040 PMCID: PMC5316841 DOI: 10.1038/ncomms14405] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 12/21/2016] [Indexed: 01/26/2023] Open
Abstract
Huntington's Disease (HD) is a neurodegenerative disease caused by poly-glutamine expansion in the Htt protein, resulting in Htt misfolding and cell death. Expression of the cellular protein folding and pro-survival machinery by heat shock transcription factor 1 (HSF1) ameliorates biochemical and neurobiological defects caused by protein misfolding. We report that HSF1 is degraded in cells and mice expressing mutant Htt, in medium spiny neurons derived from human HD iPSCs and in brain samples from patients with HD. Mutant Htt increases CK2α' kinase and Fbxw7 E3 ligase levels, phosphorylating HSF1 and promoting its proteasomal degradation. An HD mouse model heterozygous for CK2α' shows increased HSF1 and chaperone levels, maintenance of striatal excitatory synapses, clearance of Htt aggregates and preserves body mass compared with HD mice homozygous for CK2α'. These results reveal a pathway that could be modulated to prevent neuronal dysfunction and muscle wasting caused by protein misfolding in HD.
Collapse
Affiliation(s)
- Rocio Gomez-Pastor
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Eileen T. Burchfiel
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Daniel W. Neef
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Alex M. Jaeger
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Elisa Cabiscol
- Departament de Ciencies Mediques Basiques, IRB Lleida, Universitat de Lleida, Lleida 25008, Spain
| | - Spencer U. McKinstry
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Argenia Doss
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Alejandro Aballay
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Donald C. Lo
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Sergey S. Akimov
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Christopher A. Ross
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- Department of Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Cagla Eroglu
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Dennis J. Thiele
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| |
Collapse
|
581
|
Datson NA, González-Barriga A, Kourkouta E, Weij R, van de Giessen J, Mulders S, Kontkanen O, Heikkinen T, Lehtimäki K, van Deutekom JCT. The expanded CAG repeat in the huntingtin gene as target for therapeutic RNA modulation throughout the HD mouse brain. PLoS One 2017; 12:e0171127. [PMID: 28182673 PMCID: PMC5300196 DOI: 10.1371/journal.pone.0171127] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 01/16/2017] [Indexed: 01/11/2023] Open
Abstract
The aim of these studies was to demonstrate the therapeutic capacity of an antisense oligonucleotide with the sequence (CUG)7 targeting the expanded CAG repeat in huntingtin (HTT) mRNA in vivo in the R6/2 N-terminal fragment and Q175 knock-in Huntington’s disease (HD) mouse models. In a first study, R6/2 mice received six weekly intracerebroventricular infusions with a low and high dose of (CUG)7 and were sacrificed 2 weeks later. A 15–60% reduction of both soluble and aggregated mutant HTT protein was observed in striatum, hippocampus and cortex of (CUG)7-treated mice. This correction at the molecular level resulted in an improvement of performance in multiple motor tasks, increased whole brain and cortical volume, reduced levels of the gliosis marker myo-inositol, increased levels of the neuronal integrity marker N-aceyl aspartate and increased mRNA levels of the striatal marker Darpp-32. These neuroanatomical and neurochemical changes, together with the improved motor performance, suggest that treatment with (CUG)7 ameliorates basal ganglia dysfunction. The HTT-lowering was confirmed by an independent study in Q175 mice using a similar (CUG)7 AON dosing regimen, further demonstrating a lasting reduction of mutant HTT protein in striatum, hippocampus and cortex for up to 18 weeks post last infusion along with an increase in motor activity. Based on these encouraging results, (CUG)7 may thus offer an interesting alternative HTT-lowering strategy for HD.
Collapse
Affiliation(s)
| | | | | | - Rudie Weij
- BioMarin Nederland BV, Leiden, The Netherlands
| | | | | | - Outi Kontkanen
- Charles River Discovery Research Services, Kuopio, Finland
| | | | | | | |
Collapse
|
582
|
Liot G, Valette J, Pépin J, Flament J, Brouillet E. Energy defects in Huntington's disease: Why “in vivo” evidence matters. Biochem Biophys Res Commun 2017; 483:1084-1095. [DOI: 10.1016/j.bbrc.2016.09.065] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 09/13/2016] [Indexed: 01/12/2023]
|
583
|
Francelle L, Lotz C, Outeiro T, Brouillet E, Merienne K. Contribution of Neuroepigenetics to Huntington's Disease. Front Hum Neurosci 2017; 11:17. [PMID: 28194101 PMCID: PMC5276857 DOI: 10.3389/fnhum.2017.00017] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 01/10/2017] [Indexed: 12/29/2022] Open
Abstract
Unbalanced epigenetic regulation is thought to contribute to the progression of several neurodegenerative diseases, including Huntington’s disease (HD), a genetic disorder considered as a paradigm of epigenetic dysregulation. In this review, we attempt to address open questions regarding the role of epigenetic changes in HD, in the light of recent advances in neuroepigenetics. We particularly discuss studies using genome-wide scale approaches that provide insights into the relationship between epigenetic regulations, gene expression and neuronal activity in normal and diseased neurons, including HD neurons. We propose that cell-type specific techniques and 3D-based methods will advance knowledge of epigenome in the context of brain region vulnerability in neurodegenerative diseases. A better understanding of the mechanisms underlying epigenetic changes and of their consequences in neurodegenerative diseases is required to design therapeutic strategies more effective than current strategies based on histone deacetylase (HDAC) inhibitors. Researches in HD may play a driving role in this process.
Collapse
Affiliation(s)
- Laetitia Francelle
- Department of NeuroDegeneration and Restorative Research, University Medical Center Goettingen Goettingen, Germany
| | - Caroline Lotz
- CNRS UMR 7364, Laboratory of Cognitive and Adaptive Neurosciences, University of Strasbourg Strasbourg, France
| | - Tiago Outeiro
- Department of NeuroDegeneration and Restorative Research, University Medical Center Goettingen Goettingen, Germany
| | - Emmanuel Brouillet
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Département de Recherche Fondamentale, Institut d'Imagerie Biomédicale, Molecular Imaging Center, Neurodegenerative diseases Laboratory, UMR 9199, CNRS Université Paris-Sud, Université Paris-Saclay Fontenay-aux-Roses, France
| | - Karine Merienne
- CNRS UMR 7364, Laboratory of Cognitive and Adaptive Neurosciences, University of Strasbourg Strasbourg, France
| |
Collapse
|
584
|
Bulgari D, Deitcher DL, Levitan ES. Loss of Huntingtin stimulates capture of retrograde dense-core vesicles to increase synaptic neuropeptide stores. Eur J Cell Biol 2017; 96:402-406. [PMID: 28129919 DOI: 10.1016/j.ejcb.2017.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/23/2016] [Accepted: 01/04/2017] [Indexed: 02/03/2023] Open
Abstract
The Huntington's disease protein Huntingtin (Htt) regulates axonal transport of dense-core vesicles (DCVs) containing neurotrophins and neuropeptides. DCVs travel down axons to reach nerve terminals where they are either captured in synaptic boutons to support later release or reverse direction to reenter the axon as part of vesicle circulation. Currently, the impact of Htt on DCV dynamics in the terminal is unknown. Here we report that knockout of Drosophila Htt selectively reduces retrograde DCV flux at proximal boutons of motoneuron terminals. However, initiation of retrograde transport at the most distal bouton and transport velocity are unaffected suggesting that synaptic capture rate of these retrograde DCVs could be altered. In fact, tracking DCVs shows that retrograde synaptic capture efficiency is significantly elevated by Htt knockout or knockdown. Furthermore, synaptic boutons contain more neuropeptide in Htt knockout larvae even though bouton size, single DCV fluorescence intensity, neuropeptide release in response to electrical stimulation and subsequent activity-dependent capture are unaffected. Thus, loss of Htt increases synaptic capture as DCVs travel by retrograde transport through boutons resulting in reduced transport toward the axon and increased neuropeptide in the terminal. These results therefore identify native Htt as a regulator of synaptic capture and neuropeptide storage.
Collapse
Affiliation(s)
- Dinara Bulgari
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - David L Deitcher
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY 14853, USA
| | - Edwin S Levitan
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
585
|
Huntingtin-Mediated Multipolar-Bipolar Transition of Newborn Cortical Neurons Is Critical for Their Postnatal Neuronal Morphology. Neuron 2017; 93:99-114. [DOI: 10.1016/j.neuron.2016.11.035] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 09/25/2016] [Accepted: 11/16/2016] [Indexed: 01/05/2023]
|
586
|
Bryan MR, Bowman AB. Manganese and the Insulin-IGF Signaling Network in Huntington's Disease and Other Neurodegenerative Disorders. ADVANCES IN NEUROBIOLOGY 2017; 18:113-142. [PMID: 28889265 PMCID: PMC6559248 DOI: 10.1007/978-3-319-60189-2_6] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease resulting in motor impairment and death in patients. Recently, several studies have demonstrated insulin or insulin-like growth factor (IGF) treatment in models of HD, resulting in potent amelioration of HD phenotypes via modulation of the PI3K/AKT/mTOR pathways. Administration of IGF and insulin can rescue microtubule transport, metabolic function, and autophagy defects, resulting in clearance of Huntingtin (HTT) aggregates, restoration of mitochondrial function, amelioration of motor abnormalities, and enhanced survival. Manganese (Mn) is an essential metal to all biological systems but, in excess, can be toxic. Interestingly, several studies have revealed the insulin-mimetic effects of Mn-demonstrating Mn can activate several of the same metabolic kinases and increase peripheral and neuronal insulin and IGF-1 levels in rodent models. Separate studies have shown mouse and human striatal neuroprogenitor cell (NPC) models exhibit a deficit in cellular Mn uptake, indicative of a Mn deficiency. Furthermore, evidence from the literature reveals a striking overlap between cellular consequences of Mn deficiency (i.e., impaired function of Mn-dependent enzymes) and known HD endophenotypes including excitotoxicity, increased reactive oxygen species (ROS) accumulation, and decreased mitochondrial function. Here we review published evidence supporting a hypothesis that (1) the potent effect of IGF or insulin treatment on HD models, (2) the insulin-mimetic effects of Mn, and (3) the newly discovered Mn-dependent perturbations in HD may all be functionally related. Together, this review will present the intriguing possibility that intricate regulatory cross-talk exists between Mn biology and/or toxicology and the insulin/IGF signaling pathways which may be deeply connected to HD pathology and, perhaps, other neurodegenerative diseases (NDDs) and other neuropathological conditions.
Collapse
Affiliation(s)
- Miles R Bryan
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| | - Aaron B Bowman
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Center in Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| |
Collapse
|
587
|
Matilla-Dueñas A, Corral-Juan M, Rodríguez-Palmero Seuma A, Vilas D, Ispierto L, Morais S, Sequeiros J, Alonso I, Volpini V, Serrano-Munuera C, Pintos-Morell G, Álvarez R, Sánchez I. Rare Neurodegenerative Diseases: Clinical and Genetic Update. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1031:443-496. [PMID: 29214587 DOI: 10.1007/978-3-319-67144-4_25] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
More than 600 human disorders afflict the nervous system. Of these, neurodegenerative diseases are usually characterised by onset in late adulthood, progressive clinical course, and neuronal loss with regional specificity in the central nervous system. They include Alzheimer's disease and other less frequent dementias, brain cancer, degenerative nerve diseases, encephalitis, epilepsy, genetic brain disorders, head and brain malformations, hydrocephalus, stroke, Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis (ALS or Lou Gehrig's Disease), Huntington's disease, and Prion diseases, among others. Neurodegeneration usually affects, but is not limited to, the cerebral cortex, intracranial white matter, basal ganglia, thalamus, hypothalamus, brain stem, and cerebellum. Although the majority of neurodegenerative diseases are sporadic, Mendelian inheritance is well documented. Intriguingly, the clinical presentations and neuropathological findings in inherited neurodegenerative forms are often indistinguishable from those of sporadic cases, suggesting that converging genomic signatures and pathophysiologic mechanisms underlie both hereditary and sporadic neurodegenerative diseases. Unfortunately, effective therapies for these diseases are scarce to non-existent. In this chapter, we highlight the clinical and genetic features associated with the rare inherited forms of neurodegenerative diseases, including ataxias, multiple system atrophy, spastic paraplegias, Parkinson's disease, dementias, motor neuron diseases, and rare metabolic disorders.
Collapse
Affiliation(s)
- Antoni Matilla-Dueñas
- Functional and Translational Neurogenetics Unit, Department of Neurosciences, Health Sciences Research Institute Germans Trias-IGTP, Can Ruti Campus, Ctra de Can Ruti, Camí de les Escoles s/n, 08916, Badalona, Barcelona, Spain.
| | - Marc Corral-Juan
- Functional and Translational Neurogenetics Unit, Department of Neurosciences, Health Sciences Research Institute Germans Trias-IGTP, Can Ruti Campus, Ctra de Can Ruti, Camí de les Escoles s/n, 08916, Badalona, Barcelona, Spain
| | - Agustí Rodríguez-Palmero Seuma
- Department of Pediatrics, University Hospital Germans Trias i Pujol (HUGTP) and Health Sciences Research Institute, Can Ruti Campus, Ctra. de Canyet s/n, 08916, Badalona, Barcelona, Spain
| | - Dolores Vilas
- Neurodegenerative Diseases Unit, Neurology Service and Neurosciences Department, University Hospital Germans Trias i Pujol (HUGTP), Ctra. de Canyet s/n, Can Ruti Campus, 08916, Badalona, Barcelona, Spain
| | - Lourdes Ispierto
- Neurodegenerative Diseases Unit, Neurology Service and Neurosciences Department, University Hospital Germans Trias i Pujol (HUGTP), Ctra. de Canyet s/n, Can Ruti Campus, 08916, Badalona, Barcelona, Spain
| | - Sara Morais
- IBMC - Institute for Molecular and Cell Biology, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Jorge Sequeiros
- IBMC - Institute for Molecular and Cell Biology, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Isabel Alonso
- IBMC - Institute for Molecular and Cell Biology, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Víctor Volpini
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Barcelona, Spain
| | - Carmen Serrano-Munuera
- Department of Internal Medicine, Hospital St. Joan de Déu, Martorell, Spain.,Manresa University, Martorell, Barcelona, Spain
| | - Guillem Pintos-Morell
- Department of Pediatrics, University Hospital Germans Trias i Pujol (HUGTP) and Health Sciences Research Institute, Can Ruti Campus, Ctra. de Canyet s/n, 08916, Badalona, Barcelona, Spain
| | - Ramiro Álvarez
- Neurodegenerative Diseases Unit, Neurology Service and Neurosciences Department, University Hospital Germans Trias i Pujol (HUGTP), Ctra. de Canyet s/n, Can Ruti Campus, 08916, Badalona, Barcelona, Spain
| | - Ivelisse Sánchez
- Functional Biology and Experimental Therapeutics Laboratory, Functional and Translational Neurogenetics Unit, Department of Neurosciences, Health Sciences Research Institute Germans Trias-IGTP, Can Ruti Campus, Ctra de Can Ruti, Camí de les Escoles s/n, 08916, Badalona, Barcelona, Spain.
| |
Collapse
|
588
|
Atherton JF, McIver EL, Mullen MR, Wokosin DL, Surmeier DJ, Bevan MD. Early dysfunction and progressive degeneration of the subthalamic nucleus in mouse models of Huntington's disease. eLife 2016; 5. [PMID: 27995895 PMCID: PMC5199195 DOI: 10.7554/elife.21616] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Accepted: 12/08/2016] [Indexed: 01/05/2023] Open
Abstract
The subthalamic nucleus (STN) is an element of cortico-basal ganglia-thalamo-cortical circuitry critical for action suppression. In Huntington's disease (HD) action suppression is impaired, resembling the effects of STN lesioning or inactivation. To explore this potential linkage, the STN was studied in BAC transgenic and Q175 knock-in mouse models of HD. At <2 and 6 months of age autonomous STN activity was impaired due to activation of KATP channels. STN neurons exhibited prolonged NMDA receptor-mediated synaptic currents, caused by a deficit in glutamate uptake, and elevated mitochondrial oxidant stress, which was ameliorated by NMDA receptor antagonism. STN activity was rescued by NMDA receptor antagonism or the break down of hydrogen peroxide. At 12 months of age approximately 30% of STN neurons had been lost, as in HD. Together, these data argue that dysfunction within the STN is an early feature of HD that may contribute to its expression and course. DOI:http://dx.doi.org/10.7554/eLife.21616.001
Collapse
Affiliation(s)
- Jeremy F Atherton
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Eileen L McIver
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Matthew Rm Mullen
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - David L Wokosin
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - D James Surmeier
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Mark D Bevan
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| |
Collapse
|
589
|
Huntingtin polyQ Mutation Impairs the 17β-Estradiol/Neuroglobin Pathway Devoted to Neuron Survival. Mol Neurobiol 2016; 54:6634-6646. [DOI: 10.1007/s12035-016-0337-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 11/30/2016] [Indexed: 02/07/2023]
|
590
|
Ascenzi P, di Masi A, Leboffe L, Fiocchetti M, Nuzzo MT, Brunori M, Marino M. Neuroglobin: From structure to function in health and disease. Mol Aspects Med 2016; 52:1-48. [DOI: 10.1016/j.mam.2016.10.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 10/27/2016] [Accepted: 10/27/2016] [Indexed: 01/01/2023]
|
591
|
Gustincich S, Zucchelli S, Mallamaci A. The Yin and Yang of nucleic acid-based therapy in the brain. Prog Neurobiol 2016; 155:194-211. [PMID: 27887908 DOI: 10.1016/j.pneurobio.2016.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 11/16/2016] [Accepted: 11/20/2016] [Indexed: 02/06/2023]
Abstract
The post-genomic era has unveiled the existence of a large repertory of non-coding RNAs and repetitive elements that play a fundamental role in cellular homeostasis and dysfunction. These may represent unprecedented opportunities to modify gene expression at the right time in the correct space in vivo, providing an almost unlimited reservoir of new potential pharmacological agents. Hijacking their mode of actions, the druggable genome can be extended to regulatory RNAs and DNA elements in a scalable fashion. Here, we discuss the state-of-the-art of nucleic acid-based drugs to treat neurodegenerative diseases. Beneficial effects can be obtained by inhibiting (Yin) and increasing (Yang) gene expression, depending on the disease and the drug target. Together with the description of the current use of inhibitory RNAs (small inhibitory RNAs and antisense oligonucleotides) in animal models and clinical trials, we discuss the molecular basis and applications of new classes of activatory RNAs at transcriptional (RNAa) and translational (SINEUP) levels.
Collapse
Affiliation(s)
- Stefano Gustincich
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia (IIT), Genova, Italy; Area of Neuroscience, SISSA, Trieste, Italy.
| | - Silvia Zucchelli
- Area of Neuroscience, SISSA, Trieste, Italy; Department of Health Sciences, Universita' del Piemonte Orientale, Novara, Italy
| | | |
Collapse
|
592
|
Vicente Miranda H, Gomes MA, Branco-Santos J, Breda C, Lázaro DF, Lopes LV, Herrera F, Giorgini F, Outeiro TF. Glycation potentiates neurodegeneration in models of Huntington's disease. Sci Rep 2016; 6:36798. [PMID: 27857176 PMCID: PMC5114697 DOI: 10.1038/srep36798] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 10/21/2016] [Indexed: 11/19/2022] Open
Abstract
Protein glycation is an age-dependent posttranslational modification associated with several neurodegenerative disorders, including Alzheimer’s and Parkinson’s diseases. By modifying amino-groups, glycation interferes with folding of proteins, increasing their aggregation potential. Here, we studied the effect of pharmacological and genetic manipulation of glycation on huntingtin (HTT), the causative protein in Huntington’s disease (HD). We observed that glycation increased the aggregation of mutant HTT exon 1 fragments associated with HD (HTT72Q and HTT103Q) in yeast and mammalian cell models. We found that glycation impairs HTT clearance thereby promoting its intracellular accumulation and aggregation. Interestingly, under these conditions autophagy increased and the levels of mutant HTT released to the culture medium decreased. Furthermore, increased glycation enhanced HTT toxicity in human cells and neurodegeneration in fruit flies, impairing eclosion and decreasing life span. Overall, our study provides evidence that glycation modulates HTT exon-1 aggregation and toxicity, and suggests it may constitute a novel target for therapeutic intervention in HD.
Collapse
Affiliation(s)
- Hugo Vicente Miranda
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School
- Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisboa, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Marcos António Gomes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joana Branco-Santos
- Department of Genetics, University of Leicester, Leicester LE1 7RH, United Kingdom.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Estação Agronomica Nacional, Av. da República, Oeiras 2780-157, Portugal
| | - Carlo Breda
- Department of Genetics, University of Leicester, Leicester LE1 7RH, United Kingdom
| | - Diana F Lázaro
- Department of Neurodegeneration and Restorative Research, Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center Göttingen, Waldweg 33, 37073 Göttingen, Germany
| | - Luísa Vaqueiro Lopes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Federico Herrera
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Estação Agronomica Nacional, Av. da República, Oeiras 2780-157, Portugal
| | - Flaviano Giorgini
- Department of Genetics, University of Leicester, Leicester LE1 7RH, United Kingdom
| | - Tiago Fleming Outeiro
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School
- Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisboa, Portugal.,Department of Neurodegeneration and Restorative Research, Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center Göttingen, Waldweg 33, 37073 Göttingen, Germany.,Max Planck Institute for Experimental Medicine, Göttingen, Germany
| |
Collapse
|
593
|
Minegishi Y, Nakayama M, Iejima D, Kawase K, Iwata T. Significance of optineurin mutations in glaucoma and other diseases. Prog Retin Eye Res 2016; 55:149-181. [DOI: 10.1016/j.preteyeres.2016.08.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 08/18/2016] [Accepted: 08/18/2016] [Indexed: 12/12/2022]
|
594
|
Gonzalez-Riano C, Garcia A, Barbas C. Metabolomics studies in brain tissue: A review. J Pharm Biomed Anal 2016; 130:141-168. [PMID: 27451335 DOI: 10.1016/j.jpba.2016.07.008] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/05/2016] [Accepted: 07/07/2016] [Indexed: 12/11/2022]
Abstract
Brain is still an organ with a composition to be discovered but beyond that, mental disorders and especially all diseases that curse with dementia are devastating for the patient, the family and the society. Metabolomics can offer an alternative tool for unveiling new insights in the discovery of new treatments and biomarkers of mental disorders. Until now, most of metabolomic studies have been based on biofluids: serum/plasma or urine, because brain tissue accessibility is limited to animal models or post mortem studies, but even so it is crucial for understanding the pathological processes. Metabolomics studies of brain tissue imply several challenges due to sample extraction, along with brain heterogeneity, sample storage, and sample treatment for a wide coverage of metabolites with a wide range of concentrations of many lipophilic and some polar compounds. In this review, the current analytical practices for target and non-targeted metabolomics are described and discussed with emphasis on critical aspects: sample treatment (quenching, homogenization, filtration, centrifugation and extraction), analytical methods, as well as findings considering the used strategies. Besides that, the altered analytes in the different brain regions have been associated with their corresponding pathways to obtain a global overview of their dysregulation, trying to establish the link between altered biological pathways and pathophysiological conditions.
Collapse
Affiliation(s)
- Carolina Gonzalez-Riano
- Centre for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad CEU San Pablo, Campus Monteprincipe, Boadilla del Monte 28668, Madrid, Spain
| | - Antonia Garcia
- Centre for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad CEU San Pablo, Campus Monteprincipe, Boadilla del Monte 28668, Madrid, Spain.
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad CEU San Pablo, Campus Monteprincipe, Boadilla del Monte 28668, Madrid, Spain
| |
Collapse
|
595
|
Chang NC, Chevalier FP, Rudnicki MA. Satellite Cells in Muscular Dystrophy - Lost in Polarity. Trends Mol Med 2016; 22:479-496. [PMID: 27161598 PMCID: PMC4885782 DOI: 10.1016/j.molmed.2016.04.002] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 04/13/2016] [Accepted: 04/14/2016] [Indexed: 12/21/2022]
Abstract
Recent findings employing the mdx mouse model for Duchenne muscular dystrophy (DMD) have revealed that muscle satellite stem cells play a direct role in contributing to disease etiology and progression of DMD, the most common and severe form of muscular dystrophy. Lack of dystrophin expression in DMD has critical consequences in satellite cells including an inability to establish cell polarity, abrogation of asymmetric satellite stem-cell divisions, and failure to enter the myogenic program. Thus, muscle wasting in dystrophic mice is not only caused by myofiber fragility but is exacerbated by intrinsic satellite cell dysfunction leading to impaired regeneration. Despite intense research and clinical efforts, there is still no effective cure for DMD. In this review we highlight recent research advances in DMD and discuss the current state of treatment and, importantly, how we can incorporate satellite cell-targeted therapeutic strategies to correct satellite cell dysfunction in DMD.
Collapse
Affiliation(s)
- Natasha C Chang
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Fabien P Chevalier
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Michael A Rudnicki
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
596
|
N-terminal Huntingtin Knock-In Mice: Implications of Removing the N-terminal Region of Huntingtin for Therapy. PLoS Genet 2016; 12:e1006083. [PMID: 27203582 PMCID: PMC4874551 DOI: 10.1371/journal.pgen.1006083] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/04/2016] [Indexed: 02/07/2023] Open
Abstract
The Huntington’s disease (HD) protein, huntingtin (HTT), is a large protein consisting of 3144 amino acids and has conserved N-terminal sequences that are followed by a polyglutamine (polyQ) repeat. Loss of Htt is known to cause embryonic lethality in mice, whereas polyQ expansion leads to adult neuronal degeneration. Whether N-terminal HTT is essential for neuronal development or contributes only to late-onset neurodegeneration remains unknown. We established HTT knock-in mice (N160Q-KI) expressing the first 208 amino acids of HTT with 160Q, and they show age-dependent HTT aggregates in the brain and neurological phenotypes. Importantly, the N-terminal mutant HTT also preferentially accumulates in the striatum, the brain region most affected in HD, indicating the importance of N-terminal HTT in selective neuropathology. That said, homozygous N160Q-KI mice are also embryonic lethal, suggesting that N-terminal HTT alone is unable to support embryonic development. Using Htt knockout neurons, we found that loss of Htt selectively affects the survival of developing neuronal cells, but not astrocytes, in culture. This neuronal degeneration could be rescued by a truncated HTT lacking the first 237 amino acids, but not by N-terminal HTT (1–208 amino acids). Also, the rescue effect depends on the region in HTT known to be involved in intracellular trafficking. Thus, the N-terminal HTT region may not be essential for the survival of developing neurons, but when carrying a large polyQ repeat, can cause selective neuropathology. These findings imply a possible therapeutic benefit of removing the N-terminal region of HTT containing the polyQ repeat to treat the neurodegeneration in HD. The 17 amino acids in the N-terminal region of huntingtin (HTT) are conserved in a wide range of species and are followed by a polyglutamine repeat whose expansion causes selective neurodegeneration in Huntington’s disease (HD). Loss of Htt can affect developing neurons and early embryonic development in mice. Whether N-terminal HTT is important for the survival of developing neurons or contributes mainly to a gain of toxic function in HD remains unknown. In the current study, we generated N-terminal mutant HTT knock-in mice and found that N-terminal HTT with an expanded polyQ repeat is unable to support the early development of mice, but can cause age-dependent neurological phenotypes. Further, we show that a truncated HTT without the N-terminal region can rescue the Htt loss-mediated degeneration of developing neurons. Our studies suggest that removal of the N-terminal region of mutant HTT could be a strategy to abolish the neuronal toxicity of mutant HTT.
Collapse
|
597
|
Green KM, Linsalata AE, Todd PK. RAN translation-What makes it run? Brain Res 2016; 1647:30-42. [PMID: 27060770 DOI: 10.1016/j.brainres.2016.04.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/24/2016] [Accepted: 04/01/2016] [Indexed: 12/14/2022]
Abstract
Nucleotide-repeat expansions underlie a heterogeneous group of neurodegenerative and neuromuscular disorders for which there are currently no effective therapies. Recently, it was discovered that such repetitive RNA motifs can support translation initiation in the absence of an AUG start codon across a wide variety of sequence contexts, and that the products of these atypical translation initiation events contribute to neuronal toxicity. This review examines what we currently know and do not know about repeat associated non-AUG (RAN) translation in the context of established canonical and non-canonical mechanisms of translation initiation. We highlight recent findings related to RAN translation in three repeat expansion disorders: CGG repeats in fragile X-associated tremor ataxia syndrome (FXTAS), GGGGCC repeats in C9orf72 associated amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) and CAG repeats in Huntington disease. These studies suggest that mechanistic differences may exist for RAN translation dependent on repeat type, repeat reading frame, and the surrounding sequence context, but that for at least some repeats, RAN translation retains a dependence on some of the canonical translational initiation machinery. This article is part of a Special Issue entitled SI:RNA Metabolism in Disease.
Collapse
Affiliation(s)
- Katelyn M Green
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States; Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Alexander E Linsalata
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States; Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Peter K Todd
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States; Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, United States; Veterans Affairs Medical Center, Ann Arbor, MI, United States.
| |
Collapse
|